15.01.2015 Views

CST Guide:

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>CST</strong> <strong>Guide</strong>:<br />

Pathways & Protocols


<strong>CST</strong> <strong>Guide</strong>:<br />

Pathways & Protocols<br />

First edition<br />

www.cellsignal.com


Foreword<br />

The revolution in recombinant DNA that has swept through biomedical research over the past three<br />

decades has been hailed as an unalloyed advance for almost every aspect of basic biological research.<br />

The allure of nucleic acid technology, with its clear logic and mathematical precision, has proven irresistible<br />

for the young researchers who have entered into this field during this period. But it has had its<br />

downside as well: At the beginning of this era, aspiring research students studied basic biochemistry<br />

as an essential part of their training. Now, it is viewed by many as an anachronism, a vestige of early<br />

and mid-20th century experimentation that has been superseded by the far more powerful experimental<br />

approaches involving manipulation and analysis of nucleic acids. Why study complex mixtures of<br />

proteins when entire cellular genomes can be sequenced almost overnight<br />

“use of procedures like these will<br />

surely support the still-incipient<br />

campaign of returning to protein<br />

biochemistry...”<br />

Dr. Robert A. Weinberg<br />

The inconvenient truth is that nucleic acid analyses have reached their limits in terms of their ability<br />

to shed light on the subcellular processes that underlie cell physiology and thus the phenotypes of cells<br />

and organisms. We are still confronted with the complexities of signal transduction biochemistry that<br />

underlie almost all biochemical processes. Progress in understanding protein function has lagged far<br />

behind the molecular biology of nucleic acids, in no small part because studying proteins is so challenging.<br />

Consider the complexity of a cell in which almost 20,000 distinct genes are being expressed, each<br />

of which, on average, may specify five distinct protein species because of various alternative splicing<br />

and post-translational modifications. And then consider the stupefying complexity of how these proteins<br />

interact combinatorially to create biological function.<br />

These notions help explain why many have fled protein biochemistry. As is now apparent, this flight<br />

has left us with an underdeveloped sense of how the protein machinery actually operates to create<br />

phenotype. As the pressure ramps up to convert basic biomedical discoveries into useful therapeutics,<br />

our still-inadequate understanding of protein structure and function becomes increasingly apparent.<br />

Thus, the failure of many ostensibly useful compounds to enter into the clinic can be traced directly to<br />

our incomplete understanding of the wiring diagram of the cell and how it can be profitably perturbed.<br />

The present manual represents one very powerful way to reverse this tide. Many of the techniques<br />

and reagents described here—often involving monoclonal antibodies—can be wielded with the precision<br />

that nucleic acid mechanics routinely employ. The use of procedures like these will surely support the<br />

still-incipient campaign of returning to protein biochemistry so that we can indeed learn how things<br />

really work inside cells. It will require another generation, but the time has come for us to return to the<br />

old ways, to puzzle out, one protein at a time, how signals are really processed inside cells to create the<br />

marvelously functioning apparatus—the eukaryotic cell.<br />

Sincerely,<br />

Dr. Robert A. Weinberg<br />

Daniel K. Ludwig Professor<br />

for Cancer Research at MIT<br />

6<br />

7


Preface<br />

“While science is our business,<br />

as a private company, we can<br />

focus on doing things beyond<br />

making money.”<br />

Dr. Michael J. Comb, PhD<br />

As a company of scientists for scientists, all of us here at <strong>CST</strong> are working hard to help you be successful at<br />

the bench. Over the years we have built information resources to assist you in your research, including the<br />

PhosphoSitePlus ® PTM database, signaling pathway diagrams, and now this guide. As the <strong>CST</strong> website has<br />

taken over the daily function of finding, ordering, and learning about our products, we are phasing out the<br />

catalog but still wanted to continue to provide a resource to assist you in your daily research activities. We<br />

hope you find that this guide fulfills that goal. It represents the combined experience of many scientists both<br />

within and outside of <strong>CST</strong>.<br />

I hope you can appreciate that we are a different sort of company. Our goal is not to source every possible<br />

antibody product or to sell products that we have little, if any, knowledge as to whether or not they work.<br />

Instead, we focus on producing products in-house so that we can rigorously validate them to ensure that<br />

they will work as you expect them to. To do this, we have invested heavily in validation and cutting edge<br />

antibody technologies in order to bring you products of the highest possible quality. We have a large team<br />

of PhD scientists who build and validate our products, and I encourage you to reach out to them as another<br />

valuable resource we provide.<br />

As a private company, we are not driven by quarterly profits. In fact, our goals extend far beyond making<br />

money; instead, we want to help our customers advance biomedical research. This gives our employees<br />

freedom to be creative in their workplace, the ability to give back to the local community, and to embrace<br />

policies and philosophies that are consistent with environmentally responsible views of the world. We’re<br />

passionate about our mission and excited about partnering with you on the journey ahead. We believe in<br />

a shared goal—to understand biology at the cellular and molecular level in order to free us of devastating<br />

diseases like cancer and Alzheimer’s. I’m personally looking very much forward to the next chapter. I hope<br />

you’ll join us.<br />

Warm regards,<br />

Dr. Michael J. Comb, PhD<br />

CEO & President<br />

Cell Signaling Technology<br />

8 www.cellsignal.com/cstceo<br />

9


Quick<br />

Reference<br />

“We are here to help, providing you<br />

with the products and reference<br />

materials you need for every step<br />

of the experimental process.”<br />

<strong>CST</strong> Scientists<br />

Mitochondria; page 18<br />

Protein Synthesis; page 192<br />

Vesicle Trafficking; page 248<br />

Quick Reference<br />

I: Research Areas<br />

01 Gene Expression, Epigenetics, and Nuclear Function 20<br />

02 Signaling 42<br />

03 Cell Growth and Death 86<br />

04 Cell Biology 106<br />

05 Cellular Metabolism 140<br />

06 Development and Differentiation 152<br />

07 Immunology and Inflammation 172<br />

08 Neuroscience 184<br />

II: Antibody Applications<br />

09 Chromatin Immunoprecipitation (ChIP) 194<br />

10 Flow Cytometry (F) 202<br />

11 Immunofluorescence (IF) 206<br />

12 Immunohistochemistry (IHC) 214<br />

13 Sandwich ELISA 220<br />

14 Western Blotting (WB) 226<br />

15 Immunoprecipitation (IP) 234<br />

16 Control Treatments and Cell Lines 238<br />

III: Workflow Tools<br />

17 Exploration 250<br />

18 Investigation 254<br />

19 Discovery 258<br />

20 Identification 262<br />

21 Profiling 264<br />

22 Modulation 266<br />

23 Localization & Classification 269<br />

24 Screening & Quantification 272<br />

25 Monitoring 274<br />

26 Verification 277<br />

27 Customization 278<br />

Adhesion; page 280<br />

IV: Additional Information<br />

28 About <strong>CST</strong> 282<br />

29 <strong>CST</strong> Nature Conservancy 284<br />

30 Publications by <strong>CST</strong> Scientists 290<br />

31 Index 294<br />

32 Diagram and Table Keys 302<br />

33 Ordering and Contact Information 304<br />

See full Table of Contents on pages 12 & 13.<br />

10<br />

11


12<br />

Table of ContentS<br />

I: Research Areas<br />

01 Gene Expression, Epigenetics, and Nuclear Function 20<br />

Chromatin/Epigenetic Regulation 20<br />

Commonly Studied Chromatin/Epigenetic Regulation Targets 24<br />

Protein Acetylation Pathway 26<br />

Histone Lysine Methylation Pathway 27<br />

Histone Modifications Table 28<br />

Examples of Crosstalk Between Post-Translational Modifications 31<br />

Pathway<br />

Translational Control 32<br />

Commonly Studied Translational Control Targets 35<br />

Translational Control: Overview Pathway 36<br />

Translational Control: Regulation of elF4E and p70 S6K Pathway 37<br />

Translational Control: Regulation of elF Pathway 38<br />

Nuclear Receptors 39<br />

Commonly Studied Nuclear Receptor Targets 39<br />

Nuclear Receptors Signaling Pathway 41<br />

02 Signaling 42<br />

MAP Kinase Signaling 42<br />

Commonly Studied MAP Kinase Targets 45<br />

MAPK/Erk in Growth and Differentiation Pathway 44<br />

Signaling Pathways Activating p38 MAP Kinase Pathway 47<br />

SAPK/JNK Signaling Pathway 48<br />

PI3 Kinase/Akt Signaling 49<br />

Commonly Studied Akt Targets 51<br />

PI3 Kinase/Akt Signaling Pathway 53<br />

mTOR Signaling Pathway 54<br />

Akt Substrates Pathway and Table 55<br />

Additional Binding Partners Table 65<br />

Tyrosine Kinases and Associated Phosphatases 66<br />

Commonly Studied Tyrosine Kinases Targets 69<br />

Tyrosine Kinases Kinase-Disease Associations Table 71<br />

ErbB/HER Signaling Pathway 74<br />

G Protein-Coupled Receptors 75<br />

Commonly Studied GPCR Targets 77<br />

G Protein-Coupled Receptor Signaling: Overview Pathway 78<br />

G Protein-Coupled Receptor Signaling to MAP Kinase/Erk Pathway 79<br />

Calcium, cAMP, and Lipid Signaling 80<br />

Commonly Studied Calcium, cAMP, and Lipid Signaling Targets 83<br />

Calcium, cAMP, and Lipid Signaling Kinase-Disease Associations 84<br />

Table<br />

03 Cell Growth and Death 86<br />

Apoptosis 86<br />

Commonly Studied Apoptosis Targets 88<br />

Regulation of Apoptosis Overview Pathway 90<br />

Inhibition of Apoptosis Pathway 91<br />

Death Receptor Signaling Pathway 92<br />

Mitochondrial Control of Apoptosis Pathway 93<br />

Autophagy 94<br />

Commonly Studied Autophagy Targets 96<br />

Autophagy Signaling Pathway 97<br />

Cell Cycle, Checkpoint Control, and DNA Damage 98<br />

Commonly Studied Cell Cycle Targets 100<br />

Cell Cycle/Checkpoint Control Kinase-Disease Associations 102<br />

Cell Cycle Control: G1/S Checkpoint Pathway 104<br />

Cell Cycle Control: G2/M DNA Damage Checkpoint Pathway 105<br />

04 Cell Biology 106<br />

Adhesion and Extracellular Matrix 106<br />

Commonly Studied Adhesion Targets 109<br />

Adherens Junction Dynamics Pathway 110<br />

Cytoskeletal Regulation 111<br />

Commonly Studied Cytoskeletal Regulation Targets 113<br />

Regulation of Actin Dynamics Pathway 115<br />

Regulation of Microtubule Dynamics Pathway 116<br />

Protein Folding and VesIcle Trafficking 117<br />

Commonly Studied Protein Folding and Vesicle Trafficking Targets 119<br />

Ubiquitin and Ubiquitin-like Proteins 120<br />

Commonly Studied Ubiquitin Targets 123<br />

Ubiquitin/Proteasome Pathway 124<br />

Ubiquitin Ligase Table 125<br />

Deubiquitinase Table 135<br />

05 Cellular Metabolism 140<br />

Commonly Studied Cellular Metabolism Targets 142<br />

Insulin Receptor Signaling Pathway 144<br />

Warburg Effect Pathway 145<br />

AMPK Signaling Pathway 146<br />

AMPK Substrates Table 147<br />

06 Development and Differentiation 152<br />

Commonly Studied Development and Differentiation Targets 158<br />

Wnt/β-Catenin Signaling Pathway 160<br />

Notch Signaling Pathway 161<br />

Hippo Signaling Pathway 162<br />

Hedgehog Signaling Pathway 163<br />

TGF-β Signaling Pathway 164<br />

ESC Pluripotency Differentiation Pathway 165<br />

Angiogenesis 166<br />

Commonly Studied Angiogenesis Targets 170<br />

Angiogenesis Signaling in Tumor Neovascularization Pathway 171<br />

07 Immunology and Inflammation 172<br />

Commonly Studied Immunology and Inflammation Targets 174<br />

Jak and Cytokine Receptor Mutants Table 176<br />

Jak/Stat Utilization Table 177<br />

B Cell Receptor Signaling Pathway 178<br />

T Cell Receptor Signaling Pathway 179<br />

Toll-like Receptor Signaling Pathway 180<br />

Jak/Stat Signaling: IL-6 Receptor Family Pathway 181<br />

NF-κB Signaling Pathway 182<br />

Tumor Immunology Pathway 183<br />

08 Neuroscience 184<br />

Commonly Studied Neuroscience Targets 187<br />

Vesicle Trafficking in Presynaptic Neurons: Synchronous Release 189<br />

Pathway<br />

Amyloid Plaque and Neurofibrillary Tangle Formation in Alzheimer’s 190<br />

Disease Pathway<br />

Dopamine Signaling in Parkinson’s Disease Pathway 191<br />

II: Antibody Applications<br />

09 Chromatin Immunoprecipitation (ChIP) 194<br />

ChIP General Protocol 196<br />

ChIP Troubleshooting <strong>Guide</strong> 200<br />

10 Flow Cytometry (F) 202<br />

Flow Cytometry General Protocol 204<br />

Flow Cytometry Alternate Protocol (for combined staining<br />

204<br />

of intracellular proteins and cell surface markers in blood)<br />

11 Immunofluorescence (IF) 206<br />

Cultured Cells (Immunocytochemistry, IF-IC) Protocol 210<br />

Frozen/Cryostat Tissue Sections (IF-F) Protocol 211<br />

Paraffin Tissue Sections (IF-P) Protocol 212<br />

In-Cell Western Protocol 213<br />

12 Immunohistochemistry (IHC) 214<br />

IHC Paraffin Protocol (using SignalStain ® Boost Detection Reagent) 216<br />

IHC Frozen Protocol (using SignalStain ® Boost Detection Reagent) 217<br />

IHC Troubleshooting <strong>Guide</strong> 219<br />

III: Workflow Tools<br />

17 Exploration 250<br />

Using PhosphoSitePlus ® online database<br />

18 Investigation 254<br />

Using motif and post-translational modification-specific antibodies<br />

19 Discovery 258<br />

Using peptide immunoaffinity enrichment and LC-MS/MS to study<br />

post-translational modifications (PTMs)<br />

20 Identification 262<br />

Using Chromatin IP (ChIP) to identify protein-DNA interactions<br />

21 Profiling 264<br />

Using antibody arrays<br />

22 Modulation 266<br />

Using chemical activators and inhibitors, cytokines, and growth factors<br />

IV: Additional Information<br />

28 About <strong>CST</strong> 282<br />

Behind the Antibodies 282<br />

Corporate Social Responsibility 282<br />

Sustainability 283<br />

Environmental Awareness and Support 283<br />

29 <strong>CST</strong> Nature Conservancy 284<br />

Conservancy Efforts & Resources 284<br />

30 Publications by <strong>CST</strong> Scientists 290<br />

31 Index 294<br />

Pathway Diagrams and Tables 294<br />

Protocols and Troubleshooting <strong>Guide</strong>s 294<br />

Targets 295<br />

Trademarks, Terms and Conditions 301<br />

13 Sandwich ELISA 220<br />

PathScan ® Sandwich ELISA Colorimetric Protocol 221<br />

PathScan ® Sandwich ELISA Chemiluminescent Protocol 223<br />

PathScan ® Sandwich ELISA Antibody Pair Protocol 224<br />

14 Western Blotting (WB) 226<br />

WB General Protocol 228<br />

WB Fluorescent Protocol 230<br />

WB Troubleshooting <strong>Guide</strong> 232<br />

15 Immunoprecipitation (IP) 234<br />

IP Native Protein Protocol 235<br />

IP Denatured Protein Protocol 237<br />

16 Control Treatments and Cell Lines 238<br />

23 Localization & Classification 269<br />

Using conjugated primary antibodies<br />

24 Screening & Quantification 272<br />

Using ELISA to screen and quantify cellular responses at the target level<br />

25 Monitoring 274<br />

Using cell assays to monitor the health of cells and their response to<br />

multiple challenges<br />

26 Verification 277<br />

Using siRNA-mediated knockdown to verify function<br />

27 Customization 278<br />

Antibodies in a carrier-free formulation for specific assay platforms<br />

32 Diagram and Table Keys 302<br />

Pathway Diagram Key 302<br />

Application Key 303<br />

Reactivity Key 303<br />

Amino Acid Properties Table 303<br />

33 Contact Information 304<br />

Global Headquarters 304<br />

Ordering and Customer Service 304<br />

Technical Support 304<br />

13


ippo<br />

GF<br />

1011010100110101101100101<br />

0 10 1 0101<br />

0 10 10 10 1<br />

1011010100110101101010101<br />

0 10 1 0101<br />

otch<br />

Pathway<br />

Series<br />

Table of ContentS<br />

Revised Edition<br />

Angiogenesis<br />

Wnt Signaling<br />

H g<br />

Signalin<br />

GPCR Signaling<br />

R-spondin<br />

R-spondin<br />

T β g<br />

Signalin<br />

Frizzled<br />

Wnt1<br />

Wnt5A<br />

Frizzled<br />

LRP5/6<br />

mut<br />

exp<br />

mut<br />

Receptor Tyrosine Kinase (RTK)<br />

mut, trans, amp, exp<br />

ligand<br />

E-cadherin<br />

CD44<br />

FAT4<br />

BMPRI<br />

BMPs<br />

BMPRII<br />

mut<br />

GPCR<br />

GPCR<br />

GPCR<br />

α<br />

Ras<br />

γ<br />

β<br />

γ<br />

β<br />

α<br />

PLCβ<br />

ZNRF3/RNF43<br />

CKI<br />

PAR-1<br />

CKI<br />

LGR4,5,6<br />

p120<br />

AC<br />

Src<br />

β-catenin<br />

α<br />

[IP 3 ]<br />

PLCβ<br />

α-catenin<br />

γ<br />

β<br />

[Ca 2+ ]<br />

[DAG]<br />

PI3Kγ<br />

PI3K<br />

Ubiquitin<br />

Crk<br />

C3G<br />

[cAMP]<br />

Dsh Akt<br />

Dsh<br />

PAR-1<br />

GSK-3β<br />

SOS<br />

GRB2<br />

EPAC<br />

CAMKII<br />

PKC<br />

PP2A<br />

APC<br />

WTX<br />

Axin<br />

SARA<br />

Smad2/3<br />

Smad2/3<br />

TGFβ RI<br />

TGFβ<br />

TGFβ RII<br />

Smad7<br />

mut<br />

mut<br />

mut<br />

mut<br />

GPCR<br />

mut<br />

α s<br />

Ras<br />

VEGF<br />

EPO<br />

PDGF<br />

mut<br />

mut<br />

α 12/13<br />

γ<br />

β<br />

Smad1/5/8<br />

mut<br />

mut<br />

ALK<br />

ROS1<br />

EGFR<br />

HER2<br />

mut<br />

mut<br />

14-3-3<br />

mut<br />

FRMD<br />

Mer<br />

KIBRA<br />

ERK<br />

mut<br />

Smad7<br />

mut<br />

Smad1/5/8<br />

mut<br />

Ras Signaling<br />

amp, mut, exp<br />

Rap<br />

[PIP 3 ]<br />

β-catenin<br />

14-3-3<br />

YAP<br />

Mst1/2<br />

SAV1<br />

TAK<br />

Smad4<br />

PKA<br />

Ras<br />

NF1<br />

B-Raf<br />

14-3-3<br />

Raf-1<br />

GRB2<br />

SOS<br />

Normoxia<br />

Hypoxia<br />

SynGAP<br />

RasGRF<br />

RasGRP<br />

RasGAP<br />

Ubiquitin<br />

TAB1<br />

TAB2<br />

mut<br />

mut<br />

mut<br />

TAB1<br />

TAB2<br />

mut<br />

LATS1/2<br />

MOB1<br />

RasGEF<br />

Integrin<br />

RasGAP<br />

HPH<br />

Ras c-Raf<br />

Akt<br />

RAR<br />

SOX<br />

β-catenin<br />

YAP/TAZ<br />

NLK<br />

Smad1/5/8<br />

Smad4<br />

Smurf<br />

mut, del<br />

mut<br />

Smad2/3<br />

Smad4<br />

mut<br />

mut<br />

PI3K ILK<br />

Src<br />

FAK<br />

GRAF<br />

RIAM/Talin<br />

RhoGEFs<br />

KSR<br />

14-3-3 c-Raf<br />

MEK1/2<br />

ERK1/2<br />

MP1<br />

CBP<br />

TAK p38<br />

MKK3/6<br />

amp, mut, exp<br />

mut<br />

amp<br />

mut<br />

E2F<br />

C<br />

amp, exp<br />

β-catenin<br />

TCF/LEF<br />

MKK7 JNK<br />

VHL<br />

RalGEF<br />

RalB<br />

HIF-1α(-2α)<br />

HIF-1β<br />

other TFs<br />

TEAD<br />

YAP/TAZ<br />

Smad<br />

mut<br />

del<br />

ERK1/2<br />

Smad1/5/8<br />

Smad4<br />

mut<br />

mut<br />

mut<br />

myc, cyclin D, CD44<br />

exp<br />

p107<br />

E2F<br />

Smad2/3<br />

Rap<br />

RalA<br />

CDK7<br />

Cyclin H<br />

RalBP1<br />

Regulated<br />

Exocytosis<br />

Exocyst<br />

Complex<br />

Rab8a (c-Mel)<br />

Cdc42 Actin<br />

Rac<br />

PAK1<br />

Cytoskeleton<br />

JNK<br />

MEK1/2<br />

MP1<br />

ERK1/2<br />

p90 RSK<br />

p90 RSK<br />

Rho<br />

ERK1/2<br />

CREB<br />

HIF-1α(-2α)<br />

HIF-1β<br />

Fos<br />

Jun<br />

myc, cyclin D<br />

Cdc2<br />

Cyclin B<br />

p15, p21, p27<br />

mut<br />

mut<br />

exp<br />

Wee1A<br />

Miz-1<br />

Smad2/3<br />

Smad4<br />

mut, amp<br />

HPV-E7<br />

DNA Repair<br />

Cdc25<br />

B/C<br />

Myc<br />

Miz-1<br />

Rad52<br />

Rad51<br />

JNK<br />

NBS1<br />

Myc<br />

Max<br />

autophagosome<br />

Myc<br />

Max<br />

cyclin D, myc<br />

FANC<br />

D2<br />

HDAC<br />

Rb<br />

DP E2F<br />

BRCA1<br />

cyclin D, myc<br />

mut, amp<br />

vir<br />

mut<br />

mut, del<br />

Atg16/12/5<br />

mut<br />

SQSTM1<br />

PI3K<br />

class III<br />

Beclin<br />

Atg14<br />

exp<br />

Autophagy<br />

p90 RSK<br />

mut, amp<br />

Fos<br />

Jun<br />

phagopore<br />

membrane<br />

nucleation<br />

Elk-1<br />

CDK4/6<br />

Cyclin D<br />

4E-BP<br />

p15 p16 p18 p19<br />

mut<br />

UV<br />

DNA Damage<br />

Cdc2<br />

Cyclin A<br />

S6K<br />

SHP1<br />

GSK-3<br />

Cyclin D p21<br />

STAT STAT<br />

DP E2F<br />

IR<br />

* *<br />

Abl<br />

ULK1<br />

FIP200<br />

Atg13<br />

mut<br />

amp, trans<br />

Chk1<br />

ATM<br />

ATR<br />

Chk2<br />

Cdc25A<br />

DNA-<br />

PK<br />

MDM2<br />

Nutrients<br />

JAK<br />

JAK<br />

STAT<br />

SHP2<br />

GRB2<br />

STAT<br />

SOS<br />

Shc<br />

Ras<br />

(amino acids<br />

& glucose)<br />

Rb<br />

(Inactive) (Active)<br />

trans<br />

mut<br />

LC3-II<br />

mut<br />

LC3-I<br />

mut<br />

mut mut<br />

mut,del<br />

Scienstists at Ce l Signaling Technology co laborate<br />

with key opinion leaders in cancer research to create<br />

reference pathway diagrams that reflect the latest<br />

thinking in the research community. Over 40 pathway<br />

diagrams currently exist, including the pathways<br />

represented here and many others.<br />

www.cellsignal.com/<strong>CST</strong>cancer<br />

mut<br />

Atg7/3<br />

Atg4<br />

mut,del<br />

p53<br />

p21<br />

trans, mut<br />

mut<br />

p53<br />

TSC2<br />

TSC1<br />

mTOR<br />

Raptor<br />

amp<br />

Rheb<br />

AMPK<br />

p27<br />

FKHR/<br />

FOXO<br />

cyclin D<br />

mut<br />

Cytokine<br />

Receptor<br />

mTOR<br />

Rictor<br />

Cdc25A<br />

R<br />

cyclin E, A<br />

myc, cdc2<br />

p107, E2F<br />

mut<br />

The Pathways in Human Cancer poster summarizes some of the key<br />

signaling pathways implicated in tumorigenesis and tumor<br />

progression in humans. Within each pathway, gene products known<br />

to be mutated in human tumors—oncogenes and tumor suppressor<br />

genes—are coded with information on types of genetic alterations<br />

and conferred capabilities to the tumor. Proteins are shown using<br />

structural representatives. New to this revised poster edition are<br />

signaling pathways for Hippo Signaling, Autophagy, Warburg Effect,<br />

and Epigenetic Regulation.<br />

This poster was created by scientists at Cell Signaling Technology in<br />

collaboration with Robert A. Weinberg and others at the forefront of<br />

cancer research. Expanded versions of each pathway, including<br />

additional downstream signaling nodes, can be found at<br />

www.cellsignal.com/<strong>CST</strong>cancer.<br />

HIPK2<br />

ARF<br />

MDM2<br />

mut<br />

mut<br />

CDK7<br />

Cyclin H<br />

trans<br />

LKB1<br />

Aurora<br />

A<br />

HPV-E6<br />

mut<br />

amp<br />

amp, exp<br />

vir<br />

Transcription<br />

Akt<br />

PI3K<br />

PDK1<br />

Lamins<br />

PP2A<br />

14-3-3 Bim<br />

XIAP<br />

Bad<br />

MDM2 Bcl-x L<br />

p53<br />

CDK2<br />

Cyclin A<br />

amp, mut, exp<br />

amp<br />

Casp-2<br />

mut,del<br />

amp,exp<br />

CDK2<br />

Cyclin E<br />

p21<br />

p27<br />

mut<br />

myc, cyclin D, cyclin E<br />

mut<br />

mut<br />

Bax<br />

Bcl-2<br />

NOXA<br />

PUMA<br />

c-Raf<br />

Smac<br />

PARP<br />

cyclin D<br />

trans<br />

CAD<br />

Gli PKA<br />

KMT2/MLL<br />

KDM6A/UTX<br />

KMT4/DOT1L<br />

BRD4<br />

TET<br />

SirT1<br />

SWI/SNF<br />

KMT6/EZH2<br />

DNMT3<br />

HDAC<br />

Sin3<br />

CSL/CBF1<br />

CSL/CBF1<br />

amp, trans<br />

p300<br />

Epigenetic Signaling<br />

AIF<br />

cIAP<br />

NFκB1/2<br />

RelA<br />

NFκB2<br />

RelB<br />

Ce l Signaling Technology would like to thank<br />

digizyme for their co laboration on the design<br />

and concept of this poster. Please visit<br />

www.digizyme.com to see more of their work.<br />

0 101010110110110<br />

mut<br />

NICD<br />

mut,del<br />

PKM2<br />

[PIP 3 ]<br />

p53<br />

Apaf-1<br />

Casp-9<br />

Casp-3,6,7<br />

ICAD<br />

CAD<br />

IκB<br />

HAT HES1<br />

Gab1<br />

PI3K<br />

RTK<br />

Gab2<br />

IRS-1<br />

Cbl<br />

PTEN<br />

PI3K<br />

pyruvate<br />

LDHA<br />

Akt Signaling<br />

mut<br />

lactate<br />

PFK<br />

Fructose<br />

Bisphosphate<br />

NICD<br />

amp, trans<br />

Gli<br />

mut<br />

PDK1<br />

Ras<br />

Gli<br />

tBid<br />

Bcl-2<br />

Su(Fu)<br />

KIF-7<br />

Su(Fu)<br />

KIF-7<br />

Bax<br />

MEKK3<br />

JNK<br />

FLIPs<br />

NUMB<br />

Dsh<br />

Presenilin<br />

ASK<br />

Bak<br />

Bid<br />

TRADD<br />

CytC<br />

NFκB1/2<br />

RelA<br />

IκB<br />

[PIP 3 ]<br />

Hexokinase<br />

G-6-P<br />

F-6-P<br />

IDH1<br />

mut, del<br />

amp, trans<br />

Fatty acid<br />

synthesis<br />

Krebs<br />

Cycle<br />

TAK<br />

IKKα IKKβ<br />

mut<br />

citrate<br />

IDH2<br />

trans<br />

NEMO<br />

Ras<br />

TAB2<br />

TAB3<br />

IKKα<br />

IKKβ<br />

Glucose<br />

transporter<br />

citrate<br />

mut<br />

mut<br />

mut<br />

MKK7<br />

NIK<br />

Direct stimulatory modification<br />

Direct inhibitory modification<br />

Multistep stimulatory modification<br />

Tentative stimulatory modification<br />

Transcriptional contribution<br />

TYPES OF GENETIC ALTERATIONS:<br />

Point mutation<br />

Amplification<br />

Translocation<br />

Deletion<br />

Viral infection<br />

Increased expression<br />

(unknown mechanism)<br />

mut<br />

amp<br />

trans<br />

del<br />

vir<br />

exp<br />

TYPES OF CONFERRED CAPABILITIES:<br />

Evading apoptosis<br />

Self-sufficiency in growth signals<br />

Insensitivity to anti-growth signals<br />

Tissue invasion & metastasis<br />

Limitless replicative potential<br />

Sustained angiogenesis<br />

∞<br />

Ras Common protein name<br />

mut<br />

Structural representative<br />

from PDB<br />

oncogene<br />

tumor suppressor<br />

Type of genetic alteration<br />

Type of conferred capability<br />

Warburg Effect<br />

Akt<br />

FADD<br />

Casp-8,10<br />

amp, mut, exp<br />

IRAK<br />

TRAF6<br />

Myd88<br />

RIP TRADD<br />

RAIDD<br />

TRAF2<br />

TRAF<br />

2/6<br />

TRAF<br />

3<br />

Notch<br />

Furin<br />

Fringe<br />

NIC<br />

mut<br />

mut<br />

mut<br />

mut<br />

Fas/DR<br />

Ptch<br />

Cleaved<br />

Notch<br />

Smo<br />

Ptch<br />

Smo<br />

mut<br />

IL1R<br />

TNFR<br />

TNFR<br />

CDO<br />

BOC<br />

Hh<br />

trans<br />

Delta<br />

Jagged<br />

D eath Receptor / NF- κB g<br />

TACE<br />

trans<br />

CDO<br />

BOC<br />

This poster accompanies the textbook The Biology of Cancer, Second Edition<br />

by Robert A. Weinberg and published by Garland Science.<br />

Dr. Weinberg is a founding member of the Whitehead Institute for Biomedical<br />

Research and the Daniel K. Ludwig Professor of Cancer Research at the<br />

Massachusetts Institute of Technology.<br />

Signalin<br />

S ignalin<br />

N g<br />

Hardcover ISBN 978-0-8153-4219-9<br />

Paperback ISBN 978-0-8153-4220-5 www.garlandscience.com<br />

To request free copies of this poster please visit our website. www.cellsignal.com/phcposter<br />

H edgehog g<br />

Signalin<br />

Annual<br />

Signaling<br />

in Cancer<br />

Symposium<br />

Cell Signaling Technology is proud to partner with the<br />

MIT’s Koch Institute to present this Annual Symposium,<br />

which brings together cancer research thought leaders<br />

from around the world to share current findings and<br />

further research community collaboration.<br />

Speakers focus on the cancer pathways that support<br />

tumor development, the emerging research in identifying<br />

and targeting these pathways, and innovations<br />

in the development of increasingly effective cancer<br />

therapy options.<br />

<strong>CST</strong> and MIT’s Koch Institute<br />

Proudly Sponsor the Targeting<br />

Cancer Pathways Webinar Series<br />

in collaboration with Science and Science Signaling<br />

Targeting Cancer Pathways:<br />

Recent advances in our understanding of cancer<br />

have revealed that the disease cannot be understood<br />

through simple analysis of genetic mutations within<br />

cancerous cells. Instead, tumors should be considered<br />

complex tissues in which the cancer cells communicate<br />

with the surrounding cellular microenvironment<br />

and evolve traits that promote their own survival.<br />

Signaling pathways mediate the cross-talk between<br />

tumor-intrinsic factors, such as genomic instability,<br />

and tumor-extrinsic factors, such as inflammation,<br />

driving cancers to acquire the hallmark capacities to<br />

sustain proliferation, downregulate tumor suppressors,<br />

evade the immune system, resist cell death and<br />

senescence, induce angiogenesis, reprogram energy<br />

metabolism, and metastasize to secondary sites.<br />

This seminar series focuses on the cancer pathways<br />

that support tumor development, the emerging<br />

research in identifying and targeting these pathways,<br />

and innovations in the development of increasingly<br />

effective cancer therapy options.<br />

Please visit our website for more information<br />

and to register for upcoming symposiums.<br />

www.cellsignal.com/cstkoch<br />

14<br />

In collaboration with<br />

www.cellsignal.com/cstkoch<br />

15


125<br />

of Antibody Discovery and Development<br />

We celebrate the 125th anniversary<br />

of the first use of antibody-based therapy.<br />

From early explorations of vaccination to present-day clinical trials, antibody-based research<br />

and therapies have long demonstrated their enormous potential to benefit human health.<br />

1890<br />

Antibodies are<br />

shown to be active<br />

against diphtheria<br />

and tetanus, giving<br />

rise to a humoral<br />

theory of immunity<br />

(Emil von Behring<br />

and Kitasato<br />

Shibasaburo) (1)<br />

1891<br />

Observe transferrable<br />

immunity (Emil von<br />

Behring and Kitasato<br />

Shibasaburo) (2)<br />

1896<br />

Jules Bordet identifies<br />

complement as an<br />

antibacterial, heat-labile<br />

serum component (3)<br />

1940<br />

Karl Landsteiner<br />

and Alexander<br />

Weiner identify<br />

Rh antigens (8)<br />

1938<br />

John Marrack<br />

proposes Antigen-<br />

Antibody binding<br />

hypothesis (6)<br />

1942<br />

Albert Coons labels antibodies with FITC<br />

originating the field of immunofluorescence (9)<br />

Jules Freund and Katherine McDermott demonstrate<br />

use of adjuvants to stimulate antibody production (10)<br />

1957<br />

Clonal selection<br />

theory proposed by<br />

Frank MacFarlane<br />

Burnet and David<br />

W. Talmage (14,15)<br />

Free Antibody History Poster...<br />

www.cellsignal.com/abhistory<br />

1890 1900 1940 1955 1965 1975 1980 1990 1995<br />

2000<br />

2010 2015<br />

1939<br />

1948<br />

1971<br />

Arne Tiselius and Elvin<br />

Astrid Fagreaus<br />

1960<br />

ELISA assay<br />

1986<br />

2002<br />

Kabat discover the<br />

discovered antibody<br />

Radioimmunoassay<br />

developed<br />

Sean P. O’Neill and Joseph Wu<br />

<strong>CST</strong> receives patent for<br />

first antibody isotype,<br />

production in<br />

developed by Rosalyn<br />

independently by<br />

awarded patent for quantitative<br />

motif antibody technology (41)<br />

gamma-globulin (7)<br />

plasma B cells (11)<br />

Yalow and Solomon<br />

Eva Engvall and<br />

Berson (18)<br />

immunoprecipitation assay (33)<br />

Peter Perlman<br />

(21)<br />

1900<br />

Paul Erlich develops antibody<br />

formation theory (4)<br />

1909<br />

Almroth Wright publishes,<br />

“Studies on Immunisation”,<br />

a collection of papers<br />

describing opsonization in<br />

the context of therapeutic<br />

immunization (5)<br />

1944<br />

IgM is described<br />

independently by<br />

Jan Waldenström with<br />

Kai Pedersen as well<br />

as Henry Kunkel (7)<br />

1955<br />

Niels Jerne<br />

proposes<br />

naturalselection<br />

theory<br />

of antibody<br />

formation (13)<br />

1965<br />

Thomas Tomasi identifies secretory<br />

immunoglobulins (IgA) (19)<br />

First fluorescence based<br />

assay developed by<br />

Martin Fulwyler (20)<br />

1953<br />

Wallace Coulter awarded a<br />

patent on Coulter principle,<br />

enabling flow cytometry (12)<br />

1959 –1962<br />

Antibody structures independently<br />

elucidated<br />

by Gerald Edelman and<br />

Rodney Porter (16,17)<br />

1956<br />

Kappa and lambda light<br />

chains, then known as<br />

Bence Jones proteins,<br />

are shown to be two<br />

separate proteins by<br />

Leonard Korngold and<br />

Rose Lipari (7)<br />

1976<br />

Susumu Tonegawa describes<br />

somatic recombination of<br />

immunoglobulin genes<br />

to account for incredible<br />

diversity (24)<br />

1972<br />

FACS instrument<br />

developed and<br />

patented by Len<br />

Herzenberg’s<br />

lab at Stanford<br />

University (22)<br />

1966<br />

Kimishige Ishizaka et al.<br />

and S.G.O. Johansson &<br />

Hans Bennich independently<br />

identified IgE as<br />

the reaginic<br />

antibody (7)<br />

1975<br />

Georges Köhler and<br />

César Milstein develop<br />

hybridomas leading to the<br />

production of mAbs (23)<br />

1967<br />

Kimishige Ishizaka identifies<br />

IgE as the reaginic antibody,<br />

binding the molecule that<br />

induced its synthesis (7)<br />

1978<br />

Hybritech becomes the<br />

first mAb company (25)<br />

1979<br />

First patent on hybridoma<br />

technology awarded to Wistar<br />

Institute (26)<br />

Western blotting, perhaps the<br />

most widely used immunoassay<br />

in research, is invented by Harry<br />

Towbin et al. (27)<br />

1982<br />

Angus Nairn, et al.<br />

develop the first<br />

phospho-specific<br />

antibodies (30)<br />

1985<br />

John Lis and David<br />

Gilmour develop Chromatin<br />

Immunoprecipitation (ChIP)<br />

assay (32)<br />

1984<br />

hCG antibodies used to develop<br />

5 minute pregnancy test (31)<br />

1995<br />

Katherine Knight and<br />

colleagues at Loyola<br />

University, Chicago,<br />

USA published first<br />

paper on rabbit mAb<br />

development (35)<br />

1990<br />

John McCafferty et al.<br />

report the use of phage<br />

display for antibody<br />

discovery (34)<br />

1996<br />

Prostascint ® , radiolabeled<br />

anti-PSMA<br />

(prostate specific<br />

membrane antigen)<br />

imaging antibody<br />

approved by the<br />

FDA (36)<br />

1981<br />

The lab of Herman Eisen develops the first anti-pTyr antibody (28)<br />

Hybritech delivers first mAb product to measure IgE in blood to<br />

diagnose allergic reactions (29)<br />

1997<br />

Idec markets the world’s first mAb<br />

treatment for lymphoma (Rituxan ® ) (37)<br />

1998<br />

Herceptin ® approved for<br />

breast cancer treatment (38)<br />

2004<br />

Erbitux approved by<br />

FDA for treatment of<br />

colorectal cancer (42)<br />

2006<br />

<strong>CST</strong> releases its first<br />

antibody developed<br />

using the proprietary<br />

XMT method (43)<br />

2000<br />

Abgenix develops XenoMouse ®<br />

which produces fully human<br />

antibodies (40)<br />

1999<br />

<strong>CST</strong> established as an independent<br />

company and releases its first kinase<br />

substrate motif antibody (#9611) (39)<br />

2014<br />

Yervoy ® (ipilimumab), a<br />

monoclonal anti-CTLA4<br />

antibody and the first<br />

immune checkpoint<br />

cancer therapy, receives<br />

FDA approval as<br />

a late-stage melanoma<br />

treatment (46)<br />

2013<br />

Kadcyla ® (ado-trastuzumab<br />

emtansine), an antibodydrug<br />

conjugate, receives<br />

FDA approval as late-stage<br />

breast cancer treatment (45)<br />

2012<br />

<strong>CST</strong> publishes NG-XMT<br />

method, a proteomics<br />

approach to developing<br />

mAbs (44)<br />

16<br />

References:<br />

1. von Behring, E., and Shibasaburo,<br />

K. (1890) Deutsche Medizinsche<br />

Wochenschrift 16, 1113–1114.<br />

2. Deshpande, S.S., Enzyme Immunoassays:<br />

From Concept to Product<br />

Development. US: Springer; 1996.<br />

3. Jules Bordet - Biographical. Nobelprize.org.<br />

www.nobelprize.org/nobel_prizes/medicine/laureates/1919/<br />

bordet-bio.html. 26 Sep 2014.<br />

4. Abbas, A.K., Lichtman, A.H., Cellular<br />

and Molecular Immunology. 2005.<br />

Philadelphia, PA: Elsevier; 2005.<br />

5. Turk, J.L. (1994) J. R. Soc. Med. 87,<br />

576–577.<br />

6. Marrack, J. The Chemistry of<br />

Antigens and Antibodies. London,<br />

UK: H.N. Stationery Off; 1938.<br />

7. Reviewed in Black, C.A. (1997)<br />

Immunol. Cell Biol. 75, 65–68.<br />

8. Landsteiner, K. and Wiener, A.S.<br />

(1940) Proc. Soc. Exp. Biol. Med.<br />

43, 223–224.<br />

9. McDevitt, H.O. and Albert Hewett<br />

Coons 1912–1978: A Biographical<br />

Memoir. Washington D.C.: National<br />

Academies Press; 1996.<br />

10. Reviewed in Bendelac, A. and<br />

Medzhitov, R. (2002) J. Exp. Med.<br />

195, 19–23.<br />

11. Fagreaus, A. (1948) J. Immunol.<br />

58, 1–13.<br />

12. US Patent No. 3,557,352 January<br />

19th, 1971.<br />

13. Jerne, N.K. (1955) Proc. Natl.<br />

Acad. Sci. USA 41, 849–857.<br />

14. Burnet, F.M. (1957) Aust. J. Sci.<br />

20, 67–69.<br />

15. Talmage, D.W. (1957) Annu. Rev.<br />

Med. 8, 239–256.<br />

16. Edelman, G.M. and Poulik, M.D.<br />

(1961) J. Exp. Med. 113, 861–884.<br />

17. Milstein, C. (2007) Int.<br />

J. Immunogen. 13, 1–2.<br />

18. Yalow, R.S. and Berson, S.A. (1960)<br />

J. Clin. Invest. 39, 1157–1175.<br />

19. Reviewed in Tomasi, T.B. (1992)<br />

Immunol. Today 13, 416–418.<br />

20. Fulwyler, M.J. (1965) Science 150,<br />

910–911.<br />

21. Engvall, E. and Perlman, P. (1971)<br />

Immunochem. 8, 871–874.<br />

22. Reviewed in Hardy, R.R. and<br />

Roederer, M. (2013) Cell. Immun.<br />

39, 989–991.<br />

23. Köhler, G. and Milstein, C. (1975)<br />

Nature 256, 495–497.<br />

24. Reviewed in Gearhart, P.J. (2004)<br />

J. Immunol. 173, 4259.<br />

25. Chi, K.R. (2007) The Scientist, www.<br />

the-scientist.com/articles.view/<br />

articleNo/25737/title/The-Birth-of-<br />

Biotech/<br />

26. Ansell, P.R.J. (2000) Immunol.<br />

Today 21, 357–358.<br />

27. Towbin, H. et al. (1979) Proc. Nat.<br />

Acad. Sci. USA 76, 4350–4354.<br />

28. Ross, A.H., Baltimore, D., and<br />

Eisen, H.N. (1981) Nature 294, 654.<br />

29. US Patent No. 4,376,110<br />

30. Nairn, A.C. Detre, J.A., Casnellie,<br />

J.E., and Greengard, P. (1982)<br />

Nature 299, 734–736.<br />

31. Armstrong, E.G. et al. (1984) J. Clin.<br />

Endocrinol. Metab. 59, 867–874.<br />

32. Gilmour, D.S. and Lis, J.T. (1985)<br />

Mol. Cell. Biol. 5, 2009–2018.<br />

33. US Patent No. 4,604,365<br />

34. McCafferty, J., Griffiths, A.D.,<br />

Winter, D., and Chiswell, D.J.<br />

(1990) Nature 348, 552–554.<br />

35. Spieker-Polet, H., Sethupathi, P.,<br />

Yam, P.C., and Knight, K.L. (1995)<br />

Proc. Natl. Acad. Sci. USA 92,<br />

9348–9352.<br />

36. Reviewed in Taneja, S.S. (2004)<br />

Rev. Urol. 6, 19–28.<br />

37. Reviewed in Demidem, A. et al.<br />

(1997) Cancer Biother. Radiopharm.<br />

12, 177–186.<br />

38. Tolner, B. (2014) Therapeutic<br />

monoclonal antibodies approved or<br />

in review in the European Union or<br />

United States. The Antibody Society<br />

www.antibodysociety.org/news/approved_mabs.php.<br />

July 29, 2014.<br />

39. www.cellsignal.com/products/<br />

primary-antibodies/9611<br />

40. Green, L.L. (1999) J. Immunol.<br />

Methods 10, 11–23.<br />

41. US Patent No. 6,441,140<br />

42. www.fda.gov/newsevents/newsroom/pressannouncements/2004/<br />

ucm108244.htm<br />

43. www.cellsignal.com/XMT<br />

44. Cheung, W.C. et al. (2012)<br />

Nat. Biotechnol. 30, 447–452.<br />

45. www.fda.gov/newsevents/<br />

newsroom/pressannouncements/<br />

ucm340704.htm<br />

46. www.fda.gov/newsevents/<br />

newsroom/pressannouncements/<br />

ucm1193237.htm<br />

17


I<br />

Research<br />

Areas<br />

We are now at a dynamic and exciting phase of cell biology research.<br />

In the pages to follow, we hope to capture this excitement by providing<br />

our Research Area pages. Each Research Area includes overview<br />

text, images, tables, and reference pathway illustrations that provide<br />

a snapshot of information reflecting the latest thinking in the research<br />

community. The information contained within is a collaborative effort<br />

that represents contributions of both expert <strong>CST</strong> scientists and external<br />

reviewers from the global research community at the forefront of<br />

their respective fields.<br />

Research Areas are categorized into chapters covering the central<br />

themes of cell biology, so individual areas can be seen within their<br />

broader biological context.<br />

Section Includes:<br />

Introductory backgrounds and data features<br />

Reference pathways and tables<br />

Target lists for <strong>CST</strong> antibody products<br />

Citations for select targets<br />

Mitochondria<br />

Molecular landscape portraying the crosstalk between cellular metabolism (including<br />

proteins involved in the Warburg effect), mitochondrial transport, and apoptotic signaling.<br />

www.cellsignal.com/cstlandscapes<br />

19


01<br />

Section I: Research Areas<br />

Gene Expression, Epigenetics,<br />

and Nuclear Function<br />

Chromatin/Epigenetic Regulation<br />

The nucleosome, made up of four histone proteins (H2A, H2B, H3, and H4) and 147 bp of DNA, is<br />

the primary building block of chromatin. In addition to the core histone proteins, a number of histone<br />

variants exist (H2AX, H2AZ, MacroH2A, H3.3, and CENP-A) that confer different structural properties<br />

to nucleosomes and function in DNA repair, chromosome segregation during mitosis, and regulation<br />

of transcription. Histones were originally thought to function as a static scaffold for DNA packaging;<br />

however, they are now known to be dynamic proteins, undergoing multiple types of post-translational<br />

modifications (PTMs) and interacting with regulatory proteins to control gene expression in a highly<br />

regulated manner.<br />

Chromatin Modifying Enzymes<br />

Histone Acetylases and Deacetylases<br />

Protein acetylation plays a crucial role in regulating chromatin structure and transcriptional activity.<br />

Acetylation marks occur on lysine residues and are applied by histone acetyltransferases (HATs) and<br />

removed by histone deacetylases (HDACs). Acetylation within the histone tail weakens histone-DNA<br />

and histone-histone interactions and creates an open chromatin conformation that increases transcription<br />

factor access to DNA, while histone deacetylation limits gene activity by creating a closed<br />

conformation that limits transcription factor binding. In addition, acetylation creates binding sites for<br />

bromodomain-containing chromatin regulatory proteins. For example, the bromodomain-containing<br />

protein 4 (BRD4) is a chromatin-binding protein with a preference for acetyl-lysine 14 on histone H3 as<br />

well as acetyl-lysine 5 and acetyl-lysine 12 on histone H4.<br />

BRD4 binds to acetylated lysine residues within active chromatin regions.<br />

BRD4 (E2A7X) Rabbit mAb #13440: Chromatin IPs were performed with<br />

cross-linked chromatin from 4 x 10 6 MV-4-11 cells and either 10 µl of<br />

#13440 or 2 µl of Normal Rabbit IgG #2729, using SimpleChIP ® Enzymatic<br />

Chromatin IP Kit (Magnetic Beads) #9003. The enriched DNA was quantified<br />

by real-time PCR using SimpleChIP ® Human Bcl-2 Promoter Primers #12924,<br />

human c-Myc intron 1 primers, and SimpleChIP ® Human α Satellite Repeat<br />

Primers #4486. The amount of immunoprecipitated DNA in each sample is<br />

represented as a percent of the total input chromatin.<br />

BRD4 (E2A7X)<br />

Rabbit mAb #13440<br />

Normal Rabbit<br />

IgG #2729<br />

SirT6, a class III histone deacetylase, is<br />

expressed in wild-type but not knock-out MEFs.<br />

SirT6 (D8D12) Rabbit mAb #12486: WB analysis of extracts from SirT6 wild-type<br />

(WT) and knockout (KO) mouse embryonic fibroblasts (MEF) using #12486 (upper)<br />

or β-Actin (D6A8) Rabbit mAb #8457 (lower). SirT6 WT and KO MEF were kindly<br />

provided by Dr. David Lombard, University of Michigan.<br />

% of total input chromatin<br />

1.2<br />

1.0<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

Bcl-2<br />

Lanes<br />

1. SirT6 WT MEF<br />

2. SirT6 KO MEF<br />

c-Myc<br />

kDa<br />

60<br />

50<br />

40<br />

30<br />

α Satellite<br />

1 2<br />

SirT6<br />

chapter 01: GENE EXPRESSION, EPIGENETICS, AND NUCLEAR FUNCTION<br />

Histone Methyltransferases and Demethylases<br />

The histone methylation state determines active and inactive regions of the genome and is crucial for<br />

proper developmental programming. Methylation marks occur at lysine and arginine residues and are<br />

carried out by histone methyltransferases, including lysine methyltransferases EZH2, G9a, SUV39H1,<br />

and arginine methyltransferases PRMT1, PRMT4/CARM1, and PRMT5. Methylated histone residues<br />

are found within all four core histone proteins and are implicated in both transcriptional activation and<br />

silencing. Methylation facilitates binding of chromatin regulatory proteins (readers) that contain various<br />

methyl-lysine or methyl-arginine binding domains (PHD, chromo, WD40, Tudor, MBT, Ankyrin repeats,<br />

PWWP domains). Methylation marks are removed by demethylases such as Jumonji C domain family<br />

proteins (JARID, JMJD, UTX, UTY, FBXL10, FBXL11) and LSD1.<br />

A<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

1<br />

2 3<br />

Ezh2<br />

B<br />

C<br />

1.6<br />

1.4<br />

1.2<br />

1.0<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

Ezh2 (D2C9) XP ® Rabbit mAb #5246: WB analysis of extracts from MCF7, Neuro-2a, and COS-7 cell lines (A) using #5246. IHC analysis<br />

of paraffin-embedded human lymphoma (B) using #5246. Chromatin IPs were performed with cross-linked chromatin from 4 x 10 6 NCCIT<br />

cells (C) and either 5 µl of #5246 or 2 µl of Normal Rabbit IgG #2729 using SimpleChIP ® Enzymatic Chromatin IP Kit (Magnetic Beads)<br />

#9003. The enriched DNA was quantified by real-time PCR using SimpleChIP ® Human HoxA1 Intron 1 Primers #7707, SimpleChIP ® Human<br />

HoxA2 Promoter Primers #5517, and SimpleChIP ® Human α Satellite Repeat Primers #4486. The amount of immunoprecipitated DNA in<br />

each sample is represented as a percent of the total input chromatin.<br />

% of total input chromatin<br />

HoxA1<br />

HoxA2<br />

α Satellite<br />

Other Modifications: Phosphorylation,<br />

Ubiquitination, DNA Methylation<br />

Chromatin structure is also modulated through other PTMs such as phosphorylation and ubiquitination<br />

of histone proteins, which affect association with DNA-interacting proteins and have been recently<br />

identified to play a role in coordinating other histone modifications. For example, histone H2B is<br />

ubiquitinated at Lys120, which is associated with the transcribed region of active genes. Ubiquitination<br />

of H2B at Lys120 is essential for subsequent methylation of histone H3 Lys4 and 79, two additional<br />

histone modifications that regulate transcriptional initiation and elongation.<br />

In addition, methylation of DNA at cytosine residues affects chromatin folding by recruiting methyl-DNA<br />

binding proteins (MeCP2, MBD), which recruit additional chromatin modifying complexes and inhibit<br />

DNA binding of methylation-sensitive transcriptional activators. DNA methylation is critical for proper<br />

regulation of gene silencing, genomic imprinting, and development. Two families of mammalian DNA<br />

methyltransferases have been identified, DNMT1 and DNMT3, which play distinct roles in embryonic<br />

stem cells and adult somatic cells. Activity of these enzymes is regulated by accessory proteins such<br />

as DMAP1 and UHRF1, which target the associated DNMT to mediate proper methylation patterns to<br />

newly synthesized DNA during replication. DNA methylation changes are frequently associated with<br />

cancer; general hypomethylation of the genome and localized hypermethylation of CpG islands within<br />

tumor suppressor promoter regions can both be found during various stages of cancer development.<br />

Ezh2, a member of<br />

the Polycomb Group<br />

proteins, is expressed<br />

in various cell lines and<br />

cancers, and associates<br />

with HoxA genes.<br />

Lanes<br />

1. MCF7<br />

2. Neuro-2a<br />

3. COS-7<br />

Ezh2 (D2C9) XP ®<br />

Rabbit mAb #5246<br />

Normal Rabbit<br />

IgG #2729<br />

Chromatin/Epigenetics Resources<br />

Please visit our website for additional resources and products relating to the study of Chromatin/Epigenetics.<br />

www.cellsignal.com/cstchromatin<br />

60<br />

50<br />

40<br />

30<br />

β-Actin<br />

Ubiquityl-Histone H2B (Lys120) (D11) XP ® Rabbit mAb #5546:<br />

Chromatin IPs were performed with cross-linked chromatin from 4 x 10 6<br />

HeLa cells and either 10 μl of #5546 or 2 μl of Normal Rabbit IgG #2729<br />

using SimpleChIP ® Enzymatic Chromatin IP Kit (Magnetic Beads) #9003.<br />

The enriched DNA was quantified by real-time PCR using SimpleChIP ®<br />

Human γ-Actin Promoter Primers #5037, SimpleChIP ® Human γ-Actin Intron<br />

3 Primers #5047, SimpleChIP ® Human GAPDH Promoter Primers #4471,<br />

and SimpleChIP ® Human GAPDH Intron 2 Primers #4478. The amount of<br />

immunoprecipitated DNA in each sample is represented as a percent of the<br />

total input chromatin.<br />

Ubiquityl-Histone H2B (Lys120)<br />

(D11) XP ® Rabbit mAb #5546<br />

Normal Rabbit<br />

IgG #2729<br />

% of total input chromatin<br />

1.2<br />

1.0<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

γ-Actin<br />

Promoter<br />

γ-Actin<br />

Intron 3<br />

GAPDH<br />

Promoter<br />

GAPDH<br />

Intron 2<br />

Ubiquitination of<br />

histone H2B at Lys120<br />

is associated with the<br />

transcribed region of<br />

active genes.<br />

20 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstchromatin<br />

21


Section I: Research Areas<br />

chapter 01: GENE EXPRESSION, EPIGENETICS, AND NUCLEAR FUNCTION<br />

ATP-dependent Chromatin Remodeling Proteins<br />

ATP-dependent remodeling complexes, such as SWI/SNF, NuRD, and ISWI, make structural changes<br />

to chromatin by using their ATPase catalytic subunit to disrupt histone-DNA contacts and reposition<br />

nucleosomes in order to expose regions of DNA to critical regulatory proteins necessary for transcription,<br />

DNA replication, and repair.<br />

SWI/SNF Complex<br />

The SWI/SNF complex (BAF and PBAF complexes in humans) consists of multiple subunits and contains<br />

either a BRM (SMARCA2) or a BRG1 (SMARCA4) protein that acts as an ATPase. Components of the<br />

SWI/SNF complex are commonly mutated in cancer and are the focus of many research efforts as<br />

potential therapeutic targets.<br />

BAF Complex<br />

ACTL6A/B<br />

SMARCC1<br />

SMARCA<br />

2/4<br />

SMARCC2<br />

ARID1A/B<br />

Polycomb Group Proteins<br />

Polycomb group (PcG) proteins help maintain cell identity, stem cell self-renewal, cell cycle regulation,<br />

and oncogenesis by silencing genes that promote cell lineage specification, cell death, and cell cycle<br />

arrest. PcG proteins exist in two complexes: PRC2 (EED-EZH2), which methylates histone H3 on Lys27<br />

(H3K27), and the PRC1 complex, which ubiquitinylates histone H2A on Lys119 in response to H3K27<br />

methylation.<br />

Canonical PRC1<br />

PHC<br />

PCGF2/4<br />

RING1A/B<br />

CBX<br />

Blackledge, N.P., et al. (2014) Cell 157, 1445−1459.<br />

Variant PRC1<br />

PCGF1-6<br />

RING1A/B<br />

rybp/<br />

yaf2<br />

PRC2 Complex<br />

SUZ12<br />

EZH2<br />

EED<br />

RBAP46/48<br />

AEBP2<br />

Jarid2<br />

PBAF Complex<br />

SMARCD<br />

1/2/3<br />

ACTL6A/B<br />

SMARCB1<br />

SMARCA<br />

2/4<br />

SMARCE1<br />

DPF1/2/3<br />

ARID2<br />

PBRM1<br />

Wang, X., et al. (2014)<br />

Clin. Cancer Res. 20, 21−27.<br />

BRD7<br />

Bmi1 (D20B7) XP ® Rabbit mAb #6964: Chromatin IPs were performed with<br />

cross-linked chromatin from 4 x 10 6 NCCIT cells and either 10 µl of #6964 or<br />

2 µl of Normal Rabbit IgG #2729 using SimpleChIP ® Enzymatic Chromatin IP<br />

Kit (Magnetic Beads) #9003. The enriched DNA was quantified by real-time<br />

PCR using SimpleChIP ® Human HoxA1 Intron 1 Primers #7707, SimpleChIP ®<br />

Human HoxA2 Promoter Primers #5517, and SimpleChIP ® Human α Satellite<br />

Repeat Primers #4486. The amount of immunoprecipitated DNA in each<br />

sample is represented as a percent of the total input chromatin.<br />

Bmi1 (D20B7) XP ®<br />

Rabbit mAb #6964<br />

Normal Rabbit<br />

IgG #2729<br />

% of total input chromatin<br />

0.5<br />

0.4<br />

0.3<br />

0.2<br />

0.1<br />

0<br />

HoxA1<br />

HoxA2<br />

α Satellite<br />

Bmi1 (PCGF4), a<br />

component of the<br />

PRC1 complex of<br />

polycomb group proteins,<br />

associates with<br />

silenced Hox genes.<br />

SMARCC1<br />

SMARCC2<br />

SMARCD<br />

1/2/3<br />

SMARCB1<br />

SMARCE1<br />

DPF1/2/3<br />

BRM, one of two ATPase catalytic subunits for the SWI/SNF complex,<br />

associates with estrogen-responsive genes in cells treated with β-estradiol.<br />

BRM (D9E8B) XP ® Rabbit mAb #11966: Chromatin IPs were performed<br />

with cross-linked chromatin from 4 x 10 6 MCF7 cells grown in phenol red-free<br />

medium and 5% charcoal-stripped FBS for 4 d followed by treatment with<br />

β-estradiol (10 nM, 45 min) and either 5 μl of #11966 or 2 μl of Normal<br />

Rabbit IgG #2729 using SimpleChIP ® Enzymatic Chromatin IP Kit (Magnetic<br />

Beads) #9003. The enriched DNA was quantified by real-time PCR using<br />

SimpleChIP ® Human ESR1 Promoter Primers #9673, SimpleChIP ® Human<br />

pS2 Promoter Primers #9702, and SimpleChIP ® Human α Satellite Repeat<br />

Primers #4486. The amount of immunoprecipitated DNA in each sample is<br />

represented as a percent of the total input chromatin.<br />

BRM (D9E8B) XP ®<br />

Rabbit mAb #11966<br />

Normal Rabbit<br />

IgG #2729<br />

NuRD Complex<br />

The transcriptional repressor nucleosome remodeling and histone deacetylase (NuRD) complex is<br />

composed of multiple subunits, including histone deacetylases (HDAC1 and HDAC2) and the ATPdependent<br />

helicases CHD3 and CHD4. The NuRD complex plays an important role in regulating genes<br />

responsible for embryonic stem cell pluripotency and differentiation.<br />

ISWI Complex<br />

The mammalian imitation SWI (ISWI) chromatin remodeling complexes have been shown to alter<br />

nucleosome spacing in vitro. ISWI complexes are characterized by two ATPase subunits: SNF2H and<br />

SMARCA1 (SNF2L). SNF2H is part of several chromatin-remodeling complexes, including ACF1, RSF1,<br />

CHRAC, NoRC, WSTF, and WCRF180, and is important for regulation of chromatin structure, DNA<br />

damage response, and development. SMARCA1 is the catalytic subunit of the nucleosome remodeling<br />

factor (NURF) complex and plays an important role in neuronal and T cell development.<br />

% of total input chromatin<br />

0.30<br />

0.25<br />

0.20<br />

0.15<br />

0.10<br />

0.05<br />

0<br />

ESR1<br />

pS2<br />

α Satellite<br />

RNA Polymerase II Modifications<br />

RNA polymerase II (RNAPII) is a large multiprotein complex that functions as a DNA-dependent RNA<br />

polymerase, catalyzing the transcription of DNA into RNA. The largest subunit, RNAPII subunit B1<br />

(Rpb1), also known as RNAPII subunit A (POLR2A), contains a unique heptapeptide sequence (Tyr1,<br />

Ser2, Pro3, Thr4, Ser5, Pro6, Ser7), which is repeated up to 52 times in the carboxy-terminal domain<br />

(CTD) of the protein. This CTD heptapeptide repeat is subject to multiple PTMs, which dictate the<br />

functional state of the polymerase complex. Phosphorylation of the CTD during the active transcription<br />

cycle integrates transcription with chromatin remodeling and nascent RNA processing by regulating the<br />

recruitment of chromatin modifying enzymes and RNA processing proteins to the transcribed gene.<br />

Phospho-Rpb1 CTD (Ser2/Ser5) (D1G3K) Rabbit mAb # 13546:<br />

Chromatin IPs were performed with cross-linked chromatin from 4 x 10 6<br />

HeLa cells and either 10 μl of #13546 or 2 μl of Normal Rabbit IgG #2729<br />

using SimpleChIP ® Enzymatic Chromatin IP Kit (Magnetic Beads) #9003.<br />

The enriched DNA was quantified by real-time PCR using SimpleChIP ®<br />

Human β-Actin Promoter Primers #13653, human β-Actin intron 1 primers,<br />

SimpleChIP ® Human β-Actin 3ʹ UTR Primers #13669, and SimpleChIP ®<br />

Human α Satellite Repeat Primers #4486. The amount of immunoprecipitated<br />

DNA in each sample is represented as a percent of the total input chromatin.<br />

Phospho-Rpb1 CTD (Ser2/Ser5)<br />

(D1G3K) Rabbit mAb #13546<br />

Normal Rabbit<br />

IgG #2729<br />

% of total input chromatin<br />

4.0<br />

3.5<br />

3.0<br />

2.5<br />

2.0<br />

1.5<br />

1.0<br />

0.5<br />

0<br />

β-Actin<br />

Promoter<br />

β-Actin<br />

Intron 1<br />

β-Actin<br />

3’UTR<br />

α Satellite<br />

Select Reviews<br />

Del Rizzo, P.A. and Trievel, R.C. (2014) Biochim. Biophys. Acta. Jun 17 [Epub ahead of print] • Kurdistani, S.K. (2014)<br />

Curr. Opin. Genet. Dev. 26, 53−58. • Marmorstein, R. and Zhou, M.M. (2014) Cold Spring Harb. Perspect. Biol. 6, a018762.<br />

• Napolitano, G., Lania, L., and Majello, B. (2014) J. Cell Physiol. 229, 538−544. • Narlikar, G.J., Sundaramoorthy, R., and<br />

Owen-Hughes, T. (2013) Cell 154, 490−503. • Sanchez, R., Meslamani, J., and Zhou, M.M. (2014) Biochim. Biophys. Acta.<br />

1839, 676−685. • Schwartz, Y.B. and Pirrotta, V. (2013) Nat. Rev. Genet. 14, 853−864. • Swygert, S.G. and Peterson, C.L.<br />

(2014) Biochim. Biophys. Acta. 1839, 728−736. • Van Rechem, C. and Whetstine, J.R. (2014) Biochim. Biophys. Acta. May<br />

23 [Epub ahead of print]<br />

Phospho-Rpb1 CTD<br />

(Ser2/Ser5) is associated<br />

with active gene regions.<br />

22 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstchromatin<br />

23


Section I: Research Areas<br />

chapter 01: GENE EXPRESSION, EPIGENETICS, AND NUCLEAR FUNCTION<br />

These protein targets represent key<br />

nodes within chromatin/epigenetic<br />

regulation signaling pathways and<br />

are commonly studied in chromatin/<br />

epigenetic regulation research. Primary<br />

antibodies, antibody conjugates, and<br />

antibody sampler kits containing these<br />

targets are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

E PathScan ® ELISA Kits<br />

S SignalSilence ® siRNA<br />

C Antibody Conjugate<br />

Commonly Studied Chromatin/Epigenetic Regulation Targets<br />

Target M P E S C<br />

ACF1<br />

•<br />

ARID1A/BAF250A<br />

• •<br />

ASF1A<br />

•<br />

ASF1B<br />

• •<br />

ASH2L<br />

•<br />

Bmi1 • • •<br />

BORIS<br />

•<br />

BTAF1<br />

•<br />

Brd2<br />

•<br />

BRD4<br />

•<br />

Brg1<br />

•<br />

BRM<br />

• •<br />

CABIN1<br />

•<br />

CBP<br />

•<br />

Acetyl-CBP (Lys1535)/p300 (Lys1499) •<br />

CDK7<br />

• •<br />

CDK8<br />

•<br />

CDK9<br />

•<br />

Phospho-CDK9 (Thr186) •<br />

CDK12<br />

•<br />

CENP-A<br />

• •<br />

Phospho-CENPA (Ser7) •<br />

CHAF1A<br />

•<br />

CHD1 • •<br />

CHD1L<br />

•<br />

CHD2<br />

•<br />

CHD3<br />

•<br />

CHD4<br />

• •<br />

CHD7<br />

•<br />

CHD8<br />

• •<br />

CLOCK<br />

•<br />

CTCF • • •<br />

CTDSPL2<br />

•<br />

Phospho-CTDSPL2 (Ser104) •<br />

CTR9<br />

•<br />

CXXC1<br />

•<br />

Cyclin T1<br />

•<br />

DBC1<br />

• •<br />

Phospho-DBC1 (Thr454) •<br />

DMAP1<br />

•<br />

DNMT1<br />

•<br />

DNMT3A<br />

• •<br />

DNMT3L<br />

•<br />

DR1<br />

•<br />

ELP1/IKBKAP<br />

•<br />

ELP3<br />

•<br />

ESET<br />

•<br />

EWS • •<br />

Ezh2<br />

• • • • •<br />

G9a/EHMT2<br />

•<br />

GCN5L2<br />

•<br />

Histone H2A<br />

• •<br />

Acetyl-Histone H2A (pan)<br />

•<br />

Acetyl-Histone H2A (Lys5) •<br />

Ubiquityl-Histone H2A (Lys119) •<br />

MacroH2A1<br />

• •<br />

MacroH2A1.2<br />

•<br />

Histone H2A.X<br />

• •<br />

Target M P E S C<br />

Phospho-Histone H2A.X (Ser139) • • •<br />

•<br />

Phospho-Histone H2A.X (Ser139/<br />

Tyr142)<br />

Histone H2A.Z<br />

•<br />

Histone H2B • • • •<br />

Acetyl-Histone H2B (Lys5) • •<br />

Acetyl-Histone H2B (Lys12) • •<br />

Acetyl-Histone H2B (Lys15) •<br />

Acetyl-Histone H2B (Lys20) • •<br />

Phospho-Histone H2B (Ser14) •<br />

Ubiquityl-Histone H2B (Lys120) •<br />

Histone H3 • • • •<br />

Acetylated Histone H3 (pan) •<br />

Acetyl-Histone H3 (Lys9) • • •<br />

•<br />

Acetyl- and Phospho-Histone H3<br />

(Lys9/Ser10)<br />

Acetyl-Histone H3 (Lys14) •<br />

Acetyl-Histone H3 (Lys18) • • •<br />

Acetyl-Histone H3 (Lys27) • •<br />

Acetyl-Histone H3 (Lys36) •<br />

Acetyl-Histone H3 (Lys56) •<br />

Methyl-Histone H3 (Arg2) •<br />

Mono-Methyl-Histone H3 (Lys4) • • •<br />

Di-Methyl-Histone H3 (Lys4) • •<br />

Tri-Methyl-Histone H3 (Lys4) • • • •<br />

•<br />

Symmetric Di-Methyl-Histone H3<br />

(Arg8)<br />

Pan-Methyl-Histone H3 (Lys9) • •<br />

Di-Methyl-Histone H3 (Lys9) • • •<br />

Di/Tri-Methyl-Histone H3 (Lys9) •<br />

Tri-Methyl-Histone H3 (Lys9) •<br />

Di-Methyl-Histone H3 (Lys27) • •<br />

Tri-Methyl-Histone H3 (Lys27) • • •<br />

Di-Methyl-Histone H3 (Lys36) • •<br />

Tri-Methyl-Histone H3 (Lys36) • •<br />

Mono-Methyl-Histone H3 (Lys79) • •<br />

Di-Methyl-Histone H3 (Lys79) •<br />

Tri-Methyl-Histone H3 (Lys79) •<br />

Cleaved Histone H3 (Thr22) • •<br />

Phospho-Histone H3 (Thr3) • •<br />

Phospho-Histone H3 (Ser10) • • • •<br />

Phospho-Histone H3 (Thr11) • •<br />

Phospho-Histone H3 (Ser28) •<br />

Histone H4 • • •<br />

Acetyl-Histone H4 (pan)<br />

•<br />

Acetyl-Histone H4 (Lys5) • •<br />

Acetyl-Histone H4 (Lys8) • •<br />

Acetyl-Histone H4 (Lys12) • • •<br />

Acetyl-Histone H4 (Lys16) • •<br />

Mono-Methyl-Histone H4 (Lys20) •<br />

Di-Methyl-Histone H4 (Lys20) •<br />

Tri-Methyl-Histone H4 (Lys20) •<br />

HDAC1<br />

• •<br />

HDAC2<br />

• •<br />

HDAC3<br />

• •<br />

Phospho-HDAC3 (Ser424) •<br />

HDAC4 • • •<br />

•<br />

Phospho-HDAC4 (Ser246)/HDAC5<br />

(Ser259)/HDAC7 (Ser155)<br />

Target M P E S C<br />

Phospho-HDAC4 (Ser632)/HDAC5<br />

(Ser661)/HDAC7 (Ser486) •<br />

HDAC6<br />

•<br />

HELLS<br />

•<br />

HEXIM1<br />

• •<br />

HIRA<br />

•<br />

HMGA1<br />

•<br />

HMGA2<br />

• •<br />

HMGB1<br />

• •<br />

HMGB2<br />

•<br />

HMGN1<br />

• •<br />

HMGN2<br />

•<br />

HP1α<br />

•<br />

HP1α/β<br />

•<br />

HP1β<br />

• •<br />

HP1γ<br />

•<br />

Phospho-HP1γ (Ser83) •<br />

INTS9<br />

•<br />

JARID1A<br />

•<br />

JARID1B<br />

•<br />

JARID1C<br />

•<br />

JARID2<br />

•<br />

JMJD1B/JHDM2B<br />

•<br />

JMJD1B<br />

•<br />

JMJD2A<br />

•<br />

JMJD2B • • •<br />

JMJD3<br />

•<br />

LCMT1<br />

•<br />

LEDGF<br />

•<br />

LSD1<br />

• •<br />

MBD3<br />

•<br />

MeCP2<br />

•<br />

MED12<br />

•<br />

MED23<br />

•<br />

MED26<br />

•<br />

Menin<br />

•<br />

MEP50<br />

• •<br />

MLLT1/ENL<br />

•<br />

MORF4L1/MRG15 • •<br />

MTA1<br />

•<br />

NCoR1<br />

•<br />

NFI-C<br />

•<br />

Nucleomethylin<br />

•<br />

NRF2<br />

•<br />

Nucleolin<br />

•<br />

NUT1<br />

•<br />

PAF1<br />

•<br />

PCAF<br />

•<br />

PHC1<br />

•<br />

PHF2<br />

•<br />

PHF20<br />

•<br />

POLR3A<br />

•<br />

Pontin/RUVBL1<br />

•<br />

PRMT1<br />

•<br />

PRMT4/CARM1<br />

• •<br />

PRMT5/Skb1Hs Methyltransferase •<br />

RAD21<br />

•<br />

RBAP46/RBAP48<br />

• •<br />

RBBP5<br />

• •<br />

RBAP46<br />

•<br />

Target M P E S C<br />

Reptin/RuvBL2<br />

• •<br />

Ring1A<br />

• •<br />

RING1B<br />

•<br />

RNF20<br />

•<br />

RNF40<br />

• •<br />

Rpb1 CTD<br />

•<br />

Phospho-Rpb1 CTD (Ser2) •<br />

Phospho-Rpb1 CTD (Ser2/Ser5) •<br />

Phospho-Rpb1 CTD (Ser5) • •<br />

Phospho-Rpb1 CTD (Ser7) •<br />

SATB1<br />

• •<br />

Phospho-SATB1 (Ser47) •<br />

SET7/SET9<br />

• •<br />

SET8<br />

•<br />

SIN3A<br />

•<br />

SirT1 • • •<br />

Phospho-SirT1 (Ser27) •<br />

Phospho-SirT1 (Ser47) •<br />

SirT2<br />

•<br />

SirT3<br />

•<br />

SirT5<br />

•<br />

SirT6 • • •<br />

SirT7<br />

•<br />

SMARCA1<br />

• •<br />

SMARCAD1<br />

•<br />

SMARCC1/BAF155 • •<br />

SMARCC2/BAF170 • •<br />

SMYD2<br />

• •<br />

SMYD3<br />

•<br />

SNF2H<br />

•<br />

SNF5<br />

•<br />

SP1 • • •<br />

SPT5<br />

•<br />

SPT16<br />

•<br />

SRC-1<br />

•<br />

SRC-3<br />

•<br />

Phospho-SRC-3 (Thr24) •<br />

SSRP1<br />

• •<br />

SSU72<br />

•<br />

SUV39H1<br />

•<br />

SUZ12<br />

•<br />

TAF1<br />

•<br />

TAF15<br />

•<br />

TBP<br />

• •<br />

TCEB3/Elongin A<br />

•<br />

TFII-I<br />

•<br />

TFIIB<br />

•<br />

TFIIFα<br />

•<br />

TH1L<br />

• •<br />

Tip60<br />

•<br />

Topoisomerase IIα<br />

• •<br />

Phospho-Topoisomerase IIα (Ser1469) •<br />

TRIM29/ATDC<br />

•<br />

TRRAP<br />

•<br />

UHRF1<br />

•<br />

WDR5<br />

•<br />

WSTF<br />

•<br />

XPB<br />

•<br />

XPD<br />

•<br />

YY1<br />

•<br />

155<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for Histone H3 have<br />

been cited over 155 times in highimpact,<br />

peer-reviewed publications<br />

from the global research community.<br />

Select Citations:<br />

Crotti, A. et al. (2014) Mutant<br />

Huntingtin promotes autonomous<br />

microglia activation via myeloid<br />

lineage-determining factors. Nat.<br />

Neurosci. 17, 513−521.<br />

Tanaka-Nakanishi, A. et al. (2014)<br />

HTLV-1 bZIP factor suppresses<br />

apoptosis by attenuating the function<br />

of FoxO3a and altering its localization.<br />

Cancer Res. 74, 188−200.<br />

Hast, B.E. et al. (2014) Cancerderived<br />

mutations in KEAP1 impair<br />

NRF2 degradation but not ubiquitination.<br />

Cancer Res. 74, 808−817.<br />

Chen, C.H. et al. (2014) Synergistic<br />

interaction between the HDAC inhibitor,<br />

MPT0E028, and sorafenib in liver<br />

cancer cells in vitro and in vivo. Clin.<br />

Cancer Res. 20, 1274−1287.<br />

Ghare, S.S. et al. (2014) Coordinated<br />

Histone H3 Methylation and Acetylation<br />

Regulate Physiologic and Pathologic<br />

Fas Ligand Gene Expression in<br />

Human CD4+ T Cells. J. Immunol.<br />

193, 412−421.<br />

Karijolich, J. et al. (2014) Coordinated<br />

Histone H3 Methylation and<br />

Acetylation Regulate Physiologic and<br />

Pathologic Fas Ligand Gene Expression<br />

in Human CD4+ T Cells. J. Virol.<br />

88, 7024−7035.<br />

Galli, G.G. et al. (2014) Prdm5<br />

suppresses Apc(Min)-driven intestinal<br />

adenomas and regulates monoacylglycerol<br />

lipase expression. Oncogene<br />

33, 3342−3350.<br />

Khalaj, M. et al. (2014) A missense<br />

mutation in Rev7 disrupts formation of<br />

Polzeta, impairing mouse development<br />

and repair of genotoxic agent-induced<br />

DNA lesions. J. Biol. Chem. 289,<br />

3811−3824.<br />

Belozerov, V.E. et al. (2014) In vivo<br />

interaction proteomics reveal a novel<br />

p38 mitogen-activated protein kinase/<br />

Rack1 pathway regulating proteostasis<br />

in Drosophila muscle. Mol. Cell Biol.<br />

34, 474−484.<br />

Gray, C.M. et al. (2014) Noncanonical<br />

NF-kappaB signaling is limited by<br />

classical NF-kappaB activity. Sci.<br />

Signal. 7, 13.<br />

24 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstchromatin 25


Section I: Research Areas<br />

Protein Acetylation<br />

Histone<br />

Acetylation<br />

Protein<br />

Acetylation<br />

GCN5L2<br />

Cell Cycle<br />

Rb<br />

Stability<br />

DNA Damage<br />

SRC-3<br />

Tip60<br />

ATM<br />

DNA<br />

Repair<br />

TF<br />

CBP/<br />

p300<br />

TF<br />

HDAC Classes<br />

Class I: HDAC1-3, 8<br />

Class II: HDAC4-7, 9, 10<br />

Class III: SIRT1-7<br />

Class IV: HDAC11<br />

p53<br />

TF<br />

Stability<br />

Acetylases<br />

PCAF<br />

Su<br />

Sirtuins<br />

Regulatory<br />

Signaling Pathways<br />

DNA<br />

Damage<br />

AceCS1<br />

PEPCK<br />

PGC-1α<br />

Idh2<br />

TFIID<br />

TFIIA TBP<br />

HDAC<br />

NuRD<br />

NcoR<br />

Sin3<br />

26 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

Tip60<br />

Histones<br />

Signaling Pathways<br />

Metabolism<br />

Deacetylases<br />

SirT1<br />

Tip60<br />

SirT1<br />

SirT3<br />

Cytoskeletal Regulation Ribosomal Proteins<br />

Chaperones<br />

BRCA1<br />

TATA<br />

RNA Splicing<br />

Acetylation<br />

Acetyl<br />

Transferases<br />

ATF-2 MORF<br />

CBP MOZ<br />

CDY p300<br />

PCAF CLOCK PCAF<br />

CBP/<br />

p300<br />

EWI p/CIP<br />

Su<br />

Elp3 SRC-1<br />

SRC-3<br />

GCN5L2 SRC-3<br />

GRIP hTAF II<br />

250<br />

α-Importin<br />

HAT1 TFIIB<br />

HBO1 Tip60<br />

MCM3AP<br />

Nuclear Transport<br />

Transcription<br />

TFIIH<br />

Pluripotency<br />

TFIIF<br />

Development<br />

TFIIB<br />

RNA Pol II Differentiation of<br />

Neural Stem Cells<br />

TFIIE<br />

BMP, Wnt, Notch<br />

AceCS1<br />

PEPCK<br />

PGC-1α<br />

Idh2<br />

Deacetylation<br />

Fatty Acid Synthesis<br />

Histone Acetylation<br />

Gluconeogenesis<br />

Expression of<br />

Gluconeogenic Genes<br />

NADPH<br />

Lysine acetylation is a reversible post-translational modification that plays a crucial role in regulating protein function, chromatin structure, and gene expression. Many<br />

transcriptional coactivators possess intrinsic acetylase activity, while transcriptional corepressors are associated with deacetylase activity. Acetylation complexes (such as CBP/<br />

p300 and PCAF) or deacetylation complexes (such as Sin3, NuRD, NcoR, and SMRT) are recruited to DNA-bound transcription factors (TFs) in response to signaling pathways.<br />

Histone hyperacetylation by histone acetyltransferases (HATs) is associated with transcriptional activation, whereas histone deacetylation by histone deacetylases (HDACs) is<br />

associated with transcriptional repression. Histone acetylation stimulates transcription by remodeling higher order chromatin structure, weakening histone-DNA interactions,<br />

and providing binding sites for transcriptional activation complexes containing proteins that possess bromodomains, which bind acetylated lysine. Histone deacetylation<br />

represses transcription through an inverse mechanism involving the assembly of compact higher order chromatin and the exclusion of bromodomain-containing transcription<br />

activation complexes. Histone hypoacetylation is a hallmark of silent heterochromatin. Site-specific acetylation of a growing number of non-histone proteins has been shown<br />

to regulate their activity, localization, specific interactions, and stability/degradation. Remarkably, recent advances in mass spectrometry technologies allowed high resolution<br />

mapping of most of the acetylation sites in all the proteome. These studies demonstrated that the “acetylome” encompasses nearly ~3600 acetylation sites in roughly ~1750<br />

proteins, suggesting that this modification is one of the most abundant chemical modifications in nature. Indeed, it appears that this mark can influence the activity of proteins<br />

in diverse biological processes, including chromatin remodeling, cell cycle, splicing, nuclear transport, mitochondrial biology, and actin nucleation. At an organismal level,<br />

acetylation plays an important role in immunity, circadian rhythmicity, and memory formation. Protein acetylation is becoming a favorable target in drug design for numerous<br />

disease conditions.<br />

Select Reviews:<br />

Albaugh, B.N., Arnold, K.M., and Denu, J.M. (2011) Chem. Bio. Chem. 12, 290–298. • Choudhary, C., Kumar, C., Gnad, F., Nielsen, M.L., Rehman, M., Walther, T.C.,<br />

Olsen, J.V., and Mann, M. (2009) Science 325, 834–840. • Dali-Youcef, N., Lagouge, M., Froelich, S. et al. (2007) Ann. Med. 39, 335–345. • Finkel, T., Deng, C.H., and<br />

Mostoslavsky, R. (2009) Nature 460, 587–591. • Haberland, M., Montgomery, R.L., and Olson, E.N. (2009) Nat. Rev. Genet. 10, 32–42. • Peng, L. and Seto, E. (2011)<br />

Handbook Exp. Pharmac. 206, 39–56. • Spange, S., Wagner, T., Heinzel, T., and Krämer, O.H. (2009) Int. J. Biochem. Cell Biol. 41, 185–198. • Yang, X.J. and Seto, E.<br />

(2007) Oncogene 26, 5310–5318. • Yang, X.J. and Seto, E. (2008) Mol. Cell 31, 449–461.<br />

© 2002–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Raul Mostoslavsky, Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

H4K20me<br />

Methylases:<br />

SET8/KMT5A (+)<br />

NSD1/KMT3B (+,†)<br />

ASH1L/KMT2H (+,†,‡)<br />

SUV420H1/KMT5B (†,‡)<br />

SUV420H2/KMT5C (†,‡)<br />

NSD2/KMT3G (+,†)<br />

Demethylases:<br />

JHDM1D/KDM7A (+,†)<br />

PHF8/KDM7B (+,†)<br />

H3K9me<br />

Methylases:<br />

PRDM3/KMT8E (+)<br />

PPRDM16/KMT8F (+)<br />

G9a/EHMT2/KMT1C (+,†)<br />

EHMT1/KMT1D (+,†)<br />

ASH1L/KMT2H (+,†,‡)<br />

PRDM2/KMT8A (+,†,‡)<br />

PRDM8/KMT8D (†)<br />

SUV39H1/KMT1A (†,‡)<br />

SUV39H2/KMT1B (†,‡)<br />

ESET/KMT1E (†,‡)<br />

CLLD8/KMT1F (†,‡)<br />

Demethylases:<br />

AOF1/KDM1B (+,†)<br />

JMJD1A/KDM3A (+,†)<br />

JMJD1B/KDM3B (+,†)<br />

JMJD1C/KDM3C (+,†)<br />

JHDM1D/KDM7A (+,†)<br />

PHF8/KDM7B (+,†)<br />

JMJD2A/KDM4B (†,‡)<br />

JMJD2B/KDM4B (†,‡)<br />

JMJD2C/KDM4C (†,‡)<br />

JMJD2D/KDM4D (†,‡)<br />

KDM4E/KDM4DL (†,‡)<br />

H3K27me<br />

Methylases:<br />

Ezh2/KMT6 (+,†,‡)<br />

NSD2/KMT3G (+,†,‡)<br />

NSD3/KMT3F (†,‡)<br />

Demethylases:<br />

JHDM1D/KDM7A (+,†)<br />

PHF8/KDM7B (+,†)<br />

UTX/KDM6A (†,‡)<br />

JMJD3/KDM6B (†,‡)<br />

Histone Lysine Methylation<br />

SUV<br />

39h<br />

HP1<br />

Me<br />

Me<br />

Transcriptionally Inactive<br />

Chromatin<br />

SUV<br />

39h<br />

PRC1<br />

PC<br />

Me<br />

PRC1<br />

PC<br />

Me<br />

HP1<br />

Me<br />

Me<br />

SUV<br />

39h<br />

HP1<br />

Me<br />

Me<br />

SUV<br />

39h<br />

HP1<br />

Me<br />

Pericentric Heterochromatin<br />

Inactive X Chromosome<br />

Rb-Mediated Repression<br />

PRC1<br />

PC<br />

Me<br />

PRC1<br />

PC<br />

Me<br />

Me<br />

Pericentric Heterochromatin<br />

Inactive X Chromosome<br />

Hox Gene Repression<br />

Methylation<br />

H4K20me<br />

Demethylation<br />

Methylation<br />

H3K9me<br />

Demethylation<br />

Methylation Degree: + Mono † Di ‡ Tri<br />

Methylation<br />

H3K27me<br />

Demethylation<br />

chapter 01: GENE EXPRESSION, EPIGENETICS, AND NUCLEAR FUNCTION<br />

Methylation<br />

Methylation<br />

Demethylation<br />

Methylation<br />

H3K79me<br />

Demethylation<br />

H3K4me<br />

Demethylation<br />

H3K36me<br />

Transcriptionally Active<br />

Chromatin<br />

NURF<br />

BPTF<br />

Me<br />

Me<br />

Me<br />

Me<br />

NURF<br />

BPTF<br />

Me<br />

Me<br />

Me<br />

Me<br />

Me<br />

Anti-Silencing<br />

Me<br />

Me<br />

Transcription Initiation<br />

Me<br />

Transcription Elongation<br />

The nucleosome is the primary building block of chromatin containing a histone octamer composed of two sets of H3-H4 and H2A-H2B dimers.<br />

Originally thought to function as a static scaffold for DNA packaging, histones have more recently been shown to be dynamic proteins, undergoing<br />

multiple types of post-translational modifications and impacting numerous nuclear functions. Lysine methylation is one such modification and is<br />

a major determinant for genome organization and the formation of active and inactive regions of the genome. Lysines can have three different<br />

methylation states (mono-, di-, and tri-) that are associated with different nuclear features and transcriptional states. In order to establish these<br />

methylation states, cells have enzymes that both add (lysine methyltransferases- KMTs) and remove (lysine demethylases- KDMs) different degrees<br />

of methylation from specific lysines within the histones. To date, all but one histone lysine methyltransferase (DOT1L/KMT4) has a conserved<br />

catalytic SET domain that was originally identified in the Drosophila Su[var]3-9, Enhancer of zeste, and Trithorax proteins. In the case of the histone<br />

lysine demethylases, there are two different classes: the FAD-dependent amine oxidases and the JmjC-containing enzymes. Both KMTs and KDMs<br />

have specificity for specific lysine residues and degrees of methylation within the histone tails. Therefore, all KMTs and KDMs are not the same in<br />

their biological functions or roles in transcriptional output.<br />

Lysine methylation has been implicated in both transcriptional activation (H3K4, K36, K79) and silencing (H3K9, K27, H4K20). The degree of<br />

methylation is associated with different outcomes. For example, H4K20 monomethyation (H4K20me1) is observed in the bodies of active genes,<br />

while H4K20 trimethylation (H4K20me3) is affiliated with gene repression and compacted genomic regions. Gene regulation is also affected by<br />

the location of the methylated lysine residue with respect to the DNA sequence. For example, H3K9me3 at promoters is associated with gene<br />

repression, while some induced genes have H3K9me3 in the gene body. Since this modification is uncharged and chemically inert, the impact<br />

these modifications have is through recognition by other proteins with binding motifs. Lysine methylation coordinates the recruitment of chromatin<br />

modifying enzymes. Chromodomains (e.g., found in HP1, PRC1), PHD fingers (e.g., found in BPTF, ING2, SMCX/KDM5C), Tudor domains (e.g.,<br />

found in 53BP1 and JMJD2A/KDM4A), PWWP domains (e.g., found in ZMYND11) and WD-40 domains (e.g., found in WDR5) are among a<br />

growing list of methyl lysine binding modules found in histone acetyltransferases, deacetylases, methylases, demethylases and ATP-dependent<br />

chromatin remodeling enzymes. Lysine methylation provides a binding surface for these enzymes, which then regulate chromatin condensation<br />

and nucleosome mobility, active and inactive transcription as well as DNA repair and replication. In addition, lysine methylation can block binding<br />

of proteins that interact with unmethylated histones or directly inhibit catalysis of other regulatory modifications on neighboring residues.<br />

Histone methylation is crucial for proper programming of the genome during development and misregulation of the methylation machinery can<br />

lead to diseased states such as cancer. In fact, cancer genome analyses have uncovered lysine mutations in H3K27 and H3K36. These sites are<br />

enriched in subsets of cancer. Therefore, an entirely new therapeutic and biomarker space is emerging with the discovery of these enzymes, the<br />

impact modifications have on the genome and disease associated mutations.<br />

Select Reviews:<br />

Black, J.C., Van Rechem, C., and Whetstine, J.R. (2012) Mol. Cell. 48, 491–507. • Greer, E.L. and Shi, Y. (2012) Nat. Rev. Genet. 13, 343–357.<br />

• Herz, H.M., Garruss, A., and Shilatifard, A. (2013) Trends Biochem. Sci. 38, 621–639. • Kooistra, S.M. and Helin, K. (2012) Nat. Rev. Mol.<br />

Cell Biol. 13, 297–311. • Tee, W.W. and Reinberg, D. (2014) Development 141, 2376–2390. • Van Rechem, C. and Whetstine, J.R. (2014)<br />

Biophys. Acta. May 23 [Epub ahead of print].<br />

© 2006–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Jonathan Whetstine for reviewing this diagram.<br />

H3K4me H4K20me<br />

Methylases: Methylases:<br />

MLL1/KMT2A SET8/KMT5A (+,†,‡) (+)<br />

MLL2/KMT2B NSD1/KMT3B (+,†,‡) (+,†)<br />

MLL3/KMT2C ASH1L/KMT2H (+) (+,†,‡)<br />

MLL4/KMT2D (+) SUV420H1/KMT5B (†,‡)<br />

SET1A/KMT2F (+,†,‡)<br />

SUV420H2/KMT5C (†,‡)<br />

SET1B/KMT2G (+,†,‡)<br />

NSD3/KMT3F NSD2/KMT3G (+,†) (+,†)<br />

NSD2/KMT3G Demethylases: (+,†)<br />

SET7/KMT7 JHDM1D/KDM7A (†) (+,†)<br />

SMYD3/KMT3E PHF8/KDM7B (†,‡) (+,†)<br />

ASH1L/KMT2H (+,†,‡)<br />

PRDM9/KMT8B H3K9me (‡)<br />

Demethylases:<br />

Methylases:<br />

LSD1/KDM1A (+,†)<br />

PRDM3/KMT8E (+)<br />

AOF1/KDM1B (+,†)<br />

JARID1A/KDM5A PPRDM16/KMT8F (+) (†,‡)<br />

JARID1B/KDM5B G9a/EHMT2/KMT1C (†,‡) (+,†)<br />

JARID1C/KDM5C EHMT1/KMT1D (†,‡) (+,†)<br />

JARID1D/KDM5D ASH1L/KMT2H (†,‡) (+,†,‡)<br />

PRDM2/KMT8A (+,†,‡)<br />

PRDM8/KMT8D (†)<br />

H3K36me SUV39H1/KMT1A (†,‡)<br />

Methylases:<br />

SUV39H2/KMT1B (†,‡)<br />

NSD1/KMT3B (+,†)<br />

SMYD2/KMT3C ESET/KMT1E (+,†) (†,‡)<br />

NSD2/KMT3G CLLD8/KMT1F (+,†) (†,‡)<br />

SET2/KMT3A Demethylases: (‡)<br />

Demethylases: AOF1/KDM1B (+,†)<br />

JMJD1A/KDM2A JMJD1A/KDM3A (+,†) (+,†)<br />

JMJD1B/KDM2B JMJD1B/KDM3B (+,†) (+,†)<br />

JMJD2A/KDM4A (†,‡)<br />

JMJD1C/KDM3C (+,†)<br />

JMJD2B/KDM4B (†,‡)<br />

JHDM1D/KDM7A (+,†)<br />

JMJD2C/KDM4C (†,‡)<br />

JMJD2D/KDM4D PHF8/KDM7B (†,‡) (+,†)<br />

KDM4DL/KDM4E JMJD2A/KDM4B (†,‡) (†,‡)<br />

JMJD2B/KDM4B (†,‡)<br />

JMJD2C/KDM4C (†,‡)<br />

JMJD2D/KDM4D (†,‡)<br />

KDM4E/KDM4DL (†,‡)<br />

H3K79me<br />

Methylases:<br />

H3K27me<br />

DOTIL/KMT4 (+,†,‡)<br />

Demethylases: Methylases:<br />

Ezh2/KMT6 (+,†,‡)<br />

NSD2/KMT3G (+,†,‡)<br />

NSD3/KMT3F (†,‡)<br />

Demethylases:<br />

JHDM1D/KDM7A (+,†)<br />

PHF8/KDM7B (+,†)<br />

UTX/KDM6A (†,‡)<br />

JMJD3/KDM6B (†,‡)<br />

H3K4me<br />

Methylases:<br />

MLL1/KMT2A (+,†,‡)<br />

MLL2/KMT2B (+,†,‡)<br />

MLL3/KMT2C (+)<br />

MLL4/KMT2D (+)<br />

SET1A/KMT2F (+,†,‡)<br />

SET1B/KMT2G (+,†,‡)<br />

NSD3/KMT3F (+,†)<br />

NSD2/KMT3G (+,†)<br />

SET7/KMT7 (†)<br />

SMYD3/KMT3E (†,‡)<br />

ASH1L/KMT2H (+,†,‡)<br />

PRDM9/KMT8B (‡)<br />

Demethylases:<br />

LSD1/KDM1A (+,†)<br />

AOF1/KDM1B (+,†)<br />

JARID1A/KDM5A (†,‡)<br />

JARID1B/KDM5B (†,‡)<br />

JARID1C/KDM5C (†,‡)<br />

JARID1D/KDM5D (†,‡)<br />

H3K36me<br />

Methylases:<br />

NSD1/KMT3B (+,†)<br />

SMYD2/KMT3C (+,†)<br />

NSD2/KMT3G (+,†)<br />

SET2/KMT3A (‡)<br />

Demethylases:<br />

JMJD1A/KDM2A (+,†)<br />

JMJD1B/KDM2B (+,†)<br />

JMJD2A/KDM4A (†,‡)<br />

JMJD2B/KDM4B (†,‡)<br />

JMJD2C/KDM4C (†,‡)<br />

JMJD2D/KDM4D (†,‡)<br />

KDM4DL/KDM4E (†,‡)<br />

H3K79me<br />

Methylases:<br />

DOTIL/KMT4 (+,†,‡)<br />

Demethylases:<br />

<br />

www.cellsignal.com/cstpathways 27


Section I: Research Areas<br />

chapter 01: GENE EXPRESSION, EPIGENETICS, AND NUCLEAR FUNCTION<br />

This table provides<br />

a referenced list of<br />

many known histone<br />

modifications, the<br />

associated modifying<br />

enzymes, and proposed<br />

functions.<br />

References<br />

1. Clarke, A.S. et al. (1999) Mol.<br />

Cell Biol. 19, 2515–2526.<br />

2. Kimura, A. and Horikoshi, M.<br />

(1998) Genes Cells 3, 789–800.<br />

3. Schiltz, R.L. et al. (1999) J.<br />

Biol. Chem. 274, 1189–1192.<br />

4. Verreault, A. et al. (1998)<br />

Curr. Biol. 8, 96–108.<br />

5. Kawasaki, H. et al. (2000)<br />

Nature 405, 195–200.<br />

6. Suka, N. et al. (2001)<br />

Mol. Cell 8, 473–479.<br />

7. Angus-Hill, M.L. et al. (1999)<br />

J. Mol. Biol. 294, 1311–1325.<br />

8. Sobel, R.E. et al. (1995)<br />

Proc. Natl. Acad. Sci. USA<br />

92, 1237–1241.<br />

9. Grant, P.A. et al. (1999) J. Biol.<br />

Chem. 274, 5895–5900.<br />

10. Spencer, T.E. et al. (1997)<br />

Nature 389, 194–198.<br />

11. Bird, A.W. et al. (2002)<br />

Nature 419, 411–415.<br />

12. Ikura, T. et al. (2000)<br />

Cell 102, 463–473.<br />

13. Winkler, G.S. et al. (2002)<br />

Proc. Natl. Acad. Sci. USA<br />

99, 3517–3522.<br />

14. Hsieh, Y.J. et al. (1999) Mol.<br />

Cell. Biol. 19, 7697–7704.<br />

15. Mizzen, C.A. et al. (1996)<br />

Cell 87, 1261–1270.<br />

16. Sutton, A. et al. (2003) J. Biol.<br />

Chem. 278, 16887–16892.<br />

17. Howe, L. et al. (2001) Genes<br />

Dev. 15, 3144–3154.<br />

18. Daujat, S. et al. (2002)<br />

Curr. Biol. 12, 2090–2097.<br />

19. Xu, F. et al. (2005)<br />

Cell 121, 375–385.<br />

20. Masumoto, H. et al. (2005)<br />

Nature 436, 294–298.<br />

21. Parthun, M.R. et al. (1996)<br />

Cell 87, 85–94.<br />

22. Kuo, M.H. et al. (1996)<br />

Nature 383, 269–272.<br />

23. Kelly, T.J. et al. (2000) Mol.<br />

Cell. Biol. 20, 7051–7058.<br />

24. Hilfiker, A. et al. (1997)<br />

EMBO J. 16, 2054–2060.<br />

25. Ye, J. et al. (2005) Mol.<br />

Cell. 18, 123–130.<br />

26. Briggs, S.D. et al. (2001)<br />

Genes Dev. 15, 3286–3295.<br />

27. Wang, H. et al. (2001)<br />

Mol. Cell. 8, 1207–1217.<br />

Histone Modifications Table<br />

The nucleosome, made up of four core histone proteins (H2A, H2B, H3, and H4), and linker histone<br />

H1 are the primary building blocks of chromatin. Originally thought to function as a static scaffold for<br />

DNA packaging, histones have more recently been shown to be dynamic proteins, undergoing multiple<br />

types of post-translational modifications that regulate chromatin condensation and DNA accessibility.<br />

For example, acetylation of lysine residues has long been associated with histone deposition and<br />

transcriptional activation, and more recently found to be associated with DNA repair. Phosphorylation<br />

of serine and threonine residues facilitates chromatin condensation during mitosis and transcriptional<br />

activation of immediate-early genes. Methylation of lysine and arginine residues function as a major<br />

determinant for formation of transcriptionally active and inactive regions of chromatin and is crucial for<br />

proper programming of the genome during development.<br />

Acetylation<br />

Histone Site Histone-modifying Enzymes Proposed Function Ref. #<br />

H2A Lys4 (S. cerevisiae) Esa1 transcriptional activation (1)<br />

Lys5 (mammals) Tip60, p300/CBP transcriptional activation (2,3)<br />

Lys7 (S. cerevisiae) Hat1 unknown (4)<br />

Esa1 transcriptional activation (1)<br />

H2B Lys5 p300, ATF2 transcriptional activation (3,5)<br />

Lys11 (S. cerevisiae) Gcn5 transcriptional activation (6)<br />

Lys12 (mammals) p300/CBP, ATF2 transcriptional activation (3,5)<br />

Lys15 (mammals) p300/CBP, ATF2 transcriptional activation (3,5)<br />

Lys16 (S. cerevisiae) Gcn5, Esa1 transcriptional activation (6)<br />

Lys20 p300 transcriptional activation (3)<br />

H3 Lys4 (S. cerevisiae) Esa1 transcriptional activation (1)<br />

Hpa2 unknown (7)<br />

Lys9 unknown histone deposition (8)<br />

Gcn5, SRC-1 transcriptional activation (9,10)<br />

Lys14 unknown histone deposition (8)<br />

Gcn5, PCAF transcriptional activation (3,11)<br />

Esa1, Tip60 transcriptional activation (1,2)<br />

DNA repair (11,12)<br />

SRC-1 transcriptional activation (10)<br />

Elp3 transcriptional activation (elongation) (13)<br />

Hpa2 unknown (7)<br />

hTFIIIC90 RNA polymerase III transcription (14)<br />

TAF1 RNA polymerase II transcription (15)<br />

Sas2 euchromatin (16)<br />

Sas3 transcriptional activation (elongation) (17)<br />

p300 transcriptional activation (3)<br />

Lys18 Gcn5 transcriptional activation, DNA repair (9)<br />

p300/CBP DNA replication, transcriptional activation (3,18)<br />

Lys23 unknown histone deposition (8)<br />

Gcn5 transcriptional activation, DNA repair (9)<br />

Sas3 transcriptional activation (elongation) (17)<br />

p300/CBP transcriptional activation (3,18)<br />

Lys27 Gcn5 transcriptional activation (6)<br />

Lys36 Gcn5 transcriptional activation (82)<br />

Lys56 (S. cerevisiae) Spt10 transcriptional activation (19)<br />

DNA repair (20)<br />

H4 Lys5 Hat1 histone deposition (21)<br />

Esa1, Tip60 transcriptional activation (1,2)<br />

DNA repair (11,12)<br />

ATF2 transcriptional activation (5)<br />

Hpa2 unknown (7)<br />

p300 transcriptional activation (3)<br />

Acetylation<br />

Histone Site Histone-modifying Enzymes Proposed Function Ref. #<br />

Lys8 Gcn5, PCAF transcriptional activation (3,22)<br />

Esa1, Tip60 transcriptional activation (1,2)<br />

DNA repair (11,12)<br />

ATF2 transcriptional activation (5)<br />

Elp3 transcriptional activation (elongation) (13)<br />

p300 transcriptional activation (3)<br />

Lys12 Hat1 histone deposition (21)<br />

telomeric silencing (23)<br />

Esa1, Tip60 transcriptional activation (1,2)<br />

DNA repair (11,12)<br />

Hpa2 unknown (7)<br />

p300 transcriptional activation (3)<br />

Lys16 Gcn5 transcriptional activation (22)<br />

MOF (D. melanogaster) transcriptional activation (24)<br />

Esa1, Tip60 transcriptional activation (1,2)<br />

DNA repair (11,12)<br />

ATF2 transcriptional activation (5)<br />

Sas2 euchromatin (2,6)<br />

Lys91 (S. cerevisiae) Hat1/Hat2 chromatin assembly (25)<br />

Methylation<br />

Histone Site Histone-modifying Enzymes Proposed Function Ref. #<br />

H1 Lys26 Ezh2 transcriptional silencing (48,49)<br />

H2A Arg3 PRMT1/6, PRMT5/7 transcriptional activation, transcriptional<br />

repression<br />

H3 Arg2 PRMT5, PRMT6 transcriptional repression (83)<br />

Arg8 PRMT5, PRMT2/6 transcriptional activation, transcriptional (31, 88)<br />

repression<br />

Arg17 CARM1 transcriptional activation (18)<br />

Arg26 CARM1 transcriptional activation (83)<br />

Arg42 CARM1 transcriptional activation (89)<br />

Lys4 Set1 (S. cerevisiae) permissive euchromatin (di-Me) (26)<br />

Set7/9 (vertebrates) transcriptional activation (tri-Me) (27)<br />

MLL, ALL-1 transcriptional activation (28,29)<br />

Ash1 (D. melanogaster) transcriptional activation (30)<br />

Lys9 Suv39h,Clr4 transcriptional silencing (tri-Me) (32,33)<br />

G9a<br />

transcriptional repression genomic<br />

(34)<br />

imprinting<br />

SETDB1 transcriptional repression (tri-Me) (35)<br />

Dim-5 (N. crassa),<br />

DNA methylation (tri-Me) (36,37)<br />

Kryptonite (A. thaliana)<br />

Ash1 (D. melanogaster) transcriptional activation (30)<br />

Lys27 Ezh2 transcriptional silencing (38)<br />

X inactivation (tri-Me)<br />

G9a transcriptional silencing (34)<br />

Lys36 Set2 transcriptional activation (elongation) (39)<br />

Lys79 Dot1 euchromatin (40)<br />

transcriptional activation (elongation) (41)<br />

checkpoint response (42)<br />

H4 Arg3 PRMT1/6 transcriptional activation (43)<br />

PRMT5/7 transcriptional repression (31)<br />

Lys20 PR-Set7 transcriptional silencing (mono-Me) (44)<br />

Suv4-20h heterochromatin (tri-Me) (45)<br />

Ash1 (D. melanogaster) transcriptional activation (30)<br />

Set9 (S. pombe) checkpoint response (46)<br />

Lys59 unknown transcriptional silencing (47)<br />

(83)<br />

References<br />

28. Nakamura, T. et al. (2002)<br />

Mol. Cell. 10, 1119–1128.<br />

29. Sedkov, Y. et al. (2003)<br />

Nature 426, 78–83.<br />

30. Beisel, C. et al. (2002)<br />

Nature 419, 857–862.<br />

31. Pal, S. et al. (2004) Mol.<br />

Cell. Biol. 24, 9630–9645.<br />

32. Rea, S. et al. (2000) Nature<br />

406, 593–599.<br />

33. Nakayama, J. et al. (2001)<br />

Science 292, 110–113.<br />

34. Tachibana, M. et al. (2001)<br />

J. Biol. Chem. 276,<br />

25309–25317.<br />

35. Schultz, DzC. et al. (2002)<br />

Genes Dev. 16, 919–932.<br />

36. Tamaru, H. and Selker, E.U.<br />

(2001) Nature 414, 277–283.<br />

37. Johnson, L. et al. (2002)<br />

Curr. Biol. 12, 1360–1367.<br />

38. Cao, R. et al. (2002)<br />

Science 298, 1039–1043.<br />

39. Krogan, N.J. et al. (2002) Mol.<br />

Cell. Biol. 23, 4207–4218.<br />

40. Feng, Q. et al. (2002)<br />

Curr. Biol. 12, 1052–1058.<br />

41. Krogan, N.J. et al. (2003) Mol.<br />

Cell. 11, 721–729.<br />

42. Huyen, Y. et al. (2004) Nature<br />

432, 406–411.<br />

43. Strahl, B.D. et al. (2001)<br />

Curr. Biol. 11, 996–1000.<br />

44. Nishioka, K. et al. (2002)<br />

Mol. Cell. 9, 1201–1213.<br />

45. Schotta, G. et al. (2004)<br />

Genes Dev. 18, 1251–1260.<br />

46. Sanders, S.L. et al. (2004)<br />

Cell 119, 603–614.<br />

47. Zhang, L. et al. (2003)<br />

Chromosoma 112, 77–86.<br />

48. Daujat, S. et al. (2005) J. Biol.<br />

Chem. 280, 38090–38095.<br />

49. Kuzmichev, A. et al. (2004)<br />

Mol. Cell 14, 183–193.<br />

50. Barber, C.M. et al. (2004)<br />

Chromosoma 112, 360–371.<br />

51. Zhang, Y. et al. (2004) J. Biol.<br />

Chem. 279, 21866–21872.<br />

52. Aihara, H. et al. (2004)<br />

Genes Dev. 18, 877–888.<br />

53. Harvey, A.C. et al. (2005)<br />

Genetics 170, 543–553.<br />

54. Downs, J.A. et al. (2000)<br />

Nature 408, 1001–1004.<br />

55. Shroff, R. et al. (2004) Curr.<br />

Biol. 14, 1703–1711.<br />

56. Ward, I.M. and Chen, J.<br />

(2001) J. Biol. Chem. 276,<br />

47759–47762.<br />

57. Burma, S. et al. (2001) J. Biol.<br />

Chem. 276, 42462–42467.<br />

58. Park, E.J. et al. (2003) Nucleic<br />

Acids Res. 31, 6819–6827.<br />

59. Ahn, S.H. et al. (2005)<br />

Cell 120, 25–36.<br />

60. Cheung, W.L. et al. (2003)<br />

Cell 113, 507–517.<br />

28 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables 29


Section I: Research Areas<br />

chapter 01: GENE EXPRESSION, EPIGENETICS, AND NUCLEAR FUNCTION<br />

References<br />

61. Fernandez-Capetillo, O. et al.<br />

(2004) J. Exp. Med. 199,<br />

1671–1677.<br />

62. Maile, T. et al. (2004)<br />

Science 304, 1010–1014.<br />

63. Dai, J. et al. (2004)<br />

Genes Dev. 19, 472–488.<br />

64. Hendzel, M.J. et al. (1997)<br />

Chromosoma 106, 348–360.<br />

65. Hsu, J.Y. et al. (2000)<br />

Cell 102, 279–291.<br />

66. Soloaga, A. et al. (2003)<br />

EMBO J. 22, 2788–2797.<br />

67. Anest, V. et al. (2003)<br />

Nature 423, 659–663.<br />

68. Lo, W.S. et al. (2001)<br />

Science 293, 1142–1146.<br />

69. Preuss, U. et al. (2003) Nucleic<br />

Acids Res. 31, 878–885.<br />

70. Goto, H. et al. (2002)<br />

Genes Cells 7, 11–17.<br />

71. Zhong, S. et al. (2001) J. Biol.<br />

Chem. 276, 33213–33219.<br />

72. Cheung, W.L. et al. (2005)<br />

Curr. Biol. 15, 656–660.<br />

73. Wang, H. et al. (2004)<br />

Nature 431, 873–878.<br />

74. Zhu, B. et al. (2005)<br />

Mol. Cell. 20, 601–611.<br />

75. Robzyk, K. et al. (2000)<br />

Science 287, 501–504.<br />

76. Nathan, D. et al. (2006)<br />

Genes Dev. 20, 966–976.<br />

77. Shiio, Y. and Eisenman, R.N.<br />

(2003) Proc. Natl. Acad. Sci.<br />

USA 100, 13225–13230.<br />

78. Chew, Y.C. et al. (2006) J.<br />

Nutr. Biochem. 17, 225–233.<br />

79. Kobza, K. et al. (2005)<br />

FEBS J. 272, 4249–4259.<br />

80. Camporeale, G. et al. (2004)<br />

Eur. J. Biochem. 271,<br />

2257–2263.<br />

81. Kothapalli, N. and Zempleni, J.<br />

(2004) FASEB J. 18, 103–104.<br />

82. Morris, S.A. et al. (2007) J.<br />

Biol Chem. 282, 7632–7640.<br />

83. DiLorenzo, A. et al. (2011)<br />

FEBS Lett. 585, 2024–2031.<br />

84. Bungard, D. et al. (2010)<br />

Science 329, 1201–1205.<br />

85. Metzger, E. et al. (2010)<br />

Nature 464, 792–796.<br />

86. Dawson, M.A. et al. (2009)<br />

Nature 461, 819–822.<br />

87. Hurd, P.J. et al. (2009) J. Biol.<br />

Chem. 284, 16575–16583.<br />

88. Su, X. et al. (2014)<br />

Genes Dev. 28, 622–636.<br />

89. Casadio, F. et al. (2013)<br />

Proc. Natl. Acad. Sci. USA<br />

110, 14894–14899.<br />

90. Yamagishi, Y. et al. (2010)<br />

Science 330, 239–243.<br />

91. Kim, K. et al. (2013)<br />

Mol. Cell 52, 459–467.<br />

92. Xiao, A. et al. (2009)<br />

Nature 457, 57–62.<br />

Phosphorylation<br />

Histone Site Histone-modifying Enzymes Proposed Function Ref. #<br />

H1 Ser27<br />

transcriptional activation, chromatin<br />

unknown<br />

(48,49)<br />

decondensation<br />

H2A Ser1 unknown mitosis, chromatin assembly (50)<br />

MSK1 transcriptional repression (51)<br />

Ser122 (S. cerevisiae) unknown DNA repair (53)<br />

Ser129 (S. cerevisiae) Mec1, Tel1 DNA repair (54,55)<br />

Ser139 (mammalian H2A.X) ATR, ATM, DNA-PK DNA repair (56-58)<br />

Thr119 (D. melanogaster) NHK1 mitosis (52)<br />

Thr120 (mammals) Bub1, VprBP mitosis, transcriptional repression (90,91)<br />

Thr142 (mammalian H2A.X) WSTF apoptosis, DNA repair (92)<br />

H2B Ser10 (S. cerevisiae) Ste20 apoptosis (59)<br />

Ser14 (vertebrates) Mst1 apoptosis (60)<br />

unknown DNA repair (61)<br />

Ser33 (D. melanogaster) TAF1 transcriptional activation (62)<br />

Ser36 AMPK transcriptional activation (84)<br />

H3 Ser10 Aurora-B kinase mitosis, meiosis (64,65)<br />

MSK1, MSK2 immediate-early gene activation (66)<br />

IKK-α transcriptional activation (67)<br />

Snf1 transcriptional activation (68)<br />

Ser28 (mammals) Aurora-B kinase mitosis (70)<br />

MSK1, MSK2 immediate-early activation (66,71)<br />

Thr3 Haspin/Gsg2 mitosis (63)<br />

Thr6 PKCbI (85)<br />

Thr11 (mammals) Dlk/Zip mitosis (69)<br />

Tyr41 JAK2 transcriptional activation (86)<br />

Tyr45 PKCd apoptosis (87)<br />

H4 Ser1 unknown mitosis, chromatin assembly (50)<br />

CK2 DNA repair (72)<br />

Ubiquitination<br />

Histone Site Histone-modifying Enzymes Proposed Function Ref. #<br />

H2A Lys119 (mammals) Ring2 spermatogenesis (73)<br />

H2B Lys120 (mammals) UbcH6 meiosis (74)<br />

Lys123 (S. cerevisiae)<br />

transcriptional activation<br />

Rad6<br />

(75)<br />

euchromatin<br />

Sumoylation<br />

Histone Site Histone-modifying Enzymes Proposed Function Ref. #<br />

H2A Lys126 (S. cerevisiae) Ubc9 transcriptional repression (76)<br />

H2B Lys6 or Lys7 (S. cerevisiae) Ubc9 transcriptional repression (76)<br />

H4 N-terminal tail (S. cerevisiae) Ubc9 transcriptional repression (77)<br />

Biotinylation<br />

Histone Site Histone-modifying Enzymes Proposed Function Ref. #<br />

H2A Lys9 biotinidase unknown (78)<br />

Lys13 biotinidase unknown (78)<br />

H3 Lys4 biotinidase gene expression (79)<br />

Lys9 biotinidase gene expression (79)<br />

Lys18 biotinidase gene expression (79)<br />

H4 Lys12 biotinidase DNA damage response (80,81)<br />

Examples of Crosstalk Between Post-Translational Modifications<br />

Histone H3<br />

Histone H3 and H4<br />

p53<br />

MEF2A<br />

Transcriptional<br />

Repression<br />

Pim-1<br />

S10<br />

H3<br />

14-3-3<br />

HP1<br />

S10<br />

Me<br />

K9<br />

H3<br />

MOF<br />

K14<br />

Resting Neurons<br />

PIAS1<br />

K403 Su S408<br />

MEF2A<br />

K16<br />

HP1<br />

dissociation<br />

P-TEFb<br />

BRD4<br />

HATs<br />

HP1<br />

DNA Damage<br />

Aurora B, Ras<br />

H3<br />

GCN5<br />

Membrane Depolarization<br />

Calcineurin<br />

Transcription<br />

Transcriptional<br />

Activation<br />

Chk2 Set7/9<br />

MDM2<br />

TIP60<br />

Ub K372<br />

K120<br />

K372<br />

dissociation<br />

Me<br />

S20<br />

Me K372<br />

MDM2<br />

Ub K373<br />

S20<br />

CBP/<br />

p53 p53 p53 K381 p300<br />

Ub K381<br />

Ub K382<br />

K382<br />

p53 Ubiquitination/Degradation<br />

p53 Stability/Transactivation of<br />

pro-apoptotic target genes<br />

Dendric Claw<br />

Differentiation<br />

Transcriptional Repression<br />

H4<br />

S10<br />

K9<br />

H3<br />

K9<br />

S10<br />

K403 S408<br />

MEF2A<br />

K14<br />

K14<br />

RNA Pol II<br />

No Differentiation<br />

Transcriptional Activation<br />

Post-translational modifications (PTMs) are emerging as major effectors of protein function, and in turn, cellular processes. The discovery and investigation of post-translational<br />

modifications such as methylation, acetylation, phosphorylation, sumoylation, and many others has established both nuclear and non-nuclear roles for PTMs. With the<br />

awareness of PTMs, there is an ever-growing list of them and more and more research centered on their function. In recent years, there is an overwhelming appreciation for<br />

the diversity of modifications, but most importantly, the interplay between them. This interplay is essential for proper gene expression, genome organization, cell division and<br />

DNA damage response. PTMs can directly impact cell function by modifying histones, modifying enzymes and their associated activity, assembling protein complexes as well<br />

as recognition and targeting in the genome or to other cellular compartments. In the context of single modifications and gene expression, acetylation of certain lysines (i.e.,<br />

Histone 3 lysine [9-H3K9]) correlates with activation, while tri-methylation of this same residue is most often associated with compaction and gene repression. In the case of<br />

lysine methylation, lysine can be mono-, di-, or tri-methylated; while arginine can be mono- or dimethylated in an asymmetric or symmetric fashion. Each degree of methylation<br />

for lysines and arginines serves as its own PTM and impacts biological output. Most PTMs do not exist alone in the chromatin environment and the combination of these<br />

states can reinforce one another. For example, one PTM can serve as a docking site for a binding domain called a “reader” within one protein, while another “reader” within<br />

the same protein can recognize another residue. This is the case for the reader protein BPTF, which binds both H3K4me3 and H4K16 acetylation. Therefore, modulating the<br />

various types and degrees of modifications will impact output. For these reasons, the cell has developed a series of enzymes that are important for establishing and maintaining<br />

these PTMs, which are often referred to as “writers” (e.g., histone methyltransferases, acetyltransferases, etc.) or “erasers” (e.g., histone demethylases, deacetylases,<br />

etc.). Many of these enzymes have emerged as critical therapeutic targets and have been identified as key regulators of diseases such as cancer. These observations have<br />

also made their associated PTMs candidates for biomarkers in cancer and other diseases.<br />

Select Reviews:<br />

Berger, S.L. (2007) Nature 447, 407–412. • Dawson, M.A., and Kouzarides T. (2012) Cell 150, 12–27. • Gardner, K.E., Allis, C.D., and Strahl, B.D. (2011) J. Mol. Biol.<br />

409, 36–46. • Lee, J.S., Smith, E., and Shilatifard, A. (2010) Cell 142, 682–685. • Musselman, C.A. and Kutateladze, T.G. (2011) Acids Res. 39, 9061–9071. • Yang,<br />

X.J. and Seto, E. (2008) Mol. Cell 31, 449–461.<br />

Ca +2<br />

© 2006–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Jonathan Whetstine for reviewing this diagram.<br />

30 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstpathways 31


Section I: Research Areas<br />

eIF4B is expressed in<br />

HeLa cells and human<br />

lung carcinoma.<br />

Sin1, a component of<br />

the mTORC2 complex,<br />

is expressed in many<br />

cell lines.<br />

Translational Control<br />

The synthesis of new proteins is a highly regulated process that allows rapid cellular responses to a<br />

diverse set of stimuli. Two key events in the control of translational initiation are 1) the association<br />

between 5’ capped mRNA and the preinitiation complex, and 2) the binding of initiator tRNA to the start<br />

codon. Both events are mediated by multiple eukaryotic initiator factors (eIFs) that are regulated by<br />

effector kinases and inhibitors.<br />

Cap-dependent Initiation<br />

Translation initiation requires a set of factors to facilitate the association of the 40S ribosomal subunit<br />

with mRNA. The eIF4F complex, consisting of eIF4E, eIF4A, and eIF4G, binds to the 5ʹ cap structure of<br />

mRNA. eIF4A is a helicase, and together with accessory protein eIF4B, serves to unwind the secondary<br />

structure of mRNA at its 5’ untranslated region and promote formation of the preinitiation complex.<br />

A<br />

B<br />

eIF4B (1F5) Mouse mAb #13088: Confocal<br />

IF analysis of HeLa cells (A) using #13088<br />

(green). Actin filaments were labeled with<br />

DyLight 554 Phalloidin #13054 (red). Blue<br />

pseudocolor = DRAQ5 ® #4084 (fluorescent<br />

DNA dye). IHC analysis of paraffin-embedded<br />

human lung carcinoma (B) using #13088.<br />

Assembly of eIF4F is controlled by growth and survival factors that regulate activity of upstream kinase<br />

effectors, including Akt, mTOR, p70 S6 kinase, and p90RSK. mTOR kinase complexes mTORC1 and<br />

mTORC2 promote eIF4F cap-binding complex formation by activating upstream elements that favor<br />

complex assembly and inhibiting proteins that block eIF4F formation. The mTORC1 complex includes<br />

mTOR kinase bound by the adaptor raptor and several regulatory proteins (GβL, PRAS40, and DEPTOR),<br />

while the mTORC2 complex contains mTOR kinase, rictor, GβL, DEPTOR, and Sin1. mTORC1 activates<br />

p70 S6 kinase to relieve PDCD4 inhibition of eIF4A and activate eIF4B. Initiation factor eIF4B interacts<br />

with both the eIF3 scaffold protein complex and eIF4A, stimulating eIF4A RNA helicase activity. Upstream<br />

kinase pathways mediate the phosphorylation of eIF4B by p70 S6 kinase and p90RSK to increase the association<br />

between eIF4B, eIF3, and eIF4A. Inhibition of translation repressor protein 4E-BP1 by mTORC1<br />

phosphorylation causes release of cap-binding protein eIF4E and its incorporation into eIF4F.<br />

Sin1 (D7G1A) Rabbit mAb #12860: WB analysis<br />

of extracts from various cell lines using #12860.<br />

Lanes<br />

1. HeLa<br />

2. MCF7<br />

3. Hep G2<br />

4. INS-1<br />

5. KNRK<br />

6. NBT-11<br />

7. PANC-1<br />

8. Vero<br />

9. COS-7<br />

10. U-87 MG<br />

11. 293<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

1 2 3 4 5 6 7 8 9 10 11<br />

Sin1.1<br />

Sin1.2<br />

chapter 01: GENE EXPRESSION, EPIGENETICS, AND NUCLEAR FUNCTION<br />

In hypoxic environments, mTORC1 activity is inhibited, leading to down regulation of eIF4E capdependent<br />

translation. Instead, protein synthesis is driven during low oxygen conditions by eIF4E2<br />

(also known as 4EHP), which binds the 5ʹ cap and forms a complex with the hypoxia-inducible factor<br />

HIF-2α and the RNA-binding protein RBM4. This complex stimulates translation of select RNAs,<br />

including those implicated in cancer growth.<br />

The 40S ribosomal subunit then binds to the 5ʹ mRNA cap and associated initiation factors and<br />

searches along the mRNA for the initiation codon. eIF3 physically interacts with eIF4G, which may be<br />

responsible for the association of the 40S ribosomal subunit with mRNA.<br />

eIF3H is a core component of the eIF3 complex<br />

that facilitates binding of mRNA to the 40S ribosomal<br />

subunit.<br />

eIF3H (D9C1) XP ® Rabbit mAb #3413: Confocal IF analysis of SK-N-MC cells using<br />

#3413 (green). Actin filaments were labeled with DY-554 phalloidin (red). Blue pseudocolor<br />

= DRAQ5 ® #4084 (fluorescent DNA dye).<br />

Initiator tRNA and the Start Codon<br />

eIF2 mediates binding of initiator tRNA to the ribosome at the start codon to form the 43S preinitiation<br />

complex. Trimeric eIF2 is made up of regulatory (α), tRNA/mRNA interacting (β), and GTP/GDP binding<br />

(γ) proteins. Phosphorylation of eIF2α by multiple upstream kinases (including PKR, PERK, and GCN2)<br />

is stimulated by environmental stress and the presence of dsDNA, and leads to inactive eIF2 and<br />

translation inhibition. Additional control of eIF2 activity occurs through regulation of guanine nucleotide<br />

exchange, which is catalyzed by eIF2B. Exchange of GDP for GTP promotes the essential association<br />

between the eIF2 complex and tRNA. eIF2B activity is inhibited by GSK-3β phosphorylation and through<br />

interaction with eIF5, which also acts as a GDP dissociation inhibitor by stabilizing eIF2 bound by GDP.<br />

Treatment of cells with ER stress-inducing agent<br />

thapsigargin results in phosphorylation of eIF2α at Ser51.<br />

Phospho-eIF2α (Ser51) (D9G8) XP ® Rabbit mAb #3398: WB analysis of extracts from C2C12 cells,<br />

untreated or treated with Thapsigargin #12758, using #3398 (upper) or eIF2α Antibody #9722 (lower).<br />

eIF2, which transfers Met-tRNA to the 40S subunit<br />

to form the 43S preinitiation complex, is expressed<br />

in multiple cell lines.<br />

eIF2α (D7D3) XP ® Rabbit mAb #5324: WB analysis<br />

of extracts from various cell lines using #5324.<br />

Lanes<br />

1. MCF7<br />

2. Hep G2<br />

3. NIH/3T3<br />

4. COS-7<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

1 2 3 4<br />

elF2α<br />

kDa<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

– +<br />

PhosphoeIF2α<br />

(Ser51)<br />

eIF2α<br />

Thapsigargin<br />

S6 ribosomal protein<br />

is phosphorylated<br />

by p70 S6 kinase<br />

at Ser235/236 in<br />

response to growth<br />

factors and mitogens.<br />

Phospho-S6 Ribosomal Protein (Ser235/<br />

Ser236) (D57.2.2E) XP ® Rabbit mAb<br />

#4858: Confocal IF analysis of HeLa cells,<br />

rapamycin-treated (left) or 20% serum-treated<br />

(right), using #4858 (green). Actin filaments<br />

were labeled with Alexa Fluor ® 555 Phalloidin<br />

#8953 (red). Blue pseudocolor = DRAQ5 ®<br />

#4084 (fluorescent DNA dye).<br />

20<br />

32 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttranslational<br />

33


Section I: Research Areas<br />

chapter 01: GENE EXPRESSION, EPIGENETICS, AND NUCLEAR FUNCTION<br />

UV treatment<br />

results in clustering<br />

of cytosolic stress<br />

granules containing<br />

the translation repressor<br />

protein TIAR.<br />

Local Translation<br />

Some mRNAs are transported in messenger ribonucleoprotein (mRNP) granules to their subcellular<br />

locations and translated on-site in response to localized signals, known as local translation. This often<br />

occurs during development, where protein gradients and varying expression patterns are necessary for<br />

cellular differentiation. mRNPs include stress granules that store mRNA bound to stalled preinitiation<br />

complexes and the translational repressors TIA-1 and TIAR until translational initiation can begin again<br />

or the mRNA is degraded. In addition, mRNPs also include cytoplasmic processing bodies (P-bodies)<br />

that function in mRNA turnover. Together, these elements can control translation, mRNA storage, and<br />

stability in localized sites.<br />

EDC4/Ge-1 is an essential component<br />

of cytoplasmic P-bodies responsible<br />

for mRNA decapping and degradation.<br />

EDC4/Ge-1 Antibody #2548: Confocal IF analysis of HeLa cells using<br />

#2548 (green). Actin filaments were labeled with DY-554 phalloidin (red).<br />

Blue pseudocolor= DRAQ5 ® #4084 (fluorescent DNA dye).<br />

Small noncoding RNAs<br />

Small noncoding RNAs are important regulators of gene expression in higher eukaryotes. Several<br />

classes of small RNAs, including short interfering RNAs (siRNAs), microRNAs (miRNAs), and Piwiinteracting<br />

RNAs (piRNAs), have been identified. siRNAs are short segments (20–25 base pairs) of<br />

double stranded RNA that silence expression of a single gene through complementary base pairing<br />

that prevents target translation and/or promotes instability. siRNAs are commonly used in the research<br />

community for antibody validation testing or gene silencing studies.<br />

Similarly, microRNAs are about 21 nucleotides in length and have been implicated in many cellular<br />

processes such as development, differentiation, and stress response. miRNAs function together with the<br />

protein components of complexes called micro-ribonucleoproteins (miRNPs). Among the most important<br />

components in these complexes are argonaute proteins. Argonaute proteins participate in the various<br />

steps of microRNA-mediated gene silencing, such as repression of translation and mRNA turnover.<br />

Silencing of DDX5 expression using DDX5 siRNA.<br />

SignalSilence ® DDX5 siRNA I #8626 and SignalSilence ® DDX5 siRNA II #8627: WB analysis<br />

of extracts from HeLa cells, transfected with 100 nM SignalSilence ® Control siRNA (Unconjugated)<br />

#6568 (-), #8626 (+), or #8627 (+), using DDX5 (D15E10) XP ® Rabbit mAb #9877 (upper) or β-Actin<br />

(13E5) Rabbit mAb #4970 (lower). The DDX5 (D15E10) XP ® Rabbit mAb confirms silencing of DDX5<br />

expression, while the β-Actin (13E5) Rabbit mAb is used as a loading control.<br />

Mili binds to piwi-interacting RNA in male germ<br />

cells and is essential for spermatogenesis in mouse.<br />

Mili (D14F5) XP ® Rabbit mAb #5940: Confocal IF analysis of mouse testis using #5940 (green) and<br />

Pan-Keratin (C11) Mouse mAb #4545 (red). Blue pseudocolor = DRAQ5 ® #4084 (fluorescent DNA dye).<br />

TIAR (D32D3) XP ® Rabbit mAb #8509:<br />

Confocal IF analysis of HeLa cells, untreated<br />

(left) or UV-treated (right), using #8509<br />

(green). Actin filaments were labeled with<br />

DY-554 phalloidin (red). Blue pseudocolor =<br />

DRAQ5 ® #4084 (fluorescent DNA dye).<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

60<br />

50<br />

– +<br />

I<br />

II<br />

+<br />

DDX5<br />

β-actin<br />

DDX5 siRNA<br />

Select Reviews<br />

Adjibade, P. and Mazroui, R. (2014) Semin. Cell Dev. Biol. 34, 15–23. • Bar-Peled, L. and Sabatini, D.M. (2014) Trends Cell<br />

Biol. 24, 400−406. • Donnelly, N., Gorman, A.M., Gupta, S., et al. (2013) Cell Mol. Life Sci. 70, 3493−3511. • Emde, A.<br />

and Hornstein, E. (2014) EMBO J. 33,1428−1437. • Fabian, M.R., Payette, J., Holcik, M., et al. (2012) Nature 486, 126−129.<br />

• Hershey, J.W., Sonenberg, N., and Mathews, M.B. (2012) Cold Spring Harb. Perspect. Biol. 4, a011528. • Hinnebusch, A.G.<br />

and Lorsch, J.R. (2012) Cold Spring Harb. Perspect. Biol. 4, a011544. • Jung, H., Gkogkas, C.G., Sonenberg, N. et al. (2014)<br />

Cell 157, 26−40. • Kong, J. and Lasko, P. (2012) Nat. Rev. Genet. 13, 383−394. • Spilka, R., Ernst, C., Mehta, A.K., et al.<br />

(2013) Cancer Lett. 340, 9−21. • Thoreen, C.C. (2013) Biochem. Soc. Trans. 41, 913−916.<br />

Commonly Studied Translational Control Targets<br />

Target M P Target M P<br />

4E-BP1<br />

• • eIF4A<br />

• •<br />

Phospho-4E-BP1 • • eIF4B<br />

• • PABP1<br />

(Thr37/Thr46)<br />

Phospho-eIF4B (Ser406) • •<br />

Non-phospho-4E-BP1 • Phospho-eIF4B (Ser422) •<br />

(Thr46)<br />

eIF4E<br />

• • PABP2<br />

Phospho-4E-BP1 (Ser65) • •<br />

Phospho-eIF4E (Ser209) • PACT<br />

Phospho-4E-BP1 (Thr70) • •<br />

eIF4G<br />

• • Paip2A<br />

4E-BP2<br />

•<br />

Phospho-eIF4G (Ser1108) • PARN<br />

4EHP<br />

•<br />

eIF4GI<br />

• • PERK<br />

4E-T<br />

•<br />

eIF4G2/p97 • PKR<br />

ADAR1<br />

• •<br />

EIF4H<br />

• • PPIG<br />

Argonaute 1 • •<br />

eIF5<br />

•<br />

Argonaute 2 •<br />

eIF6<br />

• • PRP4K<br />

Argonaute 3 •<br />

ELAVL1/HuR • PTBP1<br />

Argonaute 4 •<br />

Exportin 5 • • Pumilio 1<br />

BRF1/2<br />

•<br />

FMRP<br />

• • Pumilio 2<br />

CLK3<br />

•<br />

FUS/TLS<br />

• RMP<br />

CNOT2<br />

•<br />

FXR1<br />

• • RPL11<br />

CNOT3<br />

•<br />

FXR2<br />

• •<br />

CNOT6<br />

•<br />

GCN2<br />

•<br />

Coilin<br />

•<br />

hnRNP A0 • •<br />

CPEB1<br />

•<br />

hnRNP A1 • •<br />

DCP1B<br />

•<br />

hnRNP C1/C2 •<br />

DDX3<br />

• •<br />

AUF1/hnRNP D •<br />

DDX4<br />

• •<br />

hnRNP E1<br />

•<br />

DDX5<br />

• •<br />

hnRNP LL<br />

•<br />

DDX6/RCK • •<br />

hnRNP K • •<br />

DGCR8<br />

•<br />

SAM68<br />

hnRNP Q/R • •<br />

DHX29<br />

• •<br />

SF2/ASF<br />

IMP1<br />

• •<br />

Dicer1<br />

• •<br />

SF3B1<br />

IWS1<br />

•<br />

Drosha<br />

•<br />

SKAR<br />

KHSRP<br />

• •<br />

EDC4/Ge-1<br />

•<br />

SKAR α/β<br />

La Antigen • •<br />

eEF1A<br />

• •<br />

SMN1<br />

LSm2<br />

•<br />

eEF2<br />

•<br />

Symplekin<br />

LysRS<br />

• •<br />

Phospho-eEF2 (Thr56) •<br />

TFEB<br />

MAPBPIP/ROBLD3/p14 •<br />

eEF2k<br />

•<br />

THEX1<br />

MAPKSP1/MP1 •<br />

Phospho-eEF2k (Ser366) •<br />

MetAP2 •<br />

eIF1<br />

•<br />

TIAR<br />

Mili<br />

• •<br />

eIF2α<br />

• •<br />

U2AF1<br />

Miwi<br />

• •<br />

Phospho-eIF2α (Ser51) • •<br />

Upf1<br />

Mnk1<br />

•<br />

eIF2B-ε<br />

•<br />

Upf2<br />

Phospho-Mnk1 •<br />

eIF3A<br />

• •<br />

XBP-1s<br />

(Thr197/202)<br />

eIF3C<br />

•<br />

XRN2<br />

MRPL11 • •<br />

eIF3H<br />

•<br />

ZPR1<br />

NCBP1/CBP80 •<br />

eIF3J<br />

• • NRF1/TCF11 •<br />

eIF4A1<br />

• NRF2<br />

•<br />

Target M P<br />

NXF1<br />

Asymmetric-Methyl-PABP1<br />

(Arg455/Arg460)<br />

Phospho-PPIG (Ser376)<br />

S6 Ribosomal Protein<br />

Phospho-S6 Ribosomal<br />

Protein (Ser235/Ser236)<br />

Phospho-S6 Ribosomal<br />

Protein (Ser240/Ser244)<br />

Ribosomal Protein L7a<br />

Ribosomal Protein L13a<br />

Ribosomal Protein L26<br />

Ribosomal Protein S3<br />

THOC4/ALY<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

• •<br />

•<br />

•<br />

• •<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

• •<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

These protein targets represent key<br />

nodes within translational control<br />

signaling pathways and are commonly<br />

studied in translational control research.<br />

Primary antibodies, antibody conjugates,<br />

and antibody sampler kits containing<br />

these targets are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

207<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for Phospho-S6<br />

Ribosomal Protein (Ser235/236)<br />

have been cited over 207 times in<br />

high-impact, peer-reviewed publications<br />

from the global research community.<br />

Select Citations:<br />

Wong, C.C. et al. (2014) Inactivating<br />

CUX1 mutations promote tumorigenesis.<br />

Nat. Genet. 46, 33−38.<br />

Kurachi, M. et al. (2014) The<br />

transcription factor BATF operates as<br />

an essential differentiation checkpoint<br />

in early effector CD8+ T cells. Nat.<br />

Immunol. 15, 373−383.<br />

Mouw, J.K. et al. (2014) Tissue<br />

mechanics modulate microRNAdependent<br />

PTEN expression to<br />

regulate malignant progression.<br />

Nat. Med. 20, 360−367.<br />

Agarwal, A. et al. (2014) Antagonism<br />

of SET using OP449 enhances the<br />

efficacy of tyrosine kinase inhibitors<br />

and overcomes drug resistance in<br />

myeloid leukemia. Clin. Cancer Res.<br />

20, 2092−2103.<br />

Koo, J. et al. (2014) Maintaining<br />

glycogen synthase kinase-3 activity<br />

is critical for mTOR kinase inhibitors<br />

to inhibit cancer cell growth. Cancer<br />

Res. 7, 2555−2568.<br />

Fay, M.M. et al. (2014) Enhanced<br />

Arginine Methylation of Programmed<br />

Cell Death 4 Protein during Nutrient<br />

Deprivation Promotes Tumor<br />

Cell Viability. J. Biol. Chem. 289,<br />

17541−17552.<br />

34 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttranslational<br />

35


Section I: Research Areas<br />

Translational Control: Overview<br />

eEF2K<br />

GTP<br />

eEF1<br />

Cap<br />

Cap<br />

eEF2<br />

GTP<br />

AUG<br />

eEF2 Control:<br />

Ca 2+<br />

cAMP<br />

mTORC1<br />

40S<br />

AUG<br />

60S<br />

eIF5B<br />

GDP<br />

Elongation<br />

40S<br />

60S<br />

Factor<br />

Release<br />

eIF3<br />

A (n)<br />

eIF1A<br />

Nascent<br />

Polypeptide<br />

ER Lumen<br />

Initiation Codon<br />

Recognition<br />

eIF2<br />

GDP<br />

A(n)<br />

eIF5<br />

Cap<br />

eIF1<br />

Polypeptide<br />

Chain<br />

eIF4 Control:<br />

Growth Factors (mTORC1),<br />

Hormones, Cytokines, Mitogens,<br />

Neuropeptides, Oxidative Stress<br />

48S<br />

AUG<br />

Cap<br />

eIF6<br />

eIF5B<br />

GTP<br />

60S<br />

A (n)<br />

Initiation<br />

Stress Granules<br />

P-bodies<br />

40S<br />

Scanning<br />

AAAAA<br />

DHX29<br />

miRNA<br />

60S<br />

48S<br />

Cap<br />

AUG<br />

eIF4B<br />

eRF1<br />

eRF3<br />

GTP<br />

A (n)<br />

ON<br />

eIF4F<br />

Cap<br />

PABP AUG<br />

A (n)<br />

Termination<br />

eIF2 Control:<br />

Viral Infection (dsRNA), (PKR)<br />

Amino Acid Starvation, UV Light (GCN2)<br />

Heme Deficiency (HRI)<br />

Heat Shock, ER Stress, Hypoxia (PERK)<br />

miRNA<br />

43S<br />

eIF2<br />

Met-tRNAi<br />

GTP<br />

eIF3<br />

40S<br />

Initiation<br />

Complex<br />

The synthesis of new proteins is a highly regulated process that allows rapid cellular responses to diverse stimuli at the post-transcriptional level. Nine key eukaryotic translation<br />

initiation factors (eIFs) catalyze the assembly of a functional ribosomal complex in two steps - first, the formation of the 48S complex from the 43S initiation complex<br />

and mRNA followed by its subsequent joining with the 60S subunit, enabling polypeptide chain formation. Of the many steps in translation, the rate-limiting step, initiation, is<br />

subjected to the most regulatory control. Many stimuli, such as growth factors and stress, either stimulate or inhibit specific eIFs. Aside from initiation, translation can also be<br />

attenuated during elongation. For instance, elevated levels of Ca 2+ or cAMP can block the action of eukaryotic elongation factor 2 (eEF2) via AMPK. Finally, upon recognition of<br />

a stop codon, eRF1 and eRF3 mediate termination of translation and ribosome disassembly and recycling.<br />

Select Reviews:<br />

Dever, T.E. and Green, R. (2012) Cold Spring Harb. Perspect. Biol. 4, a013706. • Gebauer, F., and Hentze, M.W. (2004) Nat. Rev. Mol. Cell Biol. 5, 827–835. • Hinnebusch,<br />

A.G. (2011) Microbiol. Mol. Biol. Rev. 75, 434–467. • Sonenberg, N. and Hinnebusch, A.G. (2009) Cell 136, 731–745. • Spirin, A.S. (2009) Biochemistry 48, 10688–<br />

10692. • Steitz, T.A. (2008) Nat. Rev. Mol. Cell Biol. 9, 242–253.<br />

eIF1A<br />

eIF1<br />

eIF5<br />

OFF<br />

Translational Control: Regulation of elF4E and p70 S6K<br />

Amino<br />

Acids<br />

GRB10<br />

Hormones, Growth Factors,<br />

Cytokines, Neuropeptides Mitogens Stress<br />

IRS-1<br />

PI3K<br />

RagA/B<br />

RagC/D<br />

4E-<br />

BP1 eIF4E<br />

Translation Off<br />

mTORC1<br />

GβL Raptor<br />

mTOR<br />

DEPTOR<br />

AMP:<br />

ATP<br />

LKB1<br />

AMPK<br />

PIP 3<br />

PDK1<br />

Akt<br />

TSC2<br />

TSC1 TBC1D7<br />

PRAS40 rapamycin<br />

FKBP12<br />

mTORC1<br />

mTORC2<br />

Sin1 PRR5<br />

Rictor GβL<br />

mTOR<br />

DEPTOR<br />

S6<br />

4E-<br />

BP1<br />

chapter 01: GENE EXPRESSION, EPIGENETICS, AND NUCLEAR FUNCTION<br />

PTEN<br />

mTORC2<br />

GSK-3<br />

Torin1<br />

PP242<br />

KU63794<br />

WYE354<br />

p70 S6K<br />

PDCD4<br />

eIF4B<br />

elF4A eIF4H<br />

MNK<br />

eIF4E eIF4G<br />

eIF4F<br />

Cap<br />

PABP<br />

AAAAA<br />

eIF4F<br />

LRP<br />

Wnt<br />

Frizzled<br />

Gα q/o<br />

Dvl<br />

Erk<br />

p90RSK<br />

PABP<br />

PAIP1<br />

PAIP2<br />

43S<br />

48S<br />

p38 MAPK<br />

eEF2K eEF2<br />

Translation Elongation On<br />

Translation On<br />

Translation is a tightly regulated process, and the mTORC1-S6K signaling axis plays a critical role in this control. The rate of translation initiation is predominantly determined<br />

by 5’ cap recognition by eIF4F, a trimeric protein complex composed of eIF4E, which binds the 5ʹ cap; eIF4A, a helicase necessary for unwinding complex secondary structure<br />

in the leader sequence; and eIF4G, a large scaffolding protein that delivers the mRNA to eIF3 and mediates mRNA circularization through association with polyA binding<br />

protein (PABP). Binding of eIF4F to the cap is hindered by eIF4E binding proteins (4EBPs), which, when hypophosphorylated, sequester eIF4E and prevent its association<br />

with eIF4G. However, in response to positive stimuli such as growth factors, mitogens, and amino acids, mTORC1 phosphorylates 4EBPs and relieves this inhibition, allowing<br />

the formation of eIF4F and subsequent initiation of translation. In addition, mTORC1 - alongside PDK1 - phosphorylates S6 kinase, which in turn phosphorylates numerous<br />

substrates involved in translation. These include S6 small ribosomal subunit; eIF4B, an activator of the eIF4A helicase; PDCD4, an eIF4A inhibitor that is inhibited by phosphorylation;<br />

and SKAR, an mRNA splicing factor. Aside from the mTORC1 pathway, the Ras-MAPK pathway is another major regulator of translation and is responsible for the<br />

phosphorylation of eIF4B as well as eIF4E, via MNK kinases.<br />

Select Reviews:<br />

Dowling, R.J., Topisirovic, I., Fonseca, B.D., and Sonenberg, N. (2010) Biophys. Acta. 1804, 433–439. • Fenton T.R. and Gout, I.T. (2011) Int. J. Biochem. Cell Biol. 43,<br />

47–59. • Graff, J.R., Konicek, B.W., Carter, J.H., and Marcusson, E.G. (2008) Cancer Res. 68, 631–634. • Holcik, M. and Sonenberg, N. (2005) Nat. Rev. Mol. Cell Biol. 6,<br />

318–327. • Huang, J. and Manning, B.D. (2008) Biochem. J. 412, 179–190. • Magnuson, B., Ekim, B., and Fingar, D.C. (2012) Biochem. J. 441, 1–21. • Ruvinsky, I.<br />

and Meyuhas, O. (2006) Trends Biochem. Sci. 31, 342–348. • Sonenberg, N. and Hinnebusch, A.G. (2009) Cell 136, 731–745.<br />

© 2002–2015 Cell Signaling Technology, Inc. • We would like to thank Rachel Wolfson and Prof. David Sabatini, Whitehead Institute for Biomedical Research, MIT, Cambridge, MA, for reviewing this diagram.<br />

© 2002–2015 Cell Signaling Technology, Inc. • We would like to thank Rachel Wolfson and Prof. David Sabatini, Whitehead Institute for Biomedical Research, MIT, Cambridge, MA, for reviewing this diagram.<br />

36 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstpathways<br />

37


Section I: Research Areas<br />

chapter 01: GENE EXPRESSION, EPIGENETICS, AND NUCLEAR FUNCTION<br />

Translational Control: Regulation of elF2<br />

PTEN<br />

Salubrinal<br />

ATF-4, CHOP, mRNA<br />

Translation<br />

α<br />

GDP<br />

Apoptosis, Changes in<br />

Metabolism & Redox Status<br />

SirT1<br />

eIF2B<br />

eIF2 γ<br />

Akt<br />

Growth Factors,<br />

ER Stress,<br />

Viral Infection<br />

dsRNA<br />

PKR<br />

GADD34<br />

eIF2B<br />

β<br />

Unfolded Protein<br />

Response, Hypoxia<br />

PKR<br />

PP1<br />

CreP<br />

eIF5<br />

PERK<br />

PERK<br />

PP1<br />

SirT1<br />

Global Translation Off<br />

40S<br />

eIF3<br />

eIF5<br />

BiP<br />

α<br />

GDP<br />

Glucose Deprivation,<br />

UV Light, Amino Acid<br />

Starvation<br />

α<br />

GDP<br />

GDP<br />

α<br />

GTP<br />

GCN2<br />

GCN2<br />

eIF2 γ<br />

eIF2 γ<br />

eIF2 γ<br />

β<br />

β<br />

β<br />

GTP<br />

eIF2<br />

Met-tRNAi<br />

Ternary<br />

Complex<br />

43S<br />

GTP<br />

Global Translation On<br />

eIF5<br />

HRI<br />

eIF5<br />

Met-tRNAi<br />

eIF1<br />

eIF1A<br />

Heme Deficiency,<br />

Oxidative Stress<br />

HRI<br />

NCK<br />

eIF2B<br />

GSK-3β<br />

Growth Factors,<br />

Hormones, etc.<br />

The eIF2 initiation complex integrates a diverse array of stress-related signals to regulate both global and specific mRNA translation. Under permissive conditions, eIF2 binds<br />

GTP and Met-tRNAi to form the ternary complex (TC), which then associates with the 40S ribosomal subunit, eIF1, eIF1A, eIF5, and eIF3 to form the 43S pre-initiation complex<br />

(PIC). The 43S PIC scans the mRNA UTR for an AUG start codon. Upon AUG recognition, eIF2 hydrolyzes GTP to GDP with the help of the GTPase activating protein eIF5<br />

and dissociates from the mRNA, permitting the binding of the 60S ribosomal subunit and elongation of the polypeptide chain. eIF2 remains bound to GDP in the presence of<br />

eIF5 acting as a GDI. To permit another round of initiation, eIF2B must act as both a GDI displacement factor (GDF) and a guanine exchange factor (GEF) to allow exchange of<br />

GDP for GTP on eIF2. This step is tightly regulated, and phosphorylation of eIF2α by a diverse family of four stress activated kinases—PKR (dsRNA), PERK (ER stress), GCN2<br />

(amino acid starvation), and HRI (heme deficiency)—prevents nucleotide exchange by causing eIF2 to act as a dominant negative complex to sequester eIF2B. The resulting<br />

increase in eIF2α-GDP limits the availability of the ternary complex and causes a decrease in global protein synthesis and an enhancement of the translation of specific stressrelated<br />

mRNA transcripts, such as the transcription factors ATF-4 and CHOP.<br />

Select Reviews:<br />

Hinnebusch, A.G. (2011) Microbiol. Mol. Biol. Rev. 75, 434–467. • Raven, J.F. and Koromilas, A.E. (2008) Cell Cycle 7, 1146–1150. • Schmitt, E., Naveau, M., and<br />

Mechulam, Y. (2010) FEBS Lett. 584, 405–412. • Stolboushkina, E.A. and Garber, M.B. (2011) Biochemistry 76, 283–294. • Wek, R.C., Jiang, H.Y., and Anthony, T.G.<br />

(2006) Biochem. Soc. Trans. 34, 7–11.<br />

Nuclear Receptors<br />

The nuclear receptor superfamily are ligand-activated transcription factors that play diverse roles in cell<br />

differentiation/development, proliferation, and metabolism and are associated with numerous pathologies<br />

such as cancer, cardiovascular disease, inflammation, and reproductive abnormalities. Members<br />

of this family contain an N-terminal transactivation domain, a highly conserved central region zinc-finger<br />

DNA binding domain, and a C-terminal ligand-binding domain. Ligand binding to its correlate nuclear<br />

receptor results in transactivation of specific genes within a target tissue.<br />

In addition to ligand binding, nuclear receptor activity can be modulated through the action of numerous<br />

growth factor and cytokine signaling cascades that result in receptor phosphorylation or other<br />

post-translational modifications, typically within the N-terminal transactivation domain. For example,<br />

the estrogen receptor is phosphorylated on multiple serine residues that affect receptor activity. Ser118<br />

may be the substrate of the transcription regulatory kinase CDK7, whereas Ser167 may be phosphorylated<br />

by p90RSK and Akt. Phosphorylation of Ser167 may confer resistance to tamoxifen in breast<br />

cancer patients.<br />

Type I Nuclear Receptors<br />

Type I nuclear receptors, also called steroid receptors, include the estrogen receptor, androgen<br />

receptor, progesterone receptor, mineralocorticoid receptor, and glucocorticoid receptor. Steroid<br />

hormone ligands for this subgroup of receptors travel from their respective endocrine gland through<br />

the bloodstream bound to steroid binding globulin. Some type I nuclear receptors are activated, in part,<br />

upon binding their respective ligand in the cytoplasmic compartment. The ligand-receptor complex dissociates<br />

from HSP90 and enters the nucleus where it homodimerizes and binds to hormone response<br />

elements within the promoter of a target gene. The receptor transactivation domain is responsible for<br />

interaction at the promoter with co-activators such as acetyltransferases and the general transcription<br />

machinery, resulting in transcriptional activation.<br />

Androgen receptor, a type I nuclear receptor, plays a crucial role in several<br />

stages of male development and the progression of prostate cancer.<br />

A<br />

Androgen Receptor (D6F11) XP ® Rabbit mAb #5153: IHC analysis of paraffin-embedded human prostate carcinoma (A) using #5153.<br />

Confocal IF analysis of LNCaP (positive) (B) and DU 145 (negative) (C) cells using #5153 (green). Actin filaments have been labeled with<br />

DY-554 phalloidin (red).<br />

Dexamethasone treatment results in translocation of the glucocorticoid<br />

receptor to the nucleus, where it associates with response elements within<br />

glucocorticoid-responsive genes.<br />

A<br />

B<br />

B<br />

C<br />

C<br />

Commonly Studied<br />

Nuclear Receptor<br />

Targets<br />

These protein targets represent key<br />

nodes within nuclear receptor signaling<br />

pathways and are commonly studied<br />

in nuclear receptor research. Primary<br />

antibodies, antibody conjugates, and<br />

antibody sampler kits containing these<br />

targets are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

Target M P<br />

AhR<br />

•<br />

Androgen Receptor • •<br />

Aromatase<br />

•<br />

COUP-TF1 •<br />

COUP-TF2 •<br />

Estrogen Receptor-α •<br />

Phospho-Estrogen •<br />

Receptor-α (Ser104/Ser106)<br />

Phospho-Estrogen •<br />

Receptor-α (Ser118)<br />

Phospho-Estrogen •<br />

Receptor-α (Ser167)<br />

ERRα<br />

•<br />

Glucocorticoid Receptor •<br />

Phospho-Glucocorticoid •<br />

Receptor (Ser211)<br />

NRBF-2<br />

• •<br />

Nur77<br />

•<br />

Phospho-Nur77 (Ser351) •<br />

PHB2<br />

• •<br />

PPARγ<br />

• •<br />

Progesterone Receptor •<br />

Phospho-Progesterone •<br />

Receptor (Ser190)<br />

Phospho-Progesterone •<br />

Receptor (Ser294)<br />

Phospho-Progesterone •<br />

Receptor (Ser345)<br />

Progesterone Receptor A/B • •<br />

Progesterone Receptor B • •<br />

RARα<br />

•<br />

RARγ<br />

•<br />

Rev-Erba •<br />

Phospho-Rev-erba •<br />

(Ser55/59)<br />

RXR-α<br />

• •<br />

RXRβ<br />

•<br />

RXRγ<br />

•<br />

STF-1<br />

•<br />

Vitamin D3 Receptor •<br />

© 2002–2015 Cell Signaling Technology, Inc. • We would like to thank Rachel Wolfson and Prof. David Sabatini, Whitehead Institute for Biomedical Research, MIT, Cambridge, MA, for reviewing this diagram.<br />

38 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

Glucocorticoid Receptor (D6H2L) XP ® Rabbit mAb #12041: IHC analysis of paraffin-embedded human prostate carcinoma (A) using<br />

#12041. Confocal IF analysis of HeLa cells, grown in phenol red-free media containing 5% charcoal-stripped FBS for 2 d and either untreated<br />

(B) or treated with dexamethasone (100 nM, 2 hr) (C), using #12041 (green). Actin filaments were labeled with DY-554 phalloidin (red).<br />

www.cellsignal.com/cstnuclear 39


Section I: Research Areas<br />

8<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for Androgen Receptor<br />

have been cited over 8 times in<br />

high-impact, peer-reviewed publications<br />

from the global research community.<br />

Select Citations:<br />

Cuenca-Lopez, M.D. et al. (2014)<br />

Phospho-kinase profile of triple<br />

negative breast cancer and androgen<br />

receptor signaling. BMC Cancer<br />

14, 302.<br />

Boll, K. et al. (2013) MiR-130a,<br />

miR-203 and miR-205 jointly repress<br />

key oncogenic pathways and are<br />

downregulated in prostate carcinoma.<br />

Oncogene 32, 277−285.<br />

Krycer, J.R. et al. (2013) Does changing<br />

androgen receptor status during<br />

prostate cancer development impact<br />

upon cholesterol homeostasis PLoS<br />

One 8, e54007.<br />

Nie, H. et al. (2013) Acetylcholine<br />

acts on androgen receptor to promote<br />

the migration and invasion but inhibit<br />

the apoptosis of human hepatocarcinoma.<br />

PLoS One 8, e61678.<br />

Furu, K. et al. (2013) Tzfp represses<br />

the androgen receptor in mouse<br />

testis. PLoS One 8, e62314.<br />

Li, Y. et al. (2013) Functional domains<br />

of androgen receptor coactivator p44/<br />

Mep50/WDR77and its interaction<br />

with Smad1. PLoS One 8, e64663.<br />

Ota, H. et al. (2012) Testosterone<br />

deficiency accelerates neuronal<br />

and vascular aging of SAMP8 mice:<br />

protective role of eNOS and SIRT1.<br />

PLoS One 7, e29598.<br />

Krycer, J.R. et al. (2011) Cross-talk<br />

between the androgen receptor and<br />

the liver X receptor: implications for<br />

cholesterol homeostasis. J. Biol.<br />

Chem. 286, 20637−20647.<br />

Type II Nuclear Receptors<br />

Type II nonsteroid nuclear receptors include the thyroid hormone receptors (TRα and β), retinoic acid<br />

receptors (RARα, β, and γ), vitamin D receptor (VDR), and peroxisome proliferator-activated receptors<br />

(PPARα, β, and γ). Members of this family heterodimerize with the retinoid X receptor (RXR). Prior to<br />

ligand binding, receptor heterodimers are located in the nucleus as part of complexes with histone<br />

deacetylases (HDACs) and other co-repressors that keep target DNA in a tightly wound conformation,<br />

preventing exposure to transacting factors. Ligand binding results in co-repressor dissociation,<br />

chromatin derepression, and transcriptional activation.<br />

RARγ1, a type II nuclear receptor that regulates expression<br />

of genes involved in cellular differentiation, proliferation, and<br />

apoptosis, is expressed in cancer cells and epidermal cells.<br />

RARγ1 (D3A4) XP ® Rabbit mAb #8965:<br />

WB analysis of extracts from various cell lines<br />

(A) using #8965. IHC analysis of paraffinembedded<br />

human skin (B) using #8965.<br />

Lanes<br />

1. HaCaT<br />

2. A-375<br />

3. BxPC-3<br />

4. T-47D<br />

5. SK-BR-3<br />

A<br />

kDa<br />

100<br />

80<br />

60<br />

50<br />

40<br />

1 2 3 4 5<br />

Orphan Nuclear Receptors<br />

Orphan nuclear receptors are nuclear receptors where the endogenous ligands have not been identified.<br />

Structural studies suggest that some of the orphan receptors may not bind ligands. This class of nuclear<br />

receptors includes small heterodimer partner (SHP), reverse orientation c-ErbA (Rev-Erbα and β),<br />

testicular receptor 2 and 4 (TR2 and 4), tailless homolog orphan receptor (TLX), photoreceptor-specific<br />

NR (PNR), chicken ovalbumin upstream promoter transcription factor 1 and 2 (COUP-TF1 and 2), Nur77,<br />

Nur-related protein 1 (NURR1), neuron derived orphan receptor 1 (NOR1), estrogen-related receptor<br />

(ERR α, β, and γ), and germ cell nuclear factor (GCNF). Most of these receptors regulate transcription<br />

by binding to their target DNA elements either as monomers or homodimers and recruiting chromatin<br />

modifying coactivators and the transcription machinery. Nur77 and NURR1 can also heterodimerize<br />

with RXRs and these heterodimers are able to respond to RXR ligands to regulate transcription.<br />

Rev-Erbα (E1Y6D) Rabbit mAb #13418: Chromatin IPs were performed<br />

with cross-linked chromatin from 4 x 10 6 Hep G2 cells and either 10 μl of<br />

#13418 or 2 μl of Normal Rabbit IgG #2729 using SimpleChIP ® Enzymatic<br />

Chromatin IP Kit (Magnetic Beads) #9003. The enriched DNA was quantified<br />

by real-time PCR using human BMAL1 promoter primers, SimpleChIP ® Human<br />

NR1D1 Promoter Primers #13413, and SimpleChIP ® Human α Satellite<br />

Repeat Primers #4486. The amount of immunoprecipitated DNA in each<br />

sample is represented as a percent of the total input chromatin.<br />

Rev-Erbα (E1Y6D) Rabbit<br />

mAb #13418<br />

Normal Rabbit<br />

IgG #2729<br />

% of total input chromatin<br />

RARγ1<br />

Rev-Erbα, an orphan nuclear receptor involved in<br />

cell proliferation, differentiation, and circadian rhythms.<br />

0.7<br />

0.6<br />

0.5<br />

0.4<br />

0.3<br />

0.2<br />

0.1<br />

0<br />

BMAL1<br />

NR1D1<br />

B<br />

α Satellite<br />

chapter 01: GENE EXPRESSION, EPIGENETICS, AND NUCLEAR FUNCTION<br />

Select Reviews<br />

Ahmadian, M., Suh, J.M., and Hah, N. et al. (2013) Nat. Med. 19, 557−566. • Bondesson, M., Hao, R., Lin, C.Y., Williams, C., and Gustafsson, J.A. (2014) Biochim.<br />

Biophys. Acta. Jun 17 [Epub ahead of print]. • Evans, R.M. and Mangelsdorf, D.J. (2014) Cell 157, 255−266. • Kojetin, D.J. and Burris, T.P. (2014) Nat. Rev. Drug Discov.<br />

13, 197−216. • Kurakula, K., Koenis, D.S., van Tiel, C.M., and de Vries, C.J. (2014) Biochim. Biophys. Acta. 1843, 2543–2555. • Manolagas, S.C., O’Brien, C.A., and<br />

Almeida, M. (2013) Nat. Rev. Endocrinol. 9, 699−712. • Zhou, W. and Slingerland, J.M. (2014) Nat. Rev. Cancer 14, 26−38.<br />

Nuclear Receptors Signaling<br />

HSP90<br />

Type I Steroid Receptors (Homodimers)<br />

NR<br />

NR<br />

NR<br />

NR<br />

HRE<br />

SBP<br />

HSP90<br />

Homodimerization<br />

SRC1/2 PRMT/<br />

CARM<br />

CBP/<br />

p300 PCAF<br />

RNA<br />

TBP TFIIB POLII<br />

TATA<br />

Plasma Membrane<br />

Cytoplasm<br />

Nucleus<br />

Transcriptional<br />

Activation<br />

Type IIa Non-steroid Receptors (RXR Heterodimers)<br />

Heterodimerization<br />

NcoR1<br />

SMRT<br />

RXR<br />

NR<br />

Ligand<br />

Plasma Membrane<br />

Cytoplasm<br />

Nucleus<br />

Type IIb Orphan Receptors (Monomers and Homodimers)<br />

NcoR1<br />

SMRT<br />

NR<br />

NR<br />

RXR<br />

NR<br />

HRE<br />

HDAC<br />

Complex<br />

No Ligand<br />

Transcriptional Repression<br />

NR<br />

NR<br />

HRE<br />

HDAC<br />

Complex<br />

SRC1/2<br />

PRMT/<br />

CARM<br />

CBP/<br />

p300 PCAF<br />

RNA<br />

TBP TFIIB POLII<br />

SRC1/2<br />

PRMT/<br />

CARM<br />

CBP/<br />

p300 PCAF<br />

RNA<br />

TBP TFIIB POLII<br />

Constitutive<br />

Transcriptional<br />

Repression<br />

Transcriptional<br />

Activation<br />

Nucleus<br />

Constitutive<br />

Transcriptional<br />

Activation<br />

Nur77, an orphan<br />

nuclear receptor<br />

involved in cell<br />

proliferation,<br />

differentiation,<br />

and apoptosis<br />

Nur77 (D63C5) XP ® Rabbit mAb #3960:<br />

Confocal IF analysis of Jurkat cells, untreated<br />

(left) or treated with TPA #4174 and A23187<br />

(right), using #3960 (green). Actin filaments<br />

were labeled with DY-554 phalloidin (red).<br />

© 2012–2015 Cell Signaling Technology, Inc.<br />

40 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstpathways 41


02<br />

Section I: Research Areas<br />

Activation of MAPK<br />

signaling with TPA<br />

results in phosphorylation<br />

of Erk1/2 at<br />

Thr202/Tyr204.<br />

Signaling<br />

MAP Kinase Signaling<br />

Mitogen-activated protein kinases (MAPKs) are a highly conserved family of serine/threonine protein<br />

kinases involved in a variety of fundamental cellular processes such as proliferation, differentiation,<br />

motility, stress response, apoptosis, and survival. Conventional MAPKs include the extracellular<br />

signal-regulated kinase 1 and 2 (Erk1/2 or p44/42), the c-Jun N-terminal kinases 1-3 (JNK1-3)/<br />

stress activated protein kinases (SAPK1A, 1B, 1C), the p38 isoforms (p38α, β, γ, and δ), and Erk5. The<br />

lesser-studied, atypical MAPKs include Nemo-like kinase (NLK), Erk3/4, and Erk7/8.<br />

A<br />

B<br />

C<br />

kDa<br />

80<br />

60<br />

50<br />

40<br />

30<br />

80<br />

60<br />

50<br />

40<br />

30<br />

293<br />

NIH/3T3<br />

C6<br />

Phosphop44/42<br />

MAPK<br />

(Thr202/Tyr204)<br />

+ – + – + –<br />

p44/42<br />

MAPK<br />

λ phosphatase<br />

– + – + – + TPA<br />

Phospho-p44/42 MAPK (Erk1/2) (Thr202/Tyr204) (D13.14.4E) XP ® Rabbit mAb #4370: Confocal IF analysis of C2C12 cells, treated<br />

with U0126 #9903 (10 μM for 1 hr) (A) or TPA #9905 (200 nM for 15 min) (B), using #4370 (green). Actin filaments were labeled with<br />

Alexa Fluor ® 555 Phalloidin #8953 (red). Blue pseudocolor = DRAQ5 ® #4084 (fluorescent DNA dye). WB analysis of extracts from 293,<br />

NIH/3T3, and C6 cells, treated with λ phosphatase or TPA #4174 as indicated (C), using #4370 (upper), or p44/42 MAPK (Erk1/2) (137F5)<br />

Rabbit mAb #4695 (lower).<br />

Signaling via the conventional MAPKs follows a classical<br />

three-tiered kinase cascade: MAPKKK MAPKK MAPK.<br />

CONVENTIONAL MAPKS<br />

ATYPICAL MAPKS<br />

A broad range of extracellular stimuli including mitogens, cytokines, growth factors, and environmental<br />

stressors stimulate the activation of one or more MAPKK kinases (MAPKKKs) via receptor-dependent<br />

and -independent mechanisms. MAPKKKs then phosphorylate and activate a downstream MAPK<br />

kinase (MAPKK), which in turn phosphorylates and activates MAPKs. Activation of MAPKs leads to the<br />

phosphorylation and activation of specific MAPK-activated protein kinases (MAPKAPKs), such as members<br />

of the RSK, MSK, or MNK family, and MK2/3/5. These MAPKAPKs function to amplify the signal<br />

and mediate the broad range of biological processes regulated by the different MAPKs. While most<br />

MAPKKK, MAPKK, and MAPKs display a strong preference for one set of substrates, there is significant<br />

cross-talk in a stimulus- and cell-type–dependent manner.<br />

Activation of MAPK signaling by TPA results in<br />

phosphorylation of c-Fos, a nuclear oncogene<br />

that dimerizes with c-Jun to form the AP-1<br />

transcription factor.<br />

Phospho-c-Fos (Ser32) (D82C12) XP ® Rabbit mAb (PE Conjugate) #11919:<br />

Flow cytometric analysis of HeLa cells, untreated (blue) or treated with TPA #4174<br />

(green), using #11919.<br />

Events<br />

Phospho-c-Fos (Ser32) (PE Conjugate)<br />

UV treatment activates the p38 MAPK signaling pathway, resulting<br />

in phosphorylation of MKK3 at Ser189 and MKK6 at Ser207.<br />

Phospho-MKK3 (Ser189)/MKK6<br />

(Ser207) (D8E9) Rabbit mAb #12280:<br />

Confocal IF analysis of HeLa cells,<br />

untreated (left) or UV-treated (40 mJ/<br />

cm 2 with 30 min recovery; right), using<br />

#12280 (green). Actin filaments were<br />

labeled with DY-554 phalloidin (red).<br />

Blue pseudocolor = DRAQ5 ® #4084<br />

(fluorescent DNA dye).<br />

chapter 02: Signaling<br />

Activation of MAPK<br />

signaling with TPA<br />

results in phosphorylation<br />

of MEK1/2 at<br />

Ser217/221.<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

100<br />

80<br />

60<br />

50<br />

40<br />

HeLa<br />

NIH/3T3<br />

Phospho-<br />

MEK1/2<br />

(Ser217/221)<br />

MEK1/2<br />

+ – + – TPA<br />

Phospho-MEK1/2 (Ser217/221)<br />

(41G9) Rabbit mAb #9154: WB<br />

analysis of extracts from untreated or<br />

TPA-treated HeLa and NIH/3T3 cells<br />

using #9154 (upper) or MEK1/2 Antibody<br />

#9122 (lower).<br />

Phopho-p38 MAPK<br />

(Thr180/Tyr182) is<br />

expressed in human<br />

colon carcinoma.<br />

Stimulus<br />

Growth Factors,<br />

Mitogens, GPCR,<br />

Antigen Receptors<br />

Stress, DNA Damage, GPCR,<br />

Inflammatory Cytokines,<br />

Growth Factors<br />

Stress, Mitogens, GPCR,<br />

Neurotrophic Factors,<br />

GPCR, RTKs,<br />

Neurotrophic Factors<br />

Activator<br />

MAPKKK<br />

MAPKK<br />

RAS<br />

A-Raf,<br />

B-Raf, c-Raf,<br />

Mos, Tpl2<br />

MEK1/2<br />

Ras, Rho, Rac,<br />

Cdc42, TRAFs,<br />

GADD45a<br />

MEKK3/4,<br />

ASK1/2, TAOK1/2,<br />

MLK3, Tpl2,<br />

DLK, ZAK<br />

MKK3/6,<br />

MKK4<br />

Ras, Rho, Rac,<br />

Cdc42, TRAFs,<br />

GADD45a<br />

MEKK1/2/4,<br />

MLK1-4, ASK1/2,<br />

TAOK1/2,TAK1,<br />

DLK, ZAK<br />

Gαq, Gα12/13,<br />

Ras, Rap<br />

MEKK2/3<br />

MKK4/7 MEK5 PAK1/2/3<br />

Unknown<br />

Anisomycin treatment results in activation and nuclear<br />

translocation of phospho-SAPK/JNK (Thr183/Tyr185).<br />

Phospho-SAPK/JNK (Thr183/Tyr185)<br />

(G9) Mouse mAb #9255: Confocal IF<br />

analysis of HeLa cells, untreated (left)<br />

and anisomycin-treated (right), using<br />

#9255 (green). Actin filaments were<br />

labeled with DY-554 phalloidin (red).<br />

Phospho-p38 MAPK (Thr180/Tyr182)<br />

(D3F9) XP ® Rabbit mAb #4511: IHC<br />

analysis of paraffin-embedded human<br />

colon carcinoma using #4511.<br />

MAPK<br />

Erk1/2<br />

p38-MAPK<br />

α/β/γ/δ<br />

JNK1-3 Erk5 Erk3/4 Erk7/8<br />

MAPKAPK<br />

Biological<br />

Response<br />

RSK1-4,<br />

MNK1/2,<br />

MSK1/2<br />

Growth, Differentiation,<br />

Proliferation, Development<br />

MAPKAPK-2/3,<br />

MSK1/2, MNK1<br />

MAPKAPK-2,<br />

MAPKAPK-3<br />

Inflammation, Apoptosis,<br />

Growth, Differentiation<br />

RSK1-4<br />

Growth,<br />

Differentiation, Development<br />

MAPKAPK-5<br />

Inflammation, Apoptosis,<br />

Differentiation<br />

Select Reviews<br />

Arthur, J.S. and Ley, S.C. (2013) Nat. Rev. Immunol. 13, 679–692. • Cargnello, M. and Roux, P.P. (2011) Microbiol. Mol. Biol.<br />

Rev. 75, 50–83. • Cseh, B., Doma, E., and Baccarini, M. (2014) FEBS Lett. 588, 2398–2406. • Darling, N.J. and Cook, S.J.<br />

(2014) Biochim. Biophys. Acta. 1843, 2150–2163. • Koul, H.K., Pal, M., and Koul, S. (2013) Genes Cancer 4, 342–359. •<br />

Plotnikov, A., Zehorai, E., Procaccia, S., and Seger, R. (2011) Biochim. Biophys. Acta. 1813, 1619–1633. • Sehgal, V. and<br />

Ram, P.T. (2013) Genes Cancer 4, 409–413.<br />

42 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstmapk 43


Section I: Research Areas<br />

chapter 02: Signaling<br />

These protein targets represent key<br />

nodes within MAP Kinase pathways<br />

and are commonly studied in MAP<br />

Kinase research. Primary antibodies,<br />

antibody conjugates, and antibody<br />

sampler kits containing these targets<br />

are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

E PathScan ® ELISA Kits<br />

S SignalSilence ® siRNA<br />

C Antibody Conjugate<br />

Chemical Modulators for the Study of MAP Kinase Signaling<br />

#2222 Anisomycin Inhibits protein synthesis; activates stress-activated kinases (SAPK/JNK, p38-MAPK)<br />

#11916 Chelerythrine<br />

Chloride<br />

SP600125 is a highly specific inhibitor of JNK-family kinases.<br />

SP600125 #8177: WB analysis of extracts from 293T cells, untreated or<br />

treated with Anisomycin #2222 (25 μg/ml) for the indicated times either with<br />

or without #8177 pre-treatment (50 μM, 40 min), using Phospho-JunB<br />

(Thr102/Thr104) (D3C6) Rabbit mAb #8053 (upper) or JunB (C37F9) Rabbit<br />

mAb #3753 (lower).<br />

Commonly Studied MAP Kinase Targets<br />

Target M P E S C<br />

ASK1<br />

• •<br />

Phospho-ASK1 (Ser83) •<br />

Phospho-ASK1 (Thr845) •<br />

Phospho-ASK1 (Ser967) •<br />

APS<br />

•<br />

β-Arrestin 1 • •<br />

Phospho-β-Arrestin 1 (Ser412) •<br />

β-Arrestin 1/2<br />

•<br />

β-Arrestin 2<br />

•<br />

ATF-2 • •<br />

Phospho-ATF-2 (Thr69/Thr71) •<br />

Phospho-ATF-2 (Thr71) • • • •<br />

Bcr<br />

•<br />

Phospho-Bcr (Tyr177)<br />

•<br />

Bcr-Abl (b2a2 Junction Specific) •<br />

c-Abl<br />

•<br />

Phospho-c-Abl (Tyr89) •<br />

Phospho-c-Abl (Tyr204) •<br />

Phospho-c-Abl (Tyr245) • •<br />

Phospho-c-Abl (Tyr412) • •<br />

Phospho-c-Abl (Thr735) •<br />

Csk<br />

•<br />

Dexras1<br />

Dok1<br />

Cell-permeable inhibitor of PKC; activates SAPK/JNK and p38-MAPK;<br />

Induces apoptosis in some cell lines<br />

#9900 PD98059 Highly selective inhibitor of MEK1 and MEK2; binds<br />

inactive forms and prevent activation by upstream kinases<br />

#12147 PD184352 Highly selective, noncompetitive inhibitor of MEK1 and MEK2<br />

#8158 SB202190 Cell-permeable, highly specific inhibitor of p38-MAPK (ATP-competitive inhibitor)<br />

#5633 SB203580 Cell-permeable inhibitor of p38-MAPK (inhibits PDK1 at higher concentrations)<br />

#8705 Sorafenib Inhibits VEGFR and PDGFR; inhibits Raf kinases; induces autophagy<br />

#8177 SP600125 Cell-permeable, highly specific inhibitor of JNK-family kinases<br />

#9903 U0126 Highly selective inhibitor of MEK1 and MEK2<br />

•<br />

•<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

Phospho-JunB<br />

(Thr102/Thr104)<br />

JunB<br />

0 15 30 0 15 30 Anisomycin (min)<br />

– – – + + + SP600125<br />

Target M P E S C<br />

Dok2<br />

•<br />

Phospho-Dok2 (Tyr345) •<br />

Phospho-DUSP1 (Ser359) •<br />

DUSP3/VHR<br />

•<br />

DUSP4/MKP2<br />

•<br />

DUSP6/MKP3<br />

•<br />

DUSP10/MKP5<br />

•<br />

DUSP16/MKP7<br />

•<br />

Elk-1<br />

•<br />

Phospho-Elk-1 (Ser383) • •<br />

Erk1 (p44 MAPK) • •<br />

Erk1/2 (p44/42 MAPK) • • • •<br />

Phospho-Erk1/2 (Thr202/Tyr204) • • • •<br />

Erk2 (p42 MAPK) • •<br />

Erk5 • • •<br />

Phospho-Erk5 (Thr218/Tyr220) •<br />

FAM129B<br />

•<br />

c-Fos • • •<br />

Phospho-c-Fos (Ser32) • •<br />

FosB • • •<br />

FRA1<br />

•<br />

Phospho-FRA1 (Ser265) • • •<br />

Phospho-FRS2-α (Tyr196) •<br />

Phospho-FRS2-α (Tyr436) •<br />

Target M P E S C<br />

GCK<br />

•<br />

Grb2 • •<br />

Grb10<br />

•<br />

HGK<br />

• •<br />

JIP4/SPAG9<br />

•<br />

JNK1/SAPK<br />

• • • •<br />

Phospho-JNK1/SAPK (Thr183/Tyr185) • • • •<br />

JNK2<br />

•<br />

JNK3<br />

•<br />

c-Jun • •<br />

Phospho-c-Jun (Ser63) • • •<br />

Phospho-c-Jun (Ser73) • • •<br />

Phospho-c-Jun (Thr91) •<br />

Phospho-c-Jun (Thr93) •<br />

Phospho-c-Jun (Ser243) •<br />

JunB<br />

• •<br />

JunD<br />

•<br />

KSR1<br />

•<br />

Phospho-KSR1 (Ser392) •<br />

MAPKAPK-2<br />

• •<br />

Phospho-MAPKAPK-2 (Thr222) •<br />

Phospho-MAPKAPK-2 (Thr334) • • •<br />

MAPKAPK-3<br />

• •<br />

MAPKAPK-5<br />

•<br />

MEF2A<br />

•<br />

Phospho-MEF2A (Ser408) •<br />

MEF2C<br />

•<br />

MEK1 • • • •<br />

Phospho-MEK1 (Thr286) •<br />

Phospho-MEK1 (Ser298) •<br />

MEK1/2 • • •<br />

Phospho-MEK1/2 (Ser217/221) • • • •<br />

MEK2 • • •<br />

MEKK3<br />

•<br />

Mig6<br />

•<br />

MKK3 • • •<br />

• •<br />

Phospho-MKK3 (Ser189)/MKK6<br />

(Ser207)<br />

SEK1/MKK4<br />

• •<br />

Phospho-SEK1/MKK4 (Ser80) •<br />

Phospho-SEK1/MKK4 (Ser257) •<br />

Phospho-SEK1/MKK4 (Ser257/Thr261) •<br />

Phospho-SEK1/MKK4 (Thr261) •<br />

MKK6<br />

• •<br />

MKK7 • •<br />

Phospho-MKK7 (Ser271/Thr275) •<br />

MLK1<br />

•<br />

MLK3<br />

•<br />

MSK1<br />

•<br />

Phospho-MSK1 (Ser360) •<br />

Phospho-MSK1 (Ser376) •<br />

Phospho-MSK1 (Thr581) •<br />

MSK2<br />

•<br />

p38 MAPK • • • •<br />

Phospho-p38 MAPK (Thr180/Tyr182) • • • •<br />

p38-α MAPK • • •<br />

p38-β MAPK • •<br />

Target M P E S C<br />

p38-δ MAPK • • •<br />

p38-γ MAPK • •<br />

PP2C α<br />

•<br />

Phospho-PTPα (Tyr798) •<br />

Pyk2 • • •<br />

Phospho-Pyk2 (Tyr402) • •<br />

A-Raf<br />

•<br />

Phospho-A-Raf (Ser299) •<br />

B-Raf • •<br />

Phospho-B-Raf (Ser445) •<br />

c-Raf • • •<br />

Phospho-c-Raf (Ser259) •<br />

Phospho-c-Raf<br />

(Ser289/Ser296/Ser301)<br />

Phospho-c-Raf (Ser296)<br />

Phospho-c-Raf (Ser338)<br />

Ras<br />

•<br />

•<br />

• •<br />

• •<br />

RKIP • • •<br />

p90RSK1<br />

• • • •<br />

Phospho-p90RSK (Thr359) •<br />

Phospho-p90RSK (Thr359/Ser363) •<br />

Phospho-p90RSK (Thr573) •<br />

Phospho-p90RSK (Ser380) • • • •<br />

RSK1/RSK2/RSK3<br />

• •<br />

RSK2<br />

• •<br />

Phospho-RSK2 (Ser227) •<br />

RSK3<br />

•<br />

Phospho-RSK3 (Thr356/Ser360) •<br />

Shc<br />

•<br />

Phospho-Shc (Tyr239/240) •<br />

Phospho-Shc (Tyr317)<br />

•<br />

Phospho-SHIP2 (Tyr1135) •<br />

SHP-2 • • •<br />

Phospho-SHP-2 (Tyr542) •<br />

Phospho-SHP-2 (Tyr580) • • •<br />

SOS1<br />

• •<br />

Src • • • •<br />

Phospho-Src (Ser17) • •<br />

Phospho-Src (Tyr416)<br />

•<br />

Phospho-Src (Tyr527)<br />

•<br />

Phospho-Src Family (Tyr416) • • •<br />

Non-phospho-Src (Tyr416) •<br />

Non-phospho-Src (Tyr527) •<br />

SRF<br />

•<br />

Phospho-SRF (Ser103) •<br />

TAB1<br />

•<br />

TAB2<br />

• •<br />

Phospho-TAB2 (Ser372) •<br />

TAK1 • • •<br />

Phospho-TAK1 (Thr184) •<br />

Phospho-TAK1 (Thr184/Thr187) • •<br />

Phospho-TAK1 (Thr187)<br />

Phospho-TAK1(Ser412)<br />

Tnk1<br />

Phospho-Tnk1 (Tyr277)<br />

Phospho-Tpl2/Cot (Ser400)<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

745<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for Phospho-Erk1/2<br />

(Thr202/Try204) have been cited<br />

over 745 times in high-impact, peerreviewed<br />

publications from the global<br />

research community.<br />

Select Citations:<br />

Kusumbe, A.P. et al. (2014) Coupling<br />

of angiogenesis and osteogenesis<br />

by a specific vessel subtype in bone.<br />

Nature 507, 323–328.<br />

Brady, D.C. et al. (2014) Copper is<br />

required for oncogenic BRAF signalling<br />

and tumorigenesis. Nature 509,<br />

492–496.<br />

Dhandapany, P.S. et al. (2014) RAF1<br />

mutations in childhood-onset dilated<br />

cardiomyopathy. Nat. Genet. 46,<br />

635–639.<br />

Ota, K.T. et al. (2014) REDD1 is<br />

essential for stress-induced synaptic<br />

loss and depressive behavior. Nat.<br />

Med. 20, 531–535.<br />

von Figura, G. et al. (2014) The<br />

chromatin regulator Brg1 suppresses<br />

formation of intraductal papillary<br />

mucinous neoplasm and pancreatic<br />

ductal adenocarcinoma. Nat. Cell Biol.<br />

16, 255–267.<br />

Paul, S. et al. (2014) T cell receptor<br />

signals to NF-kappaB are transmitted<br />

by a cytosolic p62-Bcl10-Malt1-IKK<br />

signalosome. Sci. Signal. 7, ra45.<br />

Barcelo, C. et al. (2014) Phosphorylation<br />

at Ser-181 of oncogenic KRAS<br />

is required for tumor growth. Cancer<br />

Res. 74, 1190–1199.<br />

Buonato, J.M. et al. (2014) ERK1/2<br />

blockade prevents epithelial-mesenchymal<br />

transition in lung cancer cells<br />

and promotes their sensitivity to EGFR<br />

inhibition. Cancer Res. 74, 309–319.<br />

Cordero, J.B. et al. (2014) c-Src<br />

drives intestinal regeneration<br />

and transformation. EMBO J. 33,<br />

1474–1491.<br />

Kabekkodu, S.P. et al. (2014) DNA<br />

promoter methylation-dependent<br />

transcription of the double C2-like<br />

domain beta (DOC2B) gene<br />

regulates tumor growth in human<br />

cervical cancer. J. Biol. Chem. 289,<br />

10637–10649.<br />

44 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstmapk 45


Section I: Research Areas<br />

chapter 02: Signaling<br />

MAPK/Erk in Growth and Differentiation<br />

Signaling Pathways Activating p38 MAP Kinase<br />

Ion Channels<br />

[Ca 2+ ]<br />

RTKs<br />

Integrins<br />

RTKs<br />

GPCR<br />

Inflammatory Cytokines, FasL<br />

DNA Damage,<br />

Oxidative Stress, UV<br />

TGF-β<br />

[Ca 2+ ]<br />

MEK1<br />

LAMTOR2<br />

MP1<br />

Erk1<br />

PLCγ<br />

PKC<br />

Ion Channels,<br />

Receptors<br />

cPLA 2<br />

Shc<br />

FRS2<br />

IRS<br />

Src<br />

PYK2<br />

GRB2<br />

SOS<br />

Spry<br />

Spred<br />

Tpl2/Cot1<br />

IMP<br />

C-TAK1<br />

Ras<br />

c-Raf<br />

PI3K<br />

PAK<br />

SOS<br />

GRB2<br />

Rac<br />

PKA<br />

c-Raf B-Raf<br />

Heterodimer<br />

KSR<br />

1/2<br />

MEK1/2<br />

Tal<br />

CAS RACK1<br />

Fyn Pax<br />

FAK<br />

[cAMP]<br />

B-Raf<br />

PD98059<br />

U0126<br />

AZD6244<br />

SOS<br />

PP1/<br />

PP2At<br />

14-3-3<br />

EPAC<br />

Bim<br />

Rap1<br />

AMPK<br />

LKB1<br />

FMK<br />

SL0101<br />

BI-D1870<br />

C3G<br />

Raptor<br />

CRK<br />

mTOR<br />

Signaling<br />

rpS6<br />

eIF4B<br />

Filamin A<br />

eEF2K<br />

DAPK<br />

TCPβ<br />

IκBα<br />

GSK-3<br />

METTL1<br />

Endocytic<br />

Trafficking<br />

Gα o<br />

TAO1/2/3<br />

EEA1<br />

Rho-GDI<br />

eEF2K<br />

DLK<br />

MKK3/6<br />

MEKK1-4<br />

p38 MAPK<br />

TRADD<br />

TRAF2<br />

RIP<br />

Daxx<br />

MLK2/3 ASK1/2 TAK1<br />

MKK4<br />

PRAK<br />

Apoptosis<br />

SB203580<br />

TAB1<br />

HSP27<br />

Late Endosome<br />

Cell Adhesion<br />

Erk1/2<br />

Translation<br />

Control<br />

PEA-15<br />

p90RSK<br />

MNK1/2<br />

MKP-3<br />

Cytoplasmic Anchoring<br />

Erk1/2<br />

WAVE-2<br />

TSC2<br />

Migration<br />

p90RSK<br />

PLD1<br />

BAD<br />

nNos<br />

Ca +2 -regulated<br />

Exocytosis<br />

Translation<br />

MNK1/2<br />

cPLA 2<br />

Tau<br />

DUSPs<br />

p90RSK<br />

MAPKAPK-3<br />

MAPKAPK-2<br />

Cytokine-induced<br />

mRNA stability<br />

TNF-α biosynthesis<br />

Cytoplasm<br />

Nucleus<br />

MKP-1/2<br />

cdc25<br />

Erk1/2<br />

PPARγ<br />

MSK1/2<br />

p27 KIP1<br />

p90RSK<br />

BUB1<br />

Cytoplasm<br />

Nucleus<br />

p38 MAPK<br />

MAPKAPK-3<br />

PRAK<br />

ER<br />

Stat1/3<br />

Ets<br />

c-Myc/<br />

N-Myc<br />

Elk-1<br />

Pax6<br />

TIF1A<br />

c-Fos<br />

FoxO3<br />

CREB<br />

ATF1<br />

Histone<br />

H3<br />

ER81<br />

HMGN1<br />

SRF<br />

ETV1<br />

c-Fos<br />

TIF1A<br />

ERα<br />

ATF4<br />

Nur77<br />

Myt1<br />

MITF<br />

Mad1<br />

C/EBPβ<br />

YB1<br />

Pax6 p53 Stat1 Max Myc Elk-1 CHOP MEF2 ATF-2 ETS1<br />

RARα<br />

MAPKAPK-2<br />

HMGN1<br />

MSK1/2<br />

Histone H3<br />

CREB<br />

NF-κB<br />

p65<br />

ATF1<br />

ER81<br />

UBF<br />

Transcription<br />

Transcription<br />

Cytokine Production,<br />

Apoptosis, etc.<br />

The MAPK/Erk signaling cascade is activated by a wide variety of receptors involved in growth and differentiation including receptor tyrosine kinases (RTKs), integrins, and ion<br />

channels. The specific components of the cascade vary greatly among different stimuli, but the architecture of the pathway usually includes a set of adaptors (Shc, GRB2, Crk,<br />

etc.) linking the receptor to a guanine nucleotide exchange factor (SOS, C3G, etc.) transducing the signal to small GTP-binding proteins (Ras, Rap1), which in turn activate the<br />

core unit of the cascade composed of a MAPKKK (Raf), a MAPKK (MEK1/2), and MAPK (Erk). An activated Erk dimer can regulate targets in the cytosol and also translocate to<br />

the nucleus where it phosphorylates a variety of transcription factors regulating gene expression.<br />

Select Reviews:<br />

Anjum, R. and Blenis, J. (2008) Nat. Rev. Mol. Cell Biol. 9, 747–758. • De Luca, A., Maiello, M.R., D’Alessio, A., Pergameno, M., and Normanno, N. (2012) Expert Opin.<br />

Ther. 2, 17–27. • Keyse, S.M. (2008) Cancer Metastasis Rev. 27, 253–261. • Kim, E.K. and Choi, E.J. (2010) Biochim. Biophys. Acta 1802, 396–405. • Mendoza,<br />

M.C., Er, E.E., and Blenis, J. (2011) Trends Biochem. Sci. 36, 320–328. • Romeo, Y., Zhang, X., and Roux, P.P. (2012) Biochem. J. 441, 553–569. • Roskoski, R. Jr.<br />

(2012) Biochem. Biophys. Res. Commun. 417, 5–10. • Tidyman, W.E. and Rauen, K.A. (2009) Curr. Opin. Genet. Dev. 19, 230–236.<br />

p38 MAPKs (α, β, γ, and δ) are members of the MAPK family that are activated by a variety of environmental stresses and inflammatory cytokines. As with other MAPK<br />

cascades, the membrane-proximal component is a MAPKKK, typically a MEKK or a mixed lineage kinase (MLK). The MAPKKK phosphorylates and activates MKK3/6, the p38<br />

MAPK kinases. MKK3/6 can also be activated directly by ASK1, which is stimulated by apoptotic stimuli. p38 MAPK is involved in regulation of HSP27, MAPKAPK-2 (MK2),<br />

MAPKAPK-3 (MK3), and several transcription factors including ATF-2, Stat1, the Max/Myc complex, MEF-2, Elk-1, and indirectly CREB via activation of MSK1.<br />

Select Reviews:<br />

Coulthard, L.R., White, D.E., Jones, D.L., McDermott, M.F., and Burchill, S.A. (2009) Trends Mol. Med. 15, 369–379. • Cuadrado, A. and Nebreda, A.R. (2010) Biochem. J.<br />

429, 403–417. • del Barco Barrantes, I. and Nebreda, A.R. (2012) Biochem. Soc. Trans. 40, 79–84. • Huang, G., Shi, L.Z., and Chi, H. (2009) Cytokine 48, 161–169. •<br />

Kostenko, S., Dumitriu, G., Lægreid, K.J., and Moens, U. (2011) World J. Biol. Chem. 2, 73–89. • Shiryaev, A. and Moens, U. (2010) Cell. Signal. 22, 1185–1192.<br />

© 2004–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. John Blenis, Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

46 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

© 2003–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. John Blenis, Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

www.cellsignal.com/cstpathways 47


Section I: Research Areas<br />

SAPK/JNK Signaling<br />

Growth Factors, UV<br />

RTKs<br />

CrkII<br />

CrkL<br />

Shc<br />

GRB2<br />

HPK1<br />

Cytoplasm<br />

Nucleus<br />

PI3K<br />

14-3-3<br />

SOS<br />

MAPKKKs :<br />

TAK1<br />

Ras<br />

MKK4/7<br />

JNK1/2/3<br />

IRS-1<br />

c-Abl 14-3-3 14-3-3<br />

DUSPs<br />

c-Abl<br />

Cellular Stress,<br />

γ Radiation<br />

Rac<br />

MEKK1/4<br />

MLK2/3<br />

DLK<br />

TpI-2<br />

TAO1/2<br />

NO<br />

Cdc42<br />

Rho<br />

MLKs<br />

POSH<br />

JNK<br />

JNK<br />

FasL, UV<br />

Inflammatory Cytokines<br />

GCK<br />

ASK1<br />

Rac<br />

c-Jun ATF-2 Elk-1 Smad4 p53 NFAT4 NFATc1 Stat3<br />

TRADD<br />

TRAF2<br />

JIP1,2,3<br />

RIP<br />

Daxx<br />

Transcription<br />

Oxidative<br />

Stress<br />

Apoptosis<br />

Cdc42<br />

MAPKKKs<br />

Bax<br />

HSF1<br />

Gα 12/13<br />

14-3-3<br />

Bax<br />

Growth<br />

Differentiation<br />

Survival<br />

Apoptosis<br />

G 12/13 -coupled<br />

Receptors<br />

Gβγ<br />

PI3Kγ<br />

Mitochondrial<br />

Translocation/<br />

Apoptosis<br />

Stress-activated protein kinases (SAPK)/Jun amino-terminal kinases (JNK) are members of the MAPK family and are activated by a variety of environmental stresses, inflammatory<br />

cytokines, growth factors, and GPCR agonists. Stress signals are delivered to this cascade by small GTPases of the Rho family (Rac, Rho, cdc42). As with the other<br />

MAPKs, the membrane proximal kinase is a MAPKKK, typically MEKK1–4, or a member of the mixed lineage kinases (MLK) that phosphorylates and activates MKK4 (SEK) or<br />

MKK7, the SAPK/JNK kinases. Alternatively, MKK4/7 can be activated by a member of the germinal center kinase (GCK) family in a GTPase-independent manner. SAPK/JNK<br />

translocates to the nucleus where it can regulate the activity of multiple transcription factors.<br />

Select Reviews:<br />

Bogoyevitch, M.A., Ngoei, K.R., Zhao, T.T., Yeap, Y.Y., and Ng, D.C. (2010) Biochim. Biophys. Acta. 1804, 463–475. • Chen, F. (2012) Cancer Res. 72, 379–386. • Davies,<br />

C. and Tournier, C. (2012) Biochem. Soc. Trans. 40, 85–89. • Engström, W., Ward, A., and Moorwood, K. (2010) Cell Prolif. 43, 56–66. • Haeusgen, W., Herdegen, T., and<br />

Waetzig, V. (2011) Eur. J. Cell Biol. 90, 536–544. • Verma, G., and Datta, M. (2012) J. Cell Physiol. 227, 1791–1795.<br />

PI3 Kinase/Akt Signaling<br />

Activation of Akt<br />

The serine/threonine kinase Akt/PKB exists as three isoforms in mammals. Akt1 and Akt2 have a wide<br />

tissue distribution, whereas Akt3 is expressed in testes and brain. Akt regulates multiple biological processes<br />

including cell survival, proliferation, growth, and glycogen metabolism. Various growth factors,<br />

hormones, and cytokines activate Akt by binding their cognate receptor tyrosine kinase (RTK), cytokine<br />

receptor, or G protein-coupled receptor (GPCR) and triggering activation of the lipid kinase PI3K, which<br />

generates PIP 3 at the plasma membrane. Akt binds PIP 3 through its pleckstrin homology (PH) domain,<br />

resulting in translocation of Akt to the membrane. Akt is activated through a dual phosphorylation<br />

mechanism. PDK1, which is also brought to the membrane through its PH domain, phosphorylates Akt<br />

within its activation loop at Thr308. A second phosphorylation at Ser473 within the C-terminus is also<br />

required for activity and is carried out by the mTOR-Rictor complex, mTORC2.<br />

Insulin stimulation results in clustering of phospho-Akt at the membrane.<br />

Phospho-Akt (Thr308) (D25E6) XP ® Rabbit<br />

mAb #13038: Confocal IF analysis of C2C12<br />

cells, insulin-treated (100 nM, 15 min; left) or<br />

treated with LY294002 #9901 (50 μM, 2 hr;<br />

right), using #13038 (green). Actin filaments<br />

were labeled with DY-554 phalloidin (red). Blue<br />

pseudocolor = DRAQ5 ® #4084 (fluorescent<br />

DNA dye).<br />

Growth factor stimulation results in phosphorylation<br />

of Akt at Thr308, one of two phosphorylations<br />

necessary for Akt activation.<br />

Phospho-Akt (Thr308) (D25E6) XP ® Rabbit mAb #13038: Western blot analysis of<br />

extracts from NIH/3T3 cells, untreated or treated with Human Platelet-Derived Growth<br />

Factor AA (hPDGF-AA) #8913 (100 ng/ml, 5 min; +), and untreated LNCaP and PC-3<br />

cells, using #1308 (upper) or Akt (pan) (C67E7) Rabbit mAb #4691 (lower).<br />

Lanes<br />

1. NIH/3T3<br />

2. NIH/3T3<br />

3. LNCaP<br />

4. PC-3<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

60<br />

50<br />

1 2 3 4<br />

Phospho-<br />

Akt (Thr308)<br />

Akt (pan)<br />

– + – – hPDGF<br />

Inhibition of Akt Activity<br />

PTEN, a lipid phosphatase that catalyzes the dephosphorylation of PIP 3 , is a major negative regulator of<br />

Akt signaling. Loss of PTEN function has been implicated in many human cancers. Akt activity is also<br />

negatively regulated by the phosphatases PP2A and PHLPP, as well as by the chemical modulators<br />

wortmannin and LY294002, both of which are inhibitors of PI3K.<br />

PTEN expression in human colon<br />

PTEN (D4.3) XP ® Rabbit mAb #9188: IHC analysis<br />

of paraffin-embedded human colon using #9188.<br />

kDa<br />

100<br />

chapter 02: Signaling<br />

Akt-activating treatments<br />

and absence<br />

of PTEN result in<br />

phosphorylation of<br />

Akt at Ser473.<br />

A<br />

80<br />

60<br />

50<br />

80<br />

60<br />

50<br />

PC-3<br />

– +<br />

– +<br />

– –<br />

NIH/3T3<br />

– –<br />

– –<br />

– +<br />

Phospho-Akt (Ser473)<br />

is expressed in human<br />

lung carcinoma.<br />

B<br />

Phospho-<br />

Akt (Ser473)<br />

Akt<br />

wortmannin<br />

LY294002<br />

PDGF<br />

Phospho-Akt (Ser473) (D9E) XP ®<br />

Rabbit mAb #4060: WB analysis of<br />

extracts from PC-3 cells (PTEN null),<br />

untreated or treated with LY294002<br />

#9901/Wortmannin #9951, and NIH/3T3<br />

cells, serum-starved or PDGF-treated<br />

(A), using #4060 (upper) or Akt (pan)<br />

(C67E7) Rabbit mAb #4691 (lower). IHC<br />

analysis of paraffin-embedded human<br />

lung carcinoma (B) using #4060.<br />

© 2002–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. John Blenis, Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

48 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstakt 49


Section I: Research Areas<br />

chapter 02: Signaling<br />

Akt Downstream Signaling<br />

Activated Akt phosphorylates a large number of downstream substrates containing the consensus sequence<br />

RXRXXS/T. One of its primary functions is to promote cell growth and protein synthesis through<br />

regulation of the mTOR signaling pathway. Akt directly phosphorylates and activates mTOR, as well as<br />

inhibits the mTOR inhibitor proteins PRAS40 and tuberin (TSC2). Combined, these actions promote cell<br />

growth and G1 cell cycle progression through signaling via p70 S6 Kinase and inhibition of 4E-BP1.<br />

GSK-3 is a primary target of Akt and inhibitory phosphorylation of GSK-3α (Ser21) or GSK-3β (Ser9)<br />

has numerous cellular effects such as promoting glycogen metabolism, cell cycle progression, regulation<br />

of Wnt signaling, and contributing to the formation of neurofibrillary tangles in Alzheimers disease.<br />

Akt promotes cell survival directly by its ability to phosphorylate and inactivate several pro-apoptotic<br />

targets, including Bad, Bim, Bax, and the Forkhead (FoxO1/3a) transcription factors. Akt also plays an<br />

important role in metabolism and insulin signaling. Insulin receptor signaling through Akt promotes<br />

Glut4 translocation through activation of AS160 and TBC1D1, resulting in increased glucose uptake.<br />

Akt regulates glycolysis through phosphorylation of PFK and hexokinase, and plays a significant role in<br />

aerobic glycolysis in cancer cells, also known as the Warburg Effect.<br />

Akt in Disease<br />

Aberrant Akt signaling is the underlying defect found in several pathologies. Akt is one of the most<br />

frequently activated kinases in human cancer as constitutively active Akt can promote unregulated<br />

cell proliferation. Abnormalities in Akt2 signaling can result in diabetes due to defects in glucose<br />

homeostasis. Akt is also a key player in cardiovascular disease through its role in cardiac growth,<br />

angiogenesis, and hypertrophy.<br />

Select Reviews<br />

Bar-Peled, L. and Sabatini, D.M. (2014) Trends Cell Biol. 24, 400−406. • Correia, N.C., Gírio, A., Antunes, I., et al. (2014)<br />

Eur. J. Cancer 50, 216−225. • Betz, C. and Hall, M.N. (2013) J. Cell. Biol. 203, 563−574. • Fruman, D.A. and Rommel, C.<br />

(2014) Nat. Rev. Drug Discov. 13, 140−156. • Kumar, A., Rajendran, V., Sethumadhavan, R., and Purohit, R. (2013) Scientific<br />

World Journal 2013, 756134. • Ortega-Molina, A. and Serrano, M. (2013) Trends Endocrinol. Metab. 24, 184−189. • Porta,<br />

C., Paglino, C., and Mosca, A. (2014) Front Oncol. 4, 64. • Shimobayashi, M. and Hall, M.N. (2014) Nat. Rev. Mol. Cell Biol.<br />

15, 155−162. • Toker, A. and Marmiroli, S. (2014) Adv. Biol. Regul. 55, 28−38.<br />

Inhibitors of<br />

PI3 Kinase/Akt/mTOR<br />

#12017 Everolimus<br />

Specific inhibitor of<br />

mTORC1 protein complex<br />

#9904 Rapamycin<br />

Specific inhibitor of<br />

mTORC1 protein complex<br />

#9901 LY294002<br />

Reversible but potent<br />

inhibitor of PI3K family<br />

members<br />

#9951 Wortmannin<br />

Irreversible, potent inhibitor<br />

of PI3K family members<br />

Akt activation by insulin<br />

treatment results in<br />

phosphorylation of<br />

mTOR at Ser2448.<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

200<br />

140<br />

100<br />

80<br />

1 2 3<br />

Phospho-mTOR<br />

(Ser2448)<br />

mTOR<br />

Phospho-mTOR (Ser2448) (D9C2)<br />

XP ® Rabbit mAb #5536: WB analysis<br />

of extracts from serum-starved NIH/3T3<br />

cells, untreated or insulin-treated (150<br />

nM, 5 min), alone or in combination with<br />

λ-phosphatase, using #5536 (upper) or<br />

mTOR (7C10) Rabbit mAb #2983.<br />

Lanes<br />

1. untreated<br />

2. insulin-treated (150 nM, 5 min)<br />

3. λ-phosphatase and insulin-treated<br />

(150 nM, 5 min)<br />

Inhibitors of Akt activation prevent phosphorylation of GSK-3β at Ser9.<br />

Phospho-GSK-3β (Ser9) (D85E12) XP ® Rabbit mAb #5558: Confocal IF analysis of PC-3 cells, untreated (A), LY294002- and<br />

Wortmannin-treated (#9901 and #9951 respectively) (B), or λ phosphatase-treated (C), using #5558 (green). Actin filaments were labeled<br />

with DY-554 phalloidin (red). Blue pseudocolor = DRAQ5 ® #4084 (fluorescent DNA dye).<br />

Active Akt phosphorylates FoxO3a at Ser253, retaining<br />

FoxO3a in the cytoplasm and preventing cell death.<br />

A<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

140<br />

100<br />

80<br />

1 2<br />

Phospho-FoxO3<br />

(Ser253)<br />

FoxO3<br />

Lanes<br />

1. untreated<br />

2. treated overnight with<br />

LY294002 #9901 (50 μM) and<br />

Wortmannin #9951 (1 μM)<br />

A<br />

B<br />

B<br />

Phospho-FoxO3a (Ser253) (D18H8) Rabbit mAb #13129: WB analysis of extracts from<br />

293T cells (A) using #13129 (upper) or FoxO3a (D19A7) Rabbit mAb #12829 (lower).<br />

FoxO3a (75D8) Rabbit mAb #2497: Confocal IF analysis of SH-SY5Y cells, IGF-I treated<br />

(B) or LY294002-treated (C), using FoxO3a (75D8) Rabbit mAb (green). Actin filaments were<br />

labeled with Alexa Fluor ® 555 Phalloidin #8953 (red). Blue pseudocolor = DRAQ5 ® #4084<br />

(fluorescent DNA dye).<br />

Akt Signaling Research<br />

Please visit our website for additional resources and products relating to the study of Akt Signaling.<br />

www.cellsignal.com/cstakt<br />

C<br />

C<br />

Commonly Studied Akt Targets<br />

Target M P E S C<br />

Akt1 • • •<br />

Phospho-Akt1 (Ser129) •<br />

Phospho-Akt1 (Ser473) • •<br />

Phospho-Drosophila Akt (Ser505) •<br />

Akt1/Akt2/Akt3 • • • •<br />

Phospho-Akt1 (Thr308)/Phospho-Akt2<br />

(Thr309)/Phospho-Akt3 (Thr305) • • • •<br />

Phospho-Akt1 (Ser473)/Phospho-Akt2<br />

(Ser474)/Phospho-Akt3 (Ser472) • • • •<br />

Phospho-Akt1 (Thr450)/Phospho-Akt2<br />

(Thr451)/Phospho-/Akt3 (Thr447) • •<br />

Akt2 • • • •<br />

Phospho-Akt2 (Ser474) • •<br />

Akt3<br />

• • •<br />

Phospho-Akt3 (Ser472)<br />

•<br />

ATP6V1B1/2<br />

•<br />

CTMP<br />

•<br />

DEPTOR/DEPDC6<br />

•<br />

DJ-1<br />

• •<br />

eNOS<br />

• •<br />

Phospho-eNOS (Ser113) •<br />

Phospho-eNOS (Thr495) •<br />

Phospho-eNOS (Ser1177) • • •<br />

FKBP5<br />

• •<br />

FLCN<br />

•<br />

FoxO1 • • • •<br />

•<br />

Phospho-FoxO1 (Thr24)/FoxO3a<br />

(Thr32)<br />

Phospho-FoxO1 (Thr24)/FoxO3a<br />

(Thr32)/FoxO4 (Thr28)<br />

Phospho-FoxO1 (Ser256)<br />

Phospho-FoxO1 (Ser319)<br />

•<br />

•<br />

•<br />

FoxO3a • •<br />

Phospho-FoxO3a (Ser253) • •<br />

Phospho-FoxO3a (Ser294) •<br />

Phospho-FoxO3a (Ser318/Ser321) •<br />

Phospho-FoxO3a (Ser413) •<br />

FoxO4<br />

• •<br />

Phospho-FoxO4 (Ser193) •<br />

Gab1<br />

•<br />

Target M P E S C<br />

Phospho-Gab1 (Tyr307) •<br />

Phospho-Gab1 (Tyr627) • •<br />

Gab2<br />

•<br />

Phospho-Gab2 (Ser159) •<br />

Phospho-Gab2 (Tyr452)<br />

• •<br />

GβL • • •<br />

GSK-3α • • •<br />

Phospho-GSK-3α (Ser21) •<br />

GSK-3α/GSK-3β • •<br />

Phospho-GSK-3α/β (Ser21/9) •<br />

GSK-3β • •<br />

Phospho-GSK-3β (Ser9) • • •<br />

Phospho-GSK-3β (Thr390)<br />

HGS<br />

LAMTOR1/C11orf59<br />

LAMTOR2/ROBLD3<br />

LAMTOR3/MAPKSP1<br />

LAMTOR4/C7orf59<br />

MAP4K3<br />

Mios<br />

mTOR<br />

Phospho-mTOR (Ser2448)<br />

Phospho-mTOR (Ser2481)<br />

p70 S6 Kinase<br />

Phospho-p70 S6 Kinase (Ser371)<br />

Phospho-p70 S6 Kinase (Thr389)<br />

Phospho-p70 S6 Kinase<br />

(Thr421/Ser424)<br />

Phospho-Drosophila p70 S6<br />

Kinase (Thr398)<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• • • • •<br />

• •<br />

• •<br />

• • • • •<br />

•<br />

• • •<br />

•<br />

•<br />

PDK1 • • •<br />

Phospho-PDK1 (Ser241) • •<br />

PI3 Kinase p55<br />

•<br />

PI3 Kinase p85 α • •<br />

PI3 Kinase p85<br />

• •<br />

Phospho-PI3 Kinase p85<br />

(Tyr458)/p55 (Tyr199)<br />

•<br />

PI3 Kinase p101<br />

•<br />

PI3 Kinase p110 α • • •<br />

These protein targets represent key<br />

nodes within Akt Signaling pathways<br />

and are commonly studied in Akt<br />

research. Primary antibodies, antibody<br />

conjugates, and antibody sampler kits<br />

containing these targets are available<br />

from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

E PathScan ® ELISA Kits<br />

S SignalSilence ® siRNA<br />

C Antibody Conjugate<br />

50 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstakt<br />

51


Other<br />

Section I: Research Areas<br />

chapter 02: Signaling<br />

Target M P E S C<br />

PI3 Kinase p110 β • •<br />

PI3 Kinase p110 γ • •<br />

PI4 Kinase<br />

•<br />

PI3 Kinase Class II α •<br />

PI3 Kinase Class III • •<br />

PNUTS<br />

•<br />

PRAS40 • • • •<br />

Phospho-PRAS40 (Ser183) •<br />

Phospho-PRAS40 (Thr246) • • •<br />

Protor2<br />

•<br />

PTEN<br />

• • • • •<br />

Phospho-PTEN (Ser380) • •<br />

Phospho-PTEN (Ser380/Thr382/383) • •<br />

non-Phospho-PTEN<br />

(Ser380/Thr382/Thr383) • •<br />

RagA<br />

•<br />

RagB<br />

•<br />

RagC<br />

• •<br />

RagD<br />

•<br />

Raptor • •<br />

Phospho-Raptor (Ser792) •<br />

REDD1<br />

•<br />

Rheb<br />

•<br />

Rictor • • • •<br />

Target M P E S C<br />

Phospho-Rictor (Thr1135) •<br />

SGK1<br />

•<br />

Phospho-SGK1 (Ser78) • •<br />

SGK2<br />

• •<br />

SGK3<br />

•<br />

Phospho-SGK3 (Thr320) •<br />

Sin1<br />

•<br />

TCL1<br />

• •<br />

Hamartin/TSC1<br />

• •<br />

Tuberin/TSC2 • • •<br />

Phospho-Tuberin/TSC2 (Ser939) •<br />

Phospho-Tuberin/TSC2 (Ser1254) •<br />

Phospho-Tuberin/TSC2 (Ser1387) •<br />

Phospho-Tuberin/TSC2 (Thr1462) • •<br />

Phospho-Tuberin/TSC2 (Tyr1571) •<br />

WNK1<br />

•<br />

Phospho-WNK1 (Thr60) •<br />

WNK4<br />

•<br />

YAP<br />

• •<br />

Phospho-YAP (Ser127) • •<br />

Phospho-YAP (Ser397) •<br />

YB1 • • •<br />

Phospho-YB1 (Ser102) • •<br />

PI3 Kinase/Akt Signaling<br />

Protein Synthesis<br />

FAK<br />

Paxillin<br />

S6<br />

FoxO1<br />

PI3K<br />

p70 S6K<br />

PDCD4<br />

Inhibits<br />

Autophagy<br />

Bim<br />

ILK<br />

Bcl-2<br />

Integrin<br />

4E-<br />

BP1<br />

ATG13<br />

Inhibits Apoptosis<br />

TRAF6<br />

mTORC1<br />

XIAP<br />

PI3K<br />

Gab1<br />

Gab2<br />

ATM/ATR<br />

DNA-PK<br />

PTEN Akt<br />

PIP<br />

PIP 3<br />

3<br />

p53<br />

IRS-1<br />

TSC2<br />

TSC1<br />

PRAS40<br />

RTK<br />

NEDD1-4<br />

TBK1<br />

Membrane PDK1<br />

Recruitment<br />

and Activation<br />

MDM2<br />

Bad<br />

14-3-3<br />

DNA Damage<br />

Jak1<br />

Akt<br />

Cytokine<br />

Receptor<br />

PI3K<br />

mTORC2<br />

PDK1<br />

GSK-3<br />

CD19<br />

PI3K<br />

Membrane<br />

Recruitment<br />

and Activation<br />

mIg<br />

BCR<br />

BCAP<br />

Syk<br />

PFKFB2<br />

Ag<br />

α/β α/β<br />

PIP5K AS160<br />

Lyn<br />

p47phox<br />

Lamin A<br />

Tpl2<br />

IKKα<br />

eNOS<br />

PI3K<br />

Gβγ<br />

Hexokinase II<br />

Gα<br />

GTP<br />

PTEN PP2A PHLPP1/2 CTMP<br />

Huntingtin<br />

Ataxin-1<br />

mIg<br />

GABAAR<br />

GPCR<br />

Neutrophil<br />

Respiratory<br />

Burst<br />

Organization<br />

of Nuclear<br />

Proteins<br />

NF-κB<br />

Pathway<br />

Cardiovascular<br />

Homeostasis<br />

Synaptic<br />

Signaling<br />

Blocks<br />

Aggregation<br />

Promotes<br />

Neuroprotection<br />

Aggregation<br />

Neurodegeneration<br />

Glycolysis<br />

Neuroscience<br />

996<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for Phospho-Akt<br />

(Ser473) have been cited over 996 times<br />

in high-impact, peer-reviewed publications<br />

from the global research community.<br />

Select Citations:<br />

Liu, P. et al. (2014) Cell-cycle-regulated activation of Akt<br />

kinase by phosphorylation at its carboxyl terminus. Nature<br />

508, 541–545.<br />

Ardestani, A. et al. (2014) MST1 is a key regulator of beta<br />

cell apoptosis and dysfunction in diabetes. Nat. Med. 20,<br />

385–397.<br />

White, A.C. et al. (2014) Stem cell quiescence acts as a<br />

tumour suppressor in squamous tumours. Nat. Cell Biol.<br />

16, 99–107.<br />

Snuderl, M. et al. (2013) Targeting placental growth factor/<br />

neuropilin 1 pathway inhibits growth and spread of medulloblastoma.<br />

Cell 152, 1065–1076.<br />

Hirabayashi, S. et al. (2013) Transformed Drosophila cells<br />

evade diet-mediated insulin resistance through wingless<br />

signaling. Cell 154, 664–675.<br />

Yan, L. et al. (2013) The ubiquitin-CXCR4 axis plays an<br />

important role in acute lung infection-enhanced lung tumor<br />

metastasis. Clin. Cancer Res. 19, 4706–4716.<br />

Keysar, S.B. et al. (2013) Hedgehog signaling alters reliance<br />

on EGF receptor signaling and mediates anti-EGFR<br />

therapeutic resistance in head and neck cancer. Cancer Res.<br />

73, 3381–3392.<br />

Romaker, D. et al. (2014) MicroRNAs are critical regulators<br />

of tuberous sclerosis complex and mTORC1 activity in the<br />

size control of the Xenopus kidney. Proc. Natl. Acad. Sci.<br />

USA 111, 6335–6340.<br />

Nguyen, A. et al. (2014) Very low density lipoprotein receptor<br />

(VLDLR) expression is a determinant factor in adipose tissue<br />

inflammation and adipocyte-macrophage interaction. J Biol.<br />

Chem. 289, 1688–1703.<br />

Xiang, M. (2014) STAT3 induction of miR-146b forms a<br />

feedback loop to inhibit the NF-kappaB to IL-6 signaling axis<br />

and STAT3-driven cancer phenotypes. Sci Signal. 7, ra11.<br />

Sakr, R.A. et al. (2014) PI3K pathway activation in highgrade<br />

ductal carcinoma in situ–implications for progression<br />

to invasive breast carcinoma. Clin. Cancer Res. 20,<br />

2326–2337.<br />

Wong, C.C. et al. (2014) Inactivating CUX1 mutations<br />

promote tumorigenesis. Nat. Genet 46, 33–38.<br />

Lee, E.Y. and Abbondante, S. (2014) Tissue-specific tumor<br />

suppression by BRCA1. Proc. Natl. Acad. Sci. USA 111,<br />

4353–4354.<br />

Li, H. et al. (2014) Lysophosphatidic acid acts as a nutrientderived<br />

developmental cue to regulate early hematopoiesis.<br />

EMBO J. 33, 1383–1396.<br />

Stanton, M.J. et al. (2013) Autophagy control by the VEGF-<br />

C/NRP-2 axis in cancer and its implication for treatment<br />

resistance. Cancer Res. 73, 160–171.<br />

Wu, W. et al. (2013) Inhibition of tumor growth and metastasis<br />

in non-small cell lung cancer by LY2801653, an inhibitor<br />

of several oncokinases, including MET. Clin. Cancer Res. 19,<br />

5699–5710.<br />

Mercan, F. et al. (2013) Novel role for SHP-2 in nutrientresponsive<br />

control of S6 kinase 1 signaling. Mol. Cell Biol.<br />

33, 293–306.<br />

Survival<br />

mTORC1<br />

GβL Raptor<br />

mTOR<br />

DEPTOR<br />

Bax<br />

Apoptosis<br />

Cytosolic<br />

Sequestration<br />

Palladin<br />

Girdin<br />

Migration<br />

Migration<br />

Wee1<br />

CDK2 Cyclin A<br />

Skp2 SCF<br />

Myt1<br />

p27 Kip1<br />

Cell Cycle<br />

Proliferation<br />

p21<br />

Waf1/Cip1<br />

GS<br />

Cyclin D1<br />

ATP-Citrate<br />

Lyase<br />

Fatty Acid<br />

Synthesis<br />

Glycogen<br />

Synthesis<br />

Glucose<br />

Transport<br />

Glucose Metabolism<br />

mTORC2<br />

Sin1 PRR5<br />

Rictor GβL<br />

mTOR<br />

DEPTOR<br />

Since its initial discovery as a proto-oncogene, the serine/threonine kinase Akt (also known as protein kinase B or PKB) has become a major focus of attention because of<br />

its critical role in regulating diverse cellular functions including metabolism, growth, proliferation, survival, transcription and protein synthesis. The Akt signaling cascade is<br />

activated by receptor tyrosine kinases, integrins, B and T cell receptors, cytokine receptors, G-protein-coupled receptors and other stimuli that induce production of phosphatidylinositol<br />

(3,4,5) trisphosphates (PIP3) by phosphoinositide 3-kinase (PI3K). These lipids serve as plasma membrane docking sites for proteins that harbor pleckstrin-homology<br />

(PH) domains, including Akt and its upstream activator PDK1. At the membrane PDK1 phosphorylates Akt at Thr308 leading to partial activation of Akt. Phosphorylation of<br />

Akt at Ser473 by mTORC2 stimulates full enzymatic activity. Members of the PI3K-related kinase (PIKK) family, including DNA-PK, can also phosphorylate Akt at Ser473. Akt<br />

is dephosphorylated by protein phosphatase 2A (PP2A) and the PH-domain leucine-rich-repeat-containing protein phosphatases (PHLPP1/2). In addition, the tumor suppressor<br />

phosphatase and tensin homolog (PTEN) inhibits Akt activity by dephosphorylating PIP3.<br />

Dysregulation of the PI3K/Akt pathway is implicated in a number of human diseases including cancer, diabetes, cardiovascular disease and neurological diseases. In cancer,<br />

two mutations that increase the intrinsic kinase activity of PI3K have been identified. In addition, PTEN is frequently mutated or lost in human tumors. Activating mutations<br />

in Akt have also been described. The frequency with which dysregulated Akt signaling contributes to human disease has culminated in the aggressive development of small<br />

molecule inhibitors of PI3K and Akt.<br />

There are three highly related isoforms of Akt (Akt1, Akt2 and Akt3), which phosphorylate substrates containing the consensus phosphorylation motif RxRxxS/T. Akt isoforms<br />

share many substrates but isoform-specific Akt substrates have also been identified. For example, all Akt isoforms are able to phosphorylate PRAS40 (proline-rich Akt substrate<br />

of 40 kDa) but only Akt1 can phosphorylate the actin-associated protein palladin.<br />

Akt regulates cell growth through its effects on the TSC1/TSC2 complex and mTORC signaling. Akt contributes to cell proliferation via phosphorylation of the CDK inhibitors<br />

p21 and p27. Akt is a major mediator of cell survival through direct inhibition of pro-apoptotic proteins like Bad or inhibition of pro-apoptotic signals generated by transcription<br />

factors like FoxO. Akt is critically involved in the regulation of metabolism through activation of AS160 and PFKFB2. In addition, Akt has been shown to regulate proteins<br />

involved in neuronal function including the GABA receptor, ataxin-1 and huntingtin proteins. Akt contributes to cell migration and invasion via phosphorylation of palladin and<br />

vimentin. Akt also regulates NF-κB signaling by phosphorylating IKKα and Tpl2. Due to the critical role of Akt/PKB in regulating diverse cellular functions it is an important<br />

therapeutic target for the treatment of human disease.<br />

Select Reviews:<br />

Bhaskar, P.T. and Hay, N. (2007) Dev. Cell 12, 487–502. • Bozulic, L. and Hemmings, B.A. (2009) Curr. Opin. Cell Biol. 21, 256–261. • Brugge, J., Hung, M.C., and Mills,<br />

G.B. (2007) Cancer Cell 12, 104–107. • Carnero, A., Blanco-Aparicio, C., Renner, O., Link, W., and Leal, J.F. (2008) Curr. Cancer Drug Targets 8, 187–198. • Hers, I.,<br />

Vincent, E.E., and Tavaré, J.M. (2011) Cell Signal. 23, 1515–1527. • Liu, P., Cheng, H., Roberts, T.M., and Zhao, J.J. (2009) Nat. Rev. Drug Discov. 8, 627–644. • Manning,<br />

B.D. and Cantley, L.C. (2007) Cell 129, 1261–1274. • Salmena, L., Carracedo, A., and Pandolfi, P.P. (2008) Cell 133, 403–414.<br />

© 2007–2015 Cell Signaling Technology, Inc. • We would like to thank Kristin Brown and Prof. Alex Toker, Beth Israel Deaconess Medical Center, Harvard Medical School for reviewing this diagram.<br />

52 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstpathways 53


Section I: Research Areas<br />

chapter 02: Signaling<br />

mTOR Signaling<br />

GRB10<br />

Sin1<br />

Torin1<br />

PP242<br />

KU63794<br />

WYE354<br />

PRR5<br />

mTORC2<br />

Rictor GβL<br />

mTOR<br />

DEPTOR<br />

SGK1<br />

rapamycin<br />

FKBP12<br />

PKCα<br />

PRAS40<br />

p70S6K<br />

Cell Growth<br />

IRS-1<br />

Akt<br />

TSC1<br />

TSC2<br />

TBC1D7<br />

Rheb<br />

mTORC1<br />

GβL<br />

Raptor<br />

mTOR<br />

DEPTOR<br />

FIP200<br />

Atg13<br />

ULK<br />

Glucose<br />

PIP 3 PIP 2<br />

AMP: ATP<br />

Growth Factors,<br />

Hormones,<br />

AICAR<br />

Cytokines, etc.<br />

Stress<br />

Hypoxia<br />

PI3K<br />

Dvl<br />

DNA<br />

Damage<br />

PDK1<br />

Ras<br />

PTEN<br />

p53<br />

Autophagy<br />

eIF4G<br />

GATOR2<br />

54 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

4E-<br />

BP1/2<br />

mRNA<br />

Translation<br />

Proliferation<br />

LRP<br />

Erk<br />

RSK<br />

GSK-3<br />

REDD1/2<br />

AMPK<br />

Lipin 1<br />

Wnt<br />

Frizzled<br />

Gα q/o<br />

mTORC1<br />

Translocation<br />

to Lysosome<br />

Lipid Synthesis<br />

LKB1<br />

Sestrin-1/2<br />

LAMTOR<br />

1/2/3/4/5<br />

Ragulator<br />

Complex<br />

Rag A/B<br />

GTP<br />

Rag C/D<br />

GDP<br />

Ribosome<br />

Biogenesis<br />

TFEB<br />

PPARα<br />

HIF-1<br />

PGC-1α<br />

PPARγ<br />

SKAR<br />

mRNA Splicing<br />

metformin<br />

Glucose,<br />

Amino Acids<br />

V-ATPase<br />

Mios<br />

WDR59<br />

Lipogenesis<br />

SREBP-1<br />

Transcription<br />

Seh1L<br />

WDR24<br />

Sec13<br />

GATOR1<br />

DEPDC5 Nprl2<br />

Nprl3<br />

Lipid<br />

Metabolism<br />

FLCN<br />

FNIP1/2<br />

Autophagy/Lysosome<br />

Biogenesis<br />

VEGF/<br />

Angiogenesis<br />

Mitochondrial<br />

Metabolism<br />

Adipogenesis<br />

The mechanistic target of rapamycin (mTOR) is an atypical serine/threonine kinase that is present in two distinct complexes. The first, mTOR complex 1 (mTORC1), is<br />

composed of mTOR, Raptor, GβL, and DEPTOR and is inhibited by rapamycin. It is a master growth regulator that senses and integrates diverse nutritional and environmental<br />

cues, including growth factors, energy levels, cellular stress, and amino acids. It couples these signals to the promotion of cellular growth by phosphorylating substrates<br />

that potentiate anabolic processes such as mRNA translation and lipid synthesis, or limit catabolic processes such as autophagy. The small GTPase Rheb, in its GTP-bound<br />

state, is a necessary and potent stimulator of mTORC1 kinase activity, which is negatively regulated by its GAP, the tuberous sclerosis heterodimer TSC1/2. Most upstream<br />

inputs are funneled through Akt and TSC1/2 to regulate the nucleotide-loading state of Rheb. In contrast, amino acids signal to mTORC1 independently of the PI3K/Akt axis to<br />

promote the translocation of mTORC1 to the lysosomal surface where it can become activated upon contact with Rheb. This process is mediated by the coordinated actions of<br />

multiple complexes, notably the v-ATPase, Ragulator, the Rag GTPases, and GATOR1/2. The second complex, mTOR complex 2 (mTORC2), is composed of mTOR, Rictor, GβL,<br />

Sin1, PRR5/Protor-1, and DEPTOR. mTORC2 promotes cellular survival by activating Akt, regulates cytoskeletal dynamics by activating PKCα, and controls ion transport and<br />

growth via SGK1 phosphorylation. Aberrant mTOR signaling is involved in many disease states including cancer, cardiovascular disease, and diabetes.<br />

Select Reviews:<br />

Dowling, R.J., Topisirovic, I., Fonseca, B.D., and Sonenberg, N. (2010) Biochim. Biophys. Acta. 1804, 433–439. • Dunlop, E.A. and Tee, A.R. (2009) Cell. Signal. 21,<br />

827–835. • Hoeffer, C.A. and Klann, E. (2010) Trends Neurosci. 33, 67–75. • Laplante, M. and Sabatini, D.M. (2013) J. Cell Sci. 126, 1713–1719. • Laplante, M. and<br />

Sabatini, D.M. (2012) Cell 149, 274–293. • Neufeld, T.P. (2010) Curr. Opin. Cell Biol. 22, 157–168. • Zoncu, R., Efeyan, A., and Sabatini, D.M. (2011) Nat. Rev. Mol. Cell<br />

Biol. 12, 21–35.<br />

© 2002–2015 Cell Signaling Technology, Inc. • We would like to thank Rachel Wolfson and Prof. David Sabatini, Whitehead Institute for Biomedical Research, MIT, Cambridge, MA, for reviewing this diagram.<br />

Akt Substrates<br />

MAPK, mTOR, and the PI3K/Akt pathways are key signaling pathways activated downstream of oncogenic receptor tyrosine<br />

kinases (RTKs). All of these pathways activate AGC kinase family members, including Akt, RSK, and p70 S6 kinases, whose<br />

protein substrates are phosphorylated at the RxRxxS/T motif.<br />

In a phosphoproteomic study co-authored by scientists in the Cell Signaling Technology (<strong>CST</strong>) Site Discovery Group (Moritz, A.<br />

et al. (2010) Sci. Signal 24,ra64), over 300 novel downstream substrates for these AGC family kinases were identified. The<br />

experimental approach involved the use of PhosphoScan ® , <strong>CST</strong>’s proprietary methodology for antibody-based peptide enrichment<br />

combined with tandem mass spectrometry for quantitative profiling of post-translational modifications. A key step was<br />

the development of a RxRxxS/T motif antibody, which was then used as an affinity reagent to selectively immunoprecipitate<br />

phosphorylated substrates of Akt, RSK, and p70 S6 kinases. The antibody was employed in PhosphoScan in three different<br />

cancer cell lines, dependent on either EGFR, c-Met, or PDGFR, allowing mapping of the signaling network downstream of these<br />

RTKs. Substrates included proteins involved in many cellular functions, including scaffolding, protein stability, metabolism, trafficking,<br />

and motility.<br />

Substrate<br />

GAP/GEF/Adaptors<br />

ARHGAP19<br />

ARHGEF12<br />

AS250<br />

TBC1D1<br />

TBC1D4<br />

Receptors/Transporters<br />

DR6 SLC20A2<br />

EPHA2 SLC9A1<br />

FGFR2<br />

SEMA4B<br />

TSC2<br />

CD2AP<br />

FRS2<br />

IRS1<br />

IRS2<br />

Adhesion/Cytoskeleton<br />

DSP PLEC1<br />

MLLT4 SVIL<br />

KIF21A PPP1R12A<br />

AMPKA<br />

Isoform<br />

RICTOR<br />

CABLES1<br />

LMO7<br />

Kinases<br />

WNK1<br />

PKD2<br />

HGK<br />

BRD2<br />

UO126<br />

EGFR<br />

Gefitinib Su11274 Gleevec<br />

Ras<br />

MEK<br />

RSK<br />

Energy/<br />

Metabolism<br />

PANK2<br />

PFKFB2<br />

OXR1<br />

GSK-3α<br />

GSK-3β<br />

PI3K<br />

Akt<br />

Met<br />

SGKs<br />

mTOR<br />

Wort<br />

S6K<br />

PDGFRα<br />

RNA Processing/<br />

Translation<br />

LARP1<br />

MEPCE<br />

RPS6<br />

EIF4ENIF1<br />

EDC3<br />

Published Data Human<br />

Organism Site Site Sequence (+/-7) PMID<br />

Rapa<br />

Chaperone/Ubiquitin<br />

CCT2<br />

DNAJC2<br />

SGTA<br />

NEDD4-2<br />

UBR4<br />

UBXN4<br />

NIPA<br />

TIF1-γ<br />

Survival<br />

AKT1S1<br />

BAD<br />

NDRG2<br />

NDRG3<br />

Transcription<br />

FOXO3<br />

GTF3C1<br />

IWS1<br />

TAF3<br />

TCF12<br />

Vesicle Trafficking<br />

C4orf16 REPS1<br />

GOLGA4 STX12<br />

NDRG1 STX7<br />

HDGF2<br />

TCF3<br />

BRD1<br />

SP100<br />

Substrate Function and<br />

Effect of Phosphorylation<br />

14-3-3 z Akt1 human S58 S58 VVGARRSsWRVVssI 11956222 A key regulatory protein in signal transduction,<br />

checkpoint control, apoptotic,<br />

and nutrient-sensing pathways; effect<br />

of phosphorylation is unknown<br />

acinus Akt1 human S1180 S1180 GPRsRsRsRDRRRKE 18559500,<br />

16177823<br />

Akt1 rat S1329 S1331 HSRSRSRsTPVRDRG 16177823<br />

Induces chromatin condensation during<br />

apoptosis; phosphorylation inhibits<br />

this process<br />

ACLY Akt1 mouse S455 S455 PAPSRtAsFsESRAD 16007201 Catalyzes the formation of acetyl-CoA<br />

and oxaloacetate (OAA) in the cytosol;<br />

phosphorylation enhances the catalytic<br />

activity of the enzyme<br />

ADRB2 Akt1 human S346 S346 LLCLRRssLKAyGNG 11809767 A receptor that binds epinephrine<br />

and norepinephrine, acting as a<br />

neuromodulator in the central nervous<br />

system and as a hormone in the<br />

vascular system; phosphorylation in<br />

response to insulin stimulation leads to<br />

sequestration of ADBR2<br />

Akt1 Akt1 human S246,<br />

T72<br />

S246,<br />

T72<br />

LSRERVFsEDRARFY,<br />

TERPRPNtFIIRCLQ<br />

Akt1 mouse S473 S473 RPHFPQFsYsAsGtA 11570877,<br />

10722653<br />

16549426 Activated by insulin and various growth<br />

and survival factors to function in a<br />

wortmannin-sensitive PI3 kinaseinvolved<br />

pathway controlling survival<br />

and apoptosis; autophosphorylation<br />

activates the kinase<br />

AMPKA1 Akt1 rat S485 S485 ATPQRSGsISNYRSC 16340011 Heterotrimeric complex that plays a<br />

key role in the regulation of energy<br />

homeostasis; phosphorylation regulates<br />

AMPK activity<br />

AMPKA2 Akt1 rat S491 S491 STPQRSCsAAGLHRP 16340011 Heterotrimeric complex that plays a<br />

key role in the regulation of energy<br />

homeostasis; phosphorylation regulates<br />

AMPK activity<br />

www.cellsignal.com/csttables 55


Section I: Research Areas<br />

chapter 02: Signaling<br />

Substrate<br />

AMPKA<br />

Isoform<br />

Published Data Human<br />

Organism Site Site Sequence (+/-7) PMID<br />

Substrate Function and<br />

Effect of Phosphorylation<br />

APS Akt1 rat S588 S598 SARSRSNsTEHLLEA 16141217 An adaptor protein recruited to the insulin<br />

receptor to signal insulin-stimulated<br />

glucose transport; phosphorylation<br />

promotes membrane localization<br />

AR Akt1 human S213,<br />

S791<br />

S213,<br />

S791<br />

SGRAREAsGAPTsSK,<br />

CVRMRHLsQEFGWLQ<br />

11404460,<br />

14555644,<br />

17470458,<br />

11156376<br />

Nuclear receptor; phosphorylation<br />

suppresses AR activation, expression<br />

of AR target genes, and AR-mediated<br />

apoptosis<br />

arfaptin 2 Akt1 human S260 S260 GTRGRLEsAQATFQA 15809304 ADP ribosylation factor-interacting<br />

protein, implicated as a factor in<br />

Huntington's disease; phosphorylation<br />

promotes neuronal cell survival and<br />

neuroprotection<br />

ARH-<br />

GAP22<br />

Akt1 human S16 S16 ARRARSKsLVMGEQS 21969604 A Rho GTPase activator that inhibits<br />

Rac1; phosphorylation allows 14-3-3<br />

binding and regulation of cell motility<br />

AS160 Akt1 human T642 T642 QFRRRAHtFsHPPss 16880201,<br />

11994271,<br />

16935857<br />

ASK1<br />

Akt1,<br />

Akt2<br />

human S83 S83 ATRGRGssVGGGSRR 11154276,<br />

15782121,<br />

15911620,<br />

14500571,<br />

12697749<br />

ataxin-1 Akt1 human S775 S775 ATRKRRWsAPESRKL 17540008,<br />

12757707<br />

B-Raf<br />

Akt1,<br />

Akt3<br />

human S365,<br />

S429<br />

S365,<br />

S429<br />

GQRDRsssAPNVHIN,<br />

PQRERKsssSsEDRN<br />

10869359,<br />

18451171<br />

BAD Akt1 human S99 S99 PFRGRsRsAPPNLWA 11020382,<br />

10558990,<br />

19667065<br />

Akt1 mouse S112,<br />

S155<br />

Bcl-10 Akt1 human S218,<br />

S231<br />

S75,<br />

S118<br />

S218,<br />

S231<br />

ETRsRHssyPAGtEE,<br />

GRELRRMsDEFEGSF<br />

EEGTCANsSEMFLPL,<br />

PLRSRtVsRQ_____<br />

Insulin stimulated Rab GTPase-activating<br />

protein, structurally and functionally<br />

similar to TBC1D1; phosphorylation<br />

results in increased Glut4 translocation<br />

MAPKKK, induces apoptosis via JNK<br />

pathway; phosphorylation inhibits activity<br />

and promotes survival<br />

14-3-3 binds to and stabilizes<br />

ataxin-1, which forms polyglutamine<br />

aggregates and neurodegeneration;<br />

phosphorylation promotes 14-3-3<br />

binding<br />

Signaling intermediate in Erk1/2 pathway;<br />

phosphorylation causes inhibition<br />

Pro-apoptotic protein; phosphorylation<br />

inhibits function and promotes survival<br />

9381178,<br />

11723239,<br />

10983986,<br />

15123689,<br />

10949026<br />

16280327 A CARD (caspase recruitment<br />

domain) containing protein shown to<br />

induce apoptosis and activate NF-κB;<br />

phosphorylation induces nuclear<br />

translocation<br />

Bcl-xL Akt1 rat S106 S106 LRYRRAFsDLTSQLH 18951975 Prevents apoptosis through binding<br />

to apoptotic proteins; phosphorylation<br />

promotes VDAC binding<br />

Bex1 Akt1 rat S105 S102 KLRERQLsHSLRAVS 16498402 A neurotrophin and nerve growth factor<br />

signaling adaptor molecule involved<br />

in promoting cell cycle progression;<br />

phosphorylation prevents degradation<br />

by the proteasome<br />

Bim Akt1 human S87 S87 FIFMRRssLLSRSss 16282323 Pro-apoptotic protein; phosphorylation<br />

promotes 14-3-3 binding/inactivation<br />

and cell survival<br />

BRCA1 Akt1 human S694,<br />

T509<br />

BRF1 Akt1 human S92,<br />

S203<br />

S694,<br />

T509<br />

S92,<br />

S203<br />

QTSKRHDsDTFPELK,<br />

LKRKRRPtsGLHPED<br />

RFRDRsFsEGGERLL,<br />

PRLQHsFsFAGFPSA<br />

20085797,<br />

10542266<br />

17030608,<br />

15538381<br />

Breast cancer susceptibility gene product,<br />

tumor suppressor; phosphorylation<br />

alters function, perhaps by preventing<br />

nuclear localization<br />

A CCCH zinc-finger protein that binds<br />

to AU-rich elements (ARE) found in the<br />

3'-untranslated regions of mRNAs and<br />

promotes de-adenylation and rapid<br />

degradation by the exosome; phosphorylation<br />

results in binding by 14-3-<br />

3 protein and inactivation of BRF1<br />

CACNB2 Akt1 rat S625 S630 KQRSRHKsKDRYCDK 15311280 Voltage-dependent calcium channel;<br />

phosphorylation regulates channel<br />

trafficking to plasma membrane<br />

CaRHSP1 Akt1 human S52 S52 tRRtRtFsAtVRASQ 15910284 RNA binding protein; phosphorylation<br />

effect currently unknown<br />

Substrate<br />

AMPKA<br />

Isoform<br />

Published Data Human<br />

Organism Site Site Sequence (+/-7) PMID<br />

Substrate Function and<br />

Effect of Phosphorylation<br />

Casp9 Akt1 human S196 S196 KLRRRFssLHFMVEV 9812896 Protease, initiates apoptosis; phosphorylation<br />

inhibits protease activity<br />

CBP Akt1 mouse T1872 T1871 LMRRRMAtMNTRNVP 17166829 Acetylates histone and non-histone<br />

proteins; phosphorylation increases<br />

activity<br />

CBY1<br />

Akt1,<br />

Akt2<br />

human S20 S20 TPPRKSAsLSNLHsL 18573912 An inhibitor of the Wnt signaling pathway;<br />

phosphorylation allows 14-3-3<br />

binding and β-catenin sequestration in<br />

the cytoplasm<br />

CCT2 Akt1 human S260 S260 GsRVRVDstAkVAEI 19332537 Member of the protein chaperone<br />

complex; effect of phosphorylation<br />

currently unknown<br />

CD34 Akt2 mouse S343 S346 yssGPGAsPETQGKA 21499536 A type I transmembrane glycophosphoprotein<br />

expressed by hematopoietic<br />

stem/progenitor cells, vascular<br />

endothelium and some fibroblasts as<br />

a negative regulator of cell adhesion;<br />

effect of phosphorylation currently<br />

unknown<br />

Cdc25B Akt1 mouse S351 S353 VQSKRRKsVtPLEEQ 17554083 Protein phosphatase responsible<br />

for cdc2 activation; phosphorylation<br />

promotes activation of M-phase<br />

promoting factor<br />

CDK2 Akt1 human T39 T39 LkKIRLDtETEGVPs 18354084 Cyclin-dependent kinase functioning<br />

in S-phase; phosphorylation increases<br />

cyclin A binding<br />

CELF1 Akt1 human S28 S28 GQVPRTWsEKDLREL 18570922 RNA-binding protein; phosphorylation<br />

enhances interaction with cyclin D1<br />

mRNA<br />

CENTB1 Akt1 human S554 S554 SIRPRPGsLRSKPEP 16256741 GTPase-activating protein (GAP) for<br />

ARF proteins; phosphorylation prevents<br />

recycling of b1-integrin containing<br />

endosomes and cell migration<br />

CENTG1 Akt1 human S985 S985 THLSRVRsLDLDDWP 19176382 A GTPase activating protein for ARF1<br />

and ARF5; phosphorylation enhances<br />

CENTG1 GTP binding and NF-κB<br />

activity<br />

CFLAR Akt1 human S273 S273 LLRDTFTsLGYEVQK 19339247 A regulator of apoptosis; phosphorylation<br />

targets CFLAR for degradation<br />

Chk1 Akt1 human S280 S280 AKRPRVtsGGVsEsP 15107605,<br />

12062056<br />

DNA damage effector that regulates<br />

G2/M transition during DNA damage;<br />

phosphorylation inhibits function by<br />

preventing phosphorylation by ATM/ATR<br />

CK1-D Akt1 rat S370 S370 MERERKVsMRLHRGA 17594292 Kinase and core component of circadian<br />

clock; phosphorylation inhibits<br />

kinase activity<br />

CLK2 Akt1 human S34,<br />

T127<br />

S34,<br />

T127<br />

HKRRRSRsWSSSSDR,<br />

RRRRRSRtFSRSSSQ<br />

20682768 A dual specificity serine/threonine and<br />

tyrosine kinase; phosphorylation<br />

increases cell survival after ionizing<br />

radiation<br />

Cot Akt1 human S400 S400 EDQPRCQsLDSALLE 12138205 Oncogene; phosphorylation induces<br />

NF-κB-dependent transcription<br />

CREB Akt1 rat S133 S133 EILsRRPsYRkILND 9829964 bZIP transcription factor that activates<br />

target genes through cAMP response<br />

elements; activated by phosphorylation<br />

CTNNB1<br />

Akt1,<br />

Akt2<br />

human S552 S552 QDtQRRtsMGGtQQQ 17287208 Wnt signaling pathway protein; phosphorylation<br />

causes nuclear localization<br />

CTNND2 Akt1 mouse T454 T457 tGTFRtstAPssPGV 17993462 Transcriptional activator, plays a role<br />

in adhesion molecule regulation;<br />

phosphorylation promotes binding to<br />

p190RhoGEF, dendritic morphogenesis<br />

Cx43 Akt1 rat S369 S369 RPssRAssRAssRPR 18163231 Gap junction protein; phosphorylation<br />

Akt1,<br />

Akt3<br />

rat S373 S373 RAssRAssRPRPDDL 17008717,<br />

18163231<br />

allows 14-3-3 binding<br />

DLC1 Akt1 rat S330 S766 VTRTRSLsTCNKRVG 16338927 Tumor suppressor and insulin stimulated<br />

phosphoprotein, may play role in<br />

Glut4 translocation; phosphorylation<br />

may inhibit its GAP activity<br />

DNAJC5 Akt1 rat S10 S10 DQRQRsLsTSGESLY 16243840 Exocytosis; phosphorylation regulates<br />

the kinetics of late stage exocytosis<br />

Akt Signaling<br />

Research<br />

Please visit our website for additional<br />

resources and products relating to the<br />

study of Akt Signaling.<br />

www.cellsignal.com/cstakt<br />

56 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables<br />

57


Section I: Research Areas<br />

chapter 02: Signaling<br />

Substrate<br />

AMPKA<br />

Isoform<br />

Published Data Human<br />

Organism Site Site Sequence (+/-7) PMID<br />

Substrate Function and<br />

Effect of Phosphorylation<br />

DNMT1 Akt1 human S143 S143 RtPRRsksDGEAkPE 21151116 A maintenance methyltransferase,<br />

transferring proper methylation<br />

patterns to newly synthesized DNA<br />

during replication; phosphorylation<br />

increases DNMT1 stability and prevents<br />

methylation<br />

EDC3<br />

Akt1,<br />

Akt2<br />

human S161 S161 sFRRRHNsWssSsRH 20051463 Involved in removal of the mRNA 5’ cap<br />

structure; phosphorylation induces 14-<br />

3-3 protein interaction and promotes<br />

ED3 mediated post-transcriptional<br />

regulation through mRNA<br />

EDG-1 Akt1 human T236 T236 RTRSRRLtFRKNISK 11583630 G protein-coupled receptor; phosphorylation<br />

activates signaling to promote<br />

cell migration<br />

eIF4B Akt1 mouse S422 S422 RERsRtGsEssQtGA 18836482 Necessary for binding of mRNA to<br />

ribosomes; phosphorylation increases<br />

transcriptional activity<br />

ENaC-a Akt1 rat S621 S594 RFRSRYWsPGRGARG 21220922 An amiloride sensitive epithelial sodium<br />

channel that mediates sodium reabsorption;<br />

phosphorylation increases<br />

ENaC specific activity<br />

eNOS Akt1 human S615,<br />

S1177<br />

S615,<br />

S1177<br />

SYKIRFNsISCSDPL,<br />

TsRIRtQsFsLQERQ<br />

12511559,<br />

12446767,<br />

10376603,<br />

18622039,<br />

12171920<br />

Enzyme that catalyzes the production<br />

of nitric oxide (NO); phosphorylation<br />

results in enzyme activation,<br />

NO production, and cardiovascular<br />

homeostasis (vasodilation, vascular<br />

remodeling, angiogenesis)<br />

EphA2 Akt1 human S897 S897 RVsIRLPstsGsEGV 19573808 Receptor tyrosine kinase that binds<br />

to a GPI-anchored ephrin A ligand for<br />

regulation of cell adhesion, cell migration,<br />

axon guidance, and homeostasis;<br />

phosphorylation regulates EphA2<br />

induced cell migration and invasion<br />

ER-a<br />

Akt1,<br />

Akt2<br />

human S167 S167 GGRERLAsTNDKGSM 11139588,<br />

16113102,<br />

11507039<br />

Akt1 human S305 S305 IkRSkkNsLALSLtA 20101208<br />

Nuclear receptor and transcription<br />

factor; phosphorylation activates the<br />

receptor and increases gene expression,<br />

causing mammary and uterine<br />

cell proliferation<br />

ER-b Akt1 mouse S236 D236 VRRQRSAsEQVHCLN 17166829 Nuclear receptor and transcription factor;<br />

phosphorylation prevents cofactor<br />

binding and decreases activity<br />

EZH2 Akt1 human S21 S21 CWRKRVKsEYMRLRQ 16224021 Methyltransferase; phosphorylation<br />

decreases histone H3 methylation of<br />

Lys27 and increases gene expression<br />

ezrin Akt2 human T567 T567 QGRDKYKtLRQIRQG 15531580 Plasma membrane/cytoskeletal linker<br />

protein; phosphorylation promotes actin<br />

binding and cytoskeletal organization<br />

FANCA Akt1 human S1149 S1149 CLRSRDPsLMVDFIL 11855836 ATPase involved in DNA repair;<br />

phosphorylation is negatively regulated<br />

by Akt<br />

FLEG1 Akt1 human S486 S486 GLEtRRLsLPSsKAK 17256767 A chaperone protein involved in directing<br />

specific histones to the centromere;<br />

phosphorylation allows binding to<br />

14-3-3<br />

FLNC<br />

Akt1,<br />

Akt2<br />

human S2233 S2233 LGRERLGsFGsItRQ 15461588 Muscle-specific filamin functioning in<br />

muscle cells; phosphorylation effect<br />

currently unknown<br />

FOXA2 Akt1 human T156 T156 KTYRRSYtHAKPPYS 14500912 Transcription factor involved in embryonic<br />

development and differentiation;<br />

phosphorylation results in nuclear<br />

exclusion and inhibition of FoxA2-<br />

dependent transcriptional activity<br />

FOXG1 Akt1 human T279 T279 KLRRRSttsRAKLAF 17435750 Transcriptional repression factor<br />

involved in brain development; phosphorylation<br />

promotes nuclear export<br />

FOXO1A Akt1 human S256,<br />

S319,<br />

T24<br />

S256,<br />

S319,<br />

T24<br />

sPrRrAAsMDNNSkF,<br />

TFRPRtssNAsTIsG,<br />

LPRPRSCtWPLPRPE<br />

15668399,<br />

10358075,<br />

11237865,<br />

16076959,<br />

11311120<br />

Transcription factor involved in cell cycle<br />

arrest, apoptosis, and glucose metabolism;<br />

phosphorylation causes export<br />

from the nucleus and inhibits activity<br />

AMPKA Published Data<br />

Substrate Isoform<br />

FOXO3A Akt1 human S253,<br />

T32<br />

FOXO4 Akt1 human S197,<br />

S262,<br />

T32<br />

Human<br />

Organism Site Site Sequence (+/-7) PMID<br />

S253,<br />

T32<br />

S197,<br />

S262,<br />

T32<br />

APRRRAVsMDNSNKY,<br />

QSRPRsCtWPLQRPE<br />

APRRRAAsMDSSSKL,<br />

TFRPRSssNASSVST,<br />

QSRPRsCtWPLPRPE<br />

10910908,<br />

10995739,<br />

10102273,<br />

11154281<br />

11313479,<br />

11313479,<br />

10217147,<br />

16272144<br />

Substrate Function and<br />

Effect of Phosphorylation<br />

Transcription factor involved in cell<br />

cycle arrest and apoptosis; phosphorylation<br />

causes export from the nucleus<br />

and inhibits activity<br />

Transcription factor involved in cell<br />

cycle arrest, apoptosis, and insulin signaling;<br />

phosphorylation causes export<br />

from the nucleus and inhibits activity<br />

Gab2 Akt1 human S159 S159 LLRERKSsAPSHsSQ 11782427 Docking/scaffolding protein, protooncogene,<br />

RTK signaling intermediate;<br />

phosphorylation inhibits activity<br />

GABRB2 Akt1 rat S434 S434 SRLRRRAsQLKITIP 12818177 Receptor that mediates fast inhibitory<br />

synaptic transmission in the brain;<br />

phosphorylation increases the number<br />

of receptors on the cell surface<br />

GAPDH Akt2 human T237 T237 GMAFRVPtANVSVVD 21979951 Catalyzes the phosphorylation of<br />

glyceraldehyde-3-phosphate during<br />

glycolysis; phosphorylation decreases<br />

nuclear translocation and GAPDH<br />

induced apoptosis<br />

GATA1 Akt1 human S310 S310 QTRNRKAsGkGkkkR 16107690 Transcription factor; phosphorylation<br />

increases activity and promotes blood<br />

cell differentiation<br />

GATA2 Akt1 human S401 S401 QTRNRKMsNKSKKSK 15837948 Transcription factor; phosphorylation<br />

inhibits activity to promote adipogenesis<br />

and reduce inflammation<br />

girdin Akt1 human S1417 S1417 INRERQKsLtLTPTR 16139227 Actin binding protein; phosphorylation<br />

promotes cell migration<br />

GOLGA3 Akt1 mouse S174,<br />

S385<br />

S174,<br />

S389<br />

VKRHRERsSQPAtKM,<br />

EVRsRRDsICsSVSM<br />

17888676 Golgi auto-antigen; phosphorylation<br />

results in reduced apoptosis<br />

Grb10 Akt1 mouse S455 S428 NAPMRsVsENsLVAM 15722337 An adaptor protein that interacts with<br />

many receptor tyrosine kinases as well<br />

as downstream signal molecules; phosphorylation<br />

allows binding to 14-3-3<br />

GSK-3a Akt1 human S21 S21 SGRARtssFAEPGGG 11340086,<br />

11563975,<br />

11577096<br />

GSK-3b Akt1 human S9 S9 SGRPRttsFAESCKP 12900420,<br />

15457186,<br />

11563975,<br />

11340086,<br />

11577096,<br />

8985174<br />

Serine/threonine protein kinase<br />

that phosphorylates and inactivates<br />

glycogen synthase; phosphorylation<br />

inhibits activity<br />

Serine/threonine protein kinase<br />

that phosphorylates and inactivates<br />

glycogen synthase; phosphorylation<br />

inhibits activity<br />

H2B Akt1 human S37 S37 RKRsRkEsysIyVyk 8985174 Core component of the nucleosome;<br />

phosphorylation effect currently<br />

unknown<br />

H3 Akt1 mouse S10 S10 tKQTARksTGGkAPR 12529330 Core component of the nucleosome;<br />

phosphorylation is correlated with<br />

chromosome condensation during<br />

mitosis and meiosis<br />

HMOX1 Akt1 human S188 S188 LYRSRMNsLEMtPAV 15581622 Heme oxygenase involved in stress<br />

response; phosphorylation regulates<br />

binding affinity<br />

hnRNP A1 Akt1 human S199 S199 sQrGrsGsGNFGGGr 18562319 Involved in pre-mRNA packaging<br />

into hnRNP particles and transport<br />

of poly(A) mRNA from cytoplasm to<br />

nucleus; phosphorylation regulates role<br />

in cyclin D1 and c-Myc IRES activity<br />

hnRNP E1 Akt1,<br />

Akt2<br />

mouse S43 S43 VKRIREEsGARINIS 20154680 Binds to single-stranded nucleic acid;<br />

phosphorylation results in disruption of<br />

BAT element binding and translational<br />

activation of Dab2 and ILEI mRNA<br />

HSP27 Akt1 human S82 S82 RALsRQLssGVSEIR 12740362 Heat shock protein that confers cellular<br />

resistance to stress and adverse<br />

environmental change; phosphorylation<br />

alters tertiary structure, modulates<br />

actin polymerization, and reorganization<br />

HTRA2<br />

Akt1,<br />

Akt2<br />

human S212 S212 RVRVRLLsGDTYEAV 17311912 Protease released during apoptosis;<br />

phosphorylation inhibits activity and<br />

attenuates its pro-apoptotic function<br />

58 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables 59


Section I: Research Areas<br />

chapter 02: Signaling<br />

Substrate<br />

Huntingtin<br />

IKK-a<br />

AMPKA<br />

Isoform<br />

Published Data Human<br />

Organism Site Site Sequence (+/-7) PMID<br />

Akt1 human S421 S421 GGRsRsGsIVELIAG 12062094,<br />

14725621,<br />

15843398,<br />

16452687<br />

Akt1,<br />

Akt2<br />

human T23 T23 EMRERLGtGGFGNVC 18515365,<br />

12048203,<br />

10485710,<br />

19609947<br />

Substrate Function and<br />

Effect of Phosphorylation<br />

Huntington’s disease; Akt phosphorylation<br />

blocks nuclear aggregation and<br />

provides neuroprotection<br />

NF-κB signaling intermediate;<br />

phosphorylation activates NF-κB and<br />

immune/stress response<br />

IP3R1 Akt1 rat S2682 S2690 FPRMRAMsLVSSDSE 16332683 Ca 2+ release and signaling; phosphorylation<br />

induces resistance to apoptosis,<br />

possibly through caspase-3 inactivation<br />

IRAK1 Akt1 human T100 T100 LRARDIItAWHPPAP 11976320 A serine/threonine-specific IL-1<br />

receptor-associated kinase involved in<br />

Toll signaling; phosphorylation inhibits<br />

IRAK mediated NK-κB activation<br />

IRS1 Akt1 human S629 S629 VPSGRKGsGDyMPMs 17640984 Insulin receptor signaling intermediate;<br />

Akt1 rat S522 S527 RFRKRTHsAGTSPTI 17579213<br />

phosphorylation inhibits function<br />

KHSRP<br />

Kv11.1<br />

iso5<br />

Lamin<br />

A/C<br />

Akt1,<br />

Akt2<br />

human S193 S193 GLPERSVsLTGAPES 17177604 Recruits degradation machinery,<br />

activates mRNA turnover, regulates<br />

splicing; phosphorylation inhibits RNA<br />

turnover by degradation<br />

Akt1 human T897 T897 SFRRRtDtDtEQPGE 18791070 Pore-forming subunit of voltage-gated<br />

potassium channels, essential for<br />

rhythmic excitability of cardiac muscle<br />

and endocrine cells; phosphorylation<br />

inhibits channels<br />

Akt1 rat S301,<br />

S404<br />

S301,<br />

S404<br />

RSRGRASsHSSQSQG 18808171 Component of nuclear lamina;<br />

phosphorylation regulates function of<br />

nuclear lamina<br />

LTB4R2 Akt1 human T355 T355 GGRsREGtMELRTTP 22044535 A low-affinity leukotriene receptor<br />

involved in chemotaxis; phosphorylation<br />

regulates activation of chemotactic<br />

responses<br />

Mad1 Akt1 human S145 S145 IERIRMDsIGSTVSS 18451027,<br />

19526459<br />

MDM2 Akt1 human S166,<br />

S186,<br />

S188<br />

S166,<br />

S186,<br />

S188<br />

SsRRRAIsEtEENsD,<br />

RQRKRHKsDsIsLsF,<br />

RKRHKsDsIsLsFDE<br />

11715018,<br />

15169778,<br />

11504915,<br />

11850850,<br />

11923280,<br />

15527798,<br />

11960368<br />

Component of spindle-assembly<br />

checkpoint; phosphorylation results in<br />

ubiquitination and degradation through<br />

26S proteasome pathway<br />

Ubiquitin ligase involved in p53<br />

degradation; phosphorylation results<br />

in translocation to the nucleus and<br />

inhibition of p53<br />

MDM4 Akt1 human S367 S367 PDCRRtIsAPVVRPK 18356162 RING-finger domain protein involved<br />

in p53 degradation and apoptosis;<br />

phosphorylation stabilizes MDM4 and<br />

MDM2<br />

METTL1 Akt1 human S27 S27 yYRQrAHsNPMADHT 15861136 Catalyzes the formation of m7G46<br />

in tRNA; phosphorylation results in<br />

inactivation<br />

MKK4 Akt1 human S80 S80 IERLRtHsIEsSGKL 15911620,<br />

11707464<br />

Signaling intermediate of the JNK/<br />

SAPK pathway involved in stress/<br />

inflammation; phosphorylation inhibits<br />

activity<br />

MLK3 Akt1 human S674 S674 PGRERGEsPTtPPTP 12458207 JNK-mediated neuronal cell death;<br />

phosphorylation inhibits activity<br />

MST1 Akt1 human T120 T120 IIRLRNktLTEDEIA 19940129 Pro-apoptotic kinase; phosphorylation<br />

inhibits kinase activity and nuclear<br />

translocation resulting in inhibition of<br />

pro-apoptotic signaling<br />

MST2 Akt1 human T117,<br />

T384<br />

mTOR Akt1 human T2446,<br />

S2448<br />

T117,<br />

T384<br />

T2446,<br />

S2448<br />

IIRLRNktLIEDEIA,<br />

GTMKRNAtsPQVQRP<br />

20231902,<br />

20086174<br />

RsRtRtDsysAGQsV 15208671,<br />

10910062,<br />

10567225<br />

Upstream activator of the MAPK<br />

pathway that regulates apoptosis,<br />

morphogenesis, and cytoskeletal rearrangements;<br />

phosphorylation inhibits<br />

pro-apoptotic activity<br />

Protein synthesis and cell growth;<br />

phosphorylation increases activity<br />

Substrate<br />

AMPKA<br />

Isoform<br />

Published Data Human<br />

Organism Site Site Sequence (+/-7) PMID<br />

Substrate Function and<br />

Effect of Phosphorylation<br />

MYO5A Akt2 mouse S1650 S1652 GLRKRtssIADEGty 17515613 Actin-based motor protein with a role<br />

in cytoplasmic vesicle transport and<br />

anchorage; phosphorylation promotes<br />

insulin-mediated Glut4 vesicle<br />

translocation<br />

Myt1 Akt1 starfish S75 S83 ESRPRAVsFRQSEPS 11802161 Wee1 family member and cell cycle<br />

regulator; phosphorylation downregulates<br />

Myt1 and initiates M-phase<br />

NDRG2 Akt1 human S332,<br />

T348<br />

S332,<br />

T348<br />

LsRsRtAsLtsAAsV,<br />

GNRsRsRtLsQssEs<br />

NFAT90 Akt1 human S647 S647 rGrGRGGsIRGRGRG 18097023,<br />

20870937<br />

NHE1 Akt1 human S648,<br />

S703,<br />

S796<br />

S648,<br />

S703,<br />

S796<br />

KTRQRLRsyNRHTLV,<br />

MsRARIGsDPLAyEP,<br />

QRIQRCLsDPGPHPE<br />

15461589 Insulin-stimulated phosphoprotein;<br />

phosphorylation promotes insulin<br />

signaling<br />

18757828,<br />

20026127<br />

Translation inhibitory protein; phosphorylation<br />

required for nuclear export<br />

Sodium/hydrogen exchanger involved<br />

in pH regulation and signal transduction;<br />

phosphorylation inhibits activity<br />

NMDAR2C Akt1 mouse S1084 S1081 GPRPRHAsLPSSVAE 19477150 Glutamate receptor channel subunit;<br />

Akt1 rat S1083 S1081 GPRPRHAsLPSSVAE 19477150<br />

phosphorylation promotes binding<br />

to 14-3-3ε and leads to increased<br />

surface expression of cerebellar NMDA<br />

receptors<br />

NuaK1 Akt1 human S600 S600 PARQRIRsCVSAENF 15060171,<br />

12409306<br />

Nur77 Akt1 human S351 S351 GRRGRLPsKPKQPPD 16434970,<br />

11274386<br />

p21 Cip1 Akt1 human S146,<br />

T145<br />

p27Kip1 Akt1 human S10,<br />

T157,<br />

T198<br />

S146,<br />

T145<br />

S10,<br />

T157,<br />

T198<br />

GRkRRQtsMTDFYHs,<br />

QGRkRRQtsMTDFYH<br />

NVRVsNGsPsLErMD,<br />

GIRkrPAtDDSSTQN,<br />

PGLRRRQt_______<br />

AMPK family member activated under<br />

glucose starvation that mediates cell<br />

survival; phosphorylation increases<br />

kinase activity<br />

A nuclear receptor and transcription<br />

factor regulating T cell apoptosis;<br />

phosphorylation inhibits transcriptional<br />

activity<br />

17855660, Regulates cell cycle and cell survival;<br />

11231573, phosphorylation increases protein<br />

11756412, stability<br />

15173090,<br />

11463845,<br />

116982699<br />

18710949,<br />

12042314,<br />

12244302<br />

p300 Akt1 human S1834 S1834 MLRRRMAsMQRTGVV 16024795,<br />

11116148<br />

p47phox Akt1 human S304,<br />

S328<br />

S304,<br />

S328<br />

GAPPRRssIRNAHSI,<br />

QDAYRRNsVRFLQQR<br />

A cyclin-dependent kinase inhibitor that<br />

enforces the G1 cell cycle restriction<br />

point; phosphorylation promotes 14-3-<br />

3 binding and cytoplasmic localization<br />

Transcriptional co-activator; phosphorylation<br />

can either activate or suppress<br />

transcriptional activity depending on<br />

cell type and physiological stimuli<br />

12734380 A component of the phagocytic NADPH<br />

oxidase multiprotein enzyme that<br />

catalyzes the reduction of oxygen to<br />

superoxide in response to pathogenic<br />

invasion; phosphorylation regulates<br />

p47hox respiratory burst activity<br />

PAK1 Akt1 mouse S21 S21 APPMRNTsTMIGAGS 14585966 A p21-activated kinase engaged in<br />

cytoskeletal reorganization, MAPK signaling,<br />

apoptotic signaling, control of<br />

phagocyte NADPH oxidase, and growth<br />

factor-induced neurite outgrowth;<br />

phosphorylation at Ser21 regulates<br />

binding with the adaptor protein Nck<br />

palladin Akt1 human S1118 S1118 VRRPRsRsRDsGDEN 20471940 Actin-bundling protein; phosphorylation<br />

promotes F-actin bundling and inhibits<br />

cell migration<br />

PAR-4 Akt1 rat S249 N257 SRHNRDTsAPANFAS 16209943 A pro-apoptotic factor that activates<br />

the Fas-FADD-caspase-8 pathway as<br />

well as inhibits the NF-κB pro-survival<br />

pathway; phosphorylation prevents<br />

nuclear translocation, promoting cell<br />

survival<br />

PDCD4 Akt1 human S67,<br />

S457<br />

PDE3A Akt1 mouse S290,<br />

S291,<br />

S292<br />

S67,<br />

S457<br />

S290,<br />

S291,<br />

S292<br />

kRRLRKNssRDsGRG,<br />

RGRKRFVsEGDGGRL<br />

GWKRRRRsssVVAGE,<br />

WKRRRRsssVVAGEM,<br />

KRRRRsssVVAGEMS<br />

16357133 Tumor suppressor protein that is<br />

strongly induced during apoptosis;<br />

phosphorylation inhibits tumor suppressor<br />

function<br />

17124499 Regulates levels of cAMP and cGMP,<br />

insulin-dependent oocyte maturation;<br />

phosphorylation increases activity<br />

Akt Signaling<br />

Research<br />

Please visit our website for additional<br />

resources and products relating to the<br />

study of Akt Signaling.<br />

www.cellsignal.com/cstakt<br />

60 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables<br />

61


Section I: Research Areas<br />

chapter 02: Signaling<br />

Substrate<br />

AMPKA<br />

Isoform<br />

Published Data Human<br />

Organism Site Site Sequence (+/-7) PMID<br />

Substrate Function and<br />

Effect of Phosphorylation<br />

PDE3B Akt1 mouse S273 S295 VIRPRRRssCVsLGE 10454575 Regulates levels of cAMP and cGMP,<br />

activated by insulin to regulate lipolysis;<br />

phosphorylation increases activity<br />

PEA-15 Akt1 human S116 S116 KDIIRQPsEEEIIKL 12808093 A phosphoprotein shown to coordinate<br />

cell growth, death, and glucose utilization;<br />

phosphorylation mediates binding<br />

to FADD or Erk and further regulates<br />

the Erk and apoptosis signaling<br />

pathways<br />

peripherin Akt1 mouse S66 S59 SSSARLGsFRAPRAG 17569669 Neuronal intermediate filament protein;<br />

phosphorylation promotes motor nerve<br />

regeneration<br />

PFKFB2 Akt1 human S466,<br />

S483<br />

S466,<br />

S483<br />

PVRMRRNsFtPLSSS,<br />

IRRPRNysVGSRPLK<br />

12853467 Glycolytic enzyme, insulin-mediated<br />

glucose metabolism; phosphorylation<br />

increases activity<br />

PFKFB3 Akt1 human S461 S461 NPLMRRNsVtPLAsP 15896703 Synthesis and degradation of fructose<br />

2,6-bisphosphate; phosphorylation<br />

decreases sensitivity to inhibition<br />

PGC-1 a<br />

Akt1,<br />

Akt2<br />

mouse S570 S571 RMRSRsRsFsRHRSC 17554339 Regulates gluconeogenesis and fatty<br />

acid oxidation; phosphorylation inhibits<br />

function<br />

PIP5K Akt1 human S307 S307 PARNRsAsItNLsLD 15546921 A protein/ lipid kinase involved in<br />

Akt1 mouse S105 S105 EELHRRSsVLENTLP 20513353<br />

membrane trafficking; phosphorylated<br />

in response to insulin<br />

PLB Akt1 rat S16 S16 RSAIRRAstIEMPQQ 18838385 A major phosphoprotein calcium regulation<br />

component of the sarcoplasmic<br />

reticulum; phosphorylation causes<br />

release of inhibition and increases<br />

calcium uptake by the sarcoplasmic<br />

reticulum<br />

PLCG1 Akt1 human S1248 S1248 HGRAREGsFEsRyQQ 16525023 Catalyzes PI 4,5 bisphosphate to IP 3<br />

and DAG, increases intracellular Ca 2+<br />

levels; phosphorylation increases<br />

activity and enhances EGF-stimulated<br />

cell motility<br />

PPP1CA Akt1 human T320 T320 NPGGRPItPPRNSAK 14633703 A serine/threonine phosphatase<br />

involved in cell cycle regulation;<br />

phosphorylation inhibits activity<br />

PRAS40 Akt1 human T246 T246 LPRPRLNtsDFQKLK 12524439,<br />

17277771,<br />

18372248<br />

Binds to and inhibits mTOR; phosphorylation<br />

causes 14-3-3 binding/<br />

inhibition and results in increased<br />

protein synthesis<br />

PRPF19 Akt1 human T193 T193 ERKKRGKtVPEELVK 20629186 A member of the splicesome that also<br />

functions in DNA double strand break<br />

repair; phosphorylation allows 14-3-3<br />

binding<br />

PRPK Akt1 human S250 S250 RLRGRKRsMVG____ 17712528 p53 binding protein and kinase;<br />

phosphorylation causes activation and<br />

results in p53 phosphorylation<br />

PTP1B Akt1 human S50 S50 RNRyRDVsPFDHsRI 11579209 Protein tyrosine phosphatase that<br />

dephosphorylates the insulin receptor;<br />

phosphorylation inhibits activity<br />

QIK Akt2 mouse S358 S358 DGRQRRPstIAEQTV 17805301 AMPK related protein; phosphorylation<br />

leads to kinase activation and promotes<br />

ubiquitination/degradation of TORC2<br />

Rac1 Akt1 human S71 S71 yDRLRPLsYPQTDVF 10617634 Rho-GTPase, actin cytoskeletal<br />

organization; phosphorylation inhibits<br />

GTP-binding activity<br />

Raf1 Akt1 mouse S259 S259 SQRQRStsTPNVHMV 12087097,<br />

12087097<br />

Akt1 rat S259 S259 SQRQRSTsTPNVHMV 11443134<br />

Signaling intermediate in Erk1/2 pathway;<br />

phosphorylation inhibits activity<br />

RANBP3 Akt1 human S126 S126 VKRERtssLtQFPPs 18280241 RAN binding protein 3 functions in<br />

nuclear transport; phosphorylation<br />

mediates Ran binding and regulates<br />

nuclear transport<br />

RARA Akt1 human S96 S96 FVCQDKSsGYHYGVS 16417524 Nuclear receptor for retinoic acid that<br />

acts as a direct regulator of gene<br />

expression, phosphorylation of the DNA<br />

binding domain inhibits RARA activity<br />

Substrate<br />

AMPKA<br />

Isoform<br />

Published Data Human<br />

Organism Site Site Sequence (+/-7) PMID<br />

Substrate Function and<br />

Effect of Phosphorylation<br />

RGC32 Akt1 human S65 S65 ERMKRRSsAsVSDSS 19162005 A regulator of cell cycle-specific<br />

kinases in response to DNA damage;<br />

phosphorylation leads to activation and<br />

regulation of growth factors<br />

RNF11 Akt1 human T135 T135 DWLMRSFtCPSCMEP 16123141 A member of a ubiquitin editing<br />

complex that modulates transient<br />

inflammatory signaling; phosphorylation<br />

allows 14-3-3 binding<br />

Ron Akt1 human S1394 S1394 VRRPRPLsEPPRPT_ 12919677,<br />

14505491<br />

Receptor tyrosine kinase for macrophage<br />

stimulating protein (MSP), cell<br />

adhesion, proliferation and migration;<br />

phosphorylation causes 14-3-3 binding<br />

RPS3 Akt1 human T70 T70 GrrIrELtAVVQkRF 20605787 A member of the 40S ribosomal subunit<br />

that also induces neuronal apoptosis<br />

and acts as an endonuclease;<br />

phosphorylation inhibits proapoptotic<br />

function, increases nuclear import/accumulation,<br />

and increases DNA repair<br />

S6 Akt1 mouse S236 S236 AKRRRLssLRAstsK 12151408 S6 ribosomal protein; phosphorylation<br />

Akt1, rat S235, S235, IAKRRRLssLRAsts, 15358595<br />

activates the protein and promotes<br />

protein synthesis<br />

Akt2<br />

S236 S236 AKRRRLssLRAstsK<br />

SFRS5 Akt2 rat S86 S86 GRGRGRYsDRFSSRR 15684423 A member of the splicesome involved<br />

in constitutive and alternative splicing;<br />

phosphorylation activates alternative<br />

splicing exon inclusion<br />

SH3BP4 Akt1 mouse S245 S246 FRSKRSysLsELsVL 19122209 Controls selective internalization of the<br />

transferrin receptor through endocytosis;<br />

phosphorylation promotes 14-3-3<br />

binding at the plasma membrane<br />

SH3RF1<br />

Akt1,<br />

Akt2<br />

human S304 S304 KNTKKRHsFtsLTMA 17535800 Scaffolding protein that binds to<br />

activated Rac and promotes apoptosis<br />

via JNK activation; phosphorylation<br />

reduces ability to bind Rac, promoting<br />

apoptosis<br />

SKI Akt1 human T458 T458 QPRKRKLtVDTPGAP 19875456 Negative regulator of TGF-b signaling<br />

by binding to Smads; phosphorylation<br />

causes its destabilization and reduces<br />

SKI-mediated inhibition of expression<br />

of Smad7<br />

SOX2 Akt1 mouse T118 T116 kYRPRRktkTLMkKD 20945330 A transcription factor required for early<br />

embryogenesis and embryonic stem<br />

cell pluripotency; phosphorylation<br />

stabilizes SOX2, increasing transcriptional<br />

activity<br />

SRPK2 Akt1 human T492 T492 PSHDRSRtVsAsstG 19592491 A protein kinase targeting the serine/<br />

arginine family of splicing factors;<br />

phosphorylation causes nuclear translocation<br />

and upregulation of targets<br />

regulating cell cycle progression and<br />

apoptosis<br />

SSB Akt1 mouse T301 T302 LLRNKKVtWKVLEGH 18836485 RNA binding protein, plays a role in<br />

processing of RNA polymerase III<br />

transcripts; phosphorylation promotes<br />

export to cytoplasm where it binds<br />

polysomes and regulates expression of<br />

a specific set of mRNAs<br />

STXBP4 Akt2 mouse S99 S99 RAKLRsEsPWEIAFI 15753124 Inhibits formation and translocation of<br />

intracellular vesicles; insulin-stimulated<br />

phosphorylation of STXBP4 releases<br />

inhibition<br />

SYTL1 Akt1 human S241 S241 RMLSSSSsVSSLNSS 15998322 A secretory factor family member that<br />

is involved in granule exocytosis; phosphorylation<br />

regulates SYTL1 subnuclear<br />

localization<br />

TAL1 Akt1 human T90 T90 EARHRVPttELCRPP 15930267,<br />

19406989<br />

Transcription factor; phosphorylation<br />

inhibits transcriptional repressor activity<br />

and regulates intracellular localization<br />

TBC1D1 Akt1 human T596 T596 AFRRRANtLsHFPIE 17995453 Rab GTPase-activating protein involved<br />

in insulin-stimulated Glut4 trafficking;<br />

phosphorylation promotes glucose<br />

transport<br />

62 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables 63


Section I: Research Areas<br />

chapter 02: Signaling<br />

AMPKA Published Data<br />

Substrate Isoform<br />

TERT Akt1 human S227,<br />

S824<br />

THOC4 Akt1 human S34,<br />

T219<br />

Human<br />

Organism Site Site Sequence (+/-7) PMID<br />

S227,<br />

S824<br />

S34,<br />

T219<br />

GARRRGGsASRSLPL,<br />

AVRIRGKsYVQCQGI<br />

RGRGRAGsQGGrGGG,<br />

GGGtrRGtRGGARGR<br />

Substrate Function and<br />

Effect of Phosphorylation<br />

10224060 Telomerase reverse transcriptase,<br />

chromosome length maintenance;<br />

phosphorylation enhances telomerase<br />

activity<br />

18562279 An RNA binding and export protein<br />

that also acts as a chaperone for<br />

dimerization of transcription factors;<br />

phosphorylation regulates THOC4<br />

subnuclear localization and activates<br />

mRNA export and cell proliferation<br />

TOPBP1 Akt1 human S1159 S1159 EERARLAsNLQWPSC 19477925 Induces a large increase in the kinase<br />

activity of ATR; phosphorylation prevents<br />

the enhanced association of ATR<br />

with TopBP1 after DNA damage<br />

TRF1 Akt1 human T273 T273 SKRTRTItSQDKPSG 19160102 Controls telomere structure; phosphorylation<br />

decreases telomere length<br />

TSC2 Akt1 human S939,<br />

S981,<br />

T1462<br />

Akt1 rat S1130,<br />

S1132<br />

S939,<br />

S981,<br />

T1462<br />

S1130,<br />

S1132<br />

sFRARstsLNERPKs,<br />

AFRCRSIsVSEHVVR,<br />

GLRPRGytIsDSAPs<br />

GARDRVRsMsGGHGL,<br />

RDRVRsMsGGHGLRV<br />

15342917,<br />

12150915,<br />

16636147<br />

12172553<br />

Tumor suppressor that inhibits mTOR;<br />

phosphorylation inhibits function and<br />

allows protein synthesis to occur<br />

TTC3 Akt1 human S378 S378 AYTPRsLsAPIFTTS 20059950 E3 ligase to Akt; phosphorylation<br />

promotes TTC3 function, such as ability<br />

to ubiquitinylate and destabilize Akt<br />

TWIST1 Akt1 human S42,<br />

S123<br />

S42,<br />

S123<br />

GGRKRRSsRRSAGGG,<br />

RERQRTQsLNEAFAA<br />

20400976 A regulatory basic helix-loop-helix<br />

anti-apoptotic transcription factor;<br />

phosphorylation activates TWIST1,<br />

causing inhibition of p53 and promotion<br />

of cell survival<br />

USP8 Akt1 mouse T907 T945 TCRRRSRtFEAFMYL 17210635 Deubiquitinating enzyme that plays a<br />

role in growth factor receptor trafficking<br />

and degradation; phosphorylation<br />

increases protein stability<br />

VCP Akt1 human S352,<br />

S746,<br />

S748<br />

S352,<br />

S746,<br />

S748<br />

AAtNRPNsIDPALRR,<br />

AMRFARRsVsDNDIR,<br />

RFARRsVsDNDIRky<br />

16551632,<br />

16027165<br />

ATPase and molecular chaperone;<br />

phosphorylation may impair its proapoptotic<br />

effects and promote cell<br />

survival<br />

Vimentin Akt1 human S39 S39 ttsTrtysLGsALRP 20856200 A cytoskeletal intermediate filament<br />

protein; phosphorylation induces cellular<br />

motility and invasion by protection<br />

from proteolysis<br />

Wee1 Akt1 human S642 S642 KKMNRsVsLTIy___ 15964826 A protein kinase that inhibits cell cycle<br />

progression by phosphorylation inhibition<br />

of cdc2 kinase; phosphorylation<br />

promotes a change in Wee1 localization<br />

from nuclear to cytoplasmic and is<br />

associated with G2/M arrest<br />

WNK1 Akt1 human T60 T60 EYRRRRHtMDKDSRG 14611643,<br />

16081417<br />

XIAP<br />

Akt1,<br />

Akt2<br />

human S87 S87 VGRHRKVsPNCRFIN 14645242,<br />

17537996<br />

Regulates ion channels; phosphorylation<br />

of WNK1 causes SGK1 activation<br />

and regulation of sodium ion transport<br />

Inhibitor of apoptosis; phosphorylation<br />

prevents ubiquitination/degradation and<br />

causes increased cell survival<br />

YAP1 Akt1 human S127 S127 PQHVRAHssPAsLQL 12535517 A transcriptional co-activator of PEBP2<br />

and other transcription factors; phosphorylation<br />

suppresses p73-mediated<br />

apoptosis<br />

YB-1 Akt1 human S102 S102 NPRKyLRsVGDGEtV 22417301 A transcription/translation factor<br />

involved in mRNA stability and expression;<br />

phosphorylation induces activation<br />

and translocation to the nucleus<br />

zyxin Akt1 human S142 S142 PQPREKVssIDLEId 17572661 A focal adhesion molecule that moves<br />

between the cytoplasm and nucleus;<br />

phosphorylation promotes an association<br />

with acinus and anti-apoptotic<br />

activity<br />

Akt Binding Partners<br />

Binding<br />

Partners Effect of Binding<br />

Effect on<br />

Akt activity References<br />

GAPDH Binds to active Akt and<br />

limits its dephosphorylation<br />

Positive Jacquin, M.A. et al. (2013) Cell Death Differ. 20,<br />

1043–1054.<br />

Jade-1 Binds to and inhibits Akt kinase activity Negative Zeng, L. et al. (2013) Cancer Res. 73, 5371–5380.<br />

Mst1 Binds to and inhibits Akt kinase activity Negative Cinar, B. et al. (2007) EMBO J. 26, 4523–4534. • Jang,<br />

S.W. et al. (2007) J. Biol. Chem. 282, 30836–30844.<br />

ArgBP2γ Binds and functions as an<br />

N/A Yuan, Z.Q. et al. (2005) J. Biol. Chem. 280, 21483–21490.<br />

adaptor for Akt and PAK1<br />

CBP Akt binds and phosphorylates<br />

N/A Liu, Y. et al. (2013) FEBS Lett. 587, 847–853.<br />

CBP to regulate CBP activity<br />

PP1 Binds Akt and dephosphorylates Negative Xiao, L. et al. (2010) Cell Death Differ. 17, 1448–1462.<br />

Akt at Thr450<br />

PLCγ1 Akt binds and phosphorylates PLCγ1 N/A Wang, Y. et al. (2006) Mol. Biol. Cell 17, 2267–2277.<br />

Skp2<br />

PEA-15<br />

PHF20<br />

PHLPP<br />

FKBP5<br />

CKIP-1<br />

Ras<br />

BTBD10<br />

KCTD20<br />

PAR-4<br />

Akt binds and phosphorylates<br />

Skp2 to regulate Skp2 activity<br />

Akt binds and phosphorylates PEA-15<br />

to regulate its anti-apoptotic function.<br />

Akt binds and phosphorylates PHF20<br />

to regulate its subcellular localization<br />

PHLPP binds Akt and dephosphorylates<br />

Akt at Ser473<br />

Binds Akt and acts as a scaffold for<br />

the interaction of Akt with PHLPP<br />

Binds Akt and inhibits<br />

Akt phosphorylation<br />

Interacts with the pleckstrin homology<br />

domain of Akt<br />

Binds Akt and inhibits<br />

its dephosphorylation<br />

Binds Akt and inhibits<br />

its dephosphorylation<br />

Akt binds and phosphorylates PAR-4<br />

to inhibit its pro-apoptotic activity<br />

N/A Lin, H. et al. (2009) Nat. Cell Biol. 11, 420–432. • Gao, D.<br />

et al. (2009) Nat. Cell Biol. 11, 397–408<br />

N/A Trencia, A. et al. (2003) Cell. Biol. 23, 4511–4521.<br />

N/A Park, S. et al. (2012) J. Biol. Chem. 287, 11151–11163.<br />

Negative Gao, T. et al. Mol. Cell 18, 13–24.<br />

Negative Pei, H. et al. (2009) Cancer Cell 16, 259–266.<br />

Negative Tokuda, E. et al. (2007) Cancer Res. 67, 9666–9676.<br />

Positive Yue, Y. et al. (2004) J. Biol. Chem. 279, 12883–12889.<br />

Positive Nawa, M. et al. (2008) Cell Signal. 20, 493–505.<br />

Positive Nawa, M. et al. (2013) BMC Biochem. 14, 27.<br />

N/A Goswami, A. et al. (2005) Mol. Cell 20, 33–44.<br />

Tpl2 Akt binds and phosphorylates Tpl2 N/A Kane, L.P. et al. (2002) Mol. Cell. Biol. 22, 5962–5974.<br />

SirT2 SirT2 interacts with Akt and is Positive Ramakrishnan, G. et al. (2014) J. Biol. Chem. 289,<br />

required for optimal Akt activation<br />

6054–6066.<br />

NPM<br />

eEF1A<br />

CLIPR-59<br />

Binds the pleckstrin homology domain<br />

of Akt to promote cell survival<br />

Interacts with Akt and contributes<br />

to Akt phosphorylation<br />

Interacts with the kinase domain<br />

of Akt and regulates the subcellular<br />

localization of Akt<br />

N/A Lee, S.B. et al. (2008) Proc. Natl. Acad. Sci. USA 105,<br />

16584–16589. • Kwon, I.S. et al. (2010) BMB Rep. 43,<br />

127–132.<br />

Positive Pecorari, L. et al. (2009) Mol. Cancer 8, 58.<br />

N/A Ding, J. et al. (2009) Mol. Cell. Biol. 29, 1459–1471.<br />

CNK1 Binds and enhances Akt activation Positive Fritz, R.D. et al. (2010) Oncogene 29, 3575–3582.<br />

Phafin2 Binds Akt in the lysosome<br />

N/A Matsuda–Lennikov, M. et al. (2014) PLoS One 9, e79795.<br />

to regulate autophagy<br />

Btk Binds Akt and promotes<br />

Akt phosphorylation<br />

Positive Lindvall, J. et al. (2002) Biochem. Biophys. Res. Commun.<br />

293, 1319–1326.<br />

β-Parvin Binds Akt and prevents the<br />

interaction of Akt with ILK<br />

Negative Kimura, M. et al. (2010) J. Cell Sci. 123, 747–755.<br />

NS1<br />

Interacts with the pleckstrin<br />

homology domain of Akt<br />

Positive<br />

Matsuda, M. et al. (2010) Biochem. Biophys. Res. Commun.<br />

395, 312–317.<br />

α-Synuclein Binds Akt and promotes Akt activation Positive Chung, J.Y. et al. (2011) Neurosignals 19, 86–96.<br />

RACK1 RACK1 interacts with Akt<br />

Negative Li, G. et al. (2012) Nat. Commun. 3, 667.<br />

in a complex with PP2A<br />

ProF Binds Akt and influences<br />

N/A Fritzius, T. et al. (2006) Biochem. J. 399, 9–20.<br />

the subcellular localization of Akt<br />

p27 Kip1 Akt binds and phosphorylates p27 N/A Liang, J. et al. (2002) Nat. Med. 8, 1153–1160. • Shin, I.<br />

et al. (2002) Nat. Med. 8, 1145–1152.<br />

FoxA2/HNF3β Akt binds and phosphorylates<br />

FoxA2/HNF3β<br />

N/A Wolfrum, C. et al. (2003) Proc. Natl. Acad. Sci. USA 100,<br />

11624–11629.<br />

DNMT1 Akt binds and phosphorylates DNMT1 N/A Estève, P.O. et al. (2011) Nat. Struct. Mol. Biol. 18, 42–48.<br />

Akt Signaling<br />

Research<br />

Please visit our website for additional<br />

resources and products relating to the<br />

study of Akt Signaling.<br />

www.cellsignal.com/cstakt<br />

64 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables<br />

65


FGFR2 FGFR3<br />

TrkC<br />

EphB2<br />

TrkB<br />

FGFR1<br />

FGFR4<br />

EphB1<br />

TrkA<br />

FLT1/VEGFR1<br />

EphA5<br />

KDR/VEGFR2<br />

MuSK ROR2<br />

Fms/CSFR<br />

ROR1<br />

EphB3<br />

Ret<br />

Kit<br />

EphA3<br />

DDR2<br />

Mer<br />

EphA4<br />

DDR1<br />

Tyro3/<br />

Axl<br />

FLT4<br />

Sky<br />

PDGFRα<br />

EphA6<br />

IGF1R IRR<br />

FLT3<br />

PDGFRβ<br />

InsR<br />

Yes<br />

EphB4<br />

Met<br />

EGFR HER2/ErbB2<br />

Src<br />

Ron<br />

MLK3<br />

Ros<br />

EphA7<br />

ALK<br />

MLK1<br />

Lyn<br />

LTK<br />

Tie2<br />

Tie1<br />

HCK<br />

Fyn<br />

RYK<br />

HER4<br />

EphA8<br />

MLK4<br />

CCK4/PTK7<br />

MLK2<br />

Lck<br />

Fgr<br />

Ack Tnk1 Tyk2<br />

Jak1<br />

HER3<br />

Jak2<br />

EphA2<br />

Jak3<br />

BLK<br />

ANKRD3 SgK288<br />

DLK<br />

Syk Zap70/SRK<br />

EphA1<br />

PYK2/FAK2<br />

LZK<br />

FAK<br />

Lmr1<br />

ALK4<br />

ITK<br />

Lmr2<br />

C-Raf/Raf1<br />

FRK<br />

TGFβR1<br />

ZAK<br />

BRaf<br />

TEC<br />

EphB6<br />

KSR<br />

Srm<br />

RIPK2<br />

KSR2<br />

TXK<br />

Brk<br />

BTK<br />

Lmr3<br />

ALK7<br />

IRAK3<br />

IRAK1<br />

LIMK1<br />

ARaf<br />

BMPR1B<br />

Etk/BMX<br />

EphA10<br />

LIMK2 TESK1 ILK<br />

BMPR1A<br />

CTK<br />

RIPK3<br />

TSK2<br />

TAK1<br />

HH498<br />

ALK1<br />

CSK<br />

ALK2<br />

Abl2/Arg<br />

IRAK2<br />

ActR2<br />

Abl Fes<br />

ActR2B<br />

Fer<br />

RIPK1<br />

Jak3~b<br />

TGFβR2<br />

LRRK2<br />

MEKK2/MAP3K2<br />

Jak2~b<br />

LRRK1<br />

MISR2<br />

MEKK3/MAP3K3<br />

Tyk2~b<br />

SuRTK106<br />

IRAK4<br />

BMPR2<br />

ASK/MAP3K5<br />

ANPα/NPR1<br />

MAP3K8<br />

Jak1~b<br />

MAP3K7<br />

ANPβ/NPR2<br />

KHS1<br />

MOS<br />

KHS2<br />

HSER<br />

SgK496<br />

WNK1<br />

WNK3<br />

MEKK6/MAP3K6<br />

DYRK2<br />

Mst4<br />

PBK<br />

MAP3K4<br />

DYRK3<br />

GUCY2D<br />

WNK2<br />

DYRK4<br />

DYRK1A<br />

GUCY2F<br />

OSR1<br />

DYRK1B<br />

WNK4<br />

MLKL<br />

PERK/PEK<br />

SgK307<br />

SLK<br />

PKR<br />

LOK<br />

HIPK1 HIPK3<br />

GCN2 SgK424<br />

TAO1<br />

SCYL3 TAO2<br />

HIPK2<br />

SCYL1<br />

Tpl2/COT<br />

SCYL2<br />

NIK<br />

TAO3<br />

PAK1<br />

HIPK4<br />

PRP4<br />

HRI<br />

PAK3 PAK4<br />

CLIK1<br />

PAK2<br />

IRE1<br />

MAP2K5<br />

PAK5/PAK7<br />

CLIK1L<br />

IRE2<br />

CLK3<br />

TBCK<br />

PAK6<br />

MAP2K7<br />

MEK1/MAP2K1<br />

RNAseL<br />

GCN2~b<br />

MEK2/MAP2K2<br />

TTK<br />

MSSK1<br />

SgK071<br />

SRPK2<br />

KIS<br />

MYT1<br />

SEK1/MAP2K4 MKK3/MKK6<br />

SRPK1<br />

CK2α1<br />

Wee1<br />

MAK<br />

CK2α2<br />

SgK196<br />

CDC7<br />

Wee1B<br />

CK1δ<br />

ICK<br />

PRPK<br />

TTBK1<br />

CK1ε<br />

GSK3β<br />

MOK<br />

TTBK2<br />

GSK3α<br />

CK1α1<br />

CDKL3<br />

CK1α2<br />

CDKL2<br />

PINK1<br />

SgK493<br />

SgK269<br />

VRK3<br />

CK1γ2<br />

CDKL1 CDKL5<br />

ERK7<br />

SgK396<br />

SgK223<br />

CDKL4 Erk4<br />

Slob<br />

CK1γ1<br />

Erk3<br />

SgK110<br />

PIK3R4<br />

CK1γ3<br />

NLK<br />

SgK069<br />

Bub1<br />

Erk5<br />

SBK<br />

BubR1<br />

Erk1/<br />

IKKα<br />

p44MAPK<br />

IKKβ<br />

VRK1<br />

CDK7<br />

IKKε<br />

VRK2<br />

Erk2/<br />

PLK4<br />

PITSLRE<br />

TBK1/NAK<br />

p42MAPK p38γ<br />

MPSK1<br />

JNK1<br />

p38δ<br />

JNK2<br />

TLK2<br />

JNK3 CDK10<br />

GAK<br />

TLK1<br />

PLK3<br />

p38β<br />

AAK1<br />

CAMKK1<br />

ULK3<br />

PLK1<br />

p38α<br />

PLK2<br />

CDK4<br />

CCRK<br />

BIKE<br />

CAMKK2<br />

BARK1/GRK2<br />

CDK6<br />

ULK1<br />

BARK2/GRK3 RHOK/GRK1<br />

Fused<br />

GRK5<br />

PFTAIRE2<br />

SgK494<br />

ULK2 ULK4<br />

GRK6 GRK4<br />

PFTAIRE1<br />

CDK9<br />

Nek6<br />

RSKL1<br />

PCTAIRE2<br />

Nek7 Nek10<br />

SgK495<br />

PASK<br />

PDK1<br />

RSKL2<br />

Nek8<br />

MSK1<br />

RSK1/p90RSK<br />

PCTAIRE1<br />

CDK5 CRK7<br />

PCTAIRE3<br />

Nek9<br />

LKB1<br />

MSK2<br />

RSK4 RSK2<br />

Chk1<br />

p70S6K<br />

RSK3<br />

Nek2<br />

Akt2/PKBβ<br />

AurA/Aur2<br />

p70S6Kβ<br />

Akt1/PKBα<br />

cdc2/CDK1<br />

Nek11<br />

Akt3/PKBγ<br />

CDK3<br />

Nek4<br />

AurB/Aur1<br />

SGK1<br />

CDK2<br />

AurC/Aur3<br />

SGK2<br />

SGK3<br />

Trb3 Pim1 Pim2<br />

PKG2<br />

PKN1/PRK1<br />

LATS1<br />

Nek3<br />

PKG1<br />

PKN2/PRK2<br />

Trb2<br />

Pim3<br />

LATS2<br />

PKN3<br />

Nek5<br />

NDR1<br />

PRKY<br />

PKCδ<br />

Trb1<br />

Obscn~b<br />

NDR2<br />

PKCθ<br />

PRKX<br />

YANK1<br />

SPEG~b<br />

MAST3<br />

PKCη<br />

Nek1<br />

MASTL<br />

PKCε<br />

Obscn<br />

STK33<br />

YANK2<br />

PKCι<br />

YANK3<br />

PKCζ<br />

SPEG<br />

PKAγ<br />

PKAα<br />

PKCγ<br />

TTN<br />

MAST2<br />

PKAβ<br />

smMLCK<br />

TSSK4<br />

ROCK1<br />

PKCα<br />

HUNK<br />

ROCK2<br />

Chk2/Rad53<br />

PKCβ<br />

skMLCK<br />

SSTK<br />

SNRK<br />

MAST4 MAST1<br />

DMPK<br />

NIM1<br />

CRIK<br />

DRAK1 TSSK3<br />

PKD2/PKCμ<br />

TSSK1<br />

SgK085<br />

TSSK2<br />

DMPK2<br />

caMLCK<br />

PKD1<br />

DCAMKL3<br />

DAPK2<br />

DAPK3<br />

DCAMKL1<br />

MELK PKD3/PKCν<br />

DAPK1<br />

DCAMKL2<br />

MRCKβ<br />

VACAMKL<br />

MRCKα<br />

MNK1<br />

PhKγ1<br />

MNK2<br />

PhKγ2<br />

PSKH1<br />

CaMKIIγ<br />

PSKH2<br />

BRSK2<br />

CaMKIIα<br />

CaMKIIβ<br />

BRSK1<br />

CaMKIIδ<br />

RSK4~b<br />

SNARK<br />

RSK1~b<br />

CaMKIV<br />

ARK5<br />

MSK2~b<br />

MSK1~b<br />

QSK<br />

RSK3~b<br />

RSK2~b<br />

CaMKIβ<br />

SIK<br />

QIK<br />

CaMKIγ<br />

MARK4<br />

CaMKIα<br />

CaMKIδ<br />

MARK3<br />

CHED<br />

DRAK2<br />

AMPKα2<br />

AMPKα1<br />

Haspin<br />

CASK<br />

MAPKAPK5<br />

MAPKAPK2<br />

MAPKAPK3<br />

STLK3<br />

STRAD/STLK5<br />

STLK6<br />

GRK7<br />

HPK1<br />

GCK<br />

NRK/ZC4<br />

Mst1<br />

Mst2<br />

TNIK/ZC2<br />

MYO3A<br />

MYO3B<br />

YSK1<br />

Mst3<br />

HGK/ZC1<br />

MINK/ZC3<br />

Section I: Research Areas<br />

chapter 02: Signaling<br />

Tyrosine Kinases and<br />

Associated Phosphatases<br />

Tyrosine Kinase Signaling<br />

Whether functioning as cell surface receptors or as internal effectors of cell signaling, tyosine kinases<br />

control multiple aspects of cell and organism growth, differentiation, and function. Approximately 20<br />

receptor tyrosine kinase (RTK) families and at least 9 distinct groups of nonreceptor tyrosine kinases<br />

have been identified in humans. Regardless of localization, all tyrosine kinases regulate target protein<br />

function through transfer of phosphate from ATP to the hydroxyl group of a target protein tyrosine.<br />

EGF stimulation results in<br />

phosphorylation of EGFR at Tyr1068.<br />

Phospho-EGF Receptor (Tyr1068) (D7A5) XP ® Rabbit mAb #3777:<br />

WB analysis of extracts of BxPC-3 cells, untreated or EGF-stimulated,<br />

using #3777 (upper) and EGF Receptor Antibody #2232 (lower).<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

200<br />

140<br />

100<br />

80<br />

+<br />

–<br />

Phospho-<br />

EGFR<br />

(Tyr1068)<br />

EGFR<br />

EGF<br />

The tyrosine kinase family, a distinct<br />

group within the human kinome<br />

CMGC<br />

CLK4<br />

CLK2 CLK1<br />

TK<br />

CDK8 CDK11<br />

MARK1<br />

MARK2<br />

Receptor Tyrosine Kinases<br />

Most RTKs are single-pass, transmembrane<br />

proteins that bind extracellular polypeptide ligands<br />

(i.e. growth factors) and cytoplasmic effector and<br />

adaptor proteins to regulate biological processes.<br />

Ligand binding promotes receptor dimerization and<br />

autophosphorylation of receptor tyrosine residues.<br />

The resultant conformational change stabilizes the<br />

active kinase, and subsequent phosphorylation<br />

events form binding sites for downstream adaptor,<br />

scaffold, and effector proteins.<br />

EGFR phosphorylation at multiple<br />

tyrosine residues provides binding<br />

sites for adaptor proteins and<br />

downstream signaling nodes.<br />

Trad Trio<br />

CAMK<br />

FGFR2 FGFR3<br />

TrkC<br />

EphB2<br />

TrkB<br />

FGFR1<br />

FGFR4<br />

EphB1<br />

TrkA<br />

FLT1/VEGFR1<br />

EphA5<br />

KDR/VEGFR2<br />

MuSK ROR2<br />

Fms/CSFR<br />

ROR1<br />

EphB3<br />

Ret<br />

Kit<br />

EphA3<br />

DDR2<br />

Mer<br />

EphA4<br />

DDR1<br />

Tyro3/<br />

Axl<br />

FLT4<br />

Sky<br />

PDGFRα<br />

EphA6<br />

IGF1R IRR<br />

FLT3<br />

PDGFRβ<br />

InsR<br />

Yes<br />

EphB4<br />

Met<br />

EGFR HER2/ErbB2<br />

Src<br />

Ron<br />

MLK3<br />

Ros<br />

EphA7<br />

ALK<br />

MLK1<br />

Lyn<br />

LTK<br />

Tie2<br />

Tie1<br />

HCK<br />

Fyn<br />

RYK<br />

HER4<br />

EphA8<br />

MLK4<br />

CCK4/PTK7<br />

MLK2<br />

Lck<br />

Fgr<br />

Ack Tnk1 Tyk2<br />

Jak1<br />

HER3<br />

Jak2<br />

EphA2<br />

Jak3<br />

BLK<br />

TKL<br />

ANKRD3 SgK288<br />

DLK<br />

Syk Zap70/SRK<br />

EphA1<br />

PYK2/FAK2<br />

LZK<br />

TKL<br />

FAK<br />

Lmr1<br />

ALK4<br />

ITK<br />

Lmr2<br />

C-Raf/Raf1<br />

FRK<br />

TGFβR1<br />

ZAK<br />

BRaf<br />

TEC<br />

EphB6<br />

KSR<br />

Srm<br />

RIPK2<br />

KSR2<br />

TXK<br />

Brk<br />

BTK<br />

Lmr3<br />

ALK7<br />

IRAK3<br />

IRAK1<br />

LIMK1<br />

ARaf<br />

BMPR1B<br />

Etk/BMX<br />

EphA10<br />

LIMK2 TESK1 ILK<br />

BMPR1A<br />

CTK<br />

RIPK3<br />

TSK2<br />

TAK1<br />

HH498<br />

ALK1<br />

CSK<br />

ALK2<br />

Abl2/Arg<br />

IRAK2<br />

ActR2<br />

Abl Fes<br />

ActR2B<br />

Fer<br />

RIPK1<br />

Jak3~b<br />

TGFβR2<br />

LRRK2<br />

MEKK2/MAP3K2<br />

Jak2~b<br />

LRRK1<br />

MISR2<br />

MEKK3/MAP3K3<br />

Tyk2~b<br />

SuRTK106<br />

IRAK4<br />

BMPR2<br />

ASK/MAP3K5<br />

ANPα/NPR1<br />

MAP3K8<br />

Jak1~b<br />

MAP3K7<br />

ANPβ/NPR2<br />

NRBP1 NRBP2 MEKK1/MAP3K1<br />

KHS1<br />

MOS<br />

KHS2<br />

HSER<br />

SgK496<br />

WNK1<br />

STE<br />

WNK3<br />

MEKK6/MAP3K6<br />

DYRK2<br />

Mst4<br />

PBK<br />

MAP3K4<br />

DYRK3<br />

GUCY2D<br />

WNK2<br />

DYRK4<br />

DYRK1A<br />

NRBP1<br />

GUCY2F<br />

NRBP2 MEKK1/MAP3K1 OSR1<br />

DYRK1B<br />

WNK4<br />

MLKL<br />

PERK/PEK<br />

SgK307<br />

SLK<br />

PKR<br />

LOK<br />

HIPK1 HIPK3<br />

GCN2 SgK424<br />

TAO1<br />

SCYL3 TAO2<br />

HIPK2<br />

SCYL1<br />

Tpl2/COT<br />

SCYL2<br />

NIK<br />

TAO3<br />

PAK1<br />

CLK4 HIPK4<br />

PRP4<br />

HRI<br />

PAK3<br />

CLIK1<br />

PAK2<br />

IRE1<br />

CLK2 CLK1<br />

PAK4<br />

MAP2K5<br />

PAK5/PAK7<br />

CLIK1L<br />

IRE2<br />

CLK3<br />

TBCK<br />

PAK6<br />

MAP2K7<br />

MEK1/MAP2K1<br />

RNAseL<br />

GCN2~b<br />

MEK2/MAP2K2<br />

CK1<br />

TTK<br />

MSSK1<br />

SgK071<br />

SRPK2<br />

KIS<br />

MYT1<br />

SEK1/MAP2K4 MKK3/MKK6<br />

SRPK1<br />

CK2α1<br />

Wee1<br />

MAK<br />

CK2α2<br />

SgK196<br />

CDC7<br />

Wee1B<br />

CK1δ<br />

ICK<br />

PRPK<br />

TTBK1<br />

CK1ε<br />

GSK3β<br />

AGC<br />

MOK<br />

TTBK2<br />

GSK3α<br />

CK1α1<br />

CDKL3<br />

CK1α2<br />

CDKL2<br />

PINK1<br />

SgK493<br />

SgK269<br />

VRK3<br />

CK1γ2<br />

CDKL1 CDKL5<br />

ERK7<br />

SgK396<br />

SgK223<br />

CDKL4 Erk4<br />

Slob<br />

CK1γ1<br />

Erk3<br />

SgK110<br />

PIK3R4<br />

CK1γ3<br />

NLK<br />

SgK069<br />

Bub1<br />

Erk5<br />

SBK<br />

BubR1<br />

Erk1/<br />

IKKα<br />

p44MAPK<br />

IKKβ<br />

VRK1<br />

CDK7<br />

IKKε<br />

VRK2<br />

Erk2/<br />

PLK4 CK1<br />

PITSLRE<br />

TBK1/NAK<br />

p42MAPK p38γ<br />

MPSK1<br />

JNK1<br />

p38δ<br />

JNK2<br />

TLK2<br />

JNK3 CDK10<br />

GAK<br />

TLK1<br />

PLK3<br />

p38β<br />

AAK1<br />

CDK8 CDK11<br />

CAMKK1<br />

ULK3<br />

PLK1<br />

p38α<br />

PLK2<br />

CDK4<br />

CCRK<br />

BIKE<br />

CAMKK2<br />

BARK1/GRK2<br />

CDK6<br />

CMGC<br />

ULK1<br />

BARK2/GRK3 RHOK/GRK1<br />

Fused<br />

GRK5<br />

PFTAIRE2<br />

SgK494<br />

ULK2 ULK4<br />

GRK6 GRK4<br />

PFTAIRE1<br />

CDK9<br />

Nek6<br />

RSKL1<br />

PCTAIRE2<br />

Nek7 Nek10<br />

SgK495<br />

PASK<br />

PDK1<br />

RSKL2<br />

Nek8<br />

MSK1<br />

RSK1/p90RSK<br />

PCTAIRE1<br />

CDK5 CRK7<br />

PCTAIRE3<br />

Nek9<br />

LKB1<br />

MSK2<br />

RSK4 RSK2<br />

Chk1<br />

p70S6K<br />

RSK3<br />

Nek2<br />

Akt2/PKBβ<br />

AurA/Aur2<br />

p70S6Kβ<br />

Akt1/PKBα<br />

cdc2/CDK1<br />

Nek11<br />

Akt3/PKBγ<br />

CDK3<br />

Nek4<br />

AurB/Aur1<br />

SGK1<br />

CDK2<br />

AurC/Aur3<br />

SGK2<br />

SGK3<br />

Trb3 Pim1 Pim2<br />

PKG2<br />

PKN1/PRK1<br />

LATS1<br />

Nek3<br />

PKG1<br />

PKN2/PRK2<br />

Trb2<br />

Pim3<br />

PKN3<br />

Nek5<br />

Trad Trio<br />

LATS2<br />

NDR1<br />

PRKY<br />

PKCδ<br />

Trb1<br />

Obscn~b<br />

NDR2<br />

PKCθ<br />

PRKX<br />

YANK1<br />

SPEG~b<br />

MAST3<br />

PKCη<br />

Nek1<br />

MASTL<br />

PKCε<br />

Obscn<br />

STK33<br />

YANK2<br />

PKCι<br />

YANK3<br />

PKCζ<br />

SPEG<br />

PKAγ<br />

PKAα<br />

PKCγ<br />

TTN<br />

MAST2<br />

PKAβ<br />

smMLCK<br />

TSSK4<br />

ROCK1<br />

PKCα<br />

HUNK<br />

ROCK2<br />

Chk2/Rad53<br />

PKCβ<br />

skMLCK<br />

SSTK<br />

SNRK<br />

MAST4 MAST1<br />

DMPK<br />

NIM1<br />

CRIK<br />

DRAK1 TSSK3<br />

PKD2/PKCμ<br />

TSSK1<br />

SgK085<br />

TSSK2<br />

DMPK2<br />

EGF<br />

caMLCK<br />

PKD1<br />

DCAMKL3<br />

DAPK2<br />

DAPK3<br />

DCAMKL1<br />

MELK PKD3/PKCν<br />

DAPK1<br />

DCAMKL2<br />

MRCKβ<br />

VACAMKL<br />

MRCKα<br />

MNK1<br />

PhKγ1<br />

MNK2<br />

PhKγ2<br />

PSKH1<br />

CaMKIIγ<br />

PSKH2<br />

BRSK2<br />

CaMKIIα<br />

CaMKIIβ<br />

NH 2<br />

BRSK1<br />

CaMKIIδ<br />

RSK4~b<br />

SNARK<br />

RSK1~b<br />

CaMKIV<br />

ARK5<br />

MSK2~b<br />

MSK1~b<br />

CaMKII<br />

CaMKII<br />

Cys<br />

rich<br />

CHED<br />

Tyr845<br />

Tyr974<br />

Tyr992<br />

Tyr1045<br />

Ser1046<br />

Ser1047<br />

Ser1057<br />

Tyr1068<br />

Tyr1086<br />

Tyr1101<br />

Ser1142<br />

Tyr1148<br />

Tyr1173<br />

TK<br />

Src phosphorylation<br />

Autophosphorylation<br />

kinase domain<br />

kinase domain<br />

COOH<br />

EGFR homodimer<br />

P<br />

P<br />

P<br />

P<br />

P<br />

P<br />

P<br />

P<br />

P<br />

P<br />

P<br />

P<br />

P<br />

STAT5b<br />

AP-2<br />

PLCγ<br />

Cbl<br />

ubiquitination<br />

Grb2<br />

Gab1<br />

ubiquitination<br />

SHC<br />

SHP1<br />

PLCγ<br />

DRAK2<br />

CAMK<br />

DNA synthesis<br />

Translocation to<br />

mitochondria survival<br />

PKC<br />

ubiquitination<br />

p85<br />

PKC<br />

AMPKα2<br />

AMPKα1<br />

QSK<br />

SIK<br />

QIK<br />

degradation<br />

degradation<br />

MARK4<br />

MARK3<br />

MARK1<br />

MARK2<br />

MAPK/ERK<br />

Cascade<br />

degradation<br />

MAPK/ERK<br />

Cascade<br />

AKT/PKB<br />

Cascade<br />

MAPK/ERK<br />

Cascade<br />

Haspin<br />

RSK3~b<br />

RSK2~b<br />

CASK<br />

MAPKAPK5<br />

MAPKAPK2<br />

MAPKAPK3<br />

STLK3<br />

STRAD/STLK5<br />

STLK6<br />

CaMKIβ<br />

CaMKIγ<br />

CaMKIα<br />

CaMKIδ<br />

GRK7<br />

HPK1<br />

GCK<br />

NRK/ZC4<br />

Mst1<br />

Mst2<br />

TNIK/ZC2<br />

MYO3A<br />

MYO3B<br />

YSK1<br />

Mst3<br />

RTKs regulate a broad range of biological effects including cell growth, proliferation, survival, migration,<br />

STE and differentiation. For this reason, altered levels and/or activities of RTKs are commonly associated<br />

with many forms of human cancer. RTK activity can be chemically manipulated through the use of<br />

targeted, small-molecule inhibitors.<br />

HGK/ZC1<br />

MINK/ZC3<br />

Phospho-EGF Receptor (Tyr1068) (D7A5)<br />

XP ® Rabbit mAb #3777: Confocal IF analysis<br />

of HeLa cells, untreated (left) or EGF-treated<br />

(right), using #3777 (green). Blue pseudocolor<br />

= DRAQ5 ® #4084 (fluorescent DNA dye).<br />

HER2, an ErbB/HER family member, is<br />

overexpressed in 40% of human breast cancers.<br />

AGC<br />

HER2/ErbB2 (D8F12) XP ® Rabbit mAb #4290: IHC analysis of paraffin-embedded<br />

human breast carcinoma using #4290.<br />

ROS1, an RTK similar in structure to ALK that<br />

forms oncogenic fusion proteins with FIG1,<br />

SLC34A2, and CD74<br />

ROS1 (D4D6 ® ) Rabbit mAb #3287: IHC analysis of paraffin-embedded human lung<br />

carcinoma using #3287. Note: Staining is of FIG-ROS1 fusion.<br />

HER3/ErbB3 is overexpressed in many forms<br />

of cancer.<br />

HER3/ErbB3 (D22C5) XP ® Rabbit mAb #12708: IHC analysis of paraffin-embedded<br />

non-small cell lung carcinoma using #12708.<br />

Ligand activation<br />

of EGFR at the cell<br />

membrane results<br />

in phosphorylation<br />

at Tyr1068.<br />

Tyrosine Kinase<br />

Inhibitors<br />

#4401 Crizotinib<br />

Inhibitor of ALK and ROS1<br />

(ATP-competitive inhibitor)<br />

#9052 Dasatinib<br />

Tyrosine kinase inhibitor<br />

(Abl, BCR/Abl, Src, c-KIT,<br />

Ephrin receptors)<br />

#5083 Erlotinib<br />

Inhibits EGFR<br />

(ATP-competitive inhibitor)<br />

#4765 Gefitinib<br />

Inhibits EGFR<br />

(active site inhibitor)<br />

#9084 Imatinib<br />

Inhibits Bcr-Abl, PDGFR,<br />

c-Kit (active site inhibitor)<br />

#12121 Lapatinib<br />

Inhibits EGFR and HER2<br />

(ATP-competitive inhibitor)<br />

#12209 Nilotinib<br />

Tyrosine kinase inhibitor<br />

(Abl, BCR/Abl, c-KIT, LCK,<br />

Ephrin receptors, DDR1/2,<br />

PDGFR-B)<br />

#9493 PKC412<br />

Very broad spectrum protein<br />

kinase inhibitor (conventional<br />

PKCs, some RTKs, Syk,<br />

Cdk1/B, c-Src)<br />

#8705 Sorafenib<br />

Inhibits VEGFR and PDGFR;<br />

inhibits Raf kinases; Induces<br />

autophagy<br />

#12328 Sunitinib<br />

Broad RTK inhibitor<br />

(PDGFR, VEGFR, c-KIT, RET,<br />

CSF-1R, FLT-3/CD135)<br />

#9842 Tyrphostin AG 1478<br />

Inhibits EGFR<br />

#12998 Vatalanib<br />

Broad RTK inhibitor<br />

(VEGFR, PDGFR-B, c-KIT)<br />

66 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttyrosine<br />

67


Section I: Research Areas<br />

chapter 02: Signaling<br />

ALK, an RTK that forms oncogenic fusion proteins with<br />

EML4, TFG, and KIF5B, is expressed in lung carcinoma.<br />

ALK (D5F3 ® ) XP ® Rabbit mAb #3633: IHC<br />

analysis of paraffin-embedded human lung<br />

carcinoma with high (left) and low (right) levels<br />

of ALK expression using #3633.<br />

Phospho-Jak2 (Tyr1008) (D4A8) Rabbit<br />

mAb #8082: WB analysis of extracts from<br />

BaF3 cells (A), untreated or treated with Mouse<br />

Interleukin-3 (mIL-3) #8923 (10 ng/ml, 5 min),<br />

using #8082 (upper) or total Jak2 (D2E12) XP ®<br />

Rabbit mAb #3230 (lower).<br />

Jak2 (D2E12) XP ® Rabbit mAb #3230:<br />

Confocal IF analysis of K-562 cells (B) using<br />

#3230 (green). Blue pseudocolor = DRAQ5 ®<br />

(fluorescent DNA dye).<br />

A<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

200<br />

140<br />

100<br />

Phospho-<br />

Jak2<br />

(Tyr1008)<br />

Jak2<br />

B<br />

Treatment with<br />

IL-3 results in<br />

phosphorylation<br />

of Jak2 at Tyr1008.<br />

80<br />

60<br />

50<br />

40<br />

Ligand activation of<br />

Met, a high affinity RTK<br />

for hepatocyte growth<br />

factor (HGF), results<br />

in phosphorylation at<br />

Tyr1234/1235.<br />

A kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

Phospho-Met<br />

(Tyr1234/5)<br />

B<br />

Phospho-Met (Tyr1234/1235) (D26) XP ® Rabbit mAb #3077: WB analysis of cell<br />

extracts from HeLa cells, untreated or stimulated with HGF (A), using #3077 (upper) and Met<br />

(25H2) Mouse mAb #3127 (lower). Confocal IF analysis of MKN-45 cells, untreated (B) or<br />

treated with SU11274 (1 μM, 3 hr) (C), using #3077. Blue pseudocolor = DRAQ5 ® #4084<br />

(fluorescent DNA dye).<br />

C<br />

Treatment with a<br />

Met inhibitor prevents<br />

Met activation and<br />

phosphorylation at<br />

Tyr1234/1235.<br />

The nonreceptor tyrosine kinase<br />

Zap-70 is expressed in T cells.<br />

30<br />

– +<br />

Zap-70 (D1C10E) XP ® Rabbit mAb (Alexa Fluor ® 488 Conjugate) #9473:<br />

Flow cytometric analysis of Ramos (B cells; blue) and Jurkat (T cells; green) cells<br />

using #9473.<br />

mIL-3<br />

Events<br />

Zap-70<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

– +<br />

Met<br />

HGF<br />

Nonreceptor Tyrosine Kinases<br />

The cytoplasmic nonreceptor tyrosine kinases transduce signals generated by RTKs and other receptors<br />

from the cell surface to the nucleus. Receptor-associated tyrosine kinases are phosphorylated within the<br />

activation loop of their kinase catalytic domain, resulting in activation and propagation of downstream<br />

signals through phosphorylation of other kinases, adaptor proteins, or transcription factors. In addition<br />

to their kinase domain, nonreceptor tyrosine kinases often contain various domains that mediate<br />

protein-protein or protein-lipid interactions, which allows association with activated receptors or downstream<br />

signaling nodes. For example, SH2 domains bind phospho-tyrosine residues, while PH domains<br />

direct interactions with membrane lipids.<br />

PDGF treatment results in phosphorylation<br />

of Src family proteins at Tyr416.<br />

Phospho-Src Family (Tyr416) (D49G4) Rabbit mAb #6943: WB analysis of extracts<br />

NIH/3T3 cells, serum-starved or treated with human Platelet-Derived Growth Factor BB<br />

hPDGF-BB #8912 (100 ng/ml, 15 min), using #6943 (upper) or Src (36D10) Rabbit mAb<br />

#2109 (lower).<br />

Select Reviews<br />

Arteaga, C.L. and Engelman, J.A. (2014) Cancer Cell 25, 282−303. • Davies, K.D. and Doebele, R.C. (2013) Clin. Cancer<br />

Res. 19, 4040−4045. • Hallberg, B. and Palmer, R.H. (2013) Nat. Rev. Cancer 13, 685−700. • Liu, S.T., Pham, H., Pandol,<br />

S.J., and Ptasznik, A. (2014) Front. Physiol. 4, 416. • Miaczynska, M. (2013) Cold Spring Harb. Perspect. Biol. 5, a009035.<br />

• Panjarian, S., Lacob, R.E., Chen, S., Engen, J.R., and Smithgall, T.E. (2013) J. Biol. Chem. 288, 5443−5450. • Shaw, A.T.,<br />

Hsu, P.P., Awad, M.M., and Engelman, J.A. (2013) Nat. Rev. Cancer 13, 772−787.<br />

kDa<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

– +<br />

Phospho-Src<br />

(Tyr416)<br />

Src<br />

hPDGF-BB<br />

Commonly Studied Tyrosine Kinases Targets<br />

Target M P E S C<br />

ACPP<br />

•<br />

ALK • • •<br />

Phospho-ALK (Tyr1078) • •<br />

Phospho-ALK (Tyr1096) • •<br />

Phospho-ALK (Tyr1278) • •<br />

Phospho-ALK (Tyr1282/Tyr1283) • •<br />

•<br />

Phospho-ALK (Tyr1278/Tyr1282/<br />

Tyr1283)<br />

Phospho-ALK (Tyr1586) • • •<br />

Phospho-ALK (Tyr1604) • • •<br />

Axl<br />

• • • •<br />

Phospho-Axl (pan Tyr)<br />

•<br />

Phospho-Axl (Tyr702) •<br />

DDR1<br />

• • •<br />

Phospho-DDR1 (pan Tyr)<br />

•<br />

Phospho-DDR1 (Tyr792) •<br />

DDR2<br />

•<br />

EGFR<br />

• • • • •<br />

Phospho-EGFR (Thr669) • •<br />

Phospho-EGFR (Tyr845) • • •<br />

Phospho-EGFR (Tyr992) •<br />

Phospho-EGFR (Tyr998) •<br />

Phospho-EGFR (Tyr1045) •<br />

Phospho-EGFR (Ser1046/1047) •<br />

Phospho-EGFR (Tyr1068) • • • •<br />

Phospho-EGFR (Tyr1086) •<br />

Phospho-EGFR (Tyr1148) •<br />

Phospho-EGFR (Tyr1173) • •<br />

• • •<br />

EGF Receptor<br />

(E746-A750del Specific)<br />

Target M P E S C<br />

EGF Receptor (L858R Mutant Specific) • •<br />

EphA2<br />

•<br />

Phospho-EphA2 (Tyr588) •<br />

Phospho-EphA2 (Tyr594) •<br />

Phospho-EphA2 (Tyr772) •<br />

Phospho-EphA2 (Ser897) •<br />

Phospho-EphA3 (Tyr779) •<br />

EphA3/EphA4/EphA5 •<br />

EphB1<br />

•<br />

EphB6<br />

•<br />

Phospho-Ephrin B (Tyr324/329) •<br />

Eps8<br />

•<br />

EREG<br />

•<br />

Etk/BMX<br />

•<br />

Phospho-Etk/BMX (Tyr40) •<br />

FGFR1 • • • •<br />

FGFR1 (pan Tyr)<br />

•<br />

FGFR1 (Tyr653/Tyr654) • • •<br />

FGFR1 (Tyr766)<br />

•<br />

FGFR2 • • •<br />

Phospho-FGFR2 (Pan Tyr)<br />

•<br />

FGFR3<br />

•<br />

FGFR4 • • • •<br />

FLT3 • •<br />

Phospho-FLT3 (pan Tyr)<br />

•<br />

Phospho-FLT3 (Tyr589/Tyr591) •<br />

Phospho-FLT3 (Tyr591) • • •<br />

Phospho-FLT3 (Tyr842) •<br />

Phospho-FLT3 (Tyr969) •<br />

HER2/ErbB2<br />

• • • • •<br />

These protein targets represent key<br />

nodes within tyrosine kinases signaling<br />

pathways and are commonly studied<br />

in tyrosine kinases research. Primary<br />

antibodies, antibody conjugates, and<br />

antibody sampler kits containing these<br />

targets are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

E PathScan ® ELISA Kits<br />

S SignalSilence ® siRNA<br />

C Antibody Conjugate<br />

68 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttyrosine 69


Section I: Research Areas<br />

chapter 02: Signaling<br />

181<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for EGFR have been<br />

cited over 181 times in high-impact,<br />

peer-reviewed publications from the<br />

global research community.<br />

Target M P E S C<br />

Phospho-HER2/ErbB2 (pan Tyr) •<br />

Phospho-HER2/ErbB2 (Tyr877) •<br />

Phospho-HER2/ErbB2 (Tyr1196) •<br />

• • •<br />

Phospho-HER2/ErbB2<br />

(Tyr1221/1222)<br />

Phospho-HER2/ErbB2 (Tyr1248)<br />

Phospho-HER2/ErbB2<br />

(Tyr1248)/EGFR (Tyr1173)<br />

•<br />

•<br />

HER3/ErbB3 • • • •<br />

Phospho-HER3/ErbB3 (pan Tyr) •<br />

Phospho-HER3/ErbB3 (Tyr1197) •<br />

Phospho-HER3/ErbB3 (Tyr1222) •<br />

Phospho-HER3/ErbB3 (Tyr1289) •<br />

Phospho-HER3/ErbB3 (Tyr1328) •<br />

HER4/ErbB4 • •<br />

Phospho-HER4/ErbB4 (Pan Tyr) •<br />

Phospho-HER4/ErbB4 (Tyr984) •<br />

Phospho-HER4/ErbB4 (Tyr1284) •<br />

c-Kit • • • •<br />

Phospho-c-Kit (pan Tyr)<br />

•<br />

Phospho-c-Kit (Tyr703) •<br />

Phospho-c-Kit (Tyr719) • •<br />

LRIG1<br />

•<br />

M-CSF Receptor<br />

• •<br />

Phospho-M-CSF Receptor (Tyr546) •<br />

Phospho-M-CSF Receptor (Tyr699) • •<br />

Phospho-M-CSF Receptor (Tyr708) •<br />

Phospho-M-CSF Receptor (Tyr723) • •<br />

Phospho-M-CSF Receptor (Tyr809) •<br />

Phospho-M-CSF Receptor (Tyr923) •<br />

Mer • •<br />

Met<br />

• • • • •<br />

Phospho-Met (pan Tyr)<br />

•<br />

Phospho-Met (Tyr1003) • •<br />

Phospho-Met (Tyr1234/1235) • • • •<br />

Phospho-Met (Tyr1349) • • •<br />

PDGFR-α • • • •<br />

Select Citations:<br />

Wu, J. et al. (2014) EGFR-STAT3 signaling promotes formation<br />

of malignant peripheral nerve sheath tumors. Oncogene<br />

33, 173–180.<br />

Sheu, J.J. et al. (2014) LRIG1 modulates aggressiveness of<br />

head and neck cancers by regulating EGFR-MAPK-SPHK1<br />

signaling and extracellular matrix remodeling. Oncogene 33,<br />

1375–1384.<br />

Bronisz, A. et al. (2014) xtracellular vesicles modulate the<br />

glioblastoma microenvironment via a tumor suppression<br />

signaling network directed by miR-1. Cancer Res. 74,<br />

738–750.<br />

De Cesare, M. et al. (2014) Synergistic antitumor activity<br />

of cetuximab and namitecan in human squamous cell<br />

carcinoma models relies on cooperative inhibition of EGFR<br />

expression and depends on high EGFR gene copy number.<br />

Clin. Cancer Res. 20, 995–1006.<br />

Muller, P.A. et al. (2014) Mutant p53 regulates Dicer through<br />

p63-dependent and -independent mechanisms to promote<br />

an invasive phenotype. J. Biol. Chem. 289, 122–132.<br />

Target M P E S C<br />

Phospho-PDGFR-α (Tyr754) •<br />

Phospho-PDGFR-α (Tyr762) •<br />

Phospho-PDGFR-α (Tyr849) •<br />

Phospho-PDGFR-α (Tyr1018) •<br />

Phospho-PDGF Receptor α/β (pan Tyr) •<br />

Phospho-PDGF Receptor α •<br />

(Tyr849)/PDGF Receptor β (Tyr857)<br />

PDGFR-β • • •<br />

Phospho-PDGFR-β (Tyr740) •<br />

Phospho-PDGFR-β (Tyr751) • • •<br />

Phospho-PDGFR-β (Tyr771) •<br />

Phospho-PDGFR-β (Tyr1009) •<br />

Phospho-PDGFR-β (Tyr1021) •<br />

PTK7<br />

•<br />

Ret<br />

• • •<br />

Phospho-Ret (pan Tyr)<br />

•<br />

Phospho-Ret (Tyr905)<br />

•<br />

Ron • •<br />

ROR1<br />

•<br />

ROR2<br />

•<br />

ROS1 • • •<br />

Phospho-ROS1 (pan Tyr)<br />

•<br />

Phospho-ROS1 (Tyr2274) •<br />

Spry1<br />

•<br />

Tie2<br />

•<br />

Phospho-Tie2 (Tyr992) •<br />

Phospho-Tie2 (Ser1119) •<br />

Tyro3<br />

•<br />

VEGFR1<br />

•<br />

VEGFR2 • • • •<br />

Phospho-VEGFR2 (Tyr951) • •<br />

Phospho-VEGFR2 (Tyr996) •<br />

Phospho-VEGFR2 (Tyr1059) •<br />

Phospho-VEGFR2 (Tyr1175) • •<br />

Phospho-VEGFR2 (Tyr1212) •<br />

VEGFR3<br />

• •<br />

Stahlschmidt, W. et al. (2014) Clathrin terminal domainligand<br />

interactions regulate sorting of mannose 6-phosphate<br />

receptors mediated by AP-1 and GGA adaptors. J Biol Chem.<br />

21, 4906–4918.<br />

Tao, J.J. et al. (2014) Antagonism of EGFR and HER3<br />

enhances the response to inhibitors of the PI3K-Akt pathway<br />

in triple-negative breast cancer. Sci. Signal. 7, ra29.<br />

Zhang, F. et al. (2014) Temporal production of the signaling<br />

lipid phosphatidic acid by phospholipase D2 determines the<br />

output of extracellular signal-regulated kinase signaling in<br />

cancer cells. Mol. Cell Biol. 34, 84–95.<br />

Lee, C.Y. et al. (2014) Neuregulin autocrine signaling<br />

promotes self-renewal of breast tumor-initiating cells by triggering<br />

HER2/HER3 activation. Cancer Res. 74, 341–352.<br />

Bronisz, A. et al. (2014) Extracellular vesicles modulate the<br />

glioblastoma microenvironment via a tumor suppression<br />

signaling network directed by miR-1. Cancer Res. 74,<br />

738–750.<br />

Tyrosine Kinases Kinase-Disease Associations<br />

Name Group Disease Type Molecular Notes<br />

ALK TK Cancer Trans About one third of large-cell lymphomas are caused by a t(2;5)(p23;q35) translocation<br />

that fuses ALK to nucleophosmin (NPM1A). Other cases caused by fusions of ALK to<br />

moesin, non-muscle myosin heavy chain 9, clathrin heavy chain and other genes.<br />

Several fusions also seen in inflammatory myofibroblastic tumors and expression<br />

has been briefly noted in a range of tumors (Medline:15095281). Proposed as tumor<br />

antigen (Medline:11877285). OMIM:105590.<br />

ALK1<br />

(ACVRL1)<br />

ALK2<br />

(ACVR1)<br />

ALK4<br />

(ACVR1B)<br />

TKL Cardiovascular Mut Fourteen distinct mutations linked to hereditary hemorrhagic telangiectasia type 2<br />

(Osler-Rendu-Weber syndrome 2) [OMIM:600376], associated with intestinal bleeding,<br />

arterial hypertension and arteriovenous malformations. OMIM:601284.<br />

TKL Development Mut Single heterozygous mutation seen in many independent cases of fibrodysplasia ossificans<br />

progressiva [OMIM:135100], causing skeletal malformations and extra-skeletal<br />

bone formation. OMIM:102576.<br />

TKL Cancer Mut, Splice Two somatic truncation mutations seen in pancreatic carcinoma. Unique splice forms<br />

with predicted dominant negative activity. OMIM:601300.<br />

Axl TK Cancer OE Overexpression in tissue culture causes oncogenic transformation. Overexpressed in<br />

several cancers including thyroid (Medline:10411118), ovarian (Medline:15452374),<br />

gastric (Medline:12168903), ER+ breast cancer (Medline:11484958) and acute<br />

myeloid leukemia, where it is associated with poor prognosis (Medline:10482985).<br />

OMIM:109135.<br />

BTK TK Cancer,<br />

Immunity<br />

LOF Mut<br />

EGFR TK Cancer Amp, OE,<br />

GOF Mut<br />

Eph<br />

family<br />

TK<br />

Cancer,<br />

Sensory<br />

LOF mutations cause X-linked agammaglobulinemia [OMIM:300300], arresting<br />

development of B cells and causing recurrent bacterial infections. Truncated splice<br />

forms found in childhood leukemias may underlie radiation resistance of tumors<br />

through inhibition of apoptosis (Medline: 12854903). Inhibitors developed to target B<br />

cell maturation and function. Inhibitor: dasatinib. OMIM:300300.<br />

Overexpressed in breast, head and neck cancers (Medline:15254682) and correlated<br />

with poor survival. Activating somatic mutations seen in lung cancer, corresponding<br />

to minority of patients with strong response to EGFR inhibitor gefinitib. Mutations and<br />

amplification also seen in glioblastoma, and upregulation seen in colon cancer and<br />

neoplasms. In xenografts, inhibitors synergized with cytotoxic drugs in inhibition of many<br />

tumor types (Medline:10815932). Inhibitors: gefinitib/ZD1839 (Astra Zeneca), cetuximab<br />

(mAb, Imclone), erlotinib (OSI/Genentech) lapatinib (Glaxo Smith Kline). OMIM:131550.<br />

A 14-member family of receptor tyrosine kinases with similar functions in intercellular<br />

communication, migration, patterning and angiogenesis. Ephrins are implicated in<br />

development of tumor vasculature and intercellular contacts required for metastasis.<br />

Several members are overexpressed in cancers. Soluble forms (competitive receptors)<br />

have shown some anti-tumor activity and extracellular domains have been used as<br />

tumor-specific antigens.<br />

EphA1 TK Cancer Expr Misexpressed in several cancers and upregulated in head and neck cancer<br />

(Medline:15023838). Downregulated in invasive breast cancer cell lines (Medline:15147954)<br />

and glioblastoma (Medline:14726470). OMIM:179610.<br />

EphA2 TK Cancer OE Overexpressed in many cancers including aggressive ovarian (Medline:15297418),<br />

cervical (Medline:15297167), breast carcinomas (Medline: 15147954) and lung<br />

cancer (Medline:12576426). Expression correlates with degree of angiogenesis<br />

(Medline:14965363), metastasis (Medline: 14767510) and xenograft tumor growth<br />

(Medline:14973554). Soluble receptor inhibits tumor growth and angiogenesis in mice<br />

(Medline:12370823, 14670182). OMIM:176946.<br />

EphA3<br />

(HEK)<br />

TK Cancer Mut Two point mutations seen in a survey of colorectal tumors (Medline:12738854).<br />

Soluble receptors reduce tumor growth and angiogenesis in mouse models (Medline:12370823,<br />

14670182). Nine point mutations found in 294 colon and lung tumors<br />

(Medline:16941478) OMIM:179611.<br />

EphB2 TK Cancer OE, Mut Point mutations seen in prostate cancer (Medline:15300251). Overexpressed and<br />

required for migration of glioblastoma (Medline:15126357). Overexpressed and correlated<br />

with poor survival in breast cancer (Medline:15029258). Overexpression and loss<br />

of heterozygosity seen in colorectal cancers (Medline:11920461, 11166921). Target<br />

for immunoconjugate drug therapy (Medline:14871799). OMIM:600997.<br />

EphB4<br />

(HTK)<br />

TK Cancer OE Required for normal development and angiogenesis of the mammary gland. Angiogenic<br />

functions may be kinase-independent by means of retrograde signaling through its<br />

ephrin-B2 ligand (Medline:15067119). High expression correlates with malignancy in<br />

breast, ovarian and other cancers, but appears to be a survival factor (Medline:16816380,<br />

17353927). Also upregulated in head and neck (Medline:14661437),<br />

endometrial (Medline:12562648) and colon carcinomas (Medline:11801186).<br />

OMIM:600011.<br />

FGFR1 TK Cancer,<br />

Development<br />

Mut, Trans<br />

Point mutations cause Pfeffer syndrome [OMIM:101600] (finger and toe malformations<br />

and other skeletal errors) and dominant Kallmann syndrome 2 [OMIM:147950]. Stem<br />

cell leukemia lymphoma syndrome (SCLL) may be caused by a t(8;13)(p12;q12)<br />

translocation that fuses a zinc finger gene (ZNF198) to FGFR1. Various myeloproliferative<br />

disorders have been linked to translocations that fuse FGFR1 to FOP, FIM, CEP1 or<br />

the atypical kinase, BCR. Inhibitor: SU5402. OMIM:136350.<br />

70 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables 71


Section I: Research Areas<br />

chapter 02: Signaling<br />

Name Group Disease Type Molecular Notes<br />

FGFR2 TK Cancer,<br />

Development<br />

Mut, Amp Mutations cause syndromes with defects in facial and limb development, including<br />

Crouzon syndrome [OMIM:123500], Beare-Stevenson cutis gyrata syndrome<br />

[OMIM:123790], Pfeiffer syndrome [OMIM:101600], Apert syndrome [OMIM:101200],<br />

and Jackson-Weiss syndrome [OMIM:123150]. Somatic mutations seen in gastric<br />

cancer (Medline:11325814). Amplified in gastric (Medline:14595756), breast<br />

(Medline:11564899) and some B cell cancers (Medline:12203778), but deleted in<br />

glioblastoma (Medline:14756442). Promoter SNP associated with breast cancer occurrance<br />

(Medline: 17529967). OMIM:176943.<br />

FGFR3 TK Cancer,<br />

Development<br />

GOF Mut,<br />

Trans<br />

Activating point mutations cause dwarfism, including achondroplasia [OMIM:100800],<br />

hypochrondroplasia [OMIM:146000] and thanatophoric dysplasia [OMIM:187600]; facial<br />

and other morphogenetic disorders, including Crouzon syndrome [OMIM:612247],<br />

craniosynostosis Adelaide type [OMIM:600593], San Diego skeletal displasia<br />

[OMIM:187600] and Muenke syndrome [OMIM:602849]. Translocations t(4;14) involving<br />

the IgH region are common in multiple myeloma and frequently involve FGFR3.<br />

Activated FGFR3 found in 30% of bladder cancers and several cervical cancers but not<br />

in other tumors. Two mutations found in colorectal cancer. OMIM:134934.<br />

FGFR4 TK Cancer SNP A common SNP variant associated with increased motility and progression of breast<br />

cancer (Medline:11830541, but see also Medline:14710228), head and neck cancer<br />

(Medline:15197773) and soft tissue sarcomas (Medline:14601095). Increased<br />

expression seen in pituitary adenomas, pancreatic cancer and breast cancer cell lines.<br />

OMIM:134935.<br />

FLT1<br />

(VEGFR1)<br />

TK Cancer Meth, OE Angiogenesis modulator that may both co-operate with and antagonize KDR/VEGFR2<br />

(Medline:14984769). Overexpressed in several tumor types (Medline:12681367,<br />

9582527, 10738243, 10893635) and an antagonistic soluble form is inhibited in<br />

progressive tumors (Medline:15112269, 14605010, 15173272). Downregulated<br />

by hypermethylation in prostate cancer (Medline:12824880). The soluble receptor<br />

and mutant forms have anti-tumor activity in model systems (Medline:15221961,<br />

15126877). Inhibitors: SU11248, PKC412, CEP-5214. OMIM:165070.<br />

FLT3 TK Cancer GOF Mut Activating mutations found in one third of cases of acute myeloid leukemia (AML), as<br />

well as in acute lymphoblastic leukemia, acute promyelocytic leukemia and myelodysplastic<br />

syndrome. Inhibitors: SU11248 and PKC412. OMIM:136351.<br />

FLT4<br />

(VEGFR3)<br />

TK<br />

Lymphangiogenesis<br />

Fyn TK Cancer,<br />

Epilepsy<br />

HER2<br />

(ErbB2)<br />

Act, LOF<br />

Mut<br />

Lymphatic-specific VEGF receptor. LOF mutations cause hereditary lymphedema<br />

[OMIM:153100], and one case of capillary infantile hemangioma [OMIM:602089].<br />

Expression of FLT4 ligands is seen in a variety of tumors, including colorectal, prostate,<br />

gastric, breast and thyroid cancers where it is associated with lymph node metastasis<br />

(Medline:19787226, 14614015, 14534690, 10430087, 14716745, 15107801). In<br />

model systems, overexpression of ligand increases lymph node metastasis and blocking<br />

antibodies and soluble receptors have been used to reduce lymphangiogenesis<br />

(Medline:15072591, 11175849). Inhibitors: BAY 43-9006, CEP-7055. OMIM:136352.<br />

Induced expression aids in cellular transformation and xenograft metastasis (Medline:3287380,<br />

8325712). In squamous cell carcinoma, Fyn transduces signals from<br />

EGFR and Src and is required for cell migration and invasiveness (Medline:11684709).<br />

Activity linked to migration in a murine melanoma model (Medline:13129922). Appears<br />

to block late stage development of neuroblastoma (Medline:12450793). Mouse knockout<br />

deficient in kindling response, a model for human epilepsy. OMIM:137025.<br />

TK Cancer Amp, OE EGF family receptor. Overexpression induces constitutive activity and the gene is amplified<br />

or overexpressed in up to 30% of breast cancers, correlating with poor survival.<br />

The antibody trastuzumab is approved for treatment of metastatic breast cancer with<br />

HER2 amplification/overexpression. Somatic mutations seen in 4% of lung cancers and<br />

also in breast, gastric, ovarian cancer and glioblastoma. One SNP shows predisposition<br />

to breast and gastric cancer (Medline:10699071, 14520697). Inhibitors: trastuzumab<br />

(mAb, Genentech), lapatinib (Glaxo Smith Kline), PKI-166 (Novartis), EKB-569, CI-1033.<br />

OMIM:164870.<br />

Name Group Disease Type Molecular Notes<br />

Met TK Cancer GOF Mut,<br />

OE, Trans<br />

PDGFRα TK Cancer,<br />

Development<br />

Trans, Del,<br />

Mut<br />

Activating point mutations cause hereditary papillary renal carcinoma [OMIM:605074].<br />

Mutations also seen in sporadic renal cell carcinoma and childhood hepatocellular<br />

carcinoma. Upregulation in carcinomas and sarcomas correlates with metastasis and<br />

poor outcome (Medline:14617781). Some gastric carcinomas harbor a translocation<br />

that creates an activated TPR-Met fusion protein (Medline:2052572). A small molecule<br />

inhibitor (PHA-665752) shows an effect in gastric carcinoma xenografts (Medline:14612533).<br />

Inhibitors: SU11274, PHA-665752, MGCD265 mAbs. OMIM:164860.<br />

Chromosomal rearrangments activate PDGFRa by fusion to Bcr causing atypical chronic<br />

myelogenous leukemia (CML), and to FIP1L1 causing idiopathic hypereosinophilic<br />

syndrome [OMIM:607685]. Activating point mutations cause a minority of gastrointestinal<br />

stromal tumors (GIST). Promoter polymorphisms linked to neural tube defects<br />

including spina bifida (Medline:11175793) as verified by mouse mutant model<br />

(Medline:9826722). Inhibitors: imatinib, SU11248. OMIM:173490.<br />

PDGFRβ TK Cancer Trans, OE A variety of myeloproliferative disorders and cancers result from translocations that activate<br />

PDGFRb by fusion with proteins such as TEL/ETV6, H2, CEV14/TRP11, rabaptin<br />

5 and huntington interacting protein 1. Imatinib treatment of TEL fusions has been<br />

successful. PDGFRb is also overexpressed in metastatic medulloblastoma. Inhibitors:<br />

imatinib, SU11248. OMIM:173410.<br />

Ret TK Cancer,<br />

Development<br />

GOF Mut,<br />

LOF Mut,<br />

Trans<br />

Familial GOF mutations cause endocrine cancers including familial medullary<br />

thyroid carcinoma (FMTC) [OMIM:155240], multiple neoplasia type IIA (MEN2A)<br />

[OMIM:171400] and MEN2B [OMIM:162300], both of which are characterized by predisposition<br />

to FMTC and phaeochromocytoma. Translocation-mediated fusion of Ret to<br />

various genes (H4, ELE1, PKA-R1, TIF1A, TIF1G) results in papillary thyroid carcinoma<br />

[OMIM:188550]. Familial LOF mutations cause Hirschsprung disease [OMIM:142623]<br />

in which enteric (intestinal) neurons fail to develop. OMIM:164761.<br />

RON TK Cancer OE, Splice Functions in cell migration and epithelial-mesenchymal transition. Highly expressed<br />

in tumors including head and neck cancer (Medline:15023838), colon (Medline:12527888),<br />

breast (Medline:9671413) ovarian carcinoma (Medline:12915129)<br />

and renal oncocytoma (Medline:15252311). Unusual splice variants seen in<br />

cancer, including a constitutively active form lacking the extracellular domain<br />

(Medline:12527888, 15289319). Overexpression in mouse lung leads to pulmonary<br />

adenomas (Medline:12214279). OMIM:600168.<br />

SRC TK Cancer Mut, OE,<br />

Act<br />

Tie2 TK Angiogenesis,<br />

Cancer<br />

Mut, OE<br />

Homolog of Rous sarcoma virus v-src. A truncated, activated form seen in approximately<br />

12% of colon cancers in one study and in one endometrial sarcoma but not seen<br />

in several other populations (Medline:9988270, 10804287, 10704743, 10485460,<br />

11161376). Expression and kinase activity are frequently increased in a wide array of<br />

cancers, including tumors from breast, colon, pancreas, lung, ovary and CNS (Medline:9988270).<br />

Inhibitors: SU6656 (Sugen), PD173955, PD166285 (Pfizer), CGP76030<br />

(Novartis), BMS-354825 (Bristol Myers Squibb; Phase 2 cancer). OMIM:190090.<br />

Point mutations cause dominantly inherited venous malformations [OMIM:600195].<br />

Expression is increased in non-small cell lung cancer (Medline:10499626), myeloid<br />

leukemia (Medline:11755466) and hepatocellular carcinoma (Medline:11915032).<br />

Expression is prognostic of metastasis in breast cancer (Medline:12527939,<br />

15026804) and expression and activation correlate with malignancy in astrocytomas<br />

(Medline:14742253). Soluble receptor used to inhibit tumor growth in mice (Medline:14985859).<br />

OMIM:600221.<br />

Yes TK Cancer Amp, Act Ortholog of Yamaguchi sarcoma virus v-yes oncogene, which can transform fibroblasts<br />

in vitro (Medline:3303862). Amplified in one case of gastric cancer and in canine<br />

mammary tumors (Medline:3935622, 10081762). Kinase activity is increased in colon<br />

carcinoma cell lines and tumors (Medline:7690925, 7806032) and in melanoma<br />

cell lines and brain metastases (Medline:7690926, 9681823). Mouse knockouts<br />

have no strong phenotype due to compensation by Src and Fyn (Medline:7958873).<br />

OMIM:164880.<br />

HER3<br />

(ErbB3)<br />

HER4<br />

(ErbB4)<br />

TK Cancer OE EGF family receptor. Kinase domain lacks activity but heterodimerizes with other EGFRs<br />

to transduce growth signals. May be required for HER2 activity (Medline:12853564).<br />

Elevated expression in breast and other tumors is indicative of poor outcome (Medline:12866037,<br />

12896906, 14614020, 15150091, 7656248). A secreted form is<br />

expressed in metastatic prostate cancer (Medline:15141384). OMIM:190151.<br />

TK Cancer Expr Heterodimerizes and signals with other EGF receptors. May act as a tumor suppressor<br />

and is overexpressed in head and neck cancer (Medline: 15476268) but downregulated<br />

in renal cancer (Medline:15360049), papillary carcinoma (Medline:15279891), highgrade<br />

gliomas (Medline:15148612) and invasive breast cancer (Medline:15084248).<br />

OMIM:600543.<br />

Zap-70 TK Immunity Mut Mutations cause selective T cell defect [OMIM:176947], a recessive form of severe<br />

combined immunodeficiency (SCID) exhibiting selective absence of CD8+ T cells.<br />

Reduced expression predicts positive outcome in B cell chronic lymphocytic leukemia.<br />

The SKG mouse is mutated at Zap-70, producing increased numbers of self-reactive T<br />

cells and resulting in chronic arthritis. OMIM:176947.<br />

Molecular: Act Activated • Amp Amplified • Del Deleted • Expr Expression • GOF Gain-of-function • Inh Inhibitor Studies • LOF Loss-offunction<br />

• LOH Loss-of-heterozygosity • Meth Methylation • Model model organism studies • Mut Mutation OE Overexpression • SNP Single<br />

Nucleotide Polymorphism • Splice Splicing change • Trans Translocation<br />

Kit TK Cancer,<br />

Depigmentation<br />

GOF Mut,<br />

LOF Mut,<br />

Act<br />

Activating mutations cause >90% of gastrointestinal stromal tumors (GIST)<br />

[OMIM:606764]; successfully treated with inhibitors imatinib and Sutent (Sutinib,<br />

SU11248). Activating mutations also induce mastocytosis [OMIM:154800] (Medline:15507672).<br />

Autocrine/paracrine stimulation may drive some lung and other tumors<br />

(Medline:15036937). Loss of expression associated with melanoma progression (Medline:9687504).<br />

Familial loss of function mutations cause piebaldism [OMIM:172800]<br />

with defects in hair and skin pigmentation due to lack of melanocytes. OMIM:164920.<br />

LYN TK Cancer Act Mouse knockout develops monocyte/macrophage tumors, while an activated transgene<br />

does not induce tumors. Hyperactivated in acute myeloid leukemia; treatment by<br />

antisense or drug inhibitors reduces proliferation (Medline:10360372). Lyn-specific<br />

inhibitors block proliferation in three prostate cancer cell lines (Medline:14871838).<br />

OMIM:165120.<br />

PhosphoSitePlus® PTM Research<br />

Please see pages 250–253 or visit our website for resources relating to PhosphoSitePlus ® PTM.<br />

www.cellsignal.com/exploration<br />

72 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables<br />

73


HER2/ICD<br />

Section I: Research Areas<br />

ErbB/HER Signaling<br />

HER4/ErbB4<br />

PSD95<br />

Cytoplasm<br />

Nucleus<br />

Gab1<br />

Memo<br />

Cell<br />

Migration<br />

TACE<br />

γ-Sec<br />

BDP1<br />

Shc<br />

Cell<br />

Cycle<br />

Cell<br />

Adhesion<br />

HER2/ErbB2<br />

NRG4 NRG4<br />

CXCR4<br />

Invasion<br />

Metastasis<br />

Akt<br />

mTOR<br />

p70S6K<br />

HER4/ErbB4<br />

HER4/ErbB4<br />

HER2/ErbB2<br />

PI3K<br />

NRG2 NRG3<br />

Vav1<br />

Rac1<br />

NRG1<br />

HER3/ErbB3<br />

HER2/ErbB2<br />

Shc<br />

TNS3<br />

GRB2<br />

p130 Cas<br />

PAK1<br />

MEKK1<br />

JNK<br />

Importin β1<br />

Protein<br />

Nup358<br />

Synthesis<br />

Proliferation Apoptosis Survival<br />

Cellular Proliferation,<br />

DNA Repair<br />

cdc2 PCNA DNA-PK<br />

EGFR/ErbB1<br />

HER2/ErbB2<br />

Increase<br />

Cell Motility<br />

TNS4<br />

FAK<br />

Nck1<br />

Muc1<br />

cdc42<br />

β-Cat<br />

MIG6<br />

Cell<br />

Motility<br />

EGFR/ErbB1<br />

Caveolin<br />

EGF EGF<br />

Paxillin<br />

E-cadherin<br />

Cytoskeletal<br />

Regulation<br />

HB-EGF TGF-α<br />

Crk<br />

GRB2<br />

c-Abl<br />

Cbl<br />

PLC<br />

Calmodulin<br />

CaMK<br />

AREG<br />

Dok<br />

GAB2<br />

SHP-2<br />

AIP4<br />

Endosomemediated<br />

Recycling<br />

of EGFR<br />

PKC<br />

EREG<br />

GEP100<br />

Ub<br />

HRS<br />

MEK<br />

MAPK<br />

EPG<br />

Lysosomemediated<br />

Degradation<br />

of EGFR<br />

Calcineurin<br />

SOS<br />

Raf<br />

EGFR/ErbB1<br />

Ras<br />

βCEL.<br />

ARF6<br />

Yes<br />

Tumor<br />

Invasion<br />

Stat<br />

TSAd<br />

Src<br />

Syk<br />

Jak<br />

MEKK2/3<br />

MEK5<br />

ERK5<br />

MEF2<br />

G Protein-Coupled Receptors<br />

G protein-coupled receptors (GPCRs) constitute a large protein family that sense molecules outside<br />

the cell and activate signal transduction pathways inside the cell. Ligands that bind and activate these<br />

receptors include light-sensitive compounds, odors, pheromones, hormones, and neurotransmitters,<br />

and vary in size from small molecules to peptides to large proteins.<br />

While the exact size of the GPCR superfamily is unknown, nearly 800 different human genes have been<br />

predicted to be a part of the GPCR superfamily from sequence analysis. About 350 of these detect hormones,<br />

growth factors, and other endogenous ligands. Approximately 150 of the GPCRs found in the<br />

human genome have unknown functions. Based on sequence homology and functional similarity, the<br />

GPCRs can be grouped into 6 classes. Class A, Rhodopsin-like receptors, comprise the largest class of<br />

nearly 85% of the genes. Over half of these are predicted to encode olfactory receptors while the rest<br />

are bound by endogenous compounds or are classified as orphan receptors. Class B genes comprise<br />

the secretin receptor family. Class C genes encode metabotropic receptors that bind neurotransmitters<br />

such as glutamate. Class D genes are defined by their fungal mating pheromone receptor functions.<br />

Class E genes are grouped based on cyclic AMP receptor-like function, and Class F genes are grouped<br />

based on Frizzled/Smoothened genotypes.<br />

Metabotropic glutamate receptor 1 (mGluR1) is a Class C GPCR<br />

in the mammalian brain for the neurotransmitter glutamate.<br />

A<br />

kDa<br />

200<br />

140<br />

1 2<br />

mGluR1<br />

B<br />

C<br />

chapter 02: Signaling<br />

Peptide ligand<br />

Neuropeptide Y is expressed<br />

in rat retina.<br />

Neuropeptide Y (D7Y5A) XP ® Rabbit<br />

mAb #11976: Confocal IF analysis of<br />

adult rat retina using #11976 (green).<br />

Blue pseudocolor = DRAQ5 ® #4084<br />

(fluorescent DNA dye).<br />

ICD<br />

HER4<br />

Stat<br />

ICD<br />

HER4<br />

TAB2<br />

N-CoR<br />

EGFR/ErbB-1<br />

EGFR/ErbB-1<br />

E2F Stat CREB Jun Fos Elk Stat<br />

MEF2<br />

100<br />

80<br />

Lanes<br />

1. neonatal mouse brain<br />

2. rat brain<br />

mGluR1 (D5H10) Rabbit mAb #12551: WB analysis of extracts from neonatal mouse<br />

brain and rat brain using #12551 (A). Confocal IF analysis of adult mouse cerebellum<br />

tissue 4X (B) and 20X (C) using #12551 (green). Blue pseudocolor = DRAQ5 ® #4084<br />

(fluorescent DNA dye).<br />

Cell Proliferation, Inflammation,<br />

Genome Instability, Tumor Progression<br />

Cell Growth/Differentiation,<br />

Cell Shape, Chemotaxis<br />

Growth Differentiation<br />

The ErbB receptor tyrosine kinase family consists of four cell surface receptors: ErbB1/ EGFR/HER1, ErbB2/HER2, ErbB3/HER3, and ErbB4/HER4. ErbB receptors are typical<br />

cell membrane receptor tyrosine kinases that are activated following ligand binding and receptor dimerization. Ligands can either display receptor specificity (i.e. EGF, TGF-α,<br />

AR, and Epigen bind EGFR) or bind to one or more related receptors; neuregulins 1–4 bind ErbB3 and ErbB4 while HB-EGF, epiregulin, and β-cellulin activate EGFR and<br />

ErbB4. ErbB2 lacks a known ligand, but recent structural studies suggest its structure resembles a ligand-activated state and favors dimerization.<br />

The ErbB receptors signal through Akt, MAPK, and many other pathways to regulate cell proliferation, migration, differentiation, apoptosis, and cell motility. ErbB family members<br />

and some of their ligands are often over-expressed, amplified, or mutated in many forms of cancer, making them important therapeutic targets. For example, researchers<br />

have found EGFR to be amplified and/or mutated in gliomas and NSCLC while ErbB2 amplifications are seen in breast, ovarian, bladder, NSCLC, as well as several other tumor<br />

types. Preclinical and clinical studies have shown that dual targeting of ErbB receptors display better efficacy than single treatment.<br />

Besides functioning as receptors on the cell surface, ErbB family proteins are also present in the nucleus to act as both kinases and transcriptional regulators. For example,<br />

EGFR could be transported into the nucleus where it functions as a tyrosine kinase to phosphorylate and stabilize PCNA. Similarly, membrane-bound ErbB2 interacts with<br />

importin β1 and Nup358 and migrates to the nucleus via endocytic vesicles. Inside the nucleus, ErbB2 modulates the transcription of multiple downstream genes including<br />

COX-2. In addition, NRG or TPA stimulation promotes ErbB4 cleavage by γ-secretase, releasing an 80 kDa intracellular domain that translocates to the nucleus to induce differentiation<br />

or apoptosis. Upon activation and cleavage, ErbB4 can also form a complex with TAB2 and N-CoR to repress gene expression.<br />

Signaling through ErbB networks is modulated through dense positive and negative feedback and feed forward loops, including transcription-independent early loops and late<br />

loops mediated by newly synthesized proteins and miRNAs. For example, activated receptors can be switched “off” through dephosphorylation, receptor ubiquitination, or<br />

removal of active receptors from the cell surface through endosomal sorting and lysosomal degradation.<br />

Select Reviews:<br />

Arteaga, C.L. and Engelman, J.A. (2014) Cancer Cell 25, 282–303. • Avraham, R. and Yarden, Y. (2011) Nat. Rev. Mol. Cell Biol. 12, 104–117. • Baselga, J. and Swain,<br />

S.M. (2009) Nat. Rev. Cancer 9, 463–475. • Ferrer-Soler, L., Vazquez-Martin, A., Brunet, J., Menendez, J.A., De Llorens, R., and Colomer, R. (2007) Int. J. Mol. Med. 20,<br />

3–10. • Moasser, M.M. (2007) Oncogene 26, 6469–6487. • Tebbutt, N., Pedersen, M.W. and Terrance, G.J. (2014) Nat. Rev. Cancer 13, 663–673. • Yarden, Y. and<br />

Pines, G. (2012) Nat. Rev. Cancer 12, 553–563. • Yarden, Y. and Shilo, B.Z. (2007) Cell 131, 1018.<br />

Retinoic Acid-Induced Gene 1 (RAIG1) is<br />

expressed in lung carcinoma and MKN-45 cells.<br />

RAIG1 (D4S7D) XP ® Rabbit mAb<br />

#12968: IHC analysis of paraffin-embedded<br />

human lung carcinoma using #12968<br />

in the presence of control peptide (A) or<br />

antigen-specific peptide (B). Confocal<br />

IF analysis of MKN-45 (C) and 293T<br />

(D) cells using #12968 (green). Actin<br />

filaments were labeled with DyLight ® 554<br />

Phalloidin #13054. Blue pseudocolor =<br />

DRAQ5 ® #4084 (fluorescent DNA dye).<br />

A<br />

C<br />

B<br />

D<br />

© 2004–2015 Cell Signaling Technology, Inc. • We would like to thank Dr. Jinyan Du, Merrimack Pharmaceuticals Inc., Cambridge, MA, for reviewing this diagram.<br />

74 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstgprotein 75


Section I: Research Areas<br />

GPR50, an orphan<br />

GPCR that inhibits<br />

melatonin receptor<br />

activity, is expressed<br />

in 293T but not<br />

HeLa cells.<br />

Physiological Roles of GPCRs<br />

GPCRs are involved in a wide variety of physiological processes, including visual sensing (photoisomerization<br />

of electromagnetic radiation), taste (release of gustducin in response to bitter- and sweet-tasting<br />

substances), sense of smell (odorants and pheromones), behavioral and mood regulation (bound<br />

neurotransmitters: serotonin, dopamine, GABA and glutamate), regulation of the immune system and<br />

inflammatory response (involvement of chemokine receptors), autonomic nervous system transmission<br />

(both sympathetic and parasympathetic systems respond to bound ligand), cell density sensing, homeostasis<br />

modulation (water balance), and involvement in growth and metastasis of some tumor types. It is<br />

not surprising then that GPCRs are involved in many diseases, and are a significant drug target.<br />

GPR50 (D1D6I) Rabbit mAb #14032:<br />

Confocal IF analysis of 293T (left, positive),<br />

or HeLa (right, negative) cells using<br />

#14032 (green). Actin filaments were<br />

labeled with DyLight 554 Phalloidin<br />

#13054 (red). Blue pseudocolor =<br />

DRAQ5 ® #4084 (fluorescent DNA dye).<br />

GPCR Structure and Signaling<br />

GPCRs are characterized by an extracellular N-terminus, followed by seven transmembrane α-helices<br />

connected by three intracellular and three extracellular loops, and finally an intracellular C-terminus.<br />

Upon binding a ligand, the seven transmembrane helices of the GPCR arrange themselves into a<br />

tertiary structure resembling a barrel, forming a cavity within the plasma membrane, thereby exposing<br />

the external ligand-binding domain (EL-2), though ligands may also bind elsewhere, as is the case for<br />

bulky ligands for the metabotropic receptors.<br />

GPCRs are known to exist in a conformational equilibrium between active and inactive biological states.<br />

Three types of ligands exist: agonists, which shift the equilibrium in favor of active states; inverse<br />

agonists, which shift the equilibrium in the favor of inactive states; and neutral antagonists, which don’t<br />

affect the equilibrium. Agonist binding creates a conformational change in the receptor that activates<br />

an associated G protein (trimer of α, β, and γ subunits, known as Gα, Gβ, and Gγ, respectively), causing<br />

the G protein to detach from the receptor and separate into GTP-bound Gα and a Gβ/γ dimer. G protein<br />

activation initiates signaling pathways that regulate cell proliferation, apoptosis, and cytoskeletal<br />

rearrangements—see details in the signaling pathway to follow. Termination of GPCR signaling occurs<br />

through phosphorylation by GCPR kinases (GRKs) and binding of β-arrestin proteins, which leads to<br />

clathrin-mediated receptor internalization and degradation or recycling.<br />

Commonly Studied GPCR Targets<br />

Target M P<br />

AKAP1<br />

•<br />

AKAP5<br />

•<br />

β1-Adrenergic Receptor<br />

•<br />

β2-Adrenergic Receptor<br />

•<br />

β-Arrestin 1<br />

•<br />

Phospho-β-Arrestin 1 (Ser412)<br />

•<br />

β-Arrestin 1/2<br />

•<br />

β-Arrestin 2<br />

•<br />

Arrestin 1/S-Arrestin<br />

•<br />

Gα (pan)<br />

•<br />

Gα (z)<br />

•<br />

Select Citations:<br />

Li, M. et al. (2014) T-cell Immunoglobulin and ITIM Domain<br />

(TIGIT) Receptor/Poliovirus Receptor (PVR) Ligand Engagement<br />

Suppresses Interferon-gamma Production of Natural<br />

Killer Cells via beta-Arrestin 2-mediated Negative Signaling.<br />

J. Biol. Chem. 289, 17647–17657.<br />

Seitz, K. et al. (2014) beta-Arrestin interacts with the beta/<br />

gamma subunits of trimeric G-proteins and dishevelled in<br />

the Wnt/Ca(2+) pathway in xenopus gastrulation. PLoS One<br />

29, e87132.<br />

Coggins, N.L. et al. (2014) CXCR7 Controls Competition<br />

for Recruitment of beta-Arrestin 2 in Cells Expressing Both<br />

CXCR4 and CXCR7. PLoS One 4, e98328.<br />

Fraisier, C. et al. (2014) Kinetic analysis of mouse brain<br />

proteome alterations following Chikungunya virus infection<br />

before and after appearance of clinical symptoms. PLoS One<br />

9, e91397.<br />

Kriz, V., et al. (2014) beta-arrestin promotes Wnt-induced<br />

low density lipoprotein receptor-related protein 6 (Lrp6)<br />

phosphorylation via increased membrane recruitment of<br />

Amer1 protein. J. Biol. Chem. 289, 1128–1141.<br />

Target M P<br />

Gα (o)<br />

Gα (i)<br />

GNB3<br />

mGluR1<br />

mGluR2<br />

GABA(B)R1<br />

GABA(B)R2<br />

Phospho-μ-Opioid Receptor (Ser375)<br />

RAIG1<br />

RGS4<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

• •<br />

•<br />

• •<br />

•<br />

Thathiah, A. et al. (2013) beta-arrestin 2 regulates Abeta<br />

generation and gamma-secretase activity in Alzheimer’s<br />

disease. Nat. Med. 19, 43–49.<br />

Stephenson, J.R. et al. (2013) Brain-specific angiogenesis<br />

inhibitor-1 signaling, regulation, and enrichment in the<br />

postsynaptic density. J. Biol. Chem. 288, 22248–22256.<br />

Erickson, C.E. et al. (2013) The beta-blocker Nebivolol Is a<br />

GRK/beta-arrestin biased agonist. PLoS One 8, e71980.<br />

Nelson, C.D. et al. (2013) Gpr3 stimulates Abeta production<br />

via interactions with APP and beta-arrestin2. PLoS One 8,<br />

e74680.<br />

Salomonnson, E. et al. (2013) Imaging CXCL12-CXCR4<br />

signaling in ovarian cancer therapy. PLoS One 8, e51500.<br />

Canals, M. et al. (2012) Ubiquitination of CXCR7 controls<br />

receptor trafficking. PLoS One 7, e34192.<br />

Cintra, D.E. et al. (2012) Unsaturated fatty acids revert<br />

diet-induced hypothalamic inflammation in obesity. PLoS<br />

One. 7, e30571.<br />

chapter 02: Signaling<br />

These protein targets represent key<br />

nodes within GPCR signaling pathways<br />

and are commonly studied in GPCR<br />

research. Primary antibodies, antibody<br />

conjugates, and antibody sampler kits<br />

containing these targets are available<br />

from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

12<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for β-arrestin 1/2 have<br />

been cited over 12 times in high-impact,<br />

peer-reviewed publications from the<br />

global research community.<br />

Arrestin proteins<br />

function as negative<br />

regulators of GPCR<br />

signaling and are<br />

widely expressed in<br />

many cell lines.<br />

A<br />

kDa<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

1 2 3<br />

β-Arrestin 1<br />

B<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

1 2 3 4 5<br />

β-Arrestin 2<br />

β-Arrestin 1 (D8O3J) Rabbit mAb<br />

#12697: WB analysis of extracts from<br />

MDA-MB-435, Hep G2, and PANC-1<br />

cells using #12697 (A).<br />

β-Arrestin 2 (C16D9) Rabbit mAb<br />

#3857: WB analysis of extracts from<br />

various cell lines using #3857 (B).<br />

Lanes<br />

1. MDA-MB-435<br />

2. Hep G2<br />

3. PANC-1<br />

Lanes<br />

1. HeLa<br />

2. NIH/3T3<br />

3. A10<br />

4. COS<br />

5. Jurkat<br />

Select Reviews<br />

Audet, M. and Bouvier, M. (2012) Cell 151, 14−23. • Irannejad, R. and von Zastrow, M. (2014) Curr. Opin. Cell Biol. 27,<br />

109−116. • Kang, D.S., Tian, X., and Benovic, J.L. (2014) Curr. Opin. Cell Biol. 27, 63−71. • O’Hayre, M., Degese, M.S., and<br />

Gutkind, J.S. (2014) Curr. Opin. Cell Biol. 27, 126−135. • Zhou, L. and Bohn, L.M. (2014) Curr. Opin. Cell Biol. 27, 102−108.<br />

76 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstgprotein<br />

77


Section I: Research Areas<br />

chapter 02: Signaling<br />

G Protein-Coupled Receptor Signaling: Overview<br />

RhoA<br />

β-Arrestin<br />

AP2<br />

clathrin<br />

Receptor<br />

Internalization<br />

β-Arrestin<br />

PI3K<br />

Akt<br />

Examples<br />

Src<br />

Ras<br />

Raf<br />

Erk<br />

All GPCRs<br />

p90RSK<br />

Bad<br />

M1, H1,<br />

V2R, OXTR,<br />

AT 1 R, PAR<br />

Gα q<br />

GDP<br />

Gα q<br />

GTP<br />

β,γ<br />

β,γ<br />

GRK<br />

GPR55, SDF-1,<br />

PAR, S1P<br />

Gα 12/13 β,γ<br />

GDP<br />

Gα 12/13<br />

GTP<br />

β,γ<br />

β1-Adrenergic R<br />

Wnt Ligand/Frizzled<br />

Gα s<br />

GDP<br />

Gα s<br />

GTP<br />

β,γ<br />

β,γ<br />

CXCR4,<br />

GABA-B, LPA<br />

Gα i β,γ<br />

GDP<br />

Gα i β,γ<br />

GTP<br />

MMPs<br />

VEGFR, PDGFR,<br />

EGFR<br />

Transactivation<br />

Src<br />

PI3K<br />

Ion<br />

Channels<br />

Stat3<br />

PLCβ PKC Rac RhoA Axin AC<br />

CDC42 PI3K PLCβ<br />

Ras<br />

[cAMP]<br />

CDC42<br />

IP3<br />

PAK<br />

Akt<br />

Raf<br />

PKA<br />

DAG<br />

ROCK LATS1/2 GSK-3β<br />

Ca 2+<br />

SOS<br />

MLK WASP<br />

PAK<br />

PKC<br />

JNK p38 β-catenin CREB<br />

GSK-3 Erk<br />

Ras<br />

Glycogen and<br />

Actin<br />

Fatty Acid<br />

JNK<br />

Remodeling<br />

Synthesis<br />

LATS1/2<br />

Raf<br />

Bad mTOR IKK<br />

Survival Protein<br />

Bcl-2<br />

Synthesis<br />

YAP/TAZ<br />

p53<br />

Inactive<br />

NF-κB<br />

RTK<br />

G Protein-Coupled Receptor Signaling to MAP Kinase/Erk<br />

Gi-Coupled<br />

Receptor<br />

Dynamin<br />

G i<br />

βγ<br />

PLCβ<br />

β-Arrestin GRK2<br />

Src<br />

PI3Kγ<br />

Receptor<br />

Internalization<br />

endosome<br />

H+<br />

JNK3<br />

Focal Adhesions<br />

Catecholamines<br />

MP<br />

Src-like<br />

Src c-Raf<br />

β-Arrestin1 MEK<br />

Erk<br />

C-TAK1<br />

TH<br />

PKC<br />

RTKs<br />

IMP<br />

Src<br />

GRB2<br />

RasGEF<br />

SOS<br />

Ras<br />

GAP<br />

MEK1/2<br />

KSR<br />

Src<br />

FAK<br />

Gα<br />

PLCβ q<br />

PYK2<br />

GTP<br />

RACK1<br />

IP 3<br />

[Ca 2+ ]<br />

PKC<br />

Ras<br />

c-Raf<br />

Erk1/2<br />

Integrins<br />

Ras<br />

GRP<br />

CaMKII,-IV<br />

Ras<br />

GRF<br />

c-Raf B-Raf<br />

Heterodimer<br />

Gq-Coupled<br />

Receptor<br />

Syn<br />

GAP<br />

RGS<br />

EPAC<br />

Rap1<br />

B-Raf<br />

DUSP6<br />

cPLA 2<br />

Synapsins<br />

AC<br />

Gα s<br />

GTP<br />

[cAMP]<br />

PKA<br />

Gs-Coupled<br />

Receptor<br />

RGS<br />

Bcl-xL<br />

Erk<br />

YAP/TAZ<br />

Cytoplasm<br />

Erk1/2 p90RSK<br />

PEA-15<br />

p90RSK<br />

cdc25<br />

c-Myc<br />

p53<br />

c-Fos<br />

c-Jun<br />

YAP/TAZ<br />

G protein-coupled receptors (GPCRs), also known as 7-transmembrane receptors (7-TMR), form the largest family of cell membrane receptors with over 800 members.<br />

GPCRs transmit extracellular signals initiated by ligands such as neurotransmitters, hormones, chemokines, and various lipid mediators to the cell interior through coupling to<br />

heterotrimeric G proteins. G proteins consist of a GDP-bound Gα subunit complexed to β and γ subunits. Upon ligand binding, the α subunit exchanges GDP for GTP, causing<br />

Gα activation, dissociation from the β/γ subunits, and initiation of signaling pathways that regulate numerous cellular responses, including proliferation, apoptosis, and cytoskeletal<br />

rearrangements. There are four classes of Gα proteins, each with a unique signaling profile: Gαs, Gαi, Gαq, and Gα12/13. Gαs-coupled receptors activate adenylate<br />

cyclase, generating cAMP and signaling through PKA . Gαi-coupled receptors inhibit AC and activate CDC42 and PI3K/Akt. Gαq signals through phospholipase C β (PLCβ),<br />

Rac, and RhoA to regulate cell proliferation and survival by MAPK pathways and actin remodeling via Rock. Gα12/13 activates RhoA, leading to cytoskeletal rearrangements<br />

as well as activation of the JNK, p38, and Hippo pathways. The β/γ subunits, particularly those associated with Gαi, can initiate signaling independent of Gα proteins, resulting<br />

in regulation of Akt, PLCβ, and various ion channels. In certain cell types, GPCRs can transactivate receptor tyrosine kinases (RTKs) such as EGFR, PDGFR, and VEGFR,<br />

thereby activating classical pathways of cell proliferation and survival. Termination of GPCR signaling occurs through phosphorylation by GPCR kinases (GRKs) and binding of<br />

β-arrestin proteins, which leads to clathrin-mediated receptor internalization and degradation or recycling. β-arrestins can also propagate signals independent of Gα proteins,<br />

initiating MAPK/Erk, Akt, and RhoA pathways.<br />

Select Reviews:<br />

Audet, M. and Bouvier, M. (2012) Cell 151, 14−23. • Entschladen, F., Zänker, K.S., and Powe, D.G. (2011) Cell Cycle 10, 1086−1091. • Kahn, S.M., Sleno, R., Gora, S.<br />

et al. (2013) Pharmacol. Rev. 65, 545–577. • Lappano, R. and Maggiolini, M. (2012) Acta. Pharmacol. Sin. 33, 351−362. • Lappano, R. and Maggiolini, M. (2011) Nat.<br />

Rev. Drug Discov. 10, 47−60. • O’Havre, M., Degese, M.S., and Gutkind, J.S. (2014) Curr. Opin. Cell Biol. 27C, 126−135. • Raiagopal, S., Raiagopal, K., and Lefkowitz,<br />

R.J. (2010) Nat. Rev. Drug Discov. 9, 373−386. • Shukla, A.K., Xiao, K., and Lefkowitz, R.J. (2011) Trends Biochem. Sci. 36, 457−469. • Smrcka, A.V. (2008) Cell. Mol.<br />

Life Sci. 65, 2191−2214. • Yu, F.X. and Guan, K.L. (2013) Genes Dev. 27, 355−371.<br />

CREB<br />

NF-κB<br />

Erk<br />

Nucleus<br />

Nucleus<br />

p90RSK<br />

Transcription<br />

Erk1/2<br />

FoxO3<br />

Tumorigenesis<br />

MSK1/2<br />

MAPKAPK2<br />

Progression<br />

of Cell Cycle<br />

G protein-coupled receptors (GPCRs) are activated by a wide variety of external stimuli. Upon receptor activation, the G protein exchanges GDP for GTP, causing the dissociation<br />

of the GTP-bound α and β/γ subunits and triggering diverse signaling cascades. Receptors coupled to different heterotrimeric G protein subtypes can utilize different scaffolds<br />

to activate the small G protein/ MAPK cascade, employing at least three different classes of Tyr kinases. Src family kinases are recruited following activation of PI3Kγ by β/γ<br />

subunits. They are also recruited by receptor internalization, crossactivation of receptor Tyr kinases, or by signaling through an integrin scaffold involving Pyk2 and/or FAK.<br />

GPCRs can also employ PLCβ to mediate activation of PKC and CaMKII, which can have either stimulatory or inhibitory consequences for the downstream MAPK pathway.<br />

Select Reviews:<br />

Aoki, Y., Niihori, T., Narumi, Y., Kure, S., and Matsubara, Y. (2008) Hum. Mutat. 29, 992–1006. • Caunt, C.J., Finch, A.R., Sedgley, K.R., and McArdle, C.A. (2006) Trends<br />

Endocrinol. Metab. 17, 276–283. • Goldsmith, Z.G. and Dhanasekaran, D.N. (2007) Oncogene 26, 3122–3142. • Kim, E.K. and Choi, E.J. (2010) Biochim. Biophys. Acta.<br />

1802, 396–405. • McKay, M.M. and Morrison, D.K. (2007) Oncogene 26, 3113–3121.<br />

© 2014–2015 Cell Signaling Technology, Inc. • We would like to thank Dr. Jonathan Violin, Trevena Inc., King of Prussia, PA for reviewing this diagram.<br />

78 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

© 2002–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. John Blenis, Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

www.cellsignal.com/cstpathways 79


Section I: Research Areas<br />

chapter 02: Signaling<br />

Activators and<br />

Inhibitors of PKC<br />

#9841 Bisindolylmaleimide I,<br />

Hydrochloride<br />

Potent inhibitor of<br />

PKC family members<br />

#11916 Chelerythrine Chloride<br />

Cell-permeable inhibitor of<br />

PKC; activates SAPK/JNK<br />

and p38-MAPK; induces<br />

apoptosis in some cell lines<br />

#12060 Gö6976<br />

Potent inhibitor of<br />

calcium-dependent<br />

PKC family members<br />

#9995 Ionomycin, Calcium Salt<br />

Calcium ionophore;<br />

activates calcium-/calmodulin-dependent<br />

kinase and<br />

calcium-dependent PKCs<br />

#9953 Staurosporine<br />

Very broad, ATP-competitive<br />

protein kinase inhbitor (PKC,<br />

PKA, Src, CaM kinase, etc.)<br />

#4174 TPA (12-O-Tetradecanoylphorbol-13-Acetate)<br />

Cell permeable, potent<br />

activator of PKC; also used<br />

to activate MAPK.<br />

Calcium, cAMP, and Lipid Signaling<br />

Calcium is a critical regulator of a diverse set of cellular functions, and maintaining calcium homeostasis<br />

is a highly regulated mechanism involving numerous proteins and hormones. Calcium ion channels and<br />

pumps regulate calcium entry and exit from cells in response to a stimulus. Inside the cell, calcium binding<br />

proteins regulate local calcium concentrations and help transduce calcium signals. Enzymes such as<br />

PKC and PLC respond to elevated levels of calcium and transmit signals to downstream signaling nodes.<br />

Protein Kinase C (PKC)<br />

Protein kinase C (PKC) family members regulate numerous cellular responses including gene expression,<br />

protein secretion, cell proliferation, and the inflammatory response. The basic protein structure<br />

includes an N-terminal regulatory region connected to a C-terminal kinase domain by a hinge region.<br />

PKC enzymes contain an auto-inhibitory pseudosubstrate domain that binds a catalytic domain<br />

sequence to inhibit kinase activity. Differences among PKC regulatory regions allow for variable second<br />

messenger binding and are the basis for the division of the PKC family into 3 broad groups. Conventional<br />

PKC enzymes (cPKC; isoforms PKCα, PKCβ, and PKCγ) contain functional C1 and C2 regulatory<br />

domains; cPKC enzyme activation requires binding of diacylglycerol (DAG) and a phospholipid to the<br />

C1 domain, and calcium binding to the C2 domain. Novel PKC enzymes (nPKC; isoforms PKCδ, PKCε,<br />

PKCη, and PKCθ) also require DAG binding for activation but contain a novel C2 domain that does not<br />

act as a calcium sensor. Distantly related protein kinase D proteins are often associated with novel PKC<br />

enzymes as they respond to DAG but not calcium stimulation. Atypical enzymes (aPKC; isoforms PKCζ<br />

and PKCι/λ) contain a nonfunctional C1 domain and lack a C2 domain, requiring no second messenger<br />

binding for aPKC activation.<br />

The enzyme PDK1 or a close relative is responsible for PKC activation. Control of PKC activity is regulated<br />

through three distinct phosphorylation events. Phosphorylation occurs in vivo at Thr500 in the<br />

activation loop, at Thr641 through autophosphorylation, and at the C-terminal hydrophobic site Ser660.<br />

Phospholipase C (PLC)<br />

Phosphoinositide-specific phospholipase C (PLC) plays a significant role in transmembrane signaling.<br />

In response to extracellular stimuli such as hormones, growth factors, and neurotransmitters, PLC<br />

hydrolyzes phosphatidylinositol 4,5-bisphosphate (PIP 2 ) to generate the secondary messengers inositol<br />

1,4,5-triphosphate (IP3) and diacylglycerol (DAG). At least four families of PLCs have been identified:<br />

PLCβ, PLCγ, PLCδ and PLCε. PLC activity is largely regulated by phosphorylation. For example,<br />

phosphorylation of PLCβ3 at Ser1105 by PKA or PKC inhibits activity, whereas phosphorylation of PLCγ<br />

at Tyr 771, 783, and 1245 by both receptor (EGFR) and nonreceptor tyrosine kinases (Syk) results<br />

in activation. In addition, members of the PLCβ subfamily are activated by the α- or β/γ-subunits of<br />

heterotrimeric G-proteins and play an important role in GPCR signaling cascades.<br />

Growth factor stimulation results in phosphorylation of PLCγ1 at Tyr783.<br />

A<br />

Phospho-PLCγ1 (Tyr783) (D6M9S) Rabbit mAb #14008: Confocal IF analysis of A-431 cells, untreated (A), treated with hEGF #8916<br />

(100 ng/ml, 5 min) (B), or treated with hEGF #8916 (100 ng/ml, 5 min) and λ phosphatase (C), using #14008 (green). Blue pseudocolor<br />

= DRAQ5 ® #4084 (fluorescent DNA dye). WB analysis of extracts from NIH/3T3 cells, untreated (-) or stimulated with hPDGF-BB #8912<br />

(5 min; +), and from A-431 cells, untreated (-) or stimulated with hEGF #8916 (5 min; +) (D), using #14008 (upper) and β-Actin (D6A8)<br />

Rabbit mAb #8457 (lower).<br />

B<br />

C<br />

D<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

60<br />

50<br />

40<br />

30<br />

1 2<br />

– + – –<br />

– – – +<br />

Phospho-<br />

PLCγ1<br />

(Tyr783)<br />

β-Actin<br />

hPDGF-BB<br />

hEGF<br />

Akt/PKB<br />

p70S6K<br />

PDK1<br />

PKCζ/λ PKCδ PKCθ PKCµ PKCα/βII<br />

Growth Factors,<br />

Insulin, etc.<br />

PI3K<br />

Activated<br />

By TPA<br />

Receptor<br />

Calcium Binding Proteins<br />

Calcium is a critical second messenger for intracellular signaling pathways that regulate a diverse<br />

range of biological functions such as cell growth, motility, contractility, membrane trafficking, neurotransmitter<br />

release, apoptosis, and differentiation. Low molecular weight calcium binding proteins<br />

such as calmodulin, calbindin, S-100, paravalbumin, and troponin C bind to calcium via EF hand<br />

domains and help transduce intracellular signals by activating downstream target proteins or act as<br />

calcium buffering proteins to regulate local calcium concentrations. For example, the calcium binding<br />

protein calbindin is expressed in discrete neuronal populations within the CNS, including Purkinje cells,<br />

and is thought to act as an intracellular calcium buffering protein. Calbindin is highly expressed in<br />

neurons during migration and differentiation.<br />

MARCKs<br />

Calcium binding protein calbindin is expressed<br />

in discrete neuronal populations within the CNS.<br />

Phorbol ester TPA<br />

activates PKCδ, resulting<br />

in phosphorylation<br />

at Thr505.<br />

kDa<br />

105<br />

76<br />

57<br />

Phospho-<br />

PKCδ<br />

(Thr505)<br />

0 15 30 60 120 240 TPA (min)<br />

Phospho-PKCδ (Thr505) Antibody<br />

#9374: WB analysis of extracts from<br />

U-937 cells, untreated or TPA-treated<br />

(0.2 µM), using #9374.<br />

PKCθ is a novel protein kinase C predominantly expressed in T cells.<br />

CD3<br />

10 4<br />

10 3<br />

10 2<br />

10 1<br />

10 0 10 1 10 2 10 3 10 4<br />

10 0<br />

10 0 10 1 10 2 10 3 10 4<br />

Rabbit Isotype Control<br />

PKCθ<br />

PKCθ (E1I7Y) Rabbit mAb<br />

#13643: Flow cytometric analysis<br />

of mouse splenocytes using Rabbit<br />

(DA1E) mAb IgG XP ® Isotype Control<br />

#3900 (left) and #13643 (right).<br />

Splenocytes were co-stained with<br />

anti-CD3 APC and the Anti-rabbit<br />

IgG (H+L), F(ab’) 2 Fragment (PE<br />

Conjugate) #8885 was used as a<br />

secondary antibody.<br />

Calbindin (D1I4Q) XP ® Rabbit mAb<br />

#13176: Confocal IF analysis of normal rat<br />

brain (A) using #13176 (green) and GFAP<br />

(GA5) Mouse mAb #3670 (red). Blue pseudocolor<br />

= DRAQ5 ® #4084 (fluorescent DNA<br />

dye). IHC analysis of paraffin-embedded rat<br />

brain (B) using #13176.<br />

A<br />

B<br />

80 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstcalcium 81


Section I: Research Areas<br />

Calcium pump<br />

protein ATP2A2/<br />

SERCA2 is widely<br />

expressed in<br />

many cell lines.<br />

Calcium Channels and Pumps<br />

Maintaining proper calcium concentrations within the cell is critical for effective cell signaling and<br />

requires a variety of channels and pumps to transport calcium ions across intracellular and plasma<br />

membranes. Ion channels move calcium ions into the cell or out from intracellular storage compartments<br />

(with the gradient), effectively raising cytoplasmic calcium concentrations. The three types of<br />

calcium ion channels are broadly classified by their ability to open in response to a ligand, second<br />

messenger, or membrane potential (voltage-dependent calcium channels; VDCC). For example, the IP3<br />

receptor requires the second messenger inositol 1,4,5-triphosphate (IP3) for activation and is located<br />

on the endoplasmic reticulum (ER) where it regulates release of intracellular calcium stores.<br />

As cytoplasmic calcium concentrations rise, calcium can be transported back outside the cell or into<br />

storage within the sarcoplasmic reticulum or ER by calcium pumps. Calcium pump proteins are calcium-<br />

ATPases that use the energy of ATP hydrolysis to retrotransport calcium across plasma or ER membranes,<br />

thus maintaining the calcium gradient necessary for rapid signaling. For example, the calcium<br />

pump ATP2A2/SERCA2 is responsible for regulating calcium transport across sarcoplasmic reticulum and<br />

ER membranes, and its activity can be regulated through a variety of post-translational modifications.<br />

ATP2A2/SERCA2 Antibody #4388: WB analysis<br />

of extracts from various cell lines using #4388.<br />

Lanes<br />

1. Hep G2<br />

2. RD<br />

3. C2C12<br />

4. Jurkat<br />

5. NIH/3T3<br />

6. PC-12<br />

IP3 receptor, a calcium ion channel activated by<br />

second messengers, is expressed in brain tissue.<br />

IP3 Receptor 1 (D53A5) Rabbit mAb #8568: WB analysis of extracts from mouse and<br />

rat brain using #8568.<br />

82 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

kDa<br />

200<br />

140<br />

100<br />

1 2<br />

IP3<br />

Receptor 1<br />

Lanes<br />

1. mouse brain<br />

2. rat brain<br />

Mitochondrial Calcium Uniporter<br />

The mitochondrial calcium uniporter (MCU) is a calcium channel specifically located within the<br />

mitochondrial inner membrane. Mitochondrial calcium uniporter regulator 1 (MCUR1) is a multi-pass,<br />

transmembrane protein that directly interacts with MCU and plays an essential role in the regulation of<br />

calcium uptake and maintenance of mitochondrial calcium homeostasis. Regulation of MCU by MCUR1<br />

may be critical for a variety of cellular functions, including signal transduction, bioenergetics, and cell<br />

death and survival.<br />

Mitochondrial calcium uniporter<br />

is expressed in many cell lines.<br />

MCUR1 Antibody #13706: WB analysis of extracts from<br />

various tissues and cell lines using #13706 (upper) and<br />

β-Actin (D6A8) Rabbit mAb #8457 (lower).<br />

1 2 3 4 5 6<br />

kDa<br />

60<br />

50<br />

40<br />

30<br />

20<br />

50<br />

40<br />

ATP2A2/<br />

SERCA2<br />

1 2 3 4 5 6<br />

MCUR1<br />

β-Actin<br />

Lanes<br />

1. mouse kidney<br />

2. mouse testis<br />

3. human kidney<br />

4. rat kidney<br />

5. C6<br />

6. Neuro-2a<br />

Select Reviews<br />

Bublitz, M., Musgaard, M., Poulsen, H., et al. (2013) J. Biol. Chem. 288, 10759–10765. • Freeley, M., Kelleher, D., Long, A.<br />

(2011) Cell. Signal. 23, 753–762. • Newton, A.C. (2010) Am. J. Physiol. Endocrinol. Metab. 298, 395–402. • Patron, M.,<br />

Raffaello, A., Granatiero, V. et al. (2013) J. Biol. Chem. 288, 10750–10758. • Rossi, A.M., Tovey, S.C., Rahman, T., et al.<br />

(2012) Biochim. Biophys. Acta. 1820, 1214–1227. • Yáñez, M., Gil-Longo, J., and Campos-Toimil, M. (2012) Adv. Exp. Med.<br />

Biol. 740, 461–482. • Yang, Y.R., Follo, M.Y., Cocco, L., and Suh, P.G. (2013) Adv. Biol. Regul. 53, 232–241.<br />

Commonly Studied Calcium, cAMP, and Lipid Signaling Targets<br />

Target M P S Target M P S Target M P S<br />

β1-Adrenergic Receptor • PDE5<br />

• Phospho-Phospholamban<br />

(Ser16/Thr17)<br />

•<br />

AKAP1 •<br />

PIP4K2A •<br />

AKAP5 •<br />

PIP4K2B • PLCβ3 • •<br />

Annexin A1 • • PIP5K1A • Phospho-PLCβ3 •<br />

(Ser537)<br />

Annexin A2 •<br />

PIP5K1C •<br />

Phospho-PLCβ3<br />

Annexin A7 • PKA C-α • • •<br />

•<br />

(Ser1105)<br />

ApoA1 •<br />

Phospho-PKA C-α • •<br />

PLCγ1 • •<br />

ApoA4 •<br />

(Thr197)<br />

Phospho-PLCγ1<br />

ApoA5 •<br />

PKA RI-α/β •<br />

• •<br />

(Tyr783)<br />

ApoM •<br />

Phospho-PKC (pan) •<br />

Phospho-PLCγ1 • •<br />

(βII Ser660)<br />

ASM<br />

•<br />

(Ser1248)<br />

Phospho-PKC (pan)<br />

ATP2A1/SERCA1 • •<br />

•<br />

PLCγ2<br />

•<br />

(γ Thr514)<br />

ATP2A2/SERCA2 • •<br />

Phospho-PLCγ2<br />

Phospho-PKC (pan) •<br />

•<br />

Pan-Calcineurin A •<br />

(Tyr759)<br />

(ζ Thr410)<br />

Calmodulin •<br />

Phospho-PLCγ2<br />

PKCα<br />

•<br />

•<br />

(Tyr1217)<br />

Calumenin •<br />

Phospho-PKCα/β II •<br />

PLD1<br />

CBARA1/MICU1 •<br />

•<br />

(Thr638/641)<br />

Phospho-PLD1 (Thr147)<br />

CFTR<br />

• PKCδ • •<br />

•<br />

Phospho-PLD1 (Ser561)<br />

Choline Kinase α •<br />

Phospho-PKCδ (Tyr311) •<br />

•<br />

PLD2<br />

cPLA2 • • Phospho-PKCδ (Thr505) •<br />

•<br />

PRK2<br />

Phospho-cPLA2 • Phospho-PKCδ/θ •<br />

• •<br />

(Ser505)<br />

(Ser643/676)<br />

PKA RI-α •<br />

Cyclic AMP •<br />

PKCε • • RyR1 •<br />

DAG Lipase β •<br />

PKCθ • • S100A1 •<br />

Gα (pan) • Phospho-PKCθ (Thr538) • S100A4 • •<br />

Gα (z)<br />

• PKCζ • • • S100A6 •<br />

Gα (i)<br />

• Phospho-PKCζ/λ • S100A10 •<br />

Gα (o)<br />

•<br />

(Thr410/403)<br />

S100B •<br />

Gelsolin • • •<br />

PKD/PKCµ • S100P<br />

• •<br />

INPP4b • •<br />

Phospho-PKD/PKCµ • nSMase1 •<br />

(Ser744/748)<br />

IP3 Receptor • •<br />

SPHK1 • •<br />

Phospho-PKD/PKCµ<br />

Phospho-IP3 Receptor • •<br />

• STIM1 • •<br />

(Ser916)<br />

(Ser1756)<br />

STIM2<br />

•<br />

PKD2<br />

MARCKS •<br />

• •<br />

TGM2 •<br />

PKD3/PKCν<br />

Phospho-MARCKS • •<br />

•<br />

TRPV3<br />

•<br />

(Ser152/156)<br />

Phospho-PRK1 •<br />

TSPO<br />

•<br />

(Thr774)/PRK2 (Thr816)<br />

Phospho-MARCKS •<br />

WFS1<br />

(Ser167/Ser170)<br />

Phospholamban •<br />

• •<br />

NIPSNAP1 •<br />

Select Citations:<br />

Volk, L. et al. (2013) PKM-zeta is not required for hippocampal<br />

synaptic plasticity, learning and memory. Nature<br />

493, 420–423.<br />

Paul, S. et al. (2014) T cell receptor signals to NF-kappaB<br />

are transmitted by a cytosolic p62-Bcl10-Malt1-IKK signalosome.<br />

Sci. Signal. 7, ra45.<br />

Dusaban, S.S. et al. (2013) Phospholipase C epsilon links<br />

G protein-coupled receptor activation to inflammatory<br />

astrocytic responses. Proc. Natl. Acad. Sci. USA 110,<br />

3609–3614.<br />

Xiang, S.Y. et al. (2013) PLCepsilon, PKD1, and SSH1L<br />

transduce RhoA signaling to protect mitochondria from<br />

oxidative stress in the heart. Sci. Signal. 6, ra108.<br />

Varsano, T. et al. (2013) Inhibition of melanoma growth by<br />

small molecules that promote the mitochondrial localization<br />

of ATF2. Clin. Cancer Res. 19, 2710–2722.<br />

Ke, G. et al. (2013) MiR-181a confers resistance of cervical<br />

cancer to radiation therapy through targeting the proapoptotic<br />

PRKCD gene. Oncogene 32, 3019–3027.<br />

Stumpf, C.R. et al. (2013) The translational landscape of the<br />

mammalian cell cycle. Mol. Cell. 52, 574–582.<br />

Gobbi, G. et al. (2013) Proplatelet generation in the mouse<br />

requires PKCepsilon-dependent RhoA inhibition. Blood 122,<br />

1305–1311.<br />

Tuszynski, M.H. et al. (2012) Concepts and methods for<br />

the study of axonal regeneration in the CNS. Neuron 7,<br />

777–791.<br />

Qu, Y. et al. (2012) Phosphorylation of NLRC4 is critical for<br />

inflammasome activation. Nature 490, 539–542.<br />

chapter 02: Signaling<br />

These protein targets represent key<br />

nodes within calcium, cAMP, and lipid<br />

signaling pathways and are commonly<br />

studied in calcium, cAMP, and lipid<br />

signaling research. Primary antibodies,<br />

antibody conjugates, and antibody<br />

sampler kits containing these targets<br />

are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

S SignalSilence ® siRNA<br />

58<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for PKC have been<br />

cited over 58 times in high-impact, peerreviewed<br />

publications from the global<br />

research community.<br />

www.cellsignal.com/cstcalcium 83


Signalin<br />

Signalin<br />

Wnt1<br />

Wnt5A<br />

LRP5/6<br />

mut<br />

BMPRI<br />

BMPs<br />

BMPRII<br />

mut<br />

exp<br />

mut<br />

ligand<br />

GPCR<br />

GPCR<br />

GPCR [IP 3 ]<br />

[DAG]<br />

PLCβ<br />

PLCβ<br />

PAR-1<br />

p120<br />

Ras<br />

CKI<br />

CKI<br />

Dsh<br />

PAR-1<br />

SARA<br />

TGFβ RI<br />

TGFβ<br />

mut<br />

GPCR<br />

mut, trans, amp, exp<br />

mut<br />

Ras<br />

mut<br />

mut<br />

mut<br />

mut<br />

mut<br />

mut<br />

Src<br />

mut<br />

α-catenin<br />

ERK<br />

VEGF<br />

EPO<br />

PDGF<br />

AC<br />

PI3Kγ<br />

Ubiquitin<br />

mut<br />

mut<br />

14-3-3<br />

KIBRA<br />

[Ca 2+ ]<br />

PI3K<br />

Smad7<br />

Smad7<br />

Crk<br />

C3G<br />

[cAMP]<br />

EPAC<br />

CAMKII<br />

PKC<br />

PP2A<br />

GSK-3β<br />

APC<br />

WTX<br />

Axin<br />

mut<br />

ALK<br />

ROS1<br />

mut<br />

mut<br />

mut<br />

Smad4<br />

amp, mut, exp<br />

14-3-3<br />

YAP<br />

SOS<br />

GRB2<br />

[PIP 3 ]<br />

Ubiquitin<br />

Mst1/2 LATS1/2<br />

SAV1 MOB1<br />

TAB1<br />

TAB2<br />

mut<br />

TAB1<br />

TAB2<br />

mut<br />

mut<br />

Rap<br />

PKA<br />

RasGRF<br />

Ras<br />

NF1<br />

Raf-1<br />

B-Raf<br />

14-3-3<br />

HPH<br />

Ras c-Raf<br />

MEK1/2<br />

ERK1/2<br />

MP1<br />

RasGEF<br />

Smad4<br />

Smurf<br />

NLK<br />

Smad4<br />

mut<br />

MKK3/6<br />

mut<br />

Akt<br />

RAR<br />

mut<br />

mut<br />

SOX<br />

mut<br />

E2F<br />

amp<br />

mut, del<br />

mut<br />

amp, mut, exp<br />

amp, exp<br />

CBP<br />

TCF/LEF<br />

Integrin<br />

Src<br />

FAK<br />

GRB2<br />

GRAF<br />

SOS<br />

Rap<br />

RalGEF<br />

TEAD<br />

Smad<br />

mut<br />

mut<br />

KSR<br />

14-3-3 c-Raf<br />

VHL<br />

HIF-1β<br />

mut<br />

del<br />

ERK1/2<br />

Smad4<br />

p107<br />

E2F<br />

mut<br />

mut<br />

mut<br />

CDK7<br />

RalA<br />

RalB<br />

Cdc2<br />

Wee1A<br />

exp<br />

Miz-1<br />

Smad4<br />

RalBP1<br />

Cdc42 Actin<br />

Rac<br />

MEK1/2<br />

MP1<br />

ERK1/2<br />

exp<br />

ERK1/2<br />

p90 RSK<br />

HIF-1β<br />

PAK1<br />

Cdc25<br />

B/C<br />

Myc<br />

Miz-1<br />

mut, amp<br />

mut, amp<br />

Cytoskeleton<br />

CREB<br />

Rho<br />

JNK<br />

HPV-E7<br />

HDAC<br />

Rb<br />

DP E2F<br />

Rad52<br />

Rad51<br />

FANC<br />

D2<br />

mut<br />

mut, del<br />

mut<br />

vir<br />

exp<br />

Abl<br />

PI3K<br />

class I<br />

ULK1<br />

Beclin<br />

FIP200<br />

Atg14<br />

Atg13<br />

mut<br />

p90 RSK<br />

mut<br />

amp, trans<br />

trans<br />

Elk-1<br />

NBS1<br />

BRCA1<br />

Chk1<br />

Chk2<br />

mut<br />

mut, amp<br />

mut<br />

CDK4/6<br />

Cdc25A<br />

mut<br />

mut mut<br />

mut<br />

mut,del<br />

Cdc2<br />

Cyclin A<br />

LC3-II<br />

Atg7/3<br />

Atg4<br />

LC3-I<br />

mut,del<br />

4E-BP<br />

S6K<br />

DP E2F<br />

MDM2<br />

p21<br />

Rb<br />

amp<br />

GSK-3<br />

mut<br />

mut<br />

mut<br />

SHP1<br />

HIPK2<br />

ARF<br />

MDM2<br />

CDK7<br />

trans, mut<br />

(amino acids<br />

& glucose)<br />

TSC2<br />

TSC1<br />

mTOR<br />

Raptor<br />

p21 p27<br />

FKHR/<br />

FOXO<br />

JAK<br />

JAK<br />

STAT<br />

SHP2<br />

GRB2<br />

STAT<br />

SOS<br />

Shc<br />

Ras<br />

LKB1<br />

PI3K<br />

AMPK<br />

1011010100110101101100101<br />

Rheb<br />

Cdc25A<br />

cyclin E, A<br />

myc, cdc2<br />

p107, E2F<br />

0 10 1 0101<br />

0<br />

0 10 1 0101<br />

0 10 10 10 1<br />

101010110110110<br />

HPV-E6<br />

mut<br />

amp<br />

1011010100110101101010101<br />

trans<br />

vir<br />

mut<br />

amp, exp<br />

CDK2<br />

Cyclin A<br />

mut<br />

mTOR<br />

Rictor<br />

CDK2<br />

Cyclin E<br />

Akt<br />

BRD4<br />

TET<br />

SirT1<br />

DNMT3<br />

XIAP<br />

Lamins<br />

PDK1<br />

Bax<br />

Bcl-2<br />

Casp-2<br />

p21<br />

p27<br />

amp<br />

mut,del<br />

amp,exp<br />

amp, mut, exp<br />

mut<br />

amp, trans<br />

trans<br />

NOXA<br />

PUMA<br />

Smac<br />

AIF<br />

PARP<br />

CAD<br />

mut<br />

mut<br />

cIAP<br />

RelA<br />

NFκB2<br />

RelB<br />

PP2A<br />

c-Raf<br />

HDAC<br />

Sin3<br />

PKM2<br />

pyruvate<br />

LDHA<br />

14-3-3 Bim<br />

Apaf-1<br />

ICAD<br />

CAD<br />

amp, trans<br />

mut<br />

mut,del<br />

IκB<br />

p53<br />

Gab1<br />

PI3K<br />

RTK<br />

Gab2<br />

IRS-1<br />

Cbl<br />

PTEN<br />

PI3K<br />

mut<br />

PFK<br />

mut<br />

CytC<br />

IκB<br />

PDK1<br />

Ras<br />

G-6-P<br />

F-6-P<br />

Fructose<br />

Bisphosphate<br />

amp, trans<br />

IDH1<br />

mut<br />

Su(Fu)<br />

KIF-7<br />

Su(Fu)<br />

KIF-7<br />

mut, del<br />

citrate<br />

IDH2<br />

Akt<br />

Bax<br />

Bcl-2 JNK<br />

tBid<br />

ASK<br />

MKK7<br />

Bid<br />

TAB2<br />

TAB3<br />

IKKα<br />

IKKβ<br />

mut<br />

trans<br />

citrate<br />

MEKK3<br />

NUMB<br />

Hardcover<br />

Paperback<br />

Glucose<br />

mut<br />

FLIPs<br />

NIK<br />

Ras<br />

mut<br />

amp<br />

trans<br />

del<br />

vir<br />

exp<br />

mut<br />

Dsh<br />

Presenilin<br />

IRAK<br />

TRAF6<br />

Myd88<br />

RAIDD<br />

TRAF2<br />

mut<br />

mut<br />

mut<br />

mut<br />

mut<br />

Amplification<br />

Translocation<br />

Deletion<br />

Viral infection<br />

amp, mut, exp<br />

from PDB<br />

oncogene<br />

Hh<br />

Fas/DR<br />

mut<br />

trans<br />

Delta<br />

Jagged<br />

TACE<br />

trans<br />

/ NF-<br />

N otch g<br />

S ignalin<br />

Signalin<br />

Section I: Research Areas<br />

chapter 02: Signaling<br />

Calcium, cAMP,<br />

and Lipid Signaling<br />

Kinase-Disease<br />

Associations<br />

Name Group Disease Type Molecular Notes<br />

PKACα AGC Cancer Multiple mutations of regulatory subunit (PRKAR1A; OMIM:188830)<br />

cause Carney complex tumors and sporadic adrenal and thyroid<br />

tumors. Altered PRKAR1A transcript editing associated with lupus.<br />

PKACa knockout mouse has phenotypes similar to Carney complex<br />

(Medline:18413734). OMIM:601639.<br />

PKC<br />

(multi-isoform)<br />

AGC CNS Inh Multiple agonists and antagonists of the PKC family have consistent<br />

effects on inhibiting or activating manic behavior in humans and rats.<br />

Tamoxifen has secondary effect as PKC inhibitor and shown antimanic<br />

activity in rat model and a clinical trial (Medline:18316672,<br />

17641532).<br />

Pathways in<br />

PKCα AGC Cancer<br />

Cardiovascular<br />

Mut,<br />

Del, OE,<br />

Act<br />

A point mutation is seen in several pituitary and thyroid tumors (Medline:9167945).<br />

Deleted in a melanoma cell line. Complex expression<br />

pattern in breast cancer (Medline 15459489, 15454252). Therapeutic<br />

target in lung, gastric and prostate cancer (Medline:15447994,<br />

15313921, 15174974). May mediate multidrug resistance (Medline:12390766).<br />

Mouse models indicate a role in heart contractility<br />

(Medline:14966518). Inhibitors: LY-900003 (antisense. aka Affinitak/<br />

ISIS 3521/aprinocarsen), Safingol, Go6976. OMIM:176960.<br />

Human Cancer<br />

The Pathways in Human Cancer Poster summarizes the signaling nodes implicated<br />

in cancer initiation and progression and features peer-reviewed protein mapping with<br />

scientifically accurate rendering of the molecular structures of proteins. The poster<br />

accompanies the text A Biology of Cancer by Robert A. Weinberg (Second Edition).<br />

PKCβ AGC Autism<br />

Cancer<br />

Diabetes<br />

PKCγ AGC Neurodegeneration<br />

Pain<br />

PKCδ AGC Cancer<br />

Cardiovascular<br />

CNS<br />

PKCε AGC Cancer<br />

Cardiovascular<br />

CNS<br />

PKCθ AGC Cancer<br />

Immunity<br />

SNP<br />

Two promoter SNPs associated with diabetic nephropathy<br />

(Medline:12874455), correlating with induction of renal expression<br />

by high glucose, reduction in renal function by a specific PKCβ<br />

inhibitor and successful inhibitor treatment of rodent models of<br />

diabetic nephropathy (Medline:12955673). PKCβ inhibition has also<br />

been proposed to treat diabetic retinopathy (Medline:12507628)<br />

and diabetic vascular complications (Medline:11903393). Ectopic<br />

expression in mouse heart leads to cardiac hypertrophy. Elevated<br />

expression is seen in and promotes early stages of colon cancer<br />

in mouse models (Medline:11245437). May mediate multidrug<br />

resistance (Medline:12697075). Activation protects astrocytes from<br />

ischemic injury (Medline:15165841). SNPs associated with autism<br />

(Medline:16027742). Inhibitors: LY333531 (ruboxistaurin; Phase 3<br />

for diabetic neuropathy and retinopathy), LY317615 (Eli Lilly: isoform<br />

selective). OMIM:176970.<br />

Mut Point mutations linked to dominant spinocerebellar ataxia type 14<br />

[OMIM:605361]. Knockout and inhibitor studies show role in pain<br />

perception (Medline:14762097, 9323205). OMIM:176980.<br />

Expr<br />

Amp,<br />

Mut<br />

Pro-apoptotic. Reduced expression correlated with progression of colon<br />

and other cancers (Medline:15054085, 12657722, 12591726).<br />

Inhibition may drive chemo-resistant cancers to apoptosis. Activated<br />

and promotes apoptosis in cardiac and neuronal cells after ischemicreperfusion<br />

injury (Medline:14654063, 15295022). Activator:<br />

bistrane A. Inhibitors: rottlerin, KAI-9803 (KAI; Phase 2 trials for<br />

reperfusion injury), dV1-1. OMIM:176977.<br />

Amplified and rearranged in thyroid cancers (Medline:9683604,<br />

10438519, 11994357). Promotes growth of an androgen-independent<br />

prostate cancer cell line (Medline:11956106) and transforms<br />

fibroblasts in culture (Medline:11968018). Activation protects cardiac<br />

myocytes and neurons from ischemic injury (Medline:15165841)<br />

and antagonizes PKCδ. Amyloid β peptide inhibits PKCε activity<br />

and may contribute to the pathogenesis of Alzheimer's disease<br />

(Medline:15207847). Inhibitor studies indicate role in pain perception<br />

(Medline:1043272). Multi-isoform inhibitors include: Bryostatin-1<br />

(BRYO) from Aphios and Perifosine/D-21266 from Baxter while KAI-<br />

1678 is a selective inhibitor. OMIM:176975.<br />

Required for activation and proliferation of mature T cells. Proposed<br />

as a target for T cell leukemias (Medline:12188914). Knockout is<br />

resistant to fat-induced insulin resistance. OMIM:600448.<br />

Molecular: Act Activated • Amp Amplified • Del Deleted • Expr Expression • GOF Gain-of-function • Inh Inhibitor<br />

Studies • LOF Loss-of-function • LOH Loss-of-heterozygosity • Meth Methylation • Model model organism studies •<br />

Mut Mutation OE Overexpression • SNP Single Nucleotide Polymorphism • Splice Splicing change • Trans Translocation<br />

Features<br />

Peer-reviewed protein mapping and accurate molecular rendering of proteins.<br />

Summarizes the latest signaling nodes implicated in cancer signaling.<br />

Pathway-specific color-coded nodes.<br />

Revised Edition<br />

Angiogenesis<br />

Wnt Signaling<br />

H ippo g<br />

GPCR Signaling<br />

R-spondin<br />

T GF β g<br />

R-spondin<br />

Frizzled<br />

Frizzled<br />

Receptor Tyrosine Kinase (RTK)<br />

E-cadherin<br />

CD44<br />

FAT4<br />

LGR4,5,6<br />

γ<br />

β<br />

α<br />

γ<br />

β<br />

α<br />

ZNRF3/RNF43<br />

β-catenin<br />

γ<br />

β<br />

α<br />

Dsh Akt<br />

α 12/13<br />

γ<br />

β<br />

α s<br />

Smad1/5/8<br />

Smad2/3<br />

TGFβ RII<br />

EGFR<br />

HER2<br />

FRMD<br />

Mer<br />

Smad1/5/8<br />

Smad2/3<br />

Ras Signaling<br />

β-catenin<br />

TAK<br />

RasGRP<br />

RasGAP<br />

Normoxia<br />

Hypoxia<br />

SynGAP<br />

β-catenin<br />

YAP/TAZ<br />

Smad1/5/8<br />

Smad2/3<br />

TAK p38<br />

C<br />

PI3K ILK<br />

RIAM/Talin<br />

RasGAP<br />

RhoGEFs<br />

β-catenin<br />

MKK7 JNK<br />

HIF-1α(-2α)<br />

other TFs<br />

YAP/TAZ<br />

myc, cyclin D, CD44<br />

Smad1/5/8<br />

Smad2/3<br />

Cyclin H<br />

Regulated<br />

Exocytosis<br />

Exocyst<br />

Complex<br />

Atg16/12/5<br />

Rab8a (c-Mel)<br />

p90 RSK JNK<br />

HIF-1α(-2α)<br />

Fos<br />

Jun<br />

myc, cyclin D<br />

Cyclin B<br />

p15, p21, p27<br />

Smad2/3<br />

DNA Repair<br />

Myc<br />

Max<br />

autophagosome<br />

Myc<br />

Max<br />

cyclin D, myc<br />

cyclin D, myc<br />

SQSTM1<br />

Autophagy<br />

Fos<br />

Jun<br />

phagopore<br />

membrane<br />

nucleation<br />

p15 p16 p18 p19<br />

UV<br />

Cyclin D<br />

DNA Damage<br />

Cyclin D<br />

STAT STAT<br />

(Inactive) (Active)<br />

IR<br />

* *<br />

ATM<br />

ATR<br />

Scienstists at Cell Signaling Technology co laborate<br />

with key opinion leaders in cance research to create<br />

reference pathway diagrams that reflect the latest<br />

thinking in the research community. Over 40 pathway<br />

diagrams cu rently exist, including the pathways<br />

represented here and many others.<br />

www.ce lsignal.com/<strong>CST</strong>cancer<br />

DNA-<br />

PK<br />

p53<br />

p53<br />

Nutrients<br />

cyclin D<br />

The Pathways in Human Cancer poster summarize some of the key<br />

signaling pathways implicated in tumorigenesis and tumor<br />

progression in humans. Within each pathway, gene products known<br />

to be mutated in human tumors—oncogenes and tumor suppressor<br />

genes—are coded with information on types of genetic alterations<br />

and confe red capabilities to the tumor. Proteins are shown using<br />

structural representatives. New to this revised poster edition are<br />

signaling pathways for Hippo Signaling, Autophagy, Warburg E fect,<br />

and Epigenetic Regulation.<br />

This poster was created by scientists at Ce l Signaling Technology in<br />

co laboration with Robert A. Weinberg and others a the forefront of<br />

cance research. Expanded versions of each pathway, including<br />

additional downstream signaling nodes, can be found at<br />

www.ce lsignal.com/<strong>CST</strong>cancer.<br />

R<br />

Cyclin H<br />

Cytokine<br />

Receptor<br />

Aurora<br />

A<br />

Transcription<br />

MDM2 Bcl-x L<br />

Bad<br />

p53<br />

myc, cyclin D, cyclin E<br />

cyclin D<br />

CSL/CBF1<br />

Gli PKA<br />

KMT2/MLL<br />

KDM6A/UTX<br />

KMT4/DOT1L<br />

SWI/SNF<br />

KMT6/EZH2<br />

NFκB1/2<br />

CSL/CBF1<br />

p300<br />

Epigenetic Signaling<br />

Ce l Signaling Technology would like to thank<br />

digizyme for their co laboration on the design<br />

and concept of this poster. Please visit<br />

www.digizyme.com to see more of their work.<br />

NICD<br />

[PIP 3 ]<br />

lactate<br />

Casp-9<br />

Casp-3,6,7<br />

HAT HES1<br />

Akt Signaling<br />

NICD<br />

Gli<br />

[PIP 3 ]<br />

Hexokinase<br />

Krebs<br />

Cycle<br />

Bak TRADD<br />

IKKα IKKβ<br />

NFκB1/2<br />

RelA<br />

Gli<br />

Fatty acid<br />

synthesis<br />

TAK<br />

NEMO<br />

Direct stimulatory modification<br />

Direct inhibitory modification<br />

Multistep stimulatory modification<br />

Tentative stimulatory modification<br />

Transcriptional contribution<br />

TYPES OF GENETIC ALTERATIONS:<br />

Point mutation<br />

Increased expression<br />

(unknown mechanism)<br />

TYPES OF CONFERRED CAPABILITIES:<br />

Evading apoptosis<br />

Self-su ficiency in growth signals<br />

Insensitivity to anti-growth signals<br />

Tissue invasion & metastasis<br />

Limitless replicative potential<br />

Sustained angiogenesis<br />

∞<br />

transporter<br />

Ras Common protein name<br />

Structural representative<br />

tumor suppressor<br />

Type of genetic alteration<br />

Type of confe red capability<br />

Warburg Effect<br />

FADD<br />

Casp-8,10<br />

RIP TRADD<br />

TRAF<br />

2/6<br />

TRAF<br />

3<br />

Notch<br />

Furin<br />

Fringe<br />

NIC<br />

Cleaved<br />

Notch<br />

CDO<br />

BOC<br />

Ptch<br />

Smo<br />

Ptch<br />

Smo<br />

IL1R<br />

TNFR<br />

TNFR<br />

CDO<br />

BOC<br />

D eath Receptor κB g<br />

This poster accompanies the textbook The Biology of Cancer, Second Edition<br />

by Robert A. Weinberg and published by Garland Science.<br />

Dr. Weinberg is a founding member of the Whitehead Institute for Biomedical<br />

Research and the Daniel K. Ludwig Professor of Cancer Research a the<br />

Massachuse ts Institute of Technology.<br />

ISBN 978-0-8153-4219-9<br />

ISBN 978-0-8153-4220-5 www.garlandscience.com<br />

Signalin<br />

H edgehog g<br />

Pathways in<br />

Human Cancer<br />

Poster<br />

To request free copies of this<br />

poster please visit our website.<br />

www.cellsignal.com/phcposter<br />

Cancer<br />

Research<br />

84 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

Please visit our website to learn<br />

more about the scientific tools and<br />

educational resources we have online<br />

for cancer signaling and proteomic<br />

analysis, including discussion of key<br />

disease drivers.<br />

www.cellsignal.com/cancerguide<br />

85


03<br />

Section I: Research Areas<br />

kDa<br />

60<br />

50<br />

40<br />

30<br />

20<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

–<br />

Raji<br />

+<br />

MCF7<br />

– +<br />

Phospho-<br />

Bim<br />

(Ser77)<br />

Cell Growth and Death<br />

Apoptosis<br />

Apoptosis is a tightly controlled process of cell death characterized by nuclear condensation, cell<br />

shrinkage, membrane blebbing, and DNA fragmentation. The central regulators of apoptosis are the<br />

caspases, a family of cysteine proteases that fall into two broad categories. The “initiator” caspases-2,<br />

-8, -9, -10, and -12 are closely coupled to upstream, pro-apoptotic signals and act by cleaving and<br />

activating the downstream “executioner” caspases-3, -6, and -7 that modify proteins ultimately responsible<br />

for programmed cell death. Targets such as PARP, lamin A/C, α-/β-actin, GAS2, α-fodrin, acinus,<br />

and many others are cleaved by executioner caspases and serve as markers of apoptosis. There are<br />

two primary apoptotic pathways that lead to caspase activation: the intrinsic and extrinsic pathways.<br />

Apoptosis-inducing agent, staurosporine, results in cleavage/activation<br />

of caspase-3 and morphological characteristics of cell death.<br />

Cleaved Caspase-3 (Asp175)<br />

(D3E9) Rabbit mAb (Alexa Fluor ®<br />

647 Conjugate) #9602: Confocal IF<br />

analysis of HeLa cells, untreated (left)<br />

or treated with Staurosporine #9953<br />

(1 μM, 4 hr; right), using #9602 (blue<br />

pseudocolor). Actin filaments were labeled<br />

with Alexa Fluor ® 488 Phalloidin<br />

#8878 (green). Red = Propidium Iodide<br />

(PI)/RNase Staining Solution #4087.<br />

Etoposide treatment results in cleavage of PARP, a marker for apoptosis.<br />

Etoposide #2200: WB analysis of extracts from Jurkat cells, untreated (-) or etoposide-treated<br />

(25 μM, overnight; +), using Cleaved PARP (Asp214) (D64E10) XP ® Rabbit mAb #5625 (upper)<br />

or total PARP Antibody #9542 (lower).<br />

Intrinsic Apoptosis<br />

The intrinsic apoptosis pathway is activated by cell stress, DNA damage, developmental cues, and<br />

withdrawal of survival factors. This pathway is regulated by the Bcl-2 family of proteins, which consists<br />

of 3 classes: the pro-apoptotic effectors (Bax and Bak); the pro-apoptotic BH3-only proteins (Bad, Bid,<br />

Puma, Bim, Noxa, Bmf, Hrk, and Bik); and the anti-apoptotic proteins (Bcl-2, Bcl-xL, Bcl-w, and Mcl-1,<br />

A1/Bfl1). The pro-apoptotic effectors localize to the mitochondrial membrane and control mitochondrial<br />

permeability and release of cytochrome c, AIF, Smac/Diablo, HtrA2, Endo G, and others. The proapoptotic<br />

effectors are activated by BH3-only proteins either through direct interaction or antagonism of<br />

anti-apoptotic proteins. Members of the BH3-only class are generally regulated by apoptotic stimuli,<br />

either at the level of transcription or through phosphorylation or other post-translational modifications.<br />

The cytochrome c released as a result of Bcl-2 protein action binds to Apaf-1 and forms the apoptosome,<br />

a large, multiprotein, flower-like structure that binds and activates caspase-9, resulting in<br />

caspase-3 cleavage and other apoptotic endpoints.<br />

Cleaved<br />

PARP<br />

(Asp214)<br />

86 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

140<br />

100<br />

80<br />

60<br />

50<br />

PARP<br />

Cleaved<br />

PARP<br />

(Asp214)<br />

– + Etoposide<br />

Bim EL<br />

Bim L<br />

Bim S<br />

TPA treatment results in phosphorylation of Bim at Ser77, a<br />

TPA<br />

modification known to promote Bim proteasomal degradation.<br />

Phospho-Bim (Ser77) (D4H12) Rabbit mAb #12433: WB analysis of extracts from Raji and MCF7 cells, untreated (-)<br />

or treated with TPA #4174 (200 nM, 30 min; +), using #12433 (upper) or Bim (C34C5) Rabbit mAb #2933 (lower).<br />

Cytochrome c release<br />

is a key indicator of<br />

intrinsic apoptosis.<br />

Extrinsic Apoptosis<br />

Caspases can also be activated through the extrinsic, or death receptor, pathway. The death receptors<br />

consist of members of the TNFR family (TNFR1/2, Fas, and DR3/4/5/6) and their associated ligands<br />

(TNF-α, FasL, TRAIL, TWEAK). Ligand binding induces receptor activation and recruitment of the adaptor<br />

proteins FADD or TRADD via their death domains (DD). Together, the ligand-bound receptor, adaptor<br />

protein, and procaspase-8 form the death-inducing signaling complex (DISC), a multiprotein complex<br />

that activates caspase-8 and initiates downstream apoptotic signaling. Signaling through TNF-family<br />

cytokines can lead to either apoptosis through DISC or survival by activation of NF-κB and the prosurvival<br />

genes, Bcl-2 and FLIP. Cycloheximide, which inhibits NF-κB gene regulation, is commonly<br />

used with TNF-α to induce apoptosis.<br />

Treatment with TNF-α and cycloheximide results in<br />

detection of caspase-8 cleavage products, p43 and p18.<br />

Cleaved Caspase-8 (Asp387) (D5B2) XP ® Rabbit mAb (Mouse Specific) #8592: WB analysis of<br />

extracts from CTLL-2 cells, untreated or treated with cycloheximide (CHX, 10 μg/ml, overnight) followed<br />

by hTNF-α #8902 (20 ng/ml, 4 hr), using #8592.<br />

Necrosis and Necroptosis<br />

The highly regulated system of apoptotic cell death is not the only way cells can die. “Accidental”<br />

death, called necrosis, is an unregulated form of cell death characterized by cell swelling, plasma<br />

membrane rupture, and inflammation.<br />

kDa<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

– +<br />

chapter 03: Cell Growth and Death<br />

p43<br />

p18<br />

CHX/TNF-α<br />

Recently, a form of programmed necrosis has also been identified. Activation of the death receptors can<br />

lead to caspase-independent cell death termed necroptosis. Necroptosis is induced by the TNF-family<br />

of cytokines and mediated by the kinases RIP1 and RIP3. In the absence of caspase-8 activation, RIP1<br />

forms a complex with the key regulator of necroptosis, RIP3, and its downstream substrate, MLKL.<br />

Together, these proteins create complex IIb in conjunction with FADD, caspase-8, and cFLIP, forming<br />

the necroptosome.<br />

Apoptosis Assays<br />

There are many assays available for measuring cell death. Apoptosis assays measure key apoptotic<br />

markers such as activation of caspase-3 or release of cytochrome c. Assays measuring necrosis typically<br />

focus on the cell permeability that is characteristic of necrotic plasma membrane rupture. Some<br />

assays, such as Annexin V, can differentiate between apoptosis and necrosis using a dual marker<br />

system—annexin V binding to membrane-bound phosphatidylserine indicates early apoptosis whereas<br />

propidium iodide labeling identifies the leaky cell membranes associated with necrosis.<br />

Select Reviews<br />

Dickens, L.S., Powley, I.R., and Hughes, M.A., et al. (2012) Exp. Cell Res. 318, 1269–1277. • Favaloro, B., Allocati, N., and<br />

Graziano, V., et al. (2012) Aging 4, 330–349. • Giampietri, C., Starace, D., Petrungaro, S., et al. (2014) Int. J. Cell Biol. 49027.<br />

• McIlwain, D.R., Berger, T., and Mak, T.W. (2013) Cold Spring Harb. Perspect. Biol. 5, a008656. • Moriwaki, K. and Chan,<br />

F.K. (2013) Genes Dev. 27, 1640−1649. • Shamas-Din, A., Kale, J., and Leber B., et al. (2013) Cold Spring Harb. Perspect.<br />

Biol. 5, a008714.<br />

Cytochrome c (6H2.B4) Mouse<br />

mAb #12963: Confocal IF analysis of<br />

HeLa cells, untreated (left) or treated<br />

with Staurosporine (1 μM) #9953 and<br />

Z-VAD (50 μM) for 3 hr (right), using<br />

#12963 (green). Blue pseudocolor=<br />

DRAQ5 ® #4084 (fluorescent DNA dye).<br />

Assay Kits for<br />

Measuring Cell<br />

Death & Viability<br />

Annexin V-FITC Early Apoptosis<br />

Detection Kit #6592<br />

Differentiates between early apoptosis<br />

and necrosis through annexin<br />

V and propidium iodide labeling<br />

Caspase-3 Activity<br />

Assay Kit #5723<br />

Measures caspase-3 activity<br />

through cleavage of fluorescently<br />

labeled caspase-3 substrate<br />

Resazurin Cell Viability<br />

Kit #11884<br />

Measures metabolic activity as<br />

an indicator of cell viability using<br />

fluorescent detection<br />

XTT Cell Viability Kit #9095<br />

Measures metabolic activity as<br />

an indicator of cell viability using<br />

colorimetric detection<br />

Cytochrome c Release<br />

Flow Kit #13648<br />

Identifies cytochrome c release by<br />

flow cytometry using a cytochrome<br />

c antibody<br />

www.cellsignal.com/cstapoptosis 87


Section I: Research Areas<br />

chapter 03: Cell Growth and Death<br />

These protein targets represent key<br />

nodes within apoptotic signaling<br />

pathways and are commonly studied<br />

in apoptosis research. Primary<br />

antibodies, antibody conjugates, and<br />

antibody sampler kits containing<br />

these targets are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

E PathScan ® ELISA Kits<br />

S SignalSilence ® siRNA<br />

C Antibody Conjugate<br />

Inducers of Apoptosis<br />

Stimulating apoptosis with chemical modulators or cytokines is a common strategy for investigating effects on downstream<br />

signaling. These agents induce apoptosis by a variety of methods, typically through inhibition of key cellular processes or kinases.<br />

#9972 Brefeldin A Interferes with ER to Golgi transport; typically used to study vesicle trafficking but<br />

prolonged exposure causes apoptosis<br />

#2112 Cycloheximide Inhibits protein synthesis and is often used in conjunction with TNF-α to induce cell death<br />

#2200 Etoposide Inhibits Topoisomerase II<br />

#9807 Paclitaxel Binds β-tubulin and prevents microtubule depolymerization<br />

#9885 Roscovitine Inhibits CDK1, CDK2, CDK5 to cause cell cycle arrest and apoptosis<br />

#9953 Staurosporine Potently inhibits PKC; also inhibits PKA, PKG, CAMKII, and MLCK<br />

#5927 Doxorubicin Inhibits DNA and RNA synthesis through intercalating the DNA double helix<br />

#8902 Human Tumor Necrosis<br />

Factor-α (hTNF-α)<br />

#5178 Mouse Tumor Necrosis<br />

Factor-α (mTNF-α)<br />

#4698 Mouse His 6 Tumor Necrosis<br />

Factor-α (m His 6TNF-α)<br />

Commonly Studied Apoptosis Targets<br />

Target M P E S C<br />

A1/Bfl-1<br />

•<br />

APR3<br />

•<br />

Acinus<br />

•<br />

AIF • • •<br />

Alix<br />

•<br />

AP-2α<br />

• •<br />

AP-2β<br />

•<br />

AP-2g<br />

•<br />

Phospho-AP2M1 (Thr156) •<br />

Apaf-1 • • •<br />

Aven<br />

•<br />

Bad<br />

• • • •<br />

Phospho-Bad (Ser112) • • • •<br />

Phospho-Bad (Ser136) • •<br />

Phospho-Bad (Ser155) • •<br />

Bak1 • • •<br />

BAP31<br />

•<br />

Bax • • •<br />

Bcl-2<br />

• • • •<br />

Phospho-Bcl-2 (Thr56) •<br />

Phospho-Bcl-2 (Ser70) • • •<br />

Bcl-w<br />

•<br />

Bcl-xL • • • •<br />

BCL2L10<br />

•<br />

A1/Bfl-1<br />

•<br />

BID<br />

• •<br />

Bik<br />

•<br />

Bim • • • •<br />

Phospho-Bim (Ser55)<br />

•<br />

Phospho-Bim (Ser69) • •<br />

Phospho-Bim (Ser77) •<br />

BIRC6<br />

•<br />

Bit1<br />

•<br />

Bmf<br />

•<br />

c-IAP1<br />

• •<br />

Triggers extrinsic apoptosis and/or survival through death receptor signaling<br />

Triggers extrinsic apoptosis and/or survival through death receptor signaling<br />

Triggers extrinsic apoptosis and/or survival through death receptor signaling<br />

#5452 Fas Ligand Triggers extrinsic apoptosis through death receptor signaling<br />

Target M P E S C<br />

c-IAP2<br />

•<br />

Caspase Cleavage Motif •<br />

Caspase-1<br />

•<br />

Cleaved Caspase-1 (Asp297) •<br />

Caspase-2<br />

•<br />

Caspase-3 • • •<br />

Cleaved Caspase-3 (Asp175) • • • •<br />

Caspase-4<br />

•<br />

Caspase-5<br />

•<br />

Caspase-6<br />

•<br />

Cleaved Caspase-6 (Asp162) •<br />

Caspase-7<br />

• •<br />

Cleaved Caspase-7 (Asp198) • •<br />

Caspase-8<br />

•<br />

Cleaved Caspase-8 (Asp384) •<br />

Cleaved Caspase-8 (Asp387) • •<br />

Cleaved Caspase-8 (Asp391) • •<br />

Caspase-9<br />

• •<br />

Cleaved Caspase-9 (Asp315) •<br />

Cleaved Caspase-9 (Asp330) • •<br />

Cleaved Caspase-9 (Asp353) •<br />

Non-Phospho-Caspase-9 (Ser196)<br />

•<br />

Caspase-11<br />

•<br />

Caspase-12<br />

•<br />

Caspase-14<br />

•<br />

Cytochrome c • • •<br />

DAP1<br />

•<br />

DAPK1<br />

•<br />

DAPK3/ZIPK<br />

•<br />

Daxx<br />

•<br />

Cleaved Drosophila Dcp-1 (Asp216) •<br />

DcR1<br />

•<br />

DcR2<br />

•<br />

DcR3<br />

•<br />

DFF45/DFF35<br />

•<br />

Target M P E S C<br />

Cleaved DFF45 (Asp224) •<br />

DIDO1<br />

•<br />

DR3<br />

•<br />

DR5<br />

• •<br />

Phospho-DR6 (Ser562) •<br />

DRAK2<br />

•<br />

eIF4G2/p97<br />

•<br />

Endonuclease G<br />

•<br />

FADD<br />

•<br />

Phospho-FADD (Ser194) •<br />

FAF1<br />

•<br />

FAIM<br />

•<br />

Fas<br />

•<br />

Fas Ligand<br />

•<br />

FLIP • •<br />

α-Fodrin<br />

•<br />

Cleaved α-Fodrin (Asp1185) •<br />

Granzyme A<br />

•<br />

Granzyme B<br />

•<br />

HIPK2<br />

•<br />

HtrA2/Omi • • •<br />

Cleaved Drosophila ICE (drICE)<br />

(Asp230)<br />

•<br />

Lamin A/C • • •<br />

Cleaved Lamin A/C (Small Subunit) • •<br />

Phospho-Lamin A/C (Ser22) • • •<br />

Lamin B1<br />

•<br />

Lamin B2<br />

•<br />

LAP2α<br />

•<br />

Livin<br />

•<br />

Mad-1<br />

•<br />

Maspin<br />

•<br />

Max<br />

•<br />

Mcl-1 • • •<br />

Phospho-Mcl-1 (Ser64) •<br />

Phospho-Mcl-1 (Ser159/Thr163) •<br />

Phospho-Mcl-1 (Thr183)/<br />

MST2(Thr180)<br />

Phospho-MLKL<br />

Mst1<br />

•<br />

•<br />

•<br />

Target M P E S C<br />

Mst2<br />

•<br />

Mst3<br />

•<br />

Mst4<br />

•<br />

c-Myc • • •<br />

Phospho-c-Myc (Ser62) •<br />

N-Myc<br />

•<br />

PAR-4<br />

•<br />

Phospho-PAR-4 (Thr163) •<br />

PARP1 • •<br />

Cleaved-PARP1 (Asp214) • • • •<br />

PDCD4<br />

•<br />

PEA-15 • • •<br />

Phospho-PEA-15 (Ser104) •<br />

Perforin<br />

•<br />

PHLDA3<br />

•<br />

Puma • • •<br />

RIP1<br />

• •<br />

RIP3<br />

• •<br />

Siva-1<br />

•<br />

Smac/Diablo<br />

•<br />

Survivin<br />

• • • • •<br />

Phospho-Survivin (Thr34) • •<br />

TANK<br />

•<br />

TAX1BP1<br />

•<br />

TMS1<br />

•<br />

TP/ECGF1<br />

• •<br />

TNF-R2<br />

•<br />

TRADD<br />

• •<br />

TRAF1<br />

•<br />

TRAF2<br />

•<br />

Phospho-TRAF2 (Ser11) •<br />

TRAF3<br />

•<br />

TRAF6<br />

•<br />

TRAIL<br />

•<br />

VDAC1<br />

• •<br />

VDAC2<br />

•<br />

WWOX<br />

•<br />

XAF1<br />

• •<br />

XIAP • • •<br />

1,240<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for cleaved caspase-3<br />

(Asp175) have been cited over 1,240<br />

times in high-impact, peer-reviewed<br />

publications from the global research<br />

community.<br />

Select Citations:<br />

Bjordal, M. et al. (2014) Sensing<br />

of amino acids in a dopaminergic<br />

circuitry promotes rejection of an<br />

incomplete diet in Drosophila. Cell<br />

156, 510–521.<br />

Doitsh, G. et al. (2014) Cell death by<br />

pyroptosis drives CD4 T-cell depletion<br />

in HIV-1 infection. Nature 505,<br />

509–514.<br />

Walczynski, J. et al. (2014) Sensitisation<br />

of c-MYC-induced B-lymphoma<br />

cells to apoptosis by ATF2. Oncogene<br />

33, 1027–1036.<br />

de Poot, S.A. et al. (2014) Granzyme<br />

M targets topoisomerase II alpha to<br />

trigger cell cycle arrest and caspasedependent<br />

apoptosis. Cell Death<br />

Differ. 21, 416–426.<br />

Pencheva, N. et al. (2014) Broadspectrum<br />

therapeutic suppression of<br />

metastatic melanoma through nuclear<br />

hormone receptor activation. Cell<br />

156, 986–1001.<br />

Ardestani, A. et al. (2014) MST1 is a<br />

key regulator of beta cell apoptosis<br />

and dysfunction in diabetes. Nat.<br />

Med. 20, 385–397.<br />

von Figura, G. et al. (2014) The<br />

chromatin regulator Brg1 suppresses<br />

formation of intraductal papillary<br />

mucinous neoplasm and pancreatic<br />

ductal adenocarcinoma. Nat. Cell Biol.<br />

16, 255–267.<br />

Beug, S.T. et al. (2014) Smac<br />

mimetics and innate immune stimuli<br />

synergize to promote tumor death.<br />

Nat. Biotechnol. 32, 182–190.<br />

Gao, T. et al. (2014) Pdx1 maintains<br />

beta cell identity and function by<br />

repressing an alpha cell program. Cell<br />

Metab. 259–271.<br />

Chen, Y. et al. (2014) Hyperactivation<br />

of mammalian target of rapamycin<br />

complex 1 (mTORC1) promotes<br />

breast cancer progression through<br />

enhancing glucose starvation-induced<br />

autophagy and Akt signaling. J. Biol.<br />

Chem. 289, 1164–1173.<br />

Razorenova, O.V. et al. (2014) The<br />

apoptosis repressor with a CARD domain<br />

(ARC) gene is a direct hypoxiainducible<br />

factor 1 target gene and<br />

promotes survival and proliferation of<br />

VHL-deficient renal cancer cells. Mol.<br />

Cell. Biol. 34, 739–751.<br />

88 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstapoptosis 89


c-IAP<br />

TRADD<br />

FADD<br />

Section I: Research Areas<br />

chapter 03: Cell Growth and Death<br />

Regulation of Apoptosis Overview<br />

Inhibition of Apoptosis<br />

TNF, FasL, TRAIL<br />

TNF, FasL, TRAIL<br />

Trophic Factors<br />

TNF-α<br />

TNF-R1<br />

Survival Factors:<br />

Growth Factors,<br />

Cytokines, etc.<br />

Cytoplasm<br />

IKKα<br />

IκBα<br />

TAK1<br />

IKKγ<br />

IκBα<br />

NF-κB<br />

NF-κB<br />

Cytoplasm<br />

Nucleus<br />

IKKβ<br />

Calpain<br />

[Ca 2+ ]<br />

FLIP<br />

XIAP<br />

c-IAP<br />

TRAF2<br />

NIK Casp-8,-10<br />

RIP<br />

TRAF2<br />

Casp-12<br />

Casp-9<br />

ER Stress<br />

RIP1<br />

TRADD<br />

NF-κB<br />

FADD<br />

CYLD<br />

PARP<br />

Itch<br />

FLIP<br />

TRADD<br />

FADD<br />

BID<br />

ROCK<br />

• Cell shrinking<br />

• Membrane<br />

blebbing<br />

DFF40<br />

XIAP<br />

AIF<br />

RIP RAIDD<br />

PIDD<br />

Casp-2<br />

p53<br />

SirT2<br />

HtrA2<br />

Arts<br />

Casp-3,-6,-7<br />

Lamin<br />

A/C<br />

APP<br />

DNA<br />

Fragmentation<br />

RIP<br />

Smac/<br />

Diablo<br />

Endo G<br />

TRAF2<br />

tBID<br />

DFF40<br />

JNK<br />

Cyto c<br />

DFF45<br />

ASK1<br />

Bax<br />

DNA Damage<br />

Bak<br />

Mcl-1<br />

Casp-9<br />

Apaf-1<br />

AIF<br />

PKC<br />

Bcl-2<br />

Bim<br />

ATM/ATR<br />

p53<br />

Puma<br />

Noxa<br />

p90RSK<br />

Puma<br />

Bcl-xL<br />

Noxa<br />

HECTH9<br />

Endo G<br />

Chk1/2<br />

FoxO1<br />

p53<br />

Bad<br />

p53<br />

PI3K<br />

Cell Cycle<br />

Erk1/2<br />

cdc2<br />

14-3-3<br />

Akt<br />

Cellular Stress<br />

FoxO1<br />

JNK<br />

JNK<br />

Bim<br />

c-Jun<br />

NIK<br />

IKKγ CDC37<br />

IKKβ HSP90<br />

IκB<br />

IκB<br />

NF-κB<br />

NF-κB<br />

Cytoplasm<br />

Nucleus<br />

NF-κB<br />

cIAP<br />

TAK1<br />

TAB<br />

IKKα IKKβ<br />

IKKγ<br />

TRAF2<br />

TRADD<br />

HSP90<br />

Casp-8<br />

XIAP<br />

CYLD<br />

HSP70<br />

FADD<br />

HSP90<br />

JNK<br />

HSP27<br />

A20<br />

GSK-3<br />

FoxO1<br />

PTEN<br />

PI3K<br />

PIP 3<br />

Akt<br />

Bcl-2<br />

Casp-3,-6,-7<br />

Bax<br />

HSP27<br />

Apoptosis<br />

Casp-9<br />

Apaf-1 Cyto c HECTH9 HSP70<br />

Akt<br />

FLIP<br />

XIAP<br />

A20<br />

FLIP<br />

BID<br />

Smac/<br />

Diablo<br />

RIP1<br />

HSP70<br />

tBID<br />

HSP27<br />

HSP90<br />

Bax<br />

Bax<br />

Mcl-1<br />

Bak<br />

Bcl-xL<br />

Bim<br />

Bim<br />

PDK1<br />

p70S6K<br />

FAS<br />

Bim<br />

Bad<br />

Bim<br />

PKA<br />

[cAMP]<br />

FoxO1<br />

Ras<br />

Raf<br />

Erk1/2<br />

FoxO1<br />

PKC<br />

p90RSK<br />

Jak<br />

Src<br />

CREB Stat1 Stat3<br />

Bcl-2<br />

Bcl-xL<br />

Cytoplasm<br />

CREB<br />

Stat1<br />

Stat3<br />

HSP90<br />

Apoptosis is a regulated cellular suicide mechanism characterized by nuclear condensation, cell shrinkage, membrane blebbing, and DNA fragmentation. Caspases, a family<br />

of cysteine proteases, are the central regulators of apoptosis. Initiator caspases (including caspase-2, -8, -9, -10, -11, and -12) are closely coupled to pro-apoptotic signals.<br />

Once activated, these caspases cleave and activate downstream effector caspases (including caspase-3, -6, and -7), which in turn execute apoptosis by cleaving cellular<br />

proteins following specific Asp residues. Activation of Fas and TNFR by FasL and TNF, respectively, leads to the activation of caspase-8 and -10. DNA damage induces the<br />

expression of PIDD, which binds to RAIDD and caspase-2 and leads to the activation of caspase-2. Cytochrome c released from damaged mitochondria is coupled to the<br />

activation of caspase-9. XIAP inhibits caspase-3, -7, and -9. Mitochondria release multiple pro-apoptotic molecules, such as Smac/Diablo, AIF, HtrA2, and Endo G, in addition<br />

to cytochrome c. Smac/Diablo binds to XIAP, preventing it from inhibiting caspases. Caspase-11 is induced and activated by pathological pro-inflammatory and pro-apoptotic<br />

stimuli and leads to the activation of caspase-1, thereby promoting inflammatory response and apoptosis by directly processing caspase-3. Caspase-12 and caspase-7 are<br />

activated under ER stress conditions. Anti-apoptotic ligands, including growth factors and cytokines, activate Akt and p90RSK. Akt inhibits Bad by direct phosphorylation and<br />

prevents the expression of Bim by phosphorylating and inhibiting the Forkhead family of transcription factors (FoxO). FoxO promotes apoptosis by upregulating pro-apoptotic<br />

molecules such as FasL and Bim.<br />

Select Reviews:<br />

Degterev, A. and Yuan J. (2008) Nat. Rev. Mol. Cell Biol. 9, 378–390. • Fuchs, Y. and Steller H. (2011) Cell 147, 742–758. • Indran, I.R., Tufo, G., Pervaiz, S., and Brenner<br />

C. (2011) Biochim. Biophys. Acta. 1807, 735–745. • Kaufmann, T., Strasser, A., and Jost, P.J. (2012) Cell Death Differ. 19, 42–50. • Kurokawa, M. and Kornbluth, S.<br />

(2009) Cell 138, 838–854. • Pradelli, L.A., Bénéteau, M., and Ricci, J.E. (2010) Cell. Mol. Life Sci. 67, 1589–1597. • Van Herreweghe, F., Festjens, N., Declercq, W., and<br />

Vandenabeele, P. (2010) Cell. Mol. Life Sci. 67, 1567–1579.<br />

Cell survival requires the active inhibition of apoptosis, which is accomplished by inhibiting the expression of pro-apoptotic factors as well as promoting the expression of<br />

anti-apoptotic factors. The PI3K pathway, activated by many survival factors, leads to the activation of Akt, an important player in survival signaling. PTEN negatively regulates<br />

the PI3K/Akt pathway. Activated Akt phosphorylates and inhibits the pro-apoptotic Bcl-2 family members Bad, Bax, caspase-9, GSK-3, and FoxO1. Many growth factors and<br />

cytokines induce anti-apoptotic Bcl-2 family members. The Jaks and Src phosphorylate and activate Stat3, which in turn induces the expression of Bcl-xL and Bcl-2. Erk1/2<br />

and PKC activate p90RSK, which activates CREB and induces the expression of Bcl-xL and Bcl-2. These Bcl-2 family members protect the integrity of mitochondria, preventing<br />

cytochrome c release and the subsequent activation of caspase-9. TNF-α may activate both pro-apoptotic and anti-apoptotic pathways; TNF-α can induce apoptosis by<br />

activating caspase-8 and -10, but can also inhibit apoptosis via NF-κB, which induces the expression of anti-apoptotic genes such as Bcl-2. cIAP1/2 inhibit TNF-α signaling<br />

by binding to TRAF2. FLIP inhibits the activation of caspase-8.<br />

Select Reviews:<br />

Brumatti, G., Salmanidis, M., and Ekert, P.G. (2010) Cell. Mol. Life Sci. 67, 1619–1630. • Fuchs, Y. and Steller, H. (2011) Cell 147, 742–758. • Fulda, S. and Vucic, D.<br />

(2012) Nat. Rev. Drug Discov. 11, 109–124. • Kaufmann, T., Strasser, A., and Jost, P.J. (2012) Cell Death Differ. 19, 42–50. • Lopez, J. and Meier, P. (2010) Curr. Opin.<br />

Cell Biol. 22, 872–881. • Rong, Y. and Distelhorst, C.W. (2008) Annu. Rev. Physiol. 70, 73–91. • Srinivasula, S.M. and Ashwell, J.D. (2008) Mol. Cell 30, 123–135. •<br />

Zhang, X., Tang, N., Hadden, T.J., and Rishi, A.K. (2011) Biochim. Biophys. Acta. 1813, 1978–1986.<br />

© 2002–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Junying Yuan, Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

90 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

© 2002–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Junying Yuan, Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

www.cellsignal.com/cstpathways 91


Section I: Research Areas<br />

chapter 03: Cell Growth and Death<br />

Death Receptor Signaling<br />

Mitochondrial Control of Apoptosis<br />

MKK1<br />

JNR<br />

ASK1<br />

Bcl-2<br />

Casp-9<br />

Casp-6<br />

FasL<br />

Daxx<br />

Fas/<br />

CD95<br />

tBID<br />

Bcl-2<br />

RIP<br />

BID<br />

Apaf1<br />

XIAP<br />

FADD<br />

Casp-8, -10<br />

FLIP<br />

Cyto c<br />

TNF-α<br />

TNF-R1<br />

TRAF2<br />

TRADD<br />

cIAP1/2<br />

Casp-8<br />

FADD<br />

Necroptosis<br />

RIP1<br />

Complex<br />

IIb<br />

RIP3<br />

MLKL<br />

CYLD<br />

RIP1<br />

XIAP<br />

IKKα/β/γ<br />

Casp-8<br />

FADD<br />

RIP1<br />

Casp-3<br />

Lamin A Actin GAS2 α-Fodrin ROCK1 DFF45 Acinus PARP<br />

ub<br />

Smac<br />

Complex<br />

IIa<br />

Active<br />

Caspase-8<br />

DFF40<br />

TNF-α<br />

TNF-R2<br />

TRAF2 TRAF1<br />

cIAP1/2<br />

IκB<br />

NF-κB<br />

TRAF3<br />

TRAF2<br />

TRAF1<br />

Casp-7<br />

RIP<br />

cIAP1/2<br />

APO-3L/<br />

TWEAK<br />

DR3<br />

APO-3<br />

TRADD TRADD<br />

IκB<br />

p65/<br />

RelA<br />

FADD<br />

Casp-8,-10<br />

NF-κB<br />

p50<br />

FADD<br />

p52<br />

APO-2L/<br />

TRAIL<br />

DR4/5<br />

NF-κB<br />

p100<br />

NIK<br />

IKKα<br />

RelB<br />

RelB<br />

Processing<br />

Survival Factors:<br />

Growth Factors, Cytokines, etc.<br />

PKA<br />

14-3-3<br />

PKC<br />

Erk1/2<br />

p90RSK<br />

Bad<br />

14-3-3<br />

Bad<br />

Cytosolic<br />

Sequestration<br />

HSP60<br />

Casp-3<br />

PI3K<br />

Akt<br />

p70 S6K<br />

Calcineurin<br />

Apaf-1<br />

Casp-9<br />

ub<br />

XIAP<br />

Arts<br />

FADD<br />

BID<br />

FasL<br />

Fas/<br />

CD95<br />

Casp-8,-10<br />

tBID<br />

Bax<br />

Bak<br />

Bad<br />

Cyto c<br />

HECTH9<br />

Bcl-xL<br />

HtrA2<br />

Mcl-1<br />

Bcl-xL<br />

tBID<br />

Smac/<br />

Diablo<br />

Mcl-1<br />

Bim<br />

LC8<br />

Microtubules<br />

DLC2<br />

Bmf<br />

Puma<br />

AIF<br />

Bim<br />

Bcl-2<br />

HECTH9<br />

Endo G<br />

Bcl-2<br />

Bmf<br />

Bcl-2<br />

Noxa<br />

Death Stimuli:<br />

Survival Factor Withdrawal<br />

Hrk<br />

Bcl-2<br />

Bax<br />

Bak<br />

p53<br />

ATM/<br />

ATR<br />

JNK<br />

Hrk<br />

ING2<br />

[NAD]<br />

SirT2<br />

JNK<br />

Bax<br />

Casp-2<br />

ARD1<br />

NatA<br />

Acetyl-CoA<br />

RAIDD<br />

CaMKII<br />

PIDD<br />

Cell Shrinkage<br />

Membrane Blebbing<br />

DNA<br />

Fragmentation<br />

Apoptosis<br />

Chromatin<br />

Condensation<br />

Transcription<br />

Survival<br />

Pro-Apoptotic<br />

Pro-Survival<br />

Apoptosis<br />

DNA Damage<br />

Genotoxic Stress<br />

Apoptosis can be induced through the activation of death receptors including Fas, TNFαR, DR3, DR4, and DR5 by their respective ligands. Death receptor ligands characteristically<br />

initiate signaling via receptor oligomerization, which in turn results in the recruitment of specialized adaptor proteins and activation of caspase cascades. Binding of FasL<br />

induces Fas trimerization, which recruits initiator caspase-8 via the adaptor protein FADD. Caspase-8 then oligomerizes and is activated via autocatalysis. Activated caspase-8<br />

stimulates apoptosis via two parallel cascades: it can directly cleave and activate caspase-3, or alternatively, it can cleave Bid, a pro-apoptotic Bcl-2 family protein. Truncated<br />

Bid (tBid) translocates to mitochondria, inducing cytochrome c release, which sequentially activates caspase-9 and -3. TNF-α and DR-3L can deliver pro- or anti-apoptotic<br />

signals. TNFαR and DR3 promote apoptosis via the adaptor proteins TRADD/FADD and the activation of caspase-8. Interaction of TNF-α with TNFαR may activate the NF-κB<br />

pathway via NIK/IKK. The activation of NF-κB induces the expression of pro-survival genes including Bcl-2 and FLIP, the latter can directly inhibit the activation of caspase-8.<br />

FasL and TNF-α may also activate JNK via ASK1/MKK7. Activation of JNK may lead to the inhibition of Bcl-2 by phosphorylation. In the absence of caspase activation, stimulation<br />

of death receptors can lead to the activation of an alternative programmed cell death pathway termed necroptosis by forming complex IIb.<br />

Select Reviews:<br />

Declercq, W., Vanden Berghe, T., and Vandenabeele, P. (2009) Cell 138, 229–232. • Fuchs, Y. and Steller H. (2011) Cell 147, 742–758. • Kantari, C. and Walczak, H.<br />

(2011) Biochim. Biophys. Acta. 1813, 558–563. • Kaufmann, T., Strasser, A., and Jost, P.J. (2012) Cell Death Differ. 19, 42–50. • Lavrik, I.N. and Krammer, P.H. (2012)<br />

Cell Death Differ. 19, 36–41. • Van Herreweghe, F., Festjens, N., Declercq, W., and Vandenabeele, P. (2010) Cell. Mol. Life Sci. 67, 1567–1579. • Wajant, H. and<br />

Scheurich, P. (2011) FEBS J. 278, 862–876.<br />

The Bcl-2 family of proteins regulate apoptosis by controlling mitochondrial permeability. The anti-apoptotic proteins Bcl-2 and Bcl-xL reside in the outer mitochondrial wall<br />

and inhibit cytochrome c release. The pro-apoptotic Bcl-2 proteins Bad, Bid, Bax, and Bim may reside in the cytosol but translocate to mitochondria following death signaling,<br />

where they promote the release of cytochrome c. Bad translocates to mitochondria and forms a pro-apoptotic complex with Bcl-xL. This translocation is inhibited by survival<br />

factors that induce the phosphorylation of Bad, leading to its cytosolic sequestration. Cytosolic Bid is cleaved by caspase-8 following signaling through Fas; its active fragment<br />

(tBid) translocates to mitochondria. Bax and Bim translocate to mitochondria in response to death stimuli, including survival factor withdrawal. Activated following DNA damage,<br />

p53 induces the transcription of Bax, Noxa, and Puma. Upon release from mitochondria, cytochrome c binds to Apaf-1 and forms an activation complex with caspase-9.<br />

Although the mechanism(s) regulating mitochondrial permeability and the release of cytochrome c during apoptosis are not fully understood, Bcl-xL, Bcl-2, and Bax may influence<br />

the voltage-dependent anion channel (VDAC), which may play a role in regulating cytochrome c release. Mule/ARF-BP1 is a DNA damage-activated E3 ubiquitin ligase<br />

for p53, and Mcl-1, an anti-apoptotic member of Bcl-2.<br />

Select Reviews:<br />

Brenner, D. and Mak, T.W. (2009) Curr. Opin. Cell Biol. 21, 871–877. • Chalah, A., Khosravi-Far, R. (2008) Adv. Exp. Med. Biol. 615, 25–45. • Lindsay, J., Esposti, M.D.,<br />

and Gilmore, A.P. (2011) Biochim. Biophys. Acta. 1813, 532–539. • Ola, M.S., Nawaz, M., and Ahsan, H. (2011) Mol. Cell. Biochem. 351, 41–58. • Pradelli, L.A.,<br />

Bénéteau, M., and Ricci, J.E. (2010) Cell. Mol. Life Sci. 67, 1589–1597. • Rong, Y. and Distelhorst, C.W. (2008) Annu. Rev. Physiol. 70, 73–91. • Speidel, D. (2010)<br />

Trends Cell Biol. 20, 14–24. • Suen, D.F., Norris, K.L., and Youle, R.J. (2008) Genes Dev. 22, 1577–1590.<br />

© 2002–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Junying Yuan, Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

92 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

© 2003–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Junying Yuan, Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

www.cellsignal.com/cstpathways 93


Section I: Research Areas<br />

Nutrient deprivation<br />

results in accumulation<br />

of Atg13 at sites<br />

of autophagy.<br />

Autophagy<br />

Autophagy is a dynamic cellular recycling system that degrades cytoplasmic contents, abnormal<br />

protein aggregates, and excess or damaged organelles so that the building blocks, such as amino<br />

acids, can be used to create new cellular components. Autophagy occurs when the protein, organelle,<br />

or cytoplasmic content to be degraded is surrounded by a small portion of membrane, creating<br />

an autophagosome. The autophagosome is then fused to the lysosome, creating an autolysosome<br />

and resulting in degradation of cellular components via lysosomal enzymes. Autophagy is generally<br />

activated by conditions of nutrient deprivation but has also been associated with physiological as well<br />

as pathological processes such as development, differentiation, neurodegenerative diseases, stress,<br />

infection, obesity, and cancer.<br />

Autophagy Induction<br />

The molecular machinery of autophagy was largely discovered in yeast and is directed by a number<br />

of autophagy-related (Atg) genes. The mammalian serine/threonine kinase ULK (similar to yeast Atg1)<br />

plays a critical role in autophagy induction, acting as a convergence point for upstream signals. Activation<br />

of mTOR through Akt or MAPK signaling results in inhibition of ULK and suppression of autophagy,<br />

whereas negative regulation of mTOR through p53 and AMPK signaling relieves this inhibition and<br />

promotes autophagy. In addition, AMPK also phosphorylates and activates ULK directly. ULK forms a<br />

complex with Atg13 and the scaffold protein FIP200.<br />

Signaling through ULK1 activates phosphoinositide 3-kinase class III (PI3K class III or hVps34), a lipid<br />

kinase that produces phosphatidylinositol-3-phosphate (PIP 3 ) and forms a large complex with associated<br />

proteins p105/Vsp15, Beclin-1, UVRAG, Atg14, and Rubicon. As autophagy signaling is induced,<br />

membranes from cytoplasmic vesicles collect and fuse, forming the phagophore. The PI3K class III<br />

complex acts upon phagophore lipids, creating binding sites for autophagic proteins and initiating the<br />

formation of autophagosomes.<br />

Treatment with AMPK activators result<br />

in phosphorylation of ULK1 at Ser555.<br />

Phospho-ULK1 (Ser555) (D1H4) Rabbit mAb #5869: WB analysis of<br />

extracts from MCF7 cells, untreated or treated with Oligomycin #9996<br />

(0.5 μM, 30 min), and C2C12 cells, untreated or treated with hydrogen<br />

peroxide (10 mM, 5 min), using #5869.<br />

PI3 Kinase class III, which is a lipid kinase<br />

that initiates autophagosome formation, is<br />

expressed in mouse brain and C6 cells.<br />

PI3 Kinase Class III (D9A5) Rabbit mAb #4263: WB analysis of extracts<br />

from mouse brain and C6 cells using #4263.<br />

94 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

MCF7<br />

C2C12<br />

– + – –<br />

– – – +<br />

1 2<br />

Phospho-<br />

ULK1<br />

(Ser555)<br />

Oligomycin<br />

Hydrogen Peroxide<br />

Atg13 (E1Y9V) Rabbit mAb #13468:<br />

Confocal IF analysis of PANC-1 cells,<br />

untreated (left) or nutrient-starved<br />

with Earle’s balanced salt solution for<br />

3 hr (right), using #13468 (green).<br />

Blue pseudocolor = DRAQ5 ® #4084<br />

(fluorescent DNA dye).<br />

PI3 Kinase<br />

Class III<br />

Lanes<br />

1. mouse brain<br />

2. C6<br />

Autophagosome Formation<br />

Formation of the autophagosome requires a pair of essential ubiquitin-like conjugation systems, Atg12-<br />

Atg5 and LC3-phosphatidylethanolamine (LC3-PE). In the first conjugation reaction, Atg12 is covalently<br />

bound to Atg5 by the ubiquitin E1-like enzyme Atg7 and the E2-like enzyme Atg10. The Atg12-Atg5<br />

conjugate then forms a complex with Atg16L1 and localizes to phagophore membranes. In the<br />

second conjugation step, LC3 is first primed for lipidation through cleavage of its C-terminus by Atg4,<br />

generating LC3-I. LC3-I is then conjugated to PE in a ubiquitin-like reaction that requires Atg7 and<br />

Atg3 (E1- and E2-like enzymes, respectively). The lipidated form of LC3, known as LC3-II, is attached<br />

to the autophagosome membrane. Throughout this process, the phagophore membrane continues to<br />

expand and surround cytoplasmic components that have been designated for degradation. Sequestosome<br />

1 (SQSTM1, p62), a ubiquitin binding protein that binds LC3, facilitates this process by delivering<br />

SQSTM1-containing protein aggregates to the autophagosome for destruction.<br />

Nutrient starvation and chloroquine treatment result in<br />

expression of LC3A/B in human and mouse cells.<br />

HeLa (human)<br />

C2C12 (mouse)<br />

Mitophagy<br />

Mitophagy is a selective autophagic process specifically designed for the removal of damaged or unneeded<br />

mitochondria from a cell. Polyubiquitination of mitochondrial membrane proteins by Parkin results<br />

in the recruitment of autophagy adaptor proteins that bind to LC3 via their LC3-interacting region<br />

(LIR). A second level of ubiquitin-independent regulation occurs through the mitochondrial membrane<br />

proteins BNIP3 and BNIP3L/NIX, which also contain LIRs and directly recruit autophagic machinery to<br />

induce autophagosome formation in certain cell types.<br />

BNIP3L/Nix (D4R4B) Rabbit mAb<br />

#12396: Confocal IF analysis of A172<br />

cells, untreated (left) or cobalt chloridetreated<br />

(100 μM, overnight; right), using<br />

#12396 (green). Blue pseudocolor =<br />

DRAQ5 ® #4084 (fluorescent DNA dye).<br />

A B C<br />

Select Reviews<br />

Alers, S., Löffler, A.S., Wesselborg, S., and Stork, B. (2012) Mol. Cell. Biol. 32, 2–11. • Codogno, P., Mehrpour, M., and<br />

Proikas-Cezanne, T. (2012) Nat. Rev. Mol. Cell Biol. 13, 7–12. • Ding, W.X. and Yin, X.M. (2012) Biol. Chem. 393, 547–564.<br />

• Feng, D., Liu, L., Zhu, Y., and Chen, Q. (2013) Exp. Cell. Res. 319, 1697–1705. • Jin, M. and Klionsky, D.J. (2014) FEBS<br />

Lett. 588, 2457–2463. • Schneider, J.L. and Cuervo, A.M. (2014) Curr. Opin. Genet. Dev. 26C, 16–23. • Papinski, D. and<br />

Kraft, C. (2014) Autophagy 10, 1338–1340.<br />

chapter 03: Cell Growth and Death<br />

Atg5, which regulates<br />

the first step in autophagosome<br />

formation,<br />

is widely expressed.<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

1 2 3 4 5 6<br />

Atg5<br />

Atg5 (D5G3) Rabbit mAb #9980: WB<br />

analysis of extracts from various cell lines<br />

using #9980.<br />

Lanes<br />

1. PANC-1 4. ACHN<br />

2. K-562 5. HT-1080<br />

3. SH-SY5Y 6. COS-7<br />

LC3A/B (D3U4C) XP ® Rabbit mAb<br />

#12741: Confocal IF analysis of HeLa<br />

(upper) and C2C12 (lower) cells,<br />

chloroquine-treated (50 μM, overnight)<br />

(A), nutrient-starved with EBSS (3 hr)<br />

(B) or untreated (C) using #12741<br />

(green) and β-Actin (13E5) Rabbit mAb<br />

(Alexa Fluor ® 555 Conjugate) #8046<br />

(red). Blue pseudocolor= DRAQ5 ®<br />

#4084 (fluorescent DNA dye).<br />

Cobalt chloride,<br />

which induces the<br />

hypoxia regulator<br />

HIF-1α, results in<br />

accumulation of the<br />

mitophagy regulator<br />

protein BNIP3L/Nix.<br />

www.cellsignal.com/cstautophagy 95


Section I: Research Areas<br />

These protein targets represent key<br />

nodes within autophagy signaling<br />

pathways and are commonly studied<br />

in autophagy research. Primary<br />

antibodies, antibody conjugates, and<br />

antibody sampler kits containing<br />

these targets are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

S SignalSilence ® siRNA<br />

207<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for LC3B have been<br />

cited over 207 times in high-impact,<br />

peer-reviewed publications from the<br />

global research community.<br />

Antibody<br />

Validation<br />

Principles<br />

Please visit our website to learn more<br />

about what Antibody Validation means<br />

at Cell Signaling Technology.<br />

www.cellsignal.com/cstvalidation<br />

Commonly Studied Autophagy Targets<br />

Target M P S Target M P S<br />

Ambra1<br />

• Phospho-Beclin-1<br />

Atg3<br />

(Ser93/96) •<br />

•<br />

Atg4A<br />

Bif-1<br />

•<br />

•<br />

Atg4B<br />

BNIP3<br />

• • •<br />

•<br />

Atg4C<br />

BNIP3L/Nix<br />

• •<br />

•<br />

Atg5<br />

FIP200<br />

• • •<br />

•<br />

Atg7<br />

GABARAP<br />

• • •<br />

•<br />

Atg9A GABARAPL2<br />

• •<br />

•<br />

Atg12<br />

LC3A<br />

• •<br />

• •<br />

Atg13 LC3A/B<br />

• •<br />

• •<br />

Atg14<br />

LC3B<br />

• •<br />

• • •<br />

Atg16L1<br />

MTMR3<br />

•<br />

•<br />

Atg101<br />

MTMR14<br />

•<br />

•<br />

Beclin-1<br />

NBR1<br />

• • •<br />

•<br />

Phospho-Beclin-1 (Ser15)<br />

Rubicon<br />

•<br />

•<br />

SQSTM1/p62 • • •<br />

Select Citations:<br />

Mancias, J.D. et al. (2014) Quantitative proteomics identifies<br />

NCOA4 as the cargo receptor mediating ferritinophagy.<br />

Nature 509, 105–109.<br />

Bejarano, E. et al. (2014) Connexins modulate autophagosome<br />

biogenesis. Nat. Cell Biol. 16, 401–414.<br />

Jang, Y.H. et al. (2014) Phospholipase D-mediated autophagic<br />

regulation is a potential target for cancer therapy. Cell<br />

Death Differ. 21, 533–546.<br />

Kim, J. et al. (2014) Differential regulation of distinct Vps34<br />

complexes by AMPK in nutrient stress and autophagy. Cell<br />

152, 290–303.<br />

Mealer, R.G. et al. (2014) Rhes, a striatal-selective protein<br />

implicated in Huntington disease, binds beclin-1 and activates<br />

autophagy. J. Biol. Chem. 289, 3547–3554.<br />

Efeyan, A. et al. (2014) Regulation of mTORC1 by the Rag<br />

GTPases is necessary for neonatal autophagy and survival.<br />

Nature 93, 679–683.<br />

Brot, S. et al. (2014) Collapsin response mediator protein 5<br />

(CRMP5) induces mitophagy, thereby regulating mitochondrion<br />

numbers in dendrites. J. Biol. Chem. 289, 2261–2276.<br />

Martins, I. et al. (2014) Molecular mechanisms of ATP<br />

secretion during immunogenic cell death. Cell Death Differ.<br />

21, 79–91.<br />

Lu, B. et al. (2014) JAK/STAT1 signaling promotes HMGB1<br />

hyperacetylation and nuclear translocation. Proc. Natl. Acad.<br />

Sci. USA 111, 3068–3073.<br />

Li, Q. et al. (2014) Cited2, a transcriptional modulator<br />

protein, regulates metabolism in murine embryonic stem<br />

cells. J. Biol. Chem. 289, 251–263.<br />

Papa, L. et al. (2014) SirT3 regulates the mitochondrial<br />

unfolded protein response. Mol Cell Biol. 34, 699–710.<br />

Hong, S.W. et al. (2013) SVCT-2 in breast cancer acts<br />

as an indicator for L-ascorbate treatment. Oncogene 32,<br />

1508–1517.<br />

Zhai, H. et al. (2013) Inhibition of autophagy and tumor<br />

growth in colon cancer by miR-502. Oncogene 32,<br />

1570–1579.<br />

Brot, S. et al. (2014) Collapsin response mediator protein 5<br />

(CRMP5) induces mitophagy, thereby regulating mitochondrion<br />

numbers in dendrites. J. Biol. Chem. 289, 2261–2276.<br />

Target M P S<br />

Phospho-SQSTM1/p62<br />

(Thr269/Ser272) •<br />

Phospho-SQSTM1/p62<br />

(Ser403)<br />

•<br />

TECPR1 •<br />

TMEM49/VMP1 •<br />

ULK1 • • •<br />

Phospho-ULK1 (Ser317) • •<br />

Phospho-ULK1 (Ser467) •<br />

Phospho-ULK1 (Ser555) •<br />

Phospho-ULK1 (Ser638) • •<br />

Phospho-ULK1 (Ser757) • •<br />

UVRAG • •<br />

WIPI1<br />

• •<br />

WIPI2<br />

•<br />

Phospho-WIPI2 (Ser413) •<br />

Mealer, R.G. et al. (2014) Rhes, a striatal-selective protein<br />

implicated in Huntington disease, binds beclin-1 and activates<br />

autophagy. J. Biol. Chem. 289, 3547–3554.<br />

Huck, B. et al. (2014) Elevated protein kinase D3 (PKD3)<br />

expression supports proliferation of triple-negative breast<br />

cancer cells and contributes to mTORC1-S6K1 pathway<br />

activation. J. Biol. Chem. 289, 3138–3147.<br />

Conacci-Sorrell, M. et al. (2014) Stress-induced cleavage of<br />

Myc promotes cancer cell survival. Genes Dev. 28, 689–707.<br />

Huck, B. et al. (2014) Elevated protein kinase D3 (PKD3)<br />

expression supports proliferation of triple-negative breast<br />

cancer cells and contributes to mTORC1-S6K1 pathway<br />

activation. J. Biol. Chem. 289, 3138–3147.<br />

Talaber, G. et al. (2014) HRES-1/Rab4 promotes the formation<br />

of LC3(+) autophagosomes and the accumulation of<br />

mitochondria during autophagy. PLoS One 9, 84392.<br />

Lin, G. et al. (2014) Reduced Warburg effect in cancer cells<br />

undergoing autophagy: steady- state 1H-MRS and real-time<br />

hyperpolarized 13C-MRS studies. PLoS One 9, 92645.<br />

Nemati, F. et al. (2014) Targeting Bcl-2/Bcl-XL induces antitumor<br />

activity in uveal melanoma patient-derived xenografts.<br />

PLoS One 9, e80836.<br />

Ren, X.S. et al. (2014) Activation of the PI3K/mTOR pathway<br />

is involved in cystic proliferation of cholangiocytes of the PCK<br />

rat. PLoS One 9, e87660.<br />

Shiroto, T. et al. (2014) Caveolin-1 is a critical determinant<br />

of autophagy, metabolic switching, and oxidative stress in<br />

vascular endothelium. PLoS One, e87871.<br />

Uetake, R. et al. (2014) Adrenomedullin-RAMP2 system suppresses<br />

ER stress-induced tubule cell death and is involved<br />

in kidney protection. PLoS One 9, e87667.<br />

Chang, P.C. et al. (2014) Autophagy pathway is required for<br />

IL-6 induced neuroendocrine differentiation and chemoresistance<br />

of prostate cancer LNCaP cells. PLoS One 9, e88556.<br />

Lin, L. et al. (2014) Mechanical stress triggers cardiomyocyte<br />

autophagy through angiotensin II type 1 receptormediated<br />

p38MAP kinase independently of angiotensin II.<br />

PLoS One 9, e89629.<br />

Gu, W. et al. (2014) Ambra1 is an essential regulator of<br />

autophagy and apoptosis in SW620 cells: pro-survival role of<br />

Ambra1. PLoS One 9, e90151.<br />

Autophagy Signaling<br />

AMP:<br />

ATP<br />

AMPK<br />

Apoptosis<br />

Phagophore<br />

Atg16L1<br />

Amino<br />

Acids<br />

Atg16L1<br />

Macroautophagy<br />

PI3K-I/Akt<br />

Signaling<br />

GβL<br />

MAPK/Erk1/2<br />

Signaling<br />

Atg13 FIP200<br />

ULK1<br />

p150<br />

PI3K<br />

Class III<br />

Beclin-1<br />

Bcl-2<br />

Atg14<br />

Atg5<br />

Rubicon<br />

Atg12<br />

Atg12<br />

mTOR<br />

Ambra1<br />

Atg5<br />

Atg10<br />

Atg7<br />

p53/Genotoxic<br />

Stress<br />

Raptor<br />

PRAS40<br />

ub<br />

Cytoplasmic<br />

Contents<br />

Oxidative<br />

Stress<br />

Membrane<br />

Nucleation<br />

ub<br />

Mitophagy<br />

Mitochondrial<br />

Damage<br />

PARL<br />

BNIP3<br />

SQSTM1/p62<br />

BNIP3L/NIX<br />

+ NBR1 +<br />

ALFY<br />

Atg3<br />

Atg7<br />

Atg4<br />

Sequestration<br />

LC3-II<br />

LC3-I<br />

LC3<br />

PE<br />

PINK<br />

SQSTM1/p62<br />

NBR1<br />

Ambra1<br />

chapter 03: Cell Growth and Death<br />

ub<br />

Parkin<br />

+ LC3-II<br />

Autophagosome<br />

Lysosome<br />

ub -targets<br />

Fusion<br />

Autophagolysosome<br />

Macroautophagy, often referred to as autophagy, is a catabolic process that results in the autophagosomic-lysosomal degradation of bulk cytoplasmic contents, abnormal<br />

protein aggregates, and excess or damaged organelles. Autophagy is generally activated by conditions of nutrient deprivation but has also been associated with physiological<br />

as well as pathological processes such as development, differentiation, neurodegenerative diseases, stress, infection, and cancer. The kinase mTOR is a critical regulator of<br />

autophagy induction, with activated mTOR (Akt and MAPK signaling) suppressing autophagy, and negative regulation of mTOR (AMPK and p53 signaling) promoting it. Three<br />

related serine/threonine kinases, UNC-51-like kinase -1, -2, and -3 (ULK1, ULK2, UKL3), which play a similar role as the yeast Atg1, act downstream of the mTOR complex.<br />

ULK1 and ULK2 form a large complex with the mammalian homolog of an autophagy-related (Atg) gene product (mAtg13) and the scaffold protein FIP200 (an ortholog of<br />

yeast Atg17). Class III PI3K complex, containing hVps34, Beclin-1 (a mammalian homolog of yeast Atg6), p150 (a mammalian homolog of yeast Vps15), and Atg14-like<br />

protein (Atg14L or Barkor) or ultraviolet irradiation resistance-associated gene (UVRAG), is required for the induction of autophagy. The Atg genes control autophagosome<br />

formation through Atg12-Atg5 and LC3-II (Atg8-II) complexes. Atg12 is conjugated to Atg5 in a ubiquitin-like reaction that requires Atg7 and Atg10 (E1 and E2-like enzymes,<br />

respectively). The Atg12-Atg5 conjugate then interacts noncovalently with Atg16 to form a large complex. LC3/Atg8 is cleaved at its C-terminus by Atg4 protease to generate<br />

the cytosolic LC3-I. LC3-I is conjugated to phosphatidylethanolamine (PE) also in a ubiquitin-like reaction that requires Atg7 and Atg3 (E1 and E2-like enzymes, respectively).<br />

The lipidated form of LC3, known as LC3-II, is attached to the autophagosome membrane. Autophagy and apoptosis are connected both positively and negatively, and extensive<br />

crosstalk exists between the two processes. During nutrient deficiency, autophagy functions as a pro-survival mechanism; however, excessive autophagy may lead to<br />

cell death, a process morphologically distinct from apoptosis. Several pro-apoptotic signals, such as TNF, TRAIL, and FADD, also induce autophagy. Additionally, Bcl-2 inhibits<br />

Beclin-1-dependent autophagy, thereby functioning both as a pro-survival and as an anti-autophagic regulator.<br />

Mitophagy is a selective autophagic process specifically designed for the removal of damaged or unneeded mitochondria from a cell. Upon mitochondrial damage, the protein<br />

PINK, which is continually degraded in the healthy state through the action of PARL, is stabilized and recruits the E3 ligase Parkin to initiate mitophagy. Polyubiquitination of<br />

mitochondrial membrane proteins by Parkin results in the recruitment of autophagy adaptor proteins SQSTM1/p62, NBR1, and Ambra1 that bind to LC3 via their LC3-<br />

interacting region (LIR). In addition, BNIP3 and BNIP3L/NIX, which also contain LIRs, directly recruit autophagic machinery by a ubiquitin-independent mechanism to induce<br />

autophagosome formation in certain cell types.<br />

Select Reviews:<br />

Alers, S., Löffler, A.S., Wesselborg, S., and Stork, B. (2012) Mol. Cell. Biol. 32, 2–11. • Codogno, P., Mehrpour, M., and Proikas-Cezanne, T. (2012) Nat. Rev. Mol. Cell Biol.<br />

13, 7–12. • Ding, W.X. and Yin, X.M. (2012) Biol. Chem. 393, 547–564. • Feng, D., Liu, L., Zhu, Y., and Chen, Q. (2013) Exp. Cell. Res. 319, 1697–1705. • Jin, M.<br />

and Klionsky, D.J. (2014) FEBS Lett. 588, 2457–2463. • Papinski, D. and Kraft, C. (2014) Autophagy 10, 1338–1340. • Schneider, J.L. and Cuervo, A.M. (2014) Curr.<br />

Opin. Genet. Dev. 26, 16–23.<br />

© 2003–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Bingren Hu, University of Maryland School of Medicine, Baltimore, MD, for reviewing this diagram.<br />

96 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstpathways<br />

97


Section I: Research Areas<br />

chapter 03: Cell Growth and Death<br />

Cell Cycle, Checkpoint<br />

Control, and DNA Damage<br />

Control of eukaryotic cell growth and division involves molecular circuits known as “checkpoints” that<br />

ensure proper timing of cellular events. Passage through a checkpoint from one cell cycle phase to the<br />

next requires a coordinated set of proteins that monitor cell growth and DNA integrity. Uncontrolled cell<br />

division or propagation of damaged DNA can contribute to genomic instability and tumorigenesis.<br />

Phases of the Cell Cycle<br />

G2/M Checkpoint<br />

The G2/M checkpoint prevents cells containing damaged DNA from entering mitosis (M). Activated<br />

CDK1 (cdc2) bound to cyclin B promotes entry into M-phase. Wee1 and Myt1 kinases and cdc25<br />

phosphatase competitively regulate CDK1 activity; Wee1 and Myt1 inhibit CDK1 and prevent entry into<br />

M-phase, while cdc25 removes inhibitory phosphates. DNA damage activates multiple kinases such<br />

as ATM/ATR, DNA-PK, HIPK2 that phosphorylate kinases Chk1/2 and tumor suppressor protein p53.<br />

Chk1/2 kinases stimulate Wee1 activity and inhibit cdc25C, preventing entry into M-phase. Phosphorylation<br />

of p53 promotes dissociation between p53 and MDM2 and allows binding of the transcription<br />

factor to DNA.<br />

UV treatment results<br />

in phosphorylation of<br />

ATM at Ser1981.<br />

kDa<br />

200<br />

Phospho-ATM<br />

(Ser1981)<br />

G2<br />

Preparation<br />

for Mitosis<br />

Prophase<br />

Metaphase<br />

Anaphase<br />

M<br />

Cyclin B1/CDK1 complex is expressed in replicating Jurkat cells.<br />

Cyclin B1 (D5C10) XP ® Rabbit mAb #12231: Flow cytometric analysis of<br />

Jurkat cells using #12231 and Propidium Iodide (PI)/RNase Staining Solution<br />

#4087 (DNA content). Anti-rabbit IgG (H+L), F(ab’) 2<br />

Fragment (Alexa Fluor ®<br />

488 Conjugate) #4412 was used as a secondary antibody.<br />

Cyclin B1<br />

10 4<br />

10 3<br />

10 2<br />

140<br />

100<br />

80<br />

200<br />

ATM<br />

Telophase<br />

10 1<br />

140<br />

G 0<br />

10 0 0 200 400 600 800 1000<br />

DNA (PI)<br />

100<br />

S<br />

DNA<br />

Replication<br />

Preparation<br />

for DNA<br />

Synthesis<br />

G1<br />

UV treatment results in nuclear translocation of Phospho-p53.<br />

Phospho-p53 (Ser15) (16G8) Mouse<br />

mAb (Alexa Fluor ® 555 Conjugate)<br />

#9481: Confocal IF analysis of HT-29<br />

cells, untreated (left) or UV-treated<br />

(right), using #9481 (red). Actin filaments<br />

were labeled with Alexa Fluor ®<br />

488 Phalloidin #8878 (green).<br />

80<br />

– + UV<br />

Phospho-ATM (Ser1981) (D6H9)<br />

Rabbit mAb #5883: WB analysis of<br />

extracts from 293 cells, untreated or<br />

UV-treated (100 mJ, 4 hr recovery),<br />

using #5883 (upper) or ATM (D2E2)<br />

Rabbit mAb #2873 (lower).<br />

CDK inhibitor p27<br />

Kip1 is expressed<br />

in quiescent cells<br />

and degraded in<br />

mitotic cells.<br />

p27 Kip1 (D69C12) XP ® Rabbit<br />

mAb #3686: Flow cytometric<br />

analysis of Jurkat cells using<br />

#3686 versus Propidium Iodide<br />

(PI)/RNase Staining Solution<br />

#4087. Anti-rabbit IgG (H+L),<br />

F(ab’) 2 Fragment (Alexa Fluor ®<br />

488 Conjugate) #4412 was<br />

used as a secondary antibody.<br />

G1/S Checkpoint<br />

The G1/S checkpoint controls progression of cells through the restriction point (R) into the DNA<br />

synthesis S-phase. During G1, the tumor suppressor Rb binds and inhibits transcription factor E2F.<br />

Phosphorylation of Rb by cyclin-bound cyclin dependent kinases (CDK) in late G1 induces dissociation<br />

of Rb and permits E2F-mediated transcription of S-phase-promoting genes. Responding to upstream<br />

signals, INK4 and Kip/Cip family inhibitors control CDK activity and prevent entry into S-phase. DNA<br />

damage activates response pathways through ATM/ATR and Chk1/2 kinases to block CDK activity,<br />

leading to cell cycle arrest and DNA repair or cell death.<br />

p27 Kip1<br />

10 4<br />

10 3<br />

10 2<br />

10 1<br />

10 0 0 200 400 600 800 1000<br />

DNA (PI)<br />

Serum treatment results in<br />

phosphorylation of Rb at Ser807/811.<br />

Phospho-Rb (Ser807/811)<br />

(D20B12) XP ® Rabbit mAb<br />

#8516: WB analysis of extracts<br />

from WI-38 cells using #8516.<br />

Lanes<br />

1. Serum-starved for 3 days (-)<br />

2. Serum-starved for 3 days<br />

followed by 10% serum for<br />

2 days (+)<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

1 2<br />

Phospho-Rb<br />

(Ser807/811)<br />

Spindle Checkpoint<br />

As mitosis proceeds, mitotic spindles are assembled that connect the kinetochore regions of sister<br />

chromatids to the microtubules within the spindle, centering the chromatids along the metaphase<br />

plate. Many proteins comprise the kinetochore and play a role in this process, including the histone H3<br />

variant CENP-A, survivin, and Aurora B kinase, which is itself regulated by the microtubule nucleating<br />

protein TPX2. The spindle checkpoint ensures proper chromatid attachment prior to progression from<br />

metaphase to anaphase. The SCF and APC/C protein complexes play prominent roles in the spindle<br />

checkpoint, with APC-cdc20 initiating the entry into anaphase by promoting ubiquitin-mediated degradation<br />

of multiple substrates, including cyclin B and the regulatory protein securin.<br />

Chemical Modulators for Studies of the Cell Cycle and DNA Damage<br />

#9886 Docetaxel Inhibits microtubule depolymerization; prevents cell division<br />

#5927 Doxorubicin Inhibits DNA and RNA synthesis by intercalating the DNA helix; inhibits topoisomerase I<br />

#2200 Etoposide Inhibits topoisomerase II resulting in DNA breakage; induces apoptosis<br />

#2194 MG-132 Potent inhibitor of the proteasome and calpain<br />

#2190 Nocodazole Inhibits microtubule polymerization; induces cell cycle arrest in G2/M-phase<br />

#9807 Paclitaxel Inhibits microtubule depolymerization; prevents cell division<br />

#9885 Roscovitine Potent and selective inhibitor of CDK1, 2, and 5 (ATP-competitive)<br />

TPX2 regulates<br />

activity of Aurora B,<br />

a kinase critical for<br />

proper chromatid<br />

attachment to the<br />

mitotic spindle.<br />

TPX2 (D2R5C) XP ® Rabbit mAb<br />

#12245: Confocal IF analysis of MCF7<br />

cells using #12245 (green). Actin filaments<br />

were labeled with DY-554 phalloidin<br />

(red). Blue pseudocolor = DRAQ5 ®<br />

#4084 (fluorescent DNA dye).<br />

98 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstcellcycle 99


Section I: Research Areas<br />

These protein targets represent key<br />

nodes within cell cycle signaling<br />

pathways and are commonly studied<br />

in cell cycle research. Primary<br />

antibodies, antibody conjugates, and<br />

antibody sampler kits containing<br />

these targets are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

E PathScan ® ELISA Kits<br />

S SignalSilence ® siRNA<br />

C Antibody Conjugate<br />

Aurora Antibody Sampler Kit #3875 contains antibodies to several<br />

targets important for investigating the G2/M phase of the cell cycle.<br />

Phospho-Aurora A (Thr288) (C39D8)<br />

Rabbit mAb #3079: Confocal IF<br />

analysis of mitotic HeLa cells during<br />

metaphase (left) and anaphase (right)<br />

using #3079 (red) and Phospho-Histone<br />

H3 (Ser10) (6G3) Mouse mAb #9706<br />

(green). Blue pseudocolor = DRAQ5 ®<br />

#4084 (fluorescent DNA dye).<br />

#3079 is a component of the<br />

Aurora Antibody Sampler Kit #3875.<br />

Select Reviews<br />

Bertoli, C., Skotheim, J.M., and de Bruin, R.A. (2013) Nat. Rev. Mol. Cell Biol. 14, 518–528. • Bretones, G., Delgado, M.D.,<br />

and León, J. (2014) Biochim. Biophys. Acta. April 1 [Epub ahead of print] • Choi, Y.J. and Anders, L. (2014) Oncogene<br />

33, 1890–1903. • Duronio, R.J. and Xiong, Y. (2013) Cold Spring Harb. Perspect. Biol. 5, a008904. • Johnson, A. and<br />

Skotheim, J.M. (2013) Curr. Opin. Cell Biol. 25, 717–723. • Lord, C.J. and Ashworth, A. (2012) Nature 481, 287–294. •<br />

Maréchal, A. and Zou, L. (2013) Cold Spring Harb. Perspect. Biol. 5, a012716. • Yasutis, K.M. and Kozminski, K.G. (2013) Cell<br />

Cycle 12, 1501–1509.<br />

Commonly Studied Cell Cycle Targets<br />

Target M P E S C<br />

APC6<br />

•<br />

Ape1<br />

•<br />

Artemis<br />

•<br />

ATM • •<br />

Phospho-ATM (Ser1981) •<br />

ATR • • •<br />

Phospho-ATR (Ser428) •<br />

ATRIP<br />

•<br />

Phospho-ATRIP (Ser224) •<br />

Aurora A • • •<br />

Phospho-Aurora A (Thr288) • •<br />

• •<br />

Phospho-Aurora A (Thr288)/Aurora B<br />

(Thr232)/Aurora C (Thr198)<br />

Aurora B • •<br />

BACH1/BRIP1<br />

•<br />

Phospho-BACH1/BRIP1 (Thr1133) •<br />

BLM<br />

•<br />

Bora<br />

•<br />

BRCA1 • • •<br />

Phospho-BRCA1 (Ser1524) •<br />

BrdU<br />

•<br />

BRE<br />

•<br />

Bub1<br />

•<br />

Bub1B<br />

• •<br />

Bub3<br />

• •<br />

Cdc2 • •<br />

Phospho-Cdc2 (Thr14) •<br />

Phospho-Cdc2 (Tyr15) • • •<br />

Phospho-Cdc2 (Thr161) •<br />

CDC20<br />

•<br />

Phospho-CDC20 (Ser51) •<br />

cdc25A<br />

•<br />

cdc25B<br />

•<br />

cdc25C<br />

•<br />

Phospho-cdc25C (Thr48) •<br />

Phospho-cdc25C (Ser198) •<br />

Target M P E S C<br />

Phospho-cdc25C (Ser216) • •<br />

Cdc6<br />

•<br />

Cdc7<br />

•<br />

CDC37<br />

•<br />

Cdc45<br />

• •<br />

CDC73<br />

• •<br />

CDK2 • •<br />

Phospho-CDK2 (Thr160) •<br />

CDK4<br />

•<br />

CDK6<br />

•<br />

CDK7<br />

• •<br />

CDK8 • •<br />

CDK9 • •<br />

Phospho-CDK9 (Thr186) •<br />

PITSLRE/CDK11<br />

•<br />

CDK12<br />

•<br />

CDT1<br />

• •<br />

CENP-T<br />

•<br />

CHFR<br />

• •<br />

Chk1 • • •<br />

Phospho-Chk1 (Ser280) •<br />

Phospho-Chk1 (Ser296) •<br />

Phospho-Chk1 (Ser317) • • •<br />

Phospho-Chk1 (Ser345) • • •<br />

Chk2 • • •<br />

Phospho-Chk2 (Ser19) •<br />

Phospho-Chk2 (Ser33/35) •<br />

Phospho-Chk2 (Thr68) • • • •<br />

Phospho-Chk2 (Ser516) •<br />

CK2α • •<br />

Claspin<br />

•<br />

Phospho-CLASP2 (Ser1234) •<br />

CP110<br />

•<br />

CtIP<br />

•<br />

CUEDC2<br />

•<br />

100 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

Target M P E S C<br />

Cyclin A2<br />

•<br />

Cyclin B1 • • •<br />

Phospho-Cyclin B1 (Ser116) •<br />

Phospho-Cyclin B1 (Ser133) •<br />

Phospho-Cyclin B1 (Ser147) •<br />

Cyclin D1 • • •<br />

Phospho-Cyclin D1 (Thr286) • •<br />

Cyclin D2<br />

• •<br />

Cyclin D3<br />

•<br />

Cyclin E1<br />

•<br />

Phospho-Cyclin E1 (Thr62) •<br />

Cyclin E2<br />

•<br />

Cyclin H<br />

•<br />

DNA-PK<br />

• •<br />

Phospho-DNA-PK (Ser2056) •<br />

DNA Polymerase γ •<br />

DNA Polymerase η •<br />

DYRK2<br />

• •<br />

E2F-1<br />

•<br />

EAPP<br />

•<br />

Eg5<br />

• •<br />

ENSA<br />

• •<br />

•<br />

Phospho-ENSA (Ser67)/ARPP19<br />

(Ser62)<br />

ERCC1<br />

• •<br />

FEN-1<br />

•<br />

FOXM1<br />

•<br />

Phospho-FOXM1 (Ser35) •<br />

GADD45 α<br />

•<br />

Geminin<br />

•<br />

HR6A/HR6B<br />

•<br />

INCENP<br />

•<br />

p18 INK4C<br />

•<br />

Ki-67 • •<br />

Ku70<br />

• •<br />

Ku80<br />

• •<br />

LATS1<br />

• •<br />

Phospho-LATS1 (Ser909) •<br />

Phospho-LATS1 (Thr1079) •<br />

LATS2<br />

• •<br />

MAD2L1<br />

•<br />

MASTL<br />

•<br />

MCM2<br />

• •<br />

Phospho-MCM2 (Ser139) • •<br />

Phospho-MDM2 (Ser166) •<br />

MCM3<br />

• •<br />

Phospho-MCM3 (Ser112) •<br />

MCM4<br />

• •<br />

MCM7<br />

• •<br />

MERIT40<br />

• •<br />

Phospho-MERIT40 (Ser29) •<br />

Microcephalin-1/BRIT1 •<br />

MGMT<br />

•<br />

MLH1<br />

•<br />

Mre11<br />

• •<br />

Phospho-Mre11 (Ser676) •<br />

MSH2<br />

•<br />

MSH6<br />

• •<br />

MYH<br />

• •<br />

Myt1<br />

•<br />

Target M P E S C<br />

Phospho-Myt1 (Ser83) •<br />

NCAPD3<br />

•<br />

NEK7<br />

•<br />

NIPA<br />

•<br />

NPM<br />

•<br />

Phospho-NPM (Ser4) •<br />

Phospho-NPM (Thr95)<br />

•<br />

Phospho-NPM (Thr199) •<br />

NuMA<br />

• •<br />

Phospho-NuMA (Ser395) •<br />

ORC1<br />

•<br />

ORC2<br />

•<br />

ORC6<br />

•<br />

p14 ARF<br />

•<br />

p21 Waf1/Cip1 • • • •<br />

p27 Kip1<br />

• • • • •<br />

p48 Primase<br />

•<br />

p53<br />

• • • • •<br />

Acetyl-p53<br />

•<br />

Acetyl-p53 (Lys379)<br />

•<br />

Acetyl-p53 (Lys382)<br />

•<br />

Phospho-p53 (Ser6)<br />

•<br />

Phospho-p53 (Ser9)<br />

•<br />

Phospho-p53 (Ser15) • • • •<br />

Phospho-p53 (Thr18)<br />

•<br />

Phospho-p53 (Ser20)<br />

•<br />

Phospho-p53 (Ser33)<br />

•<br />

Phospho-p53 (Ser37)<br />

•<br />

Phospho-p53 (Ser46)<br />

•<br />

Phospho-p53 (Thr81)<br />

•<br />

Phospho-p53 (Ser315) •<br />

Phospho-p53 (Ser392) •<br />

53BP1<br />

•<br />

Phospho-53BP1 (Ser25/29) •<br />

Phospho-53BP1 (Thr543) •<br />

Phospho-53BP1 (Ser1618) •<br />

Phospho-53BP1 (Ser1778) •<br />

p57 Kip2<br />

•<br />

Phospho-p57 Kip2 (Thr310) •<br />

p58 Primase<br />

•<br />

p63-α<br />

• •<br />

Phospho-p63 (Ser160/162) •<br />

p73<br />

•<br />

Phospho-p73 (Tyr99)<br />

•<br />

p95/NBS1<br />

•<br />

Phospho-p95/NBS1 (Ser343) •<br />

PBK/TOPK<br />

•<br />

Phospho-PBK/TOPK (Thr9) •<br />

PCNA • •<br />

Ubiquityl-PCNA (Lys164) •<br />

PCTAIRE 1<br />

•<br />

PELO<br />

•<br />

PHB1<br />

•<br />

PICH<br />

•<br />

Pin1<br />

•<br />

PLK1 • • •<br />

Phospho-PLK (Ser137) •<br />

Phospho-PLK (Thr210) • •<br />

PLK3<br />

• •<br />

Phospho-PNK1 (Ser114/Thr118)) •<br />

chapter 03: Cell Growth and Death<br />

152<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for phospho-p53<br />

(Ser15) have been cited over 152 times<br />

in high-impact, peer-reviewed publications<br />

from the global research community.<br />

Select Citations:<br />

Godinho, S.A. et al. (2014)<br />

Oncogene-like induction of cellular<br />

invasion from centrosome amplification.<br />

Nature 510, 167–171.<br />

Lee, T.H. et al. (2014) Coordinated<br />

regulation of XPA stability by ATR and<br />

HERC2 during nucleotide excision<br />

repair. Oncogene 33, 19–25.<br />

Gad, H. et al. (2014) MTH1 inhibition<br />

eradicates cancer by preventing<br />

sanitation of the dNTP pool. Nature<br />

508, 215–221.<br />

Rashi-Elkeles, S. et al. (2014) Parallel<br />

profiling of the transcriptome, cistrome,<br />

and epigenome in the cellular<br />

response to ionizing radiation. Sci.<br />

Signal. 7, rs3.<br />

Baresova, P. et al. (2014) p53 tumor<br />

suppressor protein stability and<br />

transcriptional activity are targeted<br />

by Kaposi’s sarcoma-associated<br />

herpesvirus-encoded viral interferon<br />

regulatory factor 3. Mol. Cell. Biol.<br />

34, 386–399.<br />

Cam, M. et al. (2014) p53/TAp63<br />

and AKT regulate mammalian target<br />

of rapamycin complex 1 (mTORC1)<br />

signaling through two independent<br />

parallel pathways in the presence of<br />

DNA damage. J. Biol. Chem. 289,<br />

4083–4094.<br />

Zheng, H. et al. (2014) p53 promotes<br />

repair of heterochromatin DNA by<br />

regulating JMJD2b and SUV39H1<br />

expression. Oncogene 33, 734–744.<br />

Shi, Y. et al. (2014) ROS-dependent<br />

activation of JNK converts p53 into an<br />

efficient inhibitor of oncogenes leading<br />

to robust apoptosis. Cell Death<br />

Differ. 21, 612–623.<br />

Penicud, K. et al. (2014) DMAP1 is an<br />

essential regulator of ATM activity and<br />

function. Oncogene 33, 525–531.<br />

Endo, F. et al. (2014) A compensatory<br />

role of NF-kappaB to p53 in response<br />

to 5-FU-based chemotherapy for<br />

gastric cancer cell lines. PLoS One<br />

9, e90155.<br />

Leikam, C. et al. (2014) Cystathionase<br />

mediates senescence evasion in<br />

melanocytes and melanoma cells.<br />

Oncogene 33, 771–782.<br />

www.cellsignal.com/cstcellcycle 101


Section I: Research Areas<br />

UV treatment results<br />

in ATM/ATR-mediated<br />

phosphorylation of<br />

Chk1, initiating the DNA<br />

damage checkpoint.<br />

A<br />

B<br />

C<br />

Phospho-Chk1 (Ser317) (D12H3) XP ®<br />

Rabbit mAb #12302: Confocal IF analysis<br />

of HeLa cells, untreated (A), UV-treated<br />

(B), or UV and λ phosphatase-treated (C),<br />

using #12302 (green). Actin filaments were<br />

labeled with DY-554 phalloidin (red).<br />

Target M P E S C<br />

PP1α<br />

•<br />

Phospho-PP1α (Thr320) •<br />

PP2A A subunit • • •<br />

PP2A B subunit<br />

• •<br />

PPP2R2A<br />

•<br />

PP2A C subunit<br />

• •<br />

Nonmethylated PP2A C Subunit •<br />

PPP1CB<br />

•<br />

PTPA/PPP2R4<br />

• •<br />

PPP2R5D<br />

•<br />

PP5<br />

•<br />

Rad17<br />

•<br />

Phospho-Rad17 (Ser635) •<br />

Phospho-Rad17(Ser645) •<br />

Rad18<br />

•<br />

Phospho-Rad18 (Ser403) •<br />

RAD21<br />

•<br />

Rad50<br />

•<br />

Rad51<br />

•<br />

Rb • • •<br />

Phospho-Rb (Ser608) • •<br />

Phospho-Rb (Ser780) • • •<br />

Phospho-Rb (Ser795)<br />

•<br />

Phospho-Rb (Ser807/Ser811) • • •<br />

Rb-like 1<br />

•<br />

Phospho-RCC1 (Ser11) •<br />

RecQL1<br />

•<br />

RecQ4<br />

•<br />

RecQL5<br />

•<br />

RPA70/RPA1<br />

• •<br />

RPA32/RPA2<br />

•<br />

Rpb1 CTD<br />

•<br />

Phospho-Rpb1 CTD (Ser2/5) •<br />

RRM1<br />

• •<br />

Securin<br />

•<br />

Sestrin-2<br />

•<br />

SLBP<br />

•<br />

Cell Cycle/Checkpoint Control Kinase-Disease Associations<br />

Name Group Disease Type Molecular Notes<br />

ATM Atypical Cancer<br />

CNS<br />

LOF Mut LOF mutations associated with ataxia talangiectasia [OMIM:208900], causing<br />

progressive loss of motor control (ataxia), dilation of superficial blood vessels<br />

(telangiectasia), cancer and immune deficiency. Approximately 30% of<br />

cases develop tumors, mostly lymphomas and leukemias, due to defects in<br />

DNA damage repair. Somatic mutations seen in leukemias and lymphomas.<br />

OMIM:607585.<br />

ATR Atypical Cancer<br />

Development<br />

Virology<br />

Mut, Splice<br />

Functions in DNA damage responses. A splice-altering mutation seen in<br />

cases of Seckel syndrome [OMIM:210600], featuring dwarfism and mental<br />

retardation. Heterozygous knockout mice are cancer-prone. Mutations seen<br />

in cancers of the stomach (Medline:11691784) and endometrium (Medline:12124347),<br />

tumors with high mutation rates due to microsatellite instability.<br />

May also be required for retroviral DNA integration (Medline:12679521).<br />

OMIM:601215.<br />

AurB Other Cancer OE Required for chromosome segregation and cytokinesis. Overexpressed in<br />

colorectal and other cancer cell lines (Medline:9809983) and thought to cause<br />

aneuploidy via histone phosphorylation (Medline:12884918). OMIM:604970.<br />

CDC2<br />

(CDK1)<br />

Target M P E S C<br />

SMC1<br />

• •<br />

Phospho-SMC1 (Ser360) •<br />

Phospho-SMC1 (Ser957) • •<br />

SMC2<br />

•<br />

SMC3<br />

•<br />

SMC4<br />

•<br />

SMG-1<br />

• •<br />

Spartin<br />

•<br />

STAG2<br />

• •<br />

TACC3<br />

•<br />

Phospho-TACC3 (Ser558) • •<br />

TCTP<br />

•<br />

TERF2IP<br />

•<br />

TIF1β<br />

• •<br />

Phospho-TIF1β (Ser824) •<br />

TLK1<br />

•<br />

Phospho-TLK1 (Ser743) •<br />

TPX2<br />

• •<br />

Trf1<br />

•<br />

Trf2<br />

• •<br />

TTK<br />

• •<br />

VCP<br />

•<br />

VRK1<br />

•<br />

VRK3<br />

•<br />

Wee1<br />

• •<br />

Phospho-Wee1 (Ser642) •<br />

WIP1<br />

•<br />

WRN<br />

•<br />

XLF<br />

•<br />

XPB<br />

•<br />

XPC<br />

•<br />

XPD<br />

•<br />

XPF<br />

•<br />

XRCC1<br />

•<br />

CMGC Cancer Act, Splice Cell cycle checkpoint. Activated in many cancers including colon, liver and<br />

breast (Medline:10091728, 12100577, 11091571). The delta T isoform,<br />

which lacks a regulatory region, is expressed in breast cancer. Inhibition in<br />

cancer cells may drive cells into apoptosis (Medline:12150824). May also<br />

drive cell migration (Medline:12771130). Inhibitors: BMS-265246, BMS-<br />

265246-01 (Bristol-Myers Squibb). OMIM:116940.<br />

102 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

Name Group Disease Type Molecular Notes<br />

CDK2 CMGC Cancer Cell cycle checkpoint and part of the Rb pathway disregulated in most tumors<br />

(Medline:12888290). Target of several candidate cancer drugs. However,<br />

inhibition does not always prevent cancer cell growth (Medline:12676582)<br />

due possibly to CDK redundancy. Inhibitors: BMS-265246, BMS-265246-01<br />

(Bristol-Myers Squibb), R-roscovitine (CYC200, CYC202) (Cyclacel; Ph. 2),<br />

SU9516 (Sugen), R547 (Roche), L868276. OMIM:116953.<br />

CDK4 CMGC Cancer Act, GOF<br />

Mut, Amp,<br />

Meth<br />

Point mutations found in somatic and familial melanoma. Amplified in<br />

sarcomas (Medline:9703873, 9935200), glioma (Medline:14756442) and<br />

lymphoma (Medline:12203778). Amplified, methylated or deleted in head<br />

and neck squamous cell carcinoma (Medline:14586645). Overexpression<br />

drives epithelial tumors in mice (Medline:14647432). Disruption makes<br />

mice resistant to cancer (Medline:12435633). Inhibitor: PD332991 (Onyx).<br />

OMIM:123829.<br />

CDK5 CMGC Neurodegeneration SNP Implicated in the pathology of neurofibrillary tangles and formation of senile<br />

plaques, hallmarks of Alzheimer disease (AD). Induces tau phosphorylation<br />

and aggregation and neurofibrillary tangle deposition and neurodegeneration<br />

in in vitro and in vivo animal models. Brain samples from AD patients show<br />

elevated CDK5 activity, possibly induced by Aβ amyloid. SNPs show weak AD<br />

association (Medline:18350355) OMIM:123831.<br />

CDK6 CMGC Cancer OE, Trans Overexpressed and/or disrupted by translocation in leukemias, lymphomas<br />

and other cancers; amplified in gliomas (Medline:9102208) and rodent<br />

cancers (Medline:12538879, 11719459). OMIM:603368.<br />

CDK9 CMGC Cardiovascular<br />

Viral infection<br />

CDKL5 CMGC CNS<br />

Development<br />

CHK1<br />

(CHEK1)<br />

Expr<br />

Mut, Trans<br />

Transcriptional elongation factor and cofactor for HIV Tat protein; RNAi against<br />

CDK9 blocks HIV replication and inhibitors also block varicella zoster replication.<br />

Mediates signals leading to cardiac hypertrophy (Medline:12695656).<br />

Inhibitor: Flavopiridol. (Aventis) OMIM:603251.<br />

Missense, splice and truncating mutations linked to the neurodevelopmental<br />

Rett Syndrome [OMIM:312750]. A chromosomal translocation which silences<br />

the gene is associated with severe X-linked infantile spasm syndrome<br />

[OMIM:308350]. OMIM:300203.<br />

CAMK Cancer Mut Cell cycle G2 checkpoint kinase implicated in resistance to apoptosis in<br />

response to chemotherapy. Inhibitors under development to chemosensitize<br />

tumors. Somatic mutations found in stomach tumors (Medline:11691784)<br />

and in colon and endometrial tumors, where CHK1 may be a target of<br />

microsatellite instability (Medline:14657665). Inhibitors: SB218078, UNC-01.<br />

OMIM:603078.<br />

CHK2 CAMK Cancer Mut Tumor suppressor involved in DNA damage and cell cycle arrest. LOF mutants<br />

cause Li-Fraumeni syndrome [OMIM:151623], a highly penetrant familial<br />

cancer phenotype also caused by p53 mutations. Familial mutations also associated<br />

with prostate and breast cancer, and mutations also seen in a variety<br />

of sporadic cancers and cell lines. OMIM:604373.<br />

CK1δ<br />

CK1α<br />

CK1ε<br />

(CSNK1ε)<br />

CK1 Neurodegeneration Several CK1 isoforms including CK1δ and CK1α associate with and<br />

phosphorylate tau, which forms the neurofibrillary tangles of Alzheimer’s<br />

disease. Tau phosphorylation and microtubule association is inhibited by<br />

the CK1 inhibitor IC261 (Medline:14761950). CK1 phosphorylation is<br />

involved in trafficking of the Alzheimer’s plaque component, β secretase<br />

(Medline:11278841). CK1 levels are increased in Alzheimer’s brains. CK1δ is<br />

also associated with tau tangles in several other neurodegenerative diseases<br />

(Medline:10924763). CK1δ mRNA and protein are upregulated in Alzheimer’s<br />

brain regions with most pathology (Medline:10814741). CK1 also phosphorylates<br />

the Parkinson’s-associated α synuclein protein (Medline:10617630).<br />

OMIM:600864, 60050.<br />

CK1<br />

CK2α1 Other<br />

CK2α2<br />

(CSNK2α1/2)<br />

Behavior<br />

Cancer<br />

Cancer<br />

Circadian Rhythm<br />

Neurodegeneration<br />

SNP, Mut,<br />

LOH<br />

OE, Act<br />

Mutations in hamster and Drosophila orthologs have circadian rhythm phenotypes<br />

as the circadian gene period (per) is a substrate in both human and fly.<br />

A coding SNP variant in human increases CK1e activity and is negatively associated<br />

with circadian disorder (Medline:15187983). LOF mutations and LOH<br />

seen in mammary ductal carcinoma (Medline:14871824). OMIM:600863.<br />

Two gene products (α1/α>2 or α/α) complex with each other and a regulatory<br />

β subunit. The isoforms are rarely distinguished from each other in<br />

publications. Transgene expression in mice leads to lymphoma and activation<br />

by bovine parasites leads to fatal lymphoproliferation (Medline: 7846532).<br />

Expression and activity are elevated in lung tumors (Medline:15355908,<br />

12017291) and breast tumors (Medline:11423974). Mouse transgene<br />

causes mammary gland hyperplasia. Antisense drives apoptosis of tumor<br />

cell lines (Medline:11827168) and xenografts (Medline:14965269).<br />

Involved in DNA break repair by phosphorylation of scaffold protein XRCC1<br />

(Medline:15066279), phosphorylation of BRCA1 (Medline:10403822) and<br />

phosphorylation of p53 in response to UV irradiation. The Drosophila CK2<br />

ortholog (Timekeeper) is involved in circadian regulation. Phosphorylates and<br />

binds to a major component of the inclusion bodies seen in Parkinson patients<br />

(Medline:14645218). Inhibitors: DMAT, Antisense, P15 peptide, 4,5,6,7-tetrabromobenzotriazole<br />

(TBB). OMIM:115440, 115442.<br />

Molecular: Act Activated • Amp Amplified • Del Deleted • Expr Expression • GOF Gain-of-function • Inh Inhibitor Studies • LOF Loss-offunction<br />

• LOH Loss-of-heterozygosity • Meth Methylation • Model model organism studies • Mut Mutation OE Overexpression • SNP Single<br />

Nucleotide Polymorphism • Splice Splicing change • Trans Translocation<br />

chapter 03: Cell Growth and Death<br />

Cancer<br />

Research<br />

Please visit our website to learn<br />

more about the scientific tools and<br />

educational resources we have online<br />

for cancer signaling and proteomic<br />

analysis, including discussion of key<br />

disease drivers.<br />

www.cellsignal.com/cancerguide<br />

www.cellsignal.com/csttables 103


Section I: Research Areas<br />

Cell Cycle Control: G1/S Checkpoint<br />

Ultra Violet<br />

Stress Response<br />

Growth<br />

Factor<br />

Withdrawal<br />

Ubiquitination<br />

Hormones<br />

p19 INK4D<br />

GSK-3β<br />

SCF<br />

Differentiation<br />

p18 INK4C<br />

Myc<br />

G1-PHASE<br />

Replicative<br />

Senescence<br />

p16 INK4A<br />

Cyclin D<br />

BMI1<br />

CDK4/6<br />

TGF-β<br />

p15 INK4B<br />

Myc<br />

Smad3<br />

Smad4<br />

cdc25A<br />

Growth Factor<br />

Receptor Activation<br />

R<br />

Akt<br />

FoxO1/3<br />

p27 Kip1<br />

CDK2<br />

Cyclin E<br />

Myc<br />

S-PHASE<br />

p21 Cip1<br />

Myc<br />

p53<br />

ATM/<br />

ATR<br />

Replicative<br />

Senescence<br />

SCF<br />

DNA<br />

Damage<br />

Chk1/2<br />

Ubiquitination<br />

Ubiquitination<br />

Cell Cycle Control: G2/M DNA Damage Checkpoint<br />

HIPK2<br />

Nuclear Export,<br />

Ubiquitination<br />

Nucleolar<br />

Sequestration<br />

or<br />

p53 Stabilization<br />

p19 Arf cdc25A<br />

IR UV<br />

DNA Repair<br />

DNA-PK<br />

ATM/<br />

ATR<br />

Caffeine<br />

WIP1<br />

MDM4<br />

MDM2 p53<br />

Chk2<br />

TopoII BRCA1 14-3-3σ Reprimo GADD45 p21 Cip1 CDK7<br />

Nuclear<br />

Exclusion<br />

TRIP12<br />

MDM2<br />

p53<br />

p300/<br />

PCAF<br />

WIP1<br />

BRCA1<br />

Chk1<br />

cdc25<br />

chapter 03: Cell Growth and Death<br />

Critically Short<br />

Telomeres<br />

POT1 TRF2<br />

PLK1<br />

Myt1<br />

Rad50<br />

NBS1<br />

Mre11<br />

BRCA1<br />

c-Abl<br />

14-3-3<br />

cdc25A/C<br />

cdc25A/B<br />

(MRN)<br />

SCF<br />

SCF<br />

FANCD2<br />

Rad51<br />

p90RSK<br />

DNA<br />

Repair<br />

Rad52<br />

Nuclear Exclusion<br />

Ubiquitination<br />

AurA<br />

Bora<br />

FoxO1<br />

DBE<br />

Bim<br />

FasL<br />

TRAIL<br />

Apoptosis<br />

Suv39H1<br />

Abl<br />

Rb HDAC<br />

E2F<br />

DP-1<br />

OFF<br />

Rb<br />

E2F<br />

DP-1<br />

ON<br />

E2F/DP Target Genes:<br />

Cyclin E/A, E2F-1/2/3,<br />

cdc2, c-Myc, p107,<br />

RanGAP, TK, DHFR,<br />

PCNA, H2A, etc.<br />

G2-PHASE<br />

cdc2<br />

Cyclin B<br />

Wee1<br />

M-PHASE<br />

Ubiquitination<br />

The primary G1/S cell cycle checkpoint controls the commitment of eukaryotic cells to transition through the G1 phase to enter into the DNA synthesis S phase. Two cell<br />

cycle kinase complexes, CDK4/6-Cyclin D and CDK2-Cyclin E, work in concert to relieve inhibition of a dynamic transcription complex that contains the retinoblastoma protein<br />

(Rb) and E2F. In G1-phase uncommitted cells, hypo-phosphorylated Rb binds to the E2F-DP1 transcription factors forming an inhibitory complex with HDAC to repress key<br />

downstream transcription events. Commitment to enter S-phase occurs through sequential phosphorylation of Rb by Cyclin D-CDK4/6 and Cyclin E-CDK2 that dissociates<br />

the HDAC-repressor complex, permitting transcription of genes required for DNA replication. In the presence of growth factors, Akt can phosphorylate FoxO1/3, which inhibits<br />

their function by nuclear export, thereby allowing cell survival and proliferation. Importantly, a multitude of different stimuli exert checkpoint control, including TGF-β, DNA<br />

damage, replicative senescence, and growth factor withdrawal. These stimuli act though transcription factors to induce specific members of the INK4 or Kip/Cip families of<br />

cyclin dependent kinase inhibitors (CKIs). Notably, the oncogenic polycomb protein Bmi1 acts as a negative regulator of INK4A/B expression in stem cells and human cancer.<br />

In addition to regulating CKIs, TGF-β also inhibits cdc25A transcription, a phosphatase directly required for CDK activation. At a critical convergence point with the DNAdamage<br />

checkpoint, cdc25A is ubiquitinated and targeted for degradation via the SCF ubiquitin ligase complex downstream of the ATM/ATR/Chk-pathway. However, timely<br />

degradation of cdc25A in mitosis (M-phase) via the APC ubiquitin ligase complex allows progression through mitosis. Furthermore, growth factor withdrawal activates GSK-3β<br />

to phosphorylate Cyclin D, which leads to its rapid ubiquitination and proteasomal degradation. Collectively, ubiquitin/proteasome-dependent degradation and nuclear export<br />

are mechanisms commonly used to effectively reduce the concentration of cell cycle control proteins. Importantly, Cyclin D1/CKD4/6 complexes are explored as therapeutic<br />

targets for cancer treatment as researchers have found this checkpoint to be invariantly deregulated in human tumors.<br />

Select Reviews:<br />

Besson, A., Dowdy, S.F., and Roberts, J.M. (2008) Dev. Cell. 14, 159–169. • Gil, J. and Peters, G. (2006) Nat. Rev. Mol. Cell Biol. 7, 667–677. • Malumbres, M. and<br />

Barbacid, M. (2009) Nat. Rev. Cancer 9, 153–166. • Musgrove, E.A., Caldon, C.E., Barraclough, J., Stone, A., and Sutherland, R.L. (2011) Nat. Rev. Cancer 11, 558–572.<br />

• Skaar, J.R. and Pagano, M. (2009) Curr. Opin. Cell Biol. 21, 816–824. • Sparmann, A. and van Lohuizen, M. (2006) Nat. Rev. Cancer 6, 846–856. • Tzivion, G.,<br />

Dobson, M., and Ramakrishnan, G. (2011) Biochim. Biophys. Acta. 1813, 1938–1945. • van den Heuvel, S. and Dyson, N.J. (2008) Nat. Rev. Mol. Cell Biol. 9, 713–724.<br />

• Yang, J.Y. and Hung, M.C. (2009) Clin. Cancer Res. 15, 752–757.<br />

The G2/M DNA damage checkpoint serves to prevent the cell from entering mitosis (M-phase) with genomic DNA damage. Specifically, the activity of the Cyclin B-cdc2 (CDK1)<br />

complex is pivotal in regulating the G2-phase transition wherein cdc2 is maintained in an inactive state by the tyrosine kinases Wee1 and Myt1. It is thought that coordinated<br />

action of the kinase Aurora A and the cofactor Bora activate PLK1 as cells approach the M-phase, which in turn activates the phosphatase cdc25 and downstream cdc2 activity,<br />

hence establishing a feedback amplification loop that efficiently drives the cell into mitosis. Importantly, DNA damage cues activate the sensory DNA-PK/ATM/ATR kinases, which<br />

relay two parallel cascades that ultimately serve to inactivate the Cyclin B-cdc2 complex. The first cascade rapidly inhibits progression into mitosis: the Chk kinases phosphorylate<br />

and inactivate cdc25, which prevents activation of cdc2. The slower second parallel cascade involves phosphorylation of p53 and allows for its dissociation from MDM2 and<br />

MDM4 (MdmX), which activates DNA binding and transcriptional regulatory activity, respectively. The transcriptional ability of p53 is further augmented through acetylation by the<br />

co-activator complex p300/PCAF. The second cascade constitutes the p53 downstream-regulated genes including: 14-3-3, which binds to the phosphorylated Cyclin B-cdc2<br />

complex and exports it from the nucleus; GADD45, which binds to and dissociates the Cyclin B-cdc2 complex; and p21 Cip1, an inhibitor of a subset of the cyclin-dependent<br />

kinases including cdc2. Recent data suggest an important role for the p53-regulated WIP1 phosphatase that acts as a critical dampener of DNA damage signaling in cancer.<br />

In human cancer, researchers have found p53 to be commonly mutated, indicating that this checkpoint is a critical barrier to tumor formation. In addition, sporadic and familial<br />

mutations in the DNA-repair proteins such as the BRCA-family, ATM, and the Fanconi Anemia proteins further highlight this as a key tumor suppressor checkpoint.<br />

Select Reviews:<br />

Abbas, T. and Dutta, A. (2009) Nat. Rev. Cancer 9, 400–414. • Al-Ejeh, F., Kumar, R., Wiegmans, A., Lakhani, S.R., Brown, M.P., and Khanna, K.K. (2010) Oncogene 29,<br />

6085–6098. • Boutros, R., Lobjois, V., and Ducommun, B. (2007) Nat. Rev. Cancer 7, 495–507. • Ciccia, A. and Elledge, S.J. (2010) Mol. Cell 40, 179–204. • Freed-<br />

Pastor, W.A. and Prives, C. (2012) Genes Dev. 26, 1268–1286. • Huen, M.S., Sy, S.M., and Chen, J. (2010) Nat. Rev. Mol. Cell Biol. 11, 138–148. • Junttila, M.R. and<br />

Evan, G.I. (2009) Nat. Rev. Cancer 9, 821–829. • Lens, S.M., Voest, E.E., and Medema, R.H. (2010) Nat. Rev. Cancer 10, 825–841. • Kee, Y. and D’Andrea, A.D. (2010)<br />

Genes Dev. 24,1680–1694. • Nam, E.A. and Cortez, D. (2011) Biochem. J. 436, 527–536. • Reinhardt, H.C. and Yaffe, M.B. (2009) Curr. Opin. Cell Biol. 21, 245–255.<br />

© 2002–2015 Cell Signaling Technology, Inc. • We would like to thank Dr. Hans Widlund, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

104 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

© 2002–2015 Cell Signaling Technology, Inc. • We would like to thank Dr. Hans Widlund, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

www.cellsignal.com/cstpathways 105


Section I: Research Areas<br />

VE-cadherin is<br />

expressed in<br />

adherens junctions<br />

of endothelial cells.<br />

VE-Cadherin (D87F2) XP ® Rabbit mAb<br />

#2500: Confocal IF analysis of HUVE cells<br />

(top) and HeLa cells (bottom) using #2500.<br />

Actin filaments were labeled with DY-554<br />

phalloidin (red). Blue pseudocolor =<br />

DRAQ5 ® #4084 (fluorescent DNA dye).<br />

Adhesion and Extracellular Matrix<br />

Cells can form a number of connections with the cells and matrix in their surrounding environment,<br />

including adherens junctions (cell-cell), tight junctions (impermeable cell-cell), and focal adhesions<br />

(cell-matrix).<br />

Adherens Junctions<br />

Adherens junctions are cell-cell connections mediated through a cadherin-catenin complex composed<br />

of cadherin, β-catenin, and α-catenin. The connection between cell junctions and the cytoskeleton is<br />

more dynamic than originally considered, relying on multiple, weak associations between the cadherincatenin<br />

complex and the actin cytoskeleton or on other membrane-associated proteins (i.e. nectin and<br />

afadin). Monomeric α-catenin binds β-catenin at adherens junctions, and upon release, forms α-catenin<br />

dimers that promote actin bundle formation. The transition from branched actin networks to bundled<br />

actin filaments correlates with the creation of mature, strong adherens junctions, and a decrease in<br />

membrane lamellipodia. As with most dynamic cellular systems, a collection of kinases, phosphatases,<br />

and adaptor proteins regulate the activity and localization of a few key effector proteins. p120 catenin<br />

(δ-catenin) binds and stabilizes cadherin at the plasma membrane. Membrane-bound and cytosolic<br />

tyrosine kinases phosphorylate β-catenin at weak or nascent junctions, while phosphatases remove<br />

added phosphates from β-catenin and δ-catenin at established junctions. Rho family GTPases modulate<br />

the availability and activation state of catenins and other essential adherens proteins. Together, this<br />

collection of structural proteins, enzymes, and adaptor proteins creates dynamic cell-cell junctions<br />

necessary for temporary associations during morphogenesis and maintains the integrity of complex<br />

tissues and structures following development.<br />

E-cadherin, an important component of adherens<br />

junctions, localizes to the plasma membrane.<br />

E-Cadherin (24E10) Rabbit mAb<br />

#3195: Confocal IF analysis of MCF7<br />

cells using #3195 (green, left) compared<br />

to an isotype control (right). Blue pseudocolor<br />

= DRAQ5 ® (fluorescent DNA dye).<br />

04 Cell Biology Tight Junctions<br />

Tight junctions are impermeable cell-cell junctions that form a continuous barrier to fluids across the<br />

epithelium and endothelium. They function in regulation of paracellular permeability and in the maintenance<br />

of cell polarity, blocking the movement of transmembrane proteins between the apical and the<br />

basolateral cell surfaces. The primary protein families composing tight junctions are claudin, occludins,<br />

and junctional adhesion molecules (JAMs) transmembrane proteins, which join the junctions to the<br />

cytoskeleton. Occludin is thought to be important in the assembly and maintenance of tight junctions.<br />

Differential phosphorylation of occludin at various residues may regulate its interaction with other tight<br />

junction proteins such as ZO-1.<br />

Differential expression of Claudin-1, a<br />

component of tight junctions, in IGROV-1 (high),<br />

A549 (moderate), and 293 (absent) cells<br />

Claudin-1 (D3H7C) Rabbit mAb #13995: WB analysis of extracts from IGROV-1, A549,<br />

and 293 cells using #13995 (upper) and β-Actin (D6A8) Rabbit mAb #8457 (lower).<br />

kDa<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

50<br />

40<br />

1 2 3<br />

Lanes<br />

1. IGROV-1<br />

2. A549<br />

3. 293<br />

Claudin-1<br />

β-Actin<br />

Focal Adhesions<br />

Focal adhesions are the connections that form between a cell and the extracellular matrix (ECM) and are<br />

mediated primarily through integrins. Integrins are cell surface receptors that play a pivotal role in cell<br />

adhesion, migration, invasion, growth, and survival. Integrins are α/β heterodimeric proteins composed<br />

of one α and one β subunit. The integrin family contains at least 18 α and 8 β subunits that form 24<br />

known integrin pairs with distinct tissue distribution and overlapping ligand specificities. The intracellular<br />

tail of integrins interacts with cytoskeletal proteins vinculin, talin, and α-actinin, as well as numerous<br />

signaling molecules such as focal adhesion kinase (FAK). Activation of FAK by integrin clustering leads<br />

to autophosphorylation at Tyr397, which is a binding site for the Src family kinases PI3K and PLCγ.<br />

α/β Integrin Pairs<br />

αIIb<br />

chapter 04: Cell Biology<br />

Altered expression<br />

of Claudin-1 can be<br />

found in many types<br />

of cancer.<br />

Claudin-1 (D5H1D) XP ® Rabbit mAb<br />

#13255: IHC analysis of paraffin-embedded<br />

human colon carcinoma (top) and human<br />

lung carcinoma (bottom) using #13255.<br />

α1<br />

α11<br />

α10<br />

β3<br />

αL<br />

α2<br />

α5<br />

β5<br />

α3<br />

β1<br />

αV<br />

αX β2 αD<br />

N-cadherin, a central component of adherens<br />

junctions, is up-regulated in many cancers.<br />

N-Cadherin (D4R1H) XP ® Rabbit mAb #13116: IHC analysis<br />

of paraffin-embedded human ovarian carcinoma using #13116.<br />

αE<br />

β7<br />

α4<br />

α9<br />

α6<br />

β4<br />

α8<br />

α7 β8<br />

β6<br />

αM<br />

P-cadherin, an adherens junction component<br />

expressed in epithelial cells and some cancers,<br />

localizes to the plasma membrane.<br />

P-Cadherin (C13F9) Rabbit mAb #2189: Confocal IF analysis of A-431 cells using<br />

#2189 (green). Actin filaments were labeled with Alexa Fluor ® 555 Phalloidin #8953<br />

(red). Blue pseudocolor = DRAQ5 ® (fluorescent DNA dye).<br />

On SDS-PAGE, Integrin α4 can migrate as a 150 kDa<br />

mature protein, a 140 kDa precursor protein, or as<br />

an 80 kDa or 70 kDa cleavage fragment.<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

1 2 3<br />

Full length<br />

Integrin α4<br />

Cleaved<br />

C-terminal<br />

Integrin α4<br />

Integrin α4 (D2E1) XP ® Rabbit<br />

mAb #8440: WB analysis of extracts<br />

from various cell lines using #8440.<br />

Lanes<br />

1. Jurkat<br />

2. MOLT-4<br />

3. C6<br />

106 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstadhesion 107


Section I: Research Areas<br />

Example of Integrin β3 expression in cancer<br />

Integrin β3 (D7X3P) XP ® Rabbit mAb #13166: IHC analysis of<br />

paraffin-embedded human papillary renal cell carcinoma using #13166.<br />

Vinculin is expressed in human breast ductal carcinoma in situ.<br />

Vinculin (E1E9V) XP ® Rabbit mAb #13901: IHC analysis of paraffinembedded<br />

human breast ductal carcinoma in situ using #13901.<br />

Epithelial-Mesenchymal Transition (EMT)<br />

EMT is an essential process during development whereby epithelial cells acquire mesenchymal, fibroblast-like<br />

properties and display reduced intracellular adhesion and increased motility. This is a critical<br />

feature of normal embryonic development (type I) and wound healing (type II), but it is also utilized by<br />

malignant epithelial tumors to spread beyond their origin (type III). This tightly regulated process is associated<br />

with a number of cellular and molecular events. EMT depends on a reduction in expression of<br />

several cell adhesion molecules. For example, E-cadherin is a critical component of adherens junctions<br />

and is considered an active suppressor of invasion and growth for many epithelial cancers. Research<br />

studies have shown that cancer cells typically downregulate expression of E-cadherin and upregulate<br />

expression of N-cadherin. This is referred to as the cadherin switch and is one of the hallmarks of EMT.<br />

Downregulation of E-cadherin expression occurs by binding of transcriptional repressor proteins such<br />

as Slug, Snail, and ZEB to the E-cadherin promoter region.<br />

Type III EMT: Cancer Invasion<br />

Invading Cancer Cells<br />

108 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

EMT<br />

Proliferating<br />

Tumor Cells<br />

Endothelium<br />

Extracellular<br />

Matrix<br />

Select Reviews<br />

Ciobanasu, C., Faivre, B., and Le Clainche, C. (2013) Eur. J. Cell Biol. 92, 339−348. • Dejana, E. and Orsenigo, F. (2013)<br />

J. Cell Sci. 126, 2545−2549. • Giannotta, M., Trani, M., and Dejana, E. (2013) Dev. Cell 26, 441−454. • Ivanov, A.I. and<br />

Naydenov, N.G. (2013) Int. Rev. Cell Mol. Biol. 303, 27−99. • Lamouille, S., Xu, J., and Derynck, R. (2014) Nat. Rev. Mol.<br />

Cell Biol. 15, 178−196. • Lampugnani, M.G. (2012) Cold Spring Harb. Perspect. Med. 2, a006528. • Runkle, E.A. and Mu,<br />

D. (2013) Cancer Lett. 337, 41−48. • Takeichi, M. (2014) Nat. Rev. Mol. Cell Biol. 15, 397−410. • Van Itallie, C.M. and<br />

Anderson, J.M. (2013) Tissue Barriers 1, e25247. • Zheng, H. and Kang, Y. (2014) Oncogene 33, 1755−1763.<br />

Commonly Studied Adhesion Targets<br />

Target M P<br />

ADAMTS1 •<br />

ADAM9<br />

• •<br />

ADAM10<br />

•<br />

Afadin<br />

• •<br />

Phospho-Afadin (Ser1718) •<br />

Ajuba<br />

•<br />

BSP II<br />

•<br />

Pan-Cadherin • •<br />

E-Cadherin •<br />

N-Cadherin • •<br />

OB-Cadherin • •<br />

P-Cadherin • •<br />

VE-Cadherin • •<br />

α-E-Catenin • •<br />

Phospho-α-E-Catenin •<br />

(Ser652)<br />

Phospho-α-E-Catenin •<br />

(Ser655/Thr658)<br />

α-N-Catenin • •<br />

γ-Catenin<br />

•<br />

Phospho-Catenin δ-1 •<br />

(Tyr228)<br />

Phospho-Catenin δ-1 •<br />

(Ser252)<br />

Phospho-Catenin δ-1 •<br />

(Ser320)<br />

Phospho-Catenin δ-1 •<br />

(Tyr904)<br />

Catenin δ-1<br />

•<br />

CD54/ICAM-1 •<br />

Target M P<br />

CD102/ICAM-2<br />

CEA/CD66e<br />

Claudin-1<br />

Connexin 43<br />

Phospho-Connexin 43<br />

(Ser368)<br />

CYR61<br />

Phospho-Desmoplakin<br />

(Ser165/166)<br />

EpCAM<br />

FAK<br />

Phospho-FAK (Tyr397)<br />

Phospho-FAK (Tyr576/577)<br />

GIT-1<br />

GIT2<br />

Phospho-GIT2 (Tyr392)<br />

Phospho-GIT2 (Tyr592)<br />

Hic-5<br />

ILK1<br />

βIG-H3<br />

Integrin α2b<br />

Integrin α4<br />

Integrin α5<br />

Integrin aV<br />

Integrin a6<br />

Integrin β1<br />

Integrin β3<br />

Integrin β4<br />

Integrin β5<br />

Select Citations:<br />

White, A.C. et al. (2014) Stem cell quiescence acts as a<br />

tumour suppressor in squamous tumours. Nat. Cell Biol.<br />

16, 99−107.<br />

Ardiani, A. et al. (2014) Vaccine-mediated immunotherapy<br />

directed against a transcription factor driving the metastatic<br />

process. Cancer Res. 74, 1945−1957.<br />

Reynies, A. et al. (2014) Molecular classification of malignant<br />

pleural mesothelioma: identification of a poor prognosis<br />

subgroup linked to the epithelial-to-mesenchymal transition.<br />

Clin. Cancer Res. 20, 1323−1334.<br />

Wang, Y. et al. (2014) CUL4A induces epithelial-mesenchymal<br />

transition and promotes cancer metastasis by regulating<br />

ZEB1 expression. Cancer Res. 15, 520−531.<br />

Rajabi, H. et al. (2014) MUC1-C oncoprotein activates the<br />

ZEB1/miR-200c regulatory loop and epithelial-mesenchymal<br />

transition. Oncogene 33, 1680−1689.<br />

Kao, C.J. et al. (2014) miR-30 as a tumor suppressor<br />

connects EGF/Src signal to ERG and EMT. Oncogene 33,<br />

2495−2503.<br />

Truong, H.H. et al. (2014) beta1 integrin inhibition elicits a<br />

prometastatic switch through the TGFbeta-miR-200-ZEB<br />

network in E-cadherin-positive triple-negative breast cancer.<br />

Sci. Signal. 7, ra15.<br />

Geng, Y. et al. (2014) Insulin receptor substrate 1/2 (IRS1/2)<br />

regulates Wnt/beta-catenin signaling through blocking<br />

autophagic degradation of dishevelled2. J. Biol Chem. 289,<br />

11230−11241.<br />

Ludwig, K. et al. (2013) Colon cancer cells adopt an invasive<br />

phenotype without mesenchymal transition in 3-D but not<br />

2-D culture upon combined stimulation with EGF and crypt<br />

growth factors. BMC Cancer 13, 250−257.<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

• •<br />

• •<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

• •<br />

• •<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

• •<br />

• •<br />

•<br />

• •<br />

Target M P<br />

LPP<br />

Lyric/Metadherin<br />

Maspin<br />

Mesothelin<br />

MMP-2<br />

MMP-7<br />

MMP-9<br />

MUC1<br />

NCAM (CD56)<br />

α-Parvin<br />

Paxillin<br />

Phospho-Paxillin (Tyr118)<br />

PSA/KLK3<br />

RECK<br />

Renin<br />

Talin-1<br />

Phospho-Talin (Ser425)<br />

TIMP1<br />

TIMP2<br />

TIMP3<br />

uPAR<br />

Vinculin<br />

ZO-1<br />

ZO-2<br />

ZO-3<br />

Zyxin<br />

Phospho-Zyxin<br />

(Ser142/143)<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

• •<br />

•<br />

• •<br />

• •<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

• •<br />

• •<br />

• •<br />

•<br />

•<br />

•<br />

• •<br />

Chiyomaru, T. et al. (2014) Long non-coding RNA HOTAIR is<br />

targeted and regulated by miR-141 in human cancer cells.<br />

J. Biol. Chem. 289, 12550−12565.<br />

Akalay, I. et al. (2013) Epithelial-to-mesenchymal transition<br />

and autophagy induction in breast carcinoma promote<br />

escape from T-cell-mediated lysis. Cancer Res. 73,<br />

2418−2427.<br />

Song, S. et al. (2013) Loss of TGF-beta adaptor beta2SP<br />

activates notch signaling and SOX9 expression in esophageal<br />

adenocarcinoma. Cancer Res. 73, 2159−2169.<br />

Shi, Z. et al. (2013) mTOR signaling feedback modulates<br />

mammary epithelial differentiation and restrains invasion<br />

downstream of PTEN loss. Cancer Res. 73, 5218−5231.<br />

Dasgupta, S. et al. (2013) Novel role of MDA-9/syntenin in<br />

regulating urothelial cell proliferation by modulating EGFR<br />

signaling. Clin Cancer Res. 19, 4621−4633.<br />

Smith, N.R. et al. (2013) Tumor stromal architecture can<br />

define the intrinsic tumor response to VEGF-targeted therapy.<br />

Clin Cancer Res. 19, 6943−6956.<br />

Domyan, E.T. et al. (2013) Roundabout receptors are critical<br />

for foregut separation from the body wall. Dev. Cell 24,<br />

52−63.<br />

Yu, Y.H. et al. (2013) MiR-520h-mediated FOXC2 regulation is<br />

critical for inhibition of lung cancer progression by resveratrol.<br />

Oncogene 3, 431−443.<br />

Rizvi, I. et al. (2013) Flow induces epithelial-mesenchymal<br />

transition, cellular heterogeneity and biomarker modulation<br />

in 3D ovarian cancer nodules. Proc. Natl. Acad. Sci. USA<br />

110, 1974−1983.<br />

Gumireddy, K. et al. (2013) Identification of a long noncoding<br />

RNA-associated RNP complex regulating metastasis<br />

at the translational step. EMBO J. 32, 2672−2684.<br />

chapter 04: Cell Biology<br />

These protein targets represent key<br />

nodes within adhesion signaling<br />

pathways and are commonly<br />

studied in adhesion research. Primary<br />

antibodies, antibody conjugates, and<br />

antibody sampler kits containing<br />

these targets are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

163<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for E-Cadherin<br />

have been cited over 163 times in<br />

high-impact, peer-reviewed publications<br />

from the global research community.<br />

www.cellsignal.com/cstadhesion 109


Section I: Research Areas<br />

Adherens Junction Dynamics<br />

Cytoplasm<br />

Extracellular<br />

Space<br />

Cytoplasm<br />

Adherens<br />

Junction<br />

Remodeling<br />

VE-Cadherin<br />

Occludin<br />

MLC<br />

Nucleus<br />

Cadherin<br />

IQGAP<br />

WASP<br />

Arp2/3<br />

Formation<br />

Fyn Gβ1 δ-cat<br />

p190 RhoA<br />

δ-cat<br />

SH2D1A<br />

Cytoplasm<br />

c-Cbl<br />

α-cat β-cat<br />

LAMTOR1<br />

Rac<br />

PRK1<br />

Par3/6 aPKC<br />

RhoA<br />

ARHGAP1<br />

δ-cat<br />

Ect2 Cortactin<br />

cdc42 mDIA<br />

Centralspindlin<br />

Complex<br />

Increased<br />

Actomycin<br />

Contractility<br />

• Actin Nucleation<br />

• Actin Dynamics<br />

Vinculin<br />

ROCK<br />

δ-cat<br />

Kaiso<br />

IRSp53<br />

β-cat<br />

β-cat<br />

LEF/TCF<br />

Maintenance<br />

Cadherin<br />

Rac<br />

IQGAP<br />

β-cat<br />

α-cat<br />

PI3K<br />

Akt<br />

IQGAP<br />

PTPµ<br />

δ-cat<br />

Rac<br />

β-cat<br />

δ-cat<br />

α-cat β-cat<br />

Vinculin<br />

Tiam1<br />

CK2<br />

Src<br />

• Anti-Apoptotic Signals<br />

• Apoptosis<br />

• Angiogenesis<br />

Wnt<br />

Signaling<br />

PTP1B<br />

Cadherin<br />

Recycling<br />

Ajuba<br />

α-cat<br />

Arp2/3<br />

Cadherin<br />

Degradation<br />

Cell Growth & Differentiation<br />

Nectin<br />

Afadin<br />

α-cat<br />

γ-cat<br />

Formin<br />

α-cat<br />

RaIA<br />

α<br />

Nectin<br />

Clathrin<br />

Afadin<br />

Rap1<br />

α<br />

Src<br />

Disassembly<br />

Axin1<br />

β-cat<br />

E3<br />

Actin bundles<br />

Actin<br />

Disassembly<br />

Caveolin/Clathrin<br />

Phagosome<br />

α<br />

Caveolin<br />

Ub UbUb<br />

β-cat<br />

α-actinin<br />

Cadherin Endocytosis<br />

WASP<br />

Cip4<br />

APC<br />

GSK-3<br />

β-cat<br />

APC<br />

Ub<br />

Ub<br />

Ub<br />

Ub<br />

β-cat<br />

β-catenin<br />

Degradation<br />

Adherens junctions are dynamic structures that form, strengthen and spread, degrade, and then re-form as their associated proteins create ephemeral connections with<br />

counterparts from adjacent cells. This view updates the traditional model of a stable complex composed of cadherin, β-catenin, and α-catenin bound to the actin cytoskeleton.<br />

Although cadherin does exist in a complex with β-catenin and α-catenin, this cadherin-catenin complex does not associate with the actin cytoskeleton. α-catenin does not<br />

directly anchor cell adhesion proteins to the actin cytoskeleton but acts as a regulatory protein to control actin filament dynamics.<br />

Monomeric α-catenin binds β-catenin at adherens junctions and upon release forms α-catenin dimers that promote actin bundle formation. The transition from branched actin<br />

networks to bundled actin filaments correlates with the creation of mature, strong adherens junctions and a decrease in membrane lamellipodia. The connection between cell<br />

junctions and the cytoskeleton may be more dynamic than originally considered and may rely on multiple, weak associations between the cadherin-catenin complex and the<br />

actin cytoskeleton or rely on other membrane-associated proteins (i.e. nectin and afadin).<br />

As with most dynamic cellular systems, a collection of kinases, phosphatases, and adaptor proteins regulate the activity and localization of a few key effector proteins.<br />

δ-catenin (p120 catenin) binds and stabilizes cadherin at the plasma membrane. Membrane bound and cytosolic tyrosine kinases phosphorylate β-catenin at weak or nascent<br />

junctions, while phosphatases remove added phosphates from β-catenin and δ-catenin at established junctions. Rho family GTPases modulate the availability and activation<br />

state of catenins and other essential adherens proteins. Together, this collection of structural proteins, enzymes, and adaptor proteins creates dynamic cell-cell junctions<br />

necessary for temporary associations during morphogenesis and maintains the integrity of complex tissues and structures following development.<br />

Select Reviews:<br />

Baum, B. and Georgiou, M. (2011) J. Cell Biol. 192, 907–917. • Baumann, K. (2013) Nat. Rev. Mol. Cell Biol. 14, 68. • Citi, S., Spadaro, D., Schneider, Y., Stutz, J., and<br />

Pulimeno, P. (2011) Mol. Membr. Biol. 28, 427–444. • Harris, T.J. and Tepass, U. (2010) Nat. Rev. Mol. Cell Biol. 11, 502–514. • Niessen, C.M. and Gottardi, C.J. (2008)<br />

Biochim. Biophys. Acta 1778, 562–571. • Pieters, T., van Roy, F., and van Hengel, J. (2012) Front. Biosci. 17, 1669–1694. • Yonemura, S. (2011) Curr. Opin. Cell Biol.<br />

23, 515–522.<br />

Cytoskeletal Regulation<br />

The cytoskeleton consists of three types of cytosolic fibers: microtubules, microfilaments (actin<br />

filaments), and intermediate filaments. Cytoskeletal signaling regulates several important cellular<br />

processes such as cell division, adhesion, polarity, migration, and movement through cilia and flagella.<br />

Microtubules<br />

Microtubules are composed of globular tubulin subunits, with α/β-tubulin heterodimers forming the<br />

tubulin subunit common to all eukaryotic cells. γ-tubulin is required to nucleate polymerization of<br />

tubulin subunits to form microtubule polymers. Many cell movements are mediated by microtubule<br />

action, including the beating of cilia and flagella, cytoplasmic transport of membrane vesicles, and<br />

nerve-cell axon migration. Microtubules also play a critical role in spindle assembly during mitosis/<br />

meiosis and are responsible for chromosome alignment during metaphase. Microtubules form the 9+2<br />

structure of the centriole, a critical component of the centrosome that acts as a microtubule-organizing<br />

center (MTOC) and plays a role in cell polarity. Because of their role in mitosis, microtubules have been<br />

targets of chemotherapy in cancer. Microtubules continuously undergo a process of dynamic instability,<br />

whereby microtubule polymerization on the plus end competes with depolymerization at the minus end.<br />

This process is regulated by several signaling molecules including stathmin, diap1/2, tau, and the Rho<br />

family of small GTPases.<br />

Stathmin is a microtubule destabilizing<br />

protein phosphorylated at Ser38 in cells<br />

synchronized in mitosis.<br />

Phospho-Stathmin (Ser38) (D19H10) Rabbit mAb #4191: WB analysis<br />

of extracts from HT-29 and U-2 OS cells, untreated or synchronized in<br />

mitosis, using #4191. Mitotic synchrony was performed by using either a<br />

thymidine block followed by release into nocodazole (100 ng/ml, 24 hr) or<br />

using Docetaxel #9886 (100 ng/ml, 24 hr).<br />

kDa<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

1<br />

2 3<br />

– + – + – –<br />

– – – – – +<br />

Lanes<br />

1. HT-29<br />

2. U-2 OS<br />

3. HT-29<br />

Phospho-<br />

Stathmin<br />

(Ser38)<br />

Thymidine/<br />

Nocodazole<br />

Microfilaments<br />

Microfilaments are major structural components of the cytoskeleton and consist of fibrous polymers of<br />

actin, called F-actin. Microfilaments are important for changes in cell shape, migration, proliferation,<br />

and survival. Regulation of the actin cytoskeleton begins with signaling through G protein-coupled<br />

receptors (GPCRs), integrins, receptor tyrosine kinases (RTKs), and numerous other specialized receptors<br />

such as the semaphorin 1a receptor PlexinA. These receptors initiate a large number of signaling<br />

cascades that include the Rho family of small GTPases (Rho, Rac, and Cdc42) and their activators,<br />

guanine nucleotide exchange factors (GEFs) and their downstream protein kinase effectors (ROCK and<br />

PAK), as well as through direct binding of the GTPases to several actin regulatory proteins (cortactin,<br />

diap1/2, WAVE, and WASP). These cascades converge on proteins that directly regulate the behavior<br />

and organization of the actin cytoskeleton, including actin interacting regulatory proteins such as<br />

cofilin, ADF, Arp2/3 complex, Ena/VASP, profilin, and gelsolin.<br />

Small GTPases<br />

P 1<br />

Upstream signals<br />

Rho<br />

GTP<br />

GTP<br />

Docetaxel<br />

chapter 04: Cell Biology<br />

α/β tubulin heterodimers,<br />

the building<br />

blocks of microtubules,<br />

are found throughout<br />

the cytoplasm.<br />

β-Tubulin (9F3) Rabbit mAb (Alexa<br />

Fluor ® 488 Conjugate) #3623: Confocal<br />

IF analysis of HeLa cells using #3623<br />

(green). Blue pseudocolor = DRAQ5 ®<br />

#4084 (fluorescent DNA dye).<br />

COS-7 cells express<br />

cofilin phosphorylated<br />

at Ser3, a modification<br />

known to inhibit cofilin<br />

activity.<br />

kDa<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

60<br />

50<br />

40<br />

30<br />

Phospho-<br />

Cofilin (Ser3)<br />

GTPase-activating<br />

protein<br />

GTPase<br />

cycle<br />

Guanine nucleotide<br />

exchange factor<br />

20<br />

Cofilin<br />

© 2008–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Kris DeMali of The University of Iowa Carver College of Medicine, Iowa City, IA for reviewing this diagram.<br />

H 2O<br />

Rho<br />

GTP<br />

Downstream effectors<br />

GDP<br />

10<br />

– +<br />

λ-phosphatase<br />

Phospho-Cofilin (Ser3) (77G2) Rabbit<br />

mAb #3313: WB analysis of COS-7<br />

cells, untreated or λ phosphatase-treated,<br />

using #3313 (upper) or Cofilin Antiobdy<br />

#3312 (lower).<br />

110 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

Actin cytoskeleton<br />

www.cellsignal.com/cstcytoskeletal 111


Section I: Research Areas<br />

Intermediate filament<br />

protein desmin is expressed<br />

in muscle cells.<br />

Desmin (D93F5) XP ® Rabbit mAb<br />

#5332: Confocal IF analysis of mouse<br />

heart tissue using #5332 (green). Blue<br />

pseudocolor = DRAQ5 ® #4084 (fluorescent<br />

DNA dye).<br />

Vimentin is an<br />

intermediate filament<br />

protein expressed in<br />

connective tissue<br />

and other cells of<br />

mesenchymal origin.<br />

Nonmuscle myosin IIb<br />

heavy chain isoform<br />

colocalizes with actin.<br />

Myosin IIb (D8H8) XP ® Rabbit mAb<br />

#8824: Confocal IF analysis of COS-7<br />

cells, showing Myosin (A), Actin (B),<br />

and merged (C), using #8824 (green).<br />

Actin filaments were labeled with DY-554<br />

phalloidin (red). Blue pseudocolor =<br />

DRAQ5 ® #4084 (fluorescent DNA dye).<br />

Intermediate Filaments<br />

The major types of intermediate filaments are distinguished by their cell-specific expression.<br />

Classes of<br />

Intermediate<br />

Filaments<br />

Class Protein<br />

Expression<br />

I Acidic keratins Epithelial cells<br />

II Basic keratins Epithelial cells<br />

III Desmin, GFAP, vimentin Muscle, Glial cells, Mesenchymal cells<br />

IV Neurofilaments (NFL, NFM and NFH) Neurons<br />

V Lamins Nucleus<br />

Members of this group contain a globular N-terminal head domain, a central α−helical rod domain, and<br />

a variable C-terminal tail. Intermediate filaments provide structural support for the cell, act as anchorage<br />

points for organelles and molecular motors, and function as stress proteins that provide protection<br />

from intrinsic and environmental stresses. Intermediate filaments can be regulated through several<br />

mechanisms including phosphorylation, which affects their activity or ability to assemble with other<br />

intermediate filament-interacting proteins.<br />

A<br />

A<br />

112 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

B<br />

B<br />

Vimentin (D21H3) XP ® Rabbit mAb #5741:<br />

IHC analysis of paraffin-embedded mouse<br />

colon (A) using #5741. Confocal IF analysis<br />

of SNB19 cells (B) using #5741 (green). Blue<br />

pseudocolor = DRAQ5 ® #4084 (fluorescent<br />

DNA dye).<br />

Myosins<br />

Myosins are a large superfamily of actin-binding motor proteins that use ATP hydrolysis to generate<br />

motility. Myosins are categorized in over 25 classes. Of these, class II myosins, known as conventional<br />

myosins, comprise the largest class and contain myosin proteins specific to skeletal, cardiac, and<br />

smooth muscle as well as nonmuscle isoforms. Nonmuscle myosin is essential to cell motility, cell<br />

division, migration, adhesion, and polarity. The holoenzyme consists of two identical heavy chains and<br />

two sets of light chains. The light chains (MLCs) regulate myosin II activity and stability and exist in<br />

many isoforms with varying tissue distribution. The heavy chains (NMHCs) are encoded by three genes,<br />

MYH9, MYH10, and MYH14, which generate three different nonmuscle myosin II isoforms, IIa, IIb, and<br />

IIc, respectively. While all three isoforms perform the same enzymatic tasks, binding to and contracting<br />

actin filaments coupled to ATP hydrolysis, their cellular functions do not appear to be redundant and<br />

they have different subcellular distributions.<br />

Select Reviews<br />

Blanchoin, L., Boujemaa-Paterski, R., Sykes, C., et al. (2014) Physiol. Rev. 94, 235−263. • Chircop, M. (2014) Small GTPases<br />

5, e29770. • Chung, B.M., Rotty, J.D., and Coulombe, P.A. (2013) Curr. Opin. Cell Biol. 25, 600−612. • Gurel, P.S., Hatch,<br />

A.L., and Higgs, H.N. (2014) Curr. Biol. 24, 660−672. • Julian, L. and Olson, M.F. (2014) Small GTPases 5, e29846. • Kull,<br />

F.J. and Endow, S.A. (2013) J. Cell Sci. 126, 9−19. • Parker, A.L., Kavallaris, M., and McCarroll, J.A. (2014) Front. Oncol. 4,<br />

153. • Snider, N.T. and Omary, M.B. (2014) Nat. Rev. Mol. Cell Biol. 15, 163−177. • Wojnacki, J., Quassollo, G., Marzolo,<br />

M.P., et al. (2014) Small GTPases 5, e28430.<br />

C<br />

Commonly Studied Cytoskeletal Regulation Targets<br />

Target M P Target M P Target M P<br />

14-3-3 Family • Diap1<br />

• Keratin 8/18 •<br />

14-3-3 β/α • Diap2<br />

• Keratin 17 •<br />

14-3-3 γ • DOCK180 • Phospho-Keratin 17 (Ser44) •<br />

14-3-3 ε • DRP1<br />

• Keratin 17/19 •<br />

14-3-3 ζ/δ • Phospho-DRP1 (Ser616) • • Keratin 18 •<br />

14-3-3 η • • Phospho-DRP1 (Ser637) • • Keratin 19 •<br />

14-3-3 τ • DSG2<br />

• Keratin 20 •<br />

Phospho-Ack1 (Tyr284) • EB-1<br />

• Phospho-KIF1B (Ser1487) •<br />

Pan-Actin • • Emerin<br />

• • KIFC1<br />

•<br />

α-Actinin • • EML4<br />

• • KIF3A<br />

•<br />

Afadin<br />

• • EPAC1<br />

• KIF3B<br />

•<br />

Annexin V<br />

• EPAC2<br />

• Kinectin 1 • •<br />

Phospho-AP2M1 (Thr156) • • EpCAM<br />

• • Lamin A/C • •<br />

Phospho-ARHGAP42 • EPLIN<br />

• Phospho-Lamin A/C (Ser22) • •<br />

(Tyr376)<br />

Erlin-1<br />

Cool2/αPix •<br />

• Lamin B1 •<br />

Erlin-2<br />

Cool1/βPix<br />

•<br />

• Lamin B2 •<br />

EVL<br />

ARP2<br />

• •<br />

• LAMP1<br />

•<br />

Ezrin<br />

ARP3<br />

•<br />

• LASP1<br />

•<br />

Phospho-Ezrin (Tyr353)<br />

Atlastin-1 •<br />

• • LCP1<br />

• •<br />

Ezrin/Moesin/Radixin<br />

β-Actin<br />

• •<br />

• Phospho-LCP1 (Tyr28) •<br />

Phospho-Ezrin (Thr567)/<br />

Phospho-Catenin δ-1 •<br />

• • LIMK1<br />

•<br />

Radixin (Thr564)/Moesin<br />

(Ser320)<br />

(Thr558)<br />

Phospho-LIMK1 (Thr508)/ •<br />

β2-Chimerin<br />

LIMK2 (Thr505)<br />

• Fascin<br />

• LIMK2<br />

Caldesmon-1 • •<br />

•<br />

Fer<br />

• LLGL1<br />

Caveolin-1 • •<br />

•<br />

Fes<br />

• LMAN1<br />

Phospho-Caveolin-1 (Tyr14) •<br />

•<br />

Fibrillarin • Phospho-MARK Family<br />

Caveolin-2 •<br />

•<br />

Filamin A<br />

• (Activation Loop)<br />

CD2AP<br />

• Phospho-Filamin A<br />

MARK1<br />

•<br />

•<br />

CDC37<br />

(Ser2152)<br />

• •<br />

MARK2<br />

•<br />

Filamin B<br />

Phospho-CDC37 (Ser13) •<br />

• • MARK3<br />

•<br />

Filamin C<br />

Cdc42<br />

• •<br />

• MARK4<br />

•<br />

Flotillin-1<br />

CD71<br />

•<br />

• MCF2/Dbl<br />

•<br />

Flotillin-2<br />

CdGAP<br />

•<br />

• • Moesin<br />

•<br />

FYVE-CENT<br />

Centrin-2<br />

•<br />

• M-RIP<br />

•<br />

GEF-H1<br />

Chronophin/PDXP •<br />

• • MTSS1<br />

•<br />

Gelsolin<br />

CIN85<br />

•<br />

• • Myosin IIa<br />

•<br />

Phospho-GIT2 (Tyr392)<br />

Claudin-1 • •<br />

• Phospho-Myosin IIa •<br />

GM130<br />

CLIP1/CLIP170 •<br />

• • (Ser1943)<br />

Golgin-97<br />

Myosin IIb<br />

Cofilin<br />

• •<br />

•<br />

• •<br />

HEF1/NEDD9<br />

Myosin IIc<br />

Phospho-Cofilin (Ser3) • •<br />

•<br />

• •<br />

Importin β1<br />

Myosin Va<br />

Cortactin<br />

•<br />

•<br />

•<br />

IQGAP1<br />

Myosin VI<br />

Phospho-Cortactin (Tyr421) •<br />

•<br />

•<br />

IQGAP2<br />

Myosin Light Chain 2<br />

CrkII<br />

•<br />

•<br />

• •<br />

Integrin α2b<br />

Phospho-Myosin Light Chain<br />

Phospho-CrkII (Tyr221) •<br />

•<br />

• •<br />

2 (Ser19)<br />

Pan-Keratin<br />

Dab2<br />

• •<br />

• Phospho-Myosin Light Chain •<br />

Keratin 7<br />

2 (Thr18/Ser19)<br />

Desmin<br />

• •<br />

•<br />

chapter 04: Cell Biology<br />

These protein targets represent key nodes<br />

within cytoskeletal regulation signaling<br />

pathways and are commonly studied in<br />

cytoskeletal regulation research. Primary<br />

antibodies, antibody conjugates, and<br />

antibody sampler kits containing these<br />

targets are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

www.cellsignal.com/cstcytoskeletal 113


Section I: Research Areas<br />

104<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for β-tubulin<br />

have been cited over 104 times in<br />

high-impact, peer-reviewed publications<br />

from the global research community.<br />

Select Citations:<br />

Harikumar, K.B. et al. (2014)<br />

K63-linked polyubiquitination of<br />

transcription factor IRF1 is essential<br />

for IL-1-induced production of<br />

chemokines CXCL10 and CCL5. Nat.<br />

Immunol. 15, 231−238.<br />

Kim, D.K. et al. (2014) Inverse agonist<br />

of estrogen-related receptor gamma<br />

controls Salmonella typhimurium<br />

infection by modulating host iron homeostasis.<br />

Nat. Med. 20, 419−424.<br />

Bohrer, L.R. et al. (2014) Activation<br />

of the FGFR-STAT3 pathway in breast<br />

cancer cells induces a hyaluronan-rich<br />

microenvironment that licenses tumor<br />

formation. Cancer Res. 74, 374−386.<br />

Lucas, C.L. et al. (2014) Dominantactivating<br />

germline mutations in the<br />

gene encoding the PI(3)K catalytic<br />

subunit p110delta result in T cell<br />

senescence and human immunodeficiency.<br />

Nat. Immunol. 15, 88−97.<br />

Zhu, Y. et al. (2014) Role of tumor<br />

necrosis factor alpha-induced protein<br />

1 in paclitaxel resistance. Oncogene<br />

33, 3246−3255.<br />

Sun, X. et al. (2014) p27 Protein<br />

Protects Metabolically Stressed<br />

Cardiomyocytes from Apoptosis by<br />

Promoting Autophagy. J. Biol. Chem.<br />

289, 16924−16935.<br />

Barry, E.R. et al. (2013) Restriction of<br />

intestinal stem cell expansion and the<br />

regenerative response by YAP. Nature<br />

493, 106−110.<br />

Raj, V.S .et al. (2013) Dipeptidyl<br />

peptidase 4 is a functional receptor<br />

for the emerging human coronavirus-<br />

EMC. Nature 495, 251−254.<br />

Manzanillo, P.S. et al. (2013) The<br />

ubiquitin ligase parkin mediates<br />

resistance to intracellular pathogens.<br />

Nature 501, 512−616.<br />

Chowdhury, S. et al. (2013) Flavoneresistant<br />

Leishmania donovani<br />

Overexpresses LdMRP2 Transporter in<br />

the Parasite and Activates Host MRP2<br />

on Macrophages to Circumvent the<br />

Flavone-mediated Cell Death. J. Biol.<br />

Chem. 289, 16129−16147.<br />

Wang, D. et al. (2013) MicroRNA-205<br />

controls neonatal expansion of<br />

skin stem cells by modulating the<br />

PI(3)K pathway. Nat. Cell Biol. 15,<br />

1153−1163.<br />

Target M P Target M P Target M P<br />

Myosin Light Chain 2v • PREX1<br />

• Sec31A<br />

•<br />

(Cardiac Isoform)<br />

Profilin-1<br />

MYPT1<br />

• • SPAK<br />

•<br />

• •<br />

PTP4A3<br />

Phospho-MYPT1 (Ser507)<br />

• SSH1<br />

•<br />

•<br />

PVR/CD155<br />

Phospho-MYPT1 (Ser668)<br />

• Stathmin<br />

•<br />

•<br />

R-Ras<br />

Phospho-MYPT1 (Thr696)<br />

• • Phospho-Stathmin (Ser16) •<br />

•<br />

Rac1/Cdc42<br />

Phospho-MYPT1 (Thr853)<br />

• Phospho-Stathmin (Ser38) • •<br />

•<br />

Phospho-Rac1/cdc42<br />

Na,K-ATPase<br />

• Talin-1<br />

•<br />

• (Ser71)<br />

Phospho-Talin (Ser425)<br />

Phospho-Na,K-ATPase a1 • • Rac1/Rac2/Rac3<br />

• •<br />

•<br />

(Tyr10)<br />

TCTP<br />

RACK1<br />

• •<br />

Phospho-Na,K-ATPase a1<br />

• •<br />

•<br />

Phospho-TCTP (Ser46)<br />

(Ser16)<br />

Radixin<br />

•<br />

•<br />

Phospho-Na,K-ATPase a1<br />

Tensin 2<br />

• RalA<br />

•<br />

(Ser23)<br />

• •<br />

TESK1<br />

N-WASP<br />

RalB<br />

•<br />

• •<br />

•<br />

Troponin T (Cardiac)<br />

NCK1<br />

RalBP1<br />

•<br />

•<br />

• •<br />

Tropomyosin-1<br />

NTF2<br />

Ran<br />

•<br />

•<br />

•<br />

Tropomyosin-1/3<br />

NUP88<br />

RanBP1<br />

•<br />

•<br />

•<br />

Troponin I<br />

NUP98<br />

Phospho-RanBP3 (Ser58)<br />

• •<br />

• •<br />

•<br />

Phospho-Troponin I (Cardiac)<br />

OSR1<br />

Rap1A/Rap1B<br />

•<br />

•<br />

• • (Ser23/24)<br />

PAK1/2/3<br />

Rap1B<br />

•<br />

• α-Tubulin • •<br />

PAK1<br />

RasGRP3<br />

•<br />

• Acetyl-α-Tubulin (Lys40) • •<br />

Phospho-PAK1 (Ser144)/<br />

RCC1<br />

•<br />

• • α/β-Tubulin<br />

•<br />

PAK2 (Ser141)<br />

Phospho-RCC1 (Ser11) • • β-Tubulin • •<br />

Phospho-PAK1 • RCC2<br />

(Ser199/204)/PAK2<br />

• • γ-Tubulin<br />

•<br />

(Ser192/197)<br />

Phospho-REPS1 (Ser709) • Twinfilin-1 • •<br />

Phospho-PAK1 (Thr423)/ • RhoA<br />

PAK2 (Thr402)<br />

• VASP<br />

• •<br />

PAK2<br />

• •<br />

RhoB<br />

• Phospho-VASP (Ser157) •<br />

Phospho-PAK2 (Ser20) •<br />

RhoC<br />

• Phospho-VASP (Ser239) •<br />

PAK3<br />

•<br />

RhoE<br />

• Vav1<br />

• •<br />

PAK4<br />

•<br />

p190-A RhoGAP • • Vav2<br />

•<br />

Phospho-PAK4 (Ser474)/<br />

p190-B RhoGAP<br />

•<br />

• Vav3<br />

•<br />

PAK5 (Ser602)/PAK6<br />

RhoGDI<br />

(Ser560)<br />

• Villin-1<br />

•<br />

PAR2<br />

•<br />

p115 RhoGEF • Vimentin • •<br />

PCM-1<br />

•<br />

ROCK1<br />

• Phospho-Vimentin (Ser39) •<br />

PDLIM2<br />

•<br />

ROCK2<br />

• • Phospho-Vimentin (Ser56) • •<br />

Podoplanin •<br />

SCAI<br />

• Phospho-Vimentin (Ser83) • •<br />

PKG-1<br />

•<br />

Sec23A<br />

• WASP<br />

• •<br />

PKG-1α<br />

•<br />

Sec24A<br />

• WAVE-2 •<br />

Plectin-1 • •<br />

Sec24B<br />

• • WAVE-3<br />

•<br />

PRC1<br />

•<br />

Sec24C<br />

• ZO-1<br />

• •<br />

Sec24D<br />

•<br />

Antibody Validation Principles<br />

Please visit our website to learn more about what Antibody Validation means at Cell Signaling Technology.<br />

www.cellsignal.com/cstvalidation<br />

Regulation of Actin Dynamics<br />

F-actin<br />

Bundles/<br />

Meshworks<br />

chapter 04: Cell Biology<br />

PlexinA<br />

PI3K PLCγ PIP 2<br />

G<br />

Src<br />

PI3K<br />

i G<br />

Grb2 Nck<br />

q G s<br />

FAK PIP<br />

Paxillin<br />

3 Ca 2+<br />

Mical<br />

CaM PKCα<br />

PLCβ<br />

NADPH<br />

GRB2<br />

PAK1 WASP<br />

Fascin<br />

PIP PI3Kα<br />

NADP<br />

3<br />

GEF<br />

Filopodia<br />

Ena<br />

p130 GIT1/2<br />

Ras<br />

PKA<br />

VASP<br />

PI3K<br />

PIP 3<br />

PI3K<br />

PIX<br />

Cofilin<br />

MEKK<br />

RhoGEF<br />

TIAM1<br />

PIX<br />

Vav<br />

MEKK<br />

Degradation<br />

Crk<br />

Rac<br />

cdc42 PAK1 MEK<br />

Rho<br />

Cortactin<br />

PAK1<br />

mDIA<br />

ROCK Filamin<br />

WAVE<br />

WASP<br />

ROCK<br />

PI5K<br />

MLCK<br />

MBS<br />

Arp2/3<br />

Ca 2+<br />

PIP 2<br />

IP 3 CaM<br />

LIMK MLCK mDIA Arp2/3<br />

Filamin<br />

PKD1<br />

Profilin<br />

14-3-3ζ<br />

Calcineurin SSH<br />

MLC<br />

Severed<br />

Filaments<br />

Sema1a<br />

Gelsolin<br />

Lamellipodia<br />

Integrin<br />

Receptor<br />

CIN<br />

Hsp90<br />

RTK<br />

Cofilin ADF Filopodia<br />

Barbed-end<br />

CapZ Cappers<br />

Pointed-end<br />

Tmod Cappers<br />

Actin<br />

Polymerization<br />

Tropomyosins<br />

Lamellipodia<br />

Stress Fibers<br />

Signaling to the cytoskeleton through G protein-coupled receptors (GPCRs), integrins, receptor tyrosine kinases (RTKs), and numerous other specialized receptors, such as the<br />

semaphorin 1a receptor PlexinA, can lead to diverse effects on cell activity, including changes in cell shape, migration, proliferation, and survival. Integrins, in conjuction with<br />

other components of focal adhesion complexes, serve as the link between the extracellular matrix and cytoskeleton in many cell types. Integrin activation leads to activation of<br />

focal adhesion kinase (FAK) and Src kinase, resulting in phosphorylation of other FA components such as paxillin and the Crk-associated substrate p130 Cas, as well as the<br />

recruitment of signaling adapter proteins.<br />

Intracellular regulation of the cell’s response to external cues occurs through a large number of signaling cascades that include the Rho family of small GTPases (Rho, Rac,<br />

and Cdc42) and their activators, guanine nucleotide exchange factors (GEFs), their downstream protein kinase effectors, including Rho-kinase/ROCK and p21 activated<br />

kinase (PAK), as well as through direct binding of the GTPases to several actin regulatory proteins, such as cortactin, mDia, WAVE, and WASP. These cascades converge on<br />

proteins that directly regulate the behavior and organization of the actin cytoskeleton, including actin interacting regulatory proteins such as cofilin, Arp2/3 complex, Ena/VASP,<br />

forminins, profilin, and gelsolin. Signaling through different pathways can lead to the formation of distinct actin-dependent structures whose coordinated assembly/disassembly<br />

is important for directed cell migration and other cellular behaviors. Migration is also regulated by signaling to myosin, which participates in leading edge actin dynamics<br />

and enables retraction of the rear of the cells. Tropomyosins stabilize F-actin by preventing binding of severing and dynamizing factors. Some tropomyosins may also enhance<br />

filament dynamics. Dynamic actin is required for most cellular actin-dependent processes; inhibiting actin assembly and preventing actin disassembly are equally inhibitory to<br />

most behaviors.<br />

Aberrant control of cytoskeletal signaling, which can result in a disconnection between extracellular stimuli and cellular responses, is often seen in immune pathologies,<br />

developmental defects, and cancer.<br />

Select Reviews:<br />

Bernstein, B.W. and Bamburg, J.R. (2010) Trends Cell Biol. 20, 187–195. • Lee, S.H. and Dominguez, R. (2010) Mol. Cells 29, 311–325. • Levayer, R. and Lecuit, T.<br />

(2012) Trends Cell Biol. 22, 61–81. • Poukkula, M., Kremneva, E., Serlachius, M., and Lappalainen, P. (2011) Cytoskeleton (Hoboken) 68, 471–490. • Ridley, A.J. (2011)<br />

Cell 145, 1012–1022. • Rottner, K. and Stradal, T.E. (2011) Curr. Opin. Cell Biol. 23, 569–578.<br />

© 2008–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. James Bamburg, Colorado State University, Fort Collins, CO for reviewing this diagram.<br />

G 13<br />

GPCR<br />

114 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstpathways<br />

115


Section I: Research Areas<br />

Regulation of Microtubule Dynamics<br />

Spred1<br />

TESK<br />

Cofilin<br />

LIMK<br />

TPPP<br />

Par1<br />

TAOK<br />

MARK<br />

PIP 3<br />

LL5β<br />

Rac1<br />

cdc42<br />

Par6<br />

Rac1<br />

Tau<br />

MT Stability<br />

LRP<br />

Par3<br />

mDIA<br />

Delivery to<br />

MT Plus Ends<br />

Src<br />

Tiam1<br />

aPKC<br />

Rho<br />

mDIA<br />

APC<br />

EB1<br />

CLIP<br />

c-Abl<br />

Trio<br />

Gα q/o<br />

Wnt<br />

MARK2<br />

Dvl<br />

MT<br />

Polymerization<br />

CLASP<br />

EB1<br />

CLIP<br />

Plus End<br />

Rho<br />

Plus End<br />

Proteins<br />

CLIP<br />

CLASP<br />

EB1<br />

APC<br />

mDIA1<br />

Erk<br />

MAPKAPK<br />

GSK-3β<br />

CRMP2<br />

Gα<br />

CLIP<br />

MAP1b<br />

XMAP215<br />

PI3K<br />

Akt PIP 3<br />

PTEN ROCK<br />

ICIS<br />

MCAK<br />

RTK<br />

Cdk1<br />

+ end Growth<br />

Promoting +/- end<br />

Destabilizing<br />

“Dynamic<br />

Microtubules”<br />

Focal Adhesions<br />

Rho<br />

MAP1b<br />

CaMK<br />

Aurora B<br />

Neurotrophins<br />

Actin Filaments<br />

Minus End<br />

PKA<br />

Rho<br />

PAK<br />

Stathmin<br />

MT Catastrophe<br />

RhoGEF<br />

Erk<br />

Stat3<br />

Protein Folding and<br />

VesIcle Trafficking<br />

Newly synthesized proteins must be properly folded and then directed to their correct subcellular locations<br />

in order to perform their biological functions. This highly regulated process includes molecular<br />

chaperone proteins that assist with proper folding and vesicle trafficking proteins that regulate delivery<br />

of cargo throughout the cell.<br />

Protein Folding: Heat Shock Proteins<br />

Heat Shock Proteins (HSPs) form seven families (small HSPs (sHSPs), HSP10, 40, 60, 70, 90, and<br />

100) of molecular chaperone proteins that play a central role in the cellular resistance to stress and<br />

actin organization. They are involved in the proper folding of proteins and the recognition and refolding<br />

of misfolded proteins. HSP expression is induced by a variety of environmental stresses, including heat,<br />

hypoxia, nutrient deficiency, free radicals, toxins, ischemia, and UV radiation. HSP27 is a member of<br />

the sHSP family. It is phosphorylated at Ser15, Ser78, and Ser82 by MAPKAPK-2 as a result of the<br />

activation of the p38 MAP kinase pathway. Phosphorylation and increased concentration of HSP27 has<br />

been implicated in actin polymerization and reorganization. HSP70 and HSP90 interact with unfolded<br />

proteins to prevent irreversible aggregation and catalyze the refolding of their substrates in an ATP- and<br />

co-chaperone-dependent manner. HSP70 has a broad range of substrates including newly synthesized<br />

and denatured proteins, while HSP90 tends to have a more limited subset of substrates, most of which<br />

are signaling molecules. HSP70 and HSP90 are also essential for the maturation and inactivation of<br />

nuclear hormones and other signaling molecules.<br />

Activation of the p38 MAPK pathway<br />

by UV or anisomycin results in phosphorylation<br />

of HSP27 at Ser82.<br />

Phospho-HSP27 (Ser82) (D1H2) XP ® Rabbit mAb #9709: WB<br />

analysis of extracts from HeLa or HT-29 cells, untreated (-) or treated<br />

(+) with either UV (40 mJ/cm 2 with 30 min recovery) or anisomycin<br />

(25 μg/ml, 30 min), using #9709.<br />

kDa<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

1<br />

– + – – –<br />

+ +<br />

– – –<br />

2<br />

Cells<br />

1. HeLa<br />

Phospho-<br />

HSP27<br />

(Ser82)<br />

UV<br />

Anisomycin<br />

2. HT-29<br />

chapter 04: Cell Biology<br />

HSP60, an important<br />

chaperone for folding<br />

key mitochondrial<br />

proteins, is expressed<br />

in A-204 cells.<br />

HSP60 (D6F1) XP ® Rabbit mAb<br />

#12165: Confocal IF analysis of A-204<br />

cells using #12165 (green). Actin filaments<br />

were labeled with DY-554 phalloidin<br />

(red). Blue pseudocolor = DRAQ5 ®<br />

#4084 (fluorescent DNA dye).<br />

Microtubules are required for the establishment of cell polarity, polarized migration of cells, intracellular vesicle transport, and chromosomal segregation in mitosis. Microtubules<br />

(MTs) are nonequilibrium polymers of α/β-tubulin heterodimers, in which GTP hydrolysis on the β-tubulin subunit occurs following assembly. Most microtubules are<br />

nucleated from organizing centers. The most prevalent microtubule behavior is dynamic instability, a process of slow plus end growth coupled with rapid depolymerization<br />

(“catastrophe”) and subsequent rescue. Although microtubule minus ends show dynamic instability, albeit at a lower rate than the plus ends, the minus ends are usually<br />

capped and anchored at MT organizing centers and thus often do not participate in microtubule dynamics.<br />

Maintaining a balance between dynamically unstable and stable microtubules is regulated in large part by proteins that bind either tubulin dimers or assembled microtubules.<br />

Proteins that bind tubulin dimers include stathmin, which sequesters tubulin and enhances MT dynamics by increasing catastrophe frequency, and collapsin response mediator<br />

protein (CRMP2), which increases MT growth rate by promoting addition of tubulin dimers onto microtubule plus ends. Other proteins that associate with assembled MTs include<br />

those that bundle MTs (e.g. MAP1c), those that stabilize MTs (e.g. tau), and those that maintain MTs in a dynamic state (MAP1b). A major signaling pathway that regulates MT<br />

dynamics involves GSK-3β, a kinase typically active under basal growth conditions but locally inactive in response to signals that enhance MT growth and dynamics.<br />

In addition to the above factors, many MT motor proteins, and even non-motor proteins, aid in the dynamics of MTs. Proteins such as Xenopus microtubule associated protein<br />

215 (XMAP215), promote MT assembly through binding to tubulin dimer to facilitate its incorporation in the growing plus end. XMAP215 also may compete with some of the<br />

MT plus end binding proteins (+TIPS), of which the end binding protein EB1 appears to be the master organizer. Complexes between the adenomateous polyposis coli (APC)<br />

protein and plus end binding proteins stabilize MTs by increasing the duration of the MT elongation phase. MT instability is promoted by several nonmotile kinesins from the<br />

kinesin-13 family. The mitotic centromere associated kinesin, MCAK, one of the most studied kinesin-13 family proteins, binds both plus and minus MT ends in vitro. The<br />

binding of MCAK to a MT end is thought to accelerate the transition to catastrophe by weakening the lateral interactions between the protofilaments.<br />

Tubulin undergoes several post-translational modifications such as acetylation, poly-glutamylation, and poly-glycylation, which have been shown to alter the association with<br />

certain MT motors as well as other proteins that can affect MT stability and dynamics.<br />

Select Reviews:<br />

Anitei, M. and Hoflack, B. (2011) Nat. Cell Biol. 14, 11–19. • de Forges, H., Bouissou, A., and Perez, F. (2012) Int. J. Biochem. Cell Biol. 44, 266–274. • Etienne-Manneville,<br />

S. (2010) Curr. Opin. Cell Biol. 22, 104–111. • Mimori-Kiyosue, Y. (2011) Cytoskeleton (Hoboken) 68, 603–618. • van der Vaart, B., Akhmanova, A., and Straube, A.<br />

(2009) Biochem. Soc. Trans. 37, 1007–1013.<br />

Vesicle Trafficking<br />

Types of Endosomes<br />

Endosomes are membrane-bound vesicles that transport molecules from the plasma membrane to<br />

the lysosomes for degradation and can be categorized as early, late, or recycling endosomes. Early<br />

endosomes fuse with clathrin-coated endocytic vesicles that contain extracellular particles, fluid, and<br />

membrane-bound receptors. EEA1 is an early endosome marker essential for membrane fusion and<br />

trafficking. Early endosomes increase in size and undergo a maturation process that results in their development<br />

into late endosomes, which can be identified using late endosome markers Rab7 and Rab9.<br />

Late endosomes fuse with lysosomes, which results in degradation of vesicle contents. Molecules contained<br />

within early endosomes can also be transported to the Golgi for sorting via recycling endosomes.<br />

EEA1 is a marker for early endosomes.<br />

EEA1 (C45B10) Rabbit mAb #3288: Confocal IF analysis of HeLa cells<br />

using #3288 (green). Actin filaments have been labeled with DY-554<br />

phalloidin (red). Blue pseudocolor = DRAQ5 ® (fluorescent DNA dye).<br />

© 2008–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. James Bamburg, Colorado State University, Fort Collins, CO for reviewing this diagram.<br />

116 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstproteinfolding 117


Section I: Research Areas<br />

Rab7 localizes to<br />

late endosomes.<br />

Rab7 (D95F2) XP ® Rabbit mAb<br />

#9367: Confocal IF analysis of SK-<br />

MEL-28 cells using #9367 (green). Actin<br />

filaments were labeled using DY-554<br />

phalloidin (red). Blue pseudocolor =<br />

DRAQ5 ® #4084 (fluorescent DNA dye).<br />

LAMP1 localizes<br />

to lysosomes.<br />

LAMP1 (D2D11) XP ® Rabbit mAb<br />

#9091: Confocal IF analysis of HeLa<br />

cells using #9091 (green). Actin filaments<br />

were labeled with DY-554 phalloidin<br />

(red). Blue pseudocolor = DRAQ5 ®<br />

#4084 (fluorescent DNA dye).<br />

Rab Proteins<br />

Rab proteins have been implicated in the regulation of intracellular protein trafficking in both the<br />

endocytic and biosynthetic pathways. GDP-bound Rabs are inactive and localize to the cytosol, while<br />

active GTP-bound Rabs localize to the cytoplasmic side of membrane compartments where they recruit<br />

downstream effector proteins that, along with Rabs, mediate vesicle formation, motility, docking, and<br />

fusion. Rab effector proteins include coat proteins, sorting adaptors, SNAREs, and microtubule- and<br />

actin-based motor proteins.<br />

The Rab11 subfamily (Rab11a, Rab11b, and Rab25) localizes to the endosomal recycling compartment,<br />

as well as to the apical recycling endosomes of polarized epithelial cells. Endosomal trafficking events<br />

through these compartments are mediated by Rab11 family members and their effector molecules, such<br />

as the Rab11-family interacting proteins (FIPs), Rabphilin-11/Rab11BP, myosin Vb, and Sec15.<br />

Rab proteins<br />

serve as markers<br />

for the various types<br />

of endosomes.<br />

Compartment<br />

Early Endosome<br />

Late Endosome<br />

Lysosome<br />

Recycling Endosome<br />

Perinuclear and Apical Recycling Endosome<br />

Golgi-to-Membrane Anterograde Endosome<br />

Exocytosis<br />

Endoplasmic Reticulum<br />

Golgi Apparatus<br />

ER-to-Golgi Anterograde Endosome<br />

Golgi-to-ER Retrograde Endosome<br />

Trans Golgi Network-to-Basolateral Membrane Endosome<br />

Rab25, which associates with apical recycling<br />

vesicles, is expressed in multiple cell lines.<br />

A<br />

Rab10, an early endosome marker, mediates<br />

protein transport between early endosomes<br />

and basolateral compartments.<br />

Rab10 (D36C4) XP ® Rabbit mAb #8127: Confocal IF analysis of MCF7 cells<br />

using #8127 (green). Actin filaments were labeled with DY-554 phalloidin (red).<br />

Blue pseudocolor = DRAQ5 ® #4084 (fluorescent DNA dye).<br />

Marker<br />

EEA1, Rab4, Rab5, Rab10<br />

Rab7, Rab9<br />

LAMP1<br />

Rab4, Rab11, Rab25, Rab35<br />

Rab17<br />

Rab38<br />

Rab3A<br />

PDI, Calnexin<br />

RCAS1<br />

Rab1A<br />

Rab6<br />

Rab8<br />

Rab25 (D4P6P) XP ® Rabbit mAb #13048: Confocal IF analysis of MCF7 (positive) (A) and HeLa (negative) (B) cells using #13048<br />

(green). Blue pseudocolor = DRAQ5 ® #4084 (fluorescent DNA dye). WB analysis of extracts from various cell lines (C) using #13048.<br />

Select Reviews<br />

Barr, F.A. (2013) J. Cell Biol. 202, 191−199. • Gurgis, F.M., Ziaziaris, W., and Munoz, L. (2014) Mol. Pharmacol. 85, 345−356.<br />

• Lachance, V., Angers, S., and Parent, J.L. (2014) Small GTPases 5, e29039. • Mayer, M.P. (2013) Trends Biochem. Sci. 38,<br />

507−514. • Mendoza, P., Diaz, J., Silva, P., et al. (2014) Small GTPases. 5, e28195 • Pfeffer, S.R. (2013) Curr. Opin. Cell<br />

Biol. 25, 414−419. • Saibil, H. (2013) Nat. Rev. Mol. Cell Biol. 14, 630−642. • Villarroel-Campos, D., Gastaldi, L., Conde, C.,<br />

et al. (2014) J. Neurochem. 129, 240−248.<br />

118 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

B<br />

C<br />

kDa<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

1 2 3<br />

Lanes<br />

1. HCT-15<br />

2. ZR-75-1<br />

3. MCF7<br />

Rab25<br />

Commonly Studied Protein Folding and Vesicle Trafficking Targets<br />

Target M P Target M P<br />

ACE2<br />

• Grp75<br />

• •<br />

APPL1<br />

• Grp94<br />

•<br />

Arf6<br />

• • Hip<br />

• •<br />

Bag1<br />

• Hop<br />

• •<br />

BAG6<br />

• HRS<br />

•<br />

BiP<br />

• • HSF1<br />

• •<br />

Calnexin<br />

• • HSP27<br />

• •<br />

Calpastatin<br />

• Phospho-HSP27 (Ser15) •<br />

Caveolin-1 • • Phospho-HSP27 (Ser78) •<br />

Phospho-Caveolin-1 (Tyr14) • Phospho-HSP27 (Ser82) • •<br />

CCT2<br />

• HSP40<br />

• •<br />

CDC37<br />

• • HSP60<br />

• •<br />

Phospho-CDC37 (Ser13) • HSP70<br />

• •<br />

CHOP<br />

• TRAP1/HSP75 •<br />

Clathrin Heavy Chain • • HSP90<br />

• •<br />

CRYAB<br />

• Phospho-HSP90α (Thr5/7) •<br />

Derlin-1<br />

• HSP90β<br />

• •<br />

DNAJC2/MPP11 • HSPA4/Apg-2 •<br />

Phospho-DNAJC2/MPP11 • HSPA8<br />

•<br />

(Ser47)<br />

HSPB8/HSP22 •<br />

Dynamin-I •<br />

HYOU1<br />

•<br />

Dynamin I/II<br />

•<br />

IGF-II Receptor/CI-M6PR •<br />

EEA1<br />

• •<br />

IRE1α<br />

•<br />

Eps15<br />

• •<br />

MBTPS2<br />

•<br />

Ero1-Lα<br />

•<br />

NSF<br />

• •<br />

ERp44<br />

• •<br />

OCRL1<br />

•<br />

ERp57<br />

•<br />

OS-9<br />

•<br />

ERp72<br />

• •<br />

p58IPK<br />

•<br />

FKBP4<br />

• •<br />

PDI<br />

• •<br />

GCN2<br />

•<br />

PERK<br />

•<br />

GM130<br />

• •<br />

Phospho-PERK (Thr980) •<br />

GOPC<br />

•<br />

Select Citations:<br />

Kalwa, H. et al. (2014) Central role<br />

for hydrogen peroxide in P2Y1 ADP<br />

receptor-mediated cellular responses<br />

in vascular endothelium. Proc. Natl.<br />

Acad. Sci. USA 111, 3383−3388.<br />

Gai, X. et al. (2014) Caveolin-1 is<br />

up-regulated by GLI1 and contributes<br />

to GLI1-driven EMT in hepatocellular<br />

carcinoma. PLoS One 9, e84551.<br />

Cai, B. et al. (2014) Rapid degradation<br />

of the complement regulator,<br />

CD59, by a novel inhibitor. J. Biol.<br />

Chem. 289, 12109−12125.<br />

Yi, S.L. et al. (2014) Role of<br />

caveolin-1 in atrial fibrillation as an<br />

anti-fibrotic signaling molecule in<br />

human atrial fibroblasts. PLoS One<br />

9, e85144.<br />

Huang, J. et al. (2014) Cross-talk between<br />

EphA2 and BRaf/CRaf is a key<br />

determinant of response to Dasatinib.<br />

Clin. Cancer Res. 20, 1846−1855.<br />

Mukherjee, R. et al. (2014) Sexdependent<br />

expression of caveolin 1<br />

in response to sex steroid hormones<br />

is closely associated with development<br />

of obesity in rats. PLoS One<br />

9, e90918.<br />

Meckes, D.G. Jr. et al. (2013)<br />

Epstein-Barr virus LMP1 modulates<br />

lipid raft microdomains and the<br />

vimentin cytoskeleton for signal<br />

transduction and transformation.<br />

J. Virol. 87, 1301−1311.<br />

Randazzo, D. et al. (2013) Obscurin is<br />

required for ankyrinB-dependent dystrophin<br />

localization and sarcolemma<br />

integrity. J. Cell Biol. 200, 523−536.<br />

Choi, C.H. et al. (2013) Mechanism<br />

for the endocytosis of spherical<br />

nucleic acid nanoparticle conjugates.<br />

Proc. Natl. Acad. Sci. USA 110,<br />

7625−7630.<br />

Lin, H.Y. et al. (2013) Caveolar<br />

endocytosis is required for human<br />

PSGL-1-mediated enterovirus 71<br />

infection. J. Virol. 87, 9064−9076.<br />

Pongrakhananon, V. et al. (2013)<br />

Ouabain suppresses the migratory<br />

behavior of lung cancer cells. PLoS<br />

One 8, e68623.<br />

Knowles, C.J. et al. (2013) Palmitate<br />

diet-induced loss of cardiac<br />

caveolin-3: a novel mechanism for<br />

lipid-induced contractile dysfunction.<br />

PLoS One 8, e61369.<br />

Target M P<br />

PKR<br />

Prostate Specific Membrane<br />

Antigen<br />

Rab1A<br />

Rab3A<br />

Rab4<br />

Rab5<br />

Rab6<br />

Rab7<br />

Rab8<br />

Rab9<br />

Rab10<br />

Rab11<br />

Rab11a<br />

Rab11b<br />

Rab11FIP1<br />

Rab17<br />

Rab25<br />

Rab35<br />

Rab38<br />

Rabex-5<br />

RBX1<br />

RCAS1<br />

REPS1<br />

SGTA<br />

Phospho-SGTA (Ser305)<br />

SNIP/p140Cap<br />

STAM1<br />

Syntaxin 6<br />

Tid-1<br />

VAMP8<br />

• •<br />

•<br />

•<br />

• •<br />

•<br />

• •<br />

• •<br />

• •<br />

•<br />

•<br />

• •<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

Nho, R.S. et al. (2013) FoxO3a<br />

(Forkhead Box O3a) deficiency<br />

protects Idiopathic Pulmonary<br />

Fibrosis (IPF) fibroblasts from type I<br />

polymerized collagen matrix-induced<br />

apoptosis via caveolin-1 (cav-1) and<br />

Fas. PLoS One 8, e61017.<br />

Park, W.J. et al. (2013) Protection of a<br />

ceramide synthase 2 null mouse from<br />

drug-induced liver injury: role of gap<br />

junction dysfunction and connexin 32<br />

mislocalization. J. Biol. Chem. 288,<br />

30904−30916.<br />

Demir, K. et al. (2013) RAB8B is<br />

required for activity and caveolar<br />

endocytosis of LRP6. Cell Rep. 4,<br />

1224−1234.<br />

Simone, L.C. et al. (2013) Role of<br />

phosphatidylinositol 4,5-bisphosphate<br />

in regulating EHD2 plasma membrane<br />

localization. PLoS One 8, e74519.<br />

Ekman, M. et al. (2013) Mir-29<br />

repression in bladder outlet obstruction<br />

contributes to matrix remodeling<br />

and altered stiffness. PLoS One 8,<br />

e82308.<br />

chapter 04: Cell Biology<br />

These protein targets are commonly<br />

studied in protein folding and vesicle<br />

trafficking research. Primary antibodies,<br />

antibody conjugates, and antibody<br />

sampler kits containing these targets<br />

are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

32<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for Caveolin<br />

have been cited over 32 times in<br />

high-impact, peer-reviewed publications<br />

from the global research community.<br />

www.cellsignal.com/cstproteinfolding 119


Section I: Research Areas<br />

chapter 04: Cell Biology<br />

Ubiquitin and Ubiquitin-like Proteins<br />

The ubiquitin-proteasome system (UPS) is the primary means by which cellular proteins are degraded.<br />

The UPS is a highly regulated system for elimination of misfolded or damaged proteins as well as<br />

proteins whose activity is acutely regulated by signaling pathways. This system plays a central role<br />

in cell proliferation, transcriptional regulation, apoptosis, immunity, development, and many other<br />

cellular processes (e.g., organelle biogenesis, cellular response to infection, etc.). Ubiquitin is a highly<br />

conserved 76-amino acid protein that can be covalently linked to many cellular proteins through an enzymatic<br />

cascade. Ubiquitination is an ATP-dependent process carried out by three classes of enzymes.<br />

A “ubiquitin activating enzyme” (E1), UBA1, forms a thio-ester bond with ubiquitin. This reaction allows<br />

subsequent binding of ubiquitin to “ubiquitin conjugating enzymes” (E2s), followed by the formation of<br />

an isopeptide bond between the C-terminus of ubiquitin and the ε-amino group of a lysine residue on<br />

the substrate protein. The latter reaction requires a “ubiquitin ligase” (E3). Several hundred E3 ligases<br />

exist within the eukaryotic cell; each ligase can only modify a subset of substrate proteins, thereby<br />

providing substrate specificity to the system. Ubiquitinated proteins are then targeted to the 26S<br />

proteasome for degradation or experience changes in protein location or activity.<br />

Ubiquitin Linkages<br />

Ubiquitin can be linked to a substrate as a single unit, monoubiquitination, or as a branched chain,<br />

polyubiquitination. Substrate proteins are linked to ubiquitin using seven distinct ubiquitin lysine residues<br />

(Lys6, Lys11, Lys27, Lys29, Lys33, Lys48, and Lys63). Formation of a polyubiquitin chain occurs<br />

when a lysine residue of ubiquitin is linked to the C-terminal glycine of another ubiquitin. Polyubiquitinated<br />

proteins have distinct fates depending upon the nature of the ubiquitin linkage through which<br />

they are conjugated; K48-linked polyubiquitin chains mainly target proteins for proteasomal degradation<br />

while K63-linked polyubiquitin chains typically regulate protein function, subcellular localization,<br />

and protein-protein interactions. The K63 linkage sometimes results in proteasomal degradation as<br />

well. K11-linked polyubiquitin chains regulate cell cycle targets and progression through mitosis.In<br />

addition, ubiquitin also can be linked to a target protein through its N-terminal methionine residue.<br />

This linkage, termed linear ubiquitination, is catalyzed by the linear ubiquitin chain assembly complex<br />

(LUBAC) and plays a critical role in NF-κB signaling.<br />

Distinct ubiquitin linkages can be detected using linkage-specific antibodies.<br />

Ubiquitination<br />

Ub<br />

ATP<br />

E2<br />

E3<br />

Substrate<br />

E1<br />

E2<br />

Substrate<br />

Ub<br />

Ub<br />

E3<br />

Ub<br />

Polyubiquitination<br />

via multiple cycles<br />

Proteasomal Degradation<br />

Changes in Protein Location or Activity<br />

K63-linkage Specific<br />

Polyubiquitin (D7A11)<br />

Rabbit mAb #5621: WB<br />

analysis of seven distinct<br />

recombinant polyubiquitin<br />

chains (A) using #5621<br />

(upper) and Ubiquitin<br />

Antibody #3933 (lower).<br />

K48-linkage Specific<br />

Polyubiquitin (D9D5)<br />

Rabbit mAb #8081: WB<br />

analysis of six distinct<br />

recombinant polyubiquitin<br />

chains (B) using #8081<br />

(upper) and Ubiquitin (P4D1)<br />

Mouse mAb #3936 (lower).<br />

Lanes<br />

1. K6<br />

2. K11<br />

3. K27<br />

4. K29<br />

5. K33<br />

6. K48<br />

7. K63<br />

A<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

1 2 3 4 5 6 7<br />

K63-linked<br />

Polyubiquitin<br />

Polyubiquitin<br />

B<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

1 2 4 5 6 7<br />

K48-linked<br />

Polyubiquitin<br />

Polyubiquitin<br />

UBE3A, a widely expressed E3 ubiquitin ligase<br />

UBE3A (D10D3) Rabbit mAb #7526: WB analysis<br />

of extracts from various cell lines using #7526.<br />

Lanes<br />

1. K-562<br />

2. SK-N-SH<br />

3. SK-N-MC<br />

4. A172<br />

5. T-47D<br />

6. HEK001<br />

7. T24<br />

8. KNRK<br />

9. NIH/3T3<br />

10. COS-7<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

1 2 3 4 5 6 7 8 9 10<br />

UBE3A<br />

Deubiquitinating Enzymes<br />

Deubiquitinating enzymes (DUBs) reverse the process of ubiquitination by removing ubiquitin from its<br />

substrate protein. DUB activity maintains ubiquitin recycling and ensures the cellular pool of ubiquitin<br />

molecules remains steady. DUBs are categorized into 5 subfamilies: USP, UCH, OTU, MJD, and JAMM,<br />

each with a specific tissue and ubiquitin-linkage specificity. Overexpression or misregulation of DUBs<br />

have been linked to cancer and other diseases. For example, overexpression of USP6 has been linked<br />

to bone and other mesenchymal tumors, mutations in CYLD are associated with skin tumors, and<br />

overexpression of USP33 to von Hippel-Lindau diease. In addition, other DUB mutations are associated<br />

with neurodegenerative disorders, including a role for UCHL1 in Parkinson’s disease and ATX3 in<br />

spinocerebellar ataxia.<br />

Skp2, a substrate<br />

recognition subunit of<br />

the Skp-Cullin-F-box<br />

(SCF) ubiquitin ligase<br />

complex, is expressed<br />

in many cell lines<br />

and cancers.<br />

A<br />

B<br />

kDa<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

1 2 3 4 5 6 7 8 9 10<br />

Skp2 (D3G5) XP ® Rabbit mAb #2652: IHC analysis of paraffin-embedded human ovarian carcinoma (A) using #2652.<br />

WB analysis of extracts from various cell lines (B) using #2652.<br />

Skp2<br />

Lanes<br />

1. U-2 OS<br />

2. LNCaP<br />

3. MDA-<br />

MB-468<br />

4. K-562<br />

5. U-87 MG<br />

6. WI-3B<br />

7. SK-OV-3<br />

8. MCF7<br />

9. MCF-10A<br />

10. COS-7<br />

A<br />

B<br />

kDa 1 2 3 4 5 6 7 8 9 10<br />

200<br />

140<br />

100<br />

80<br />

60<br />

USP10 (D7A5) Rabbit mAb #8501: Confocal IF analysis of HCT 116 cells (A) using #8501 (green). Blue pseudocolor<br />

= DRAQ5 ® #4084 (fluorescent DNA dye). WB analysis of extracts from various cell lines (B) using #8501.<br />

USP10<br />

Lanes<br />

1. HCT 116<br />

2. NCI-H23<br />

3. 786-0<br />

4. U-2 OS<br />

5. LNCaP<br />

6. MCF7<br />

7. Meg-01<br />

8. mIMCD-3<br />

9. H-4-11-E<br />

10. COS-7<br />

USP10, a DUB known<br />

to act on p53, Vps34,<br />

and CFTR, is found<br />

in the cytoplasm and<br />

is widely expressed in<br />

many cell lines.<br />

120 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstubiquitin<br />

121


Section I: Research Areas<br />

chapter 04: Cell Biology<br />

UCHL1 (D3T2E) XP ® Rabbit<br />

mAb #13179: WB analysis of<br />

extracts from various cell lines (A)<br />

using #13179 (upper) and GAPDH<br />

(D16H11) XP ® Rabbit mAb #5174<br />

(lower). IHC analysis of paraffinembedded<br />

human non-small cell<br />

lung carcinoma (B) using #13179.<br />

Lanes<br />

1. DU 145<br />

2. LNCaP<br />

3. NCI-H1299<br />

4. NCI-H358<br />

5. WI-38<br />

6. BT-549<br />

7. U266<br />

8. A172<br />

9. SH-SY5Y<br />

10. T98G<br />

11. SK-N-AS<br />

12. Neuro-2a<br />

13. C6<br />

14. COS-7<br />

SENP3, a SUMO<br />

protease, localizes<br />

to the nucleolus.<br />

SENP3 (D20A10) XP ® Rabbit mAb<br />

#5591: Confocal IF analysis of HeLa cells<br />

using #5591 (green). Actin filaments<br />

were labeled with DY-554 phalloidin (red).<br />

Blue pseudocolor = DRAQ5 ® #4084<br />

(fluorescent DNA dye).<br />

Interferon-γ<br />

induces expression<br />

of PSMB8, a core<br />

particle subunit of the<br />

immunoproteasome.<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

60<br />

50<br />

40<br />

30<br />

1<br />

– +<br />

– +<br />

PSMB8/LMP7<br />

(precursor)<br />

PSMB8/LMP7<br />

(mature)<br />

GAPDH<br />

hIFN-γ<br />

PSMB8/LMP7 (1A5) Mouse mAb<br />

#13726: WB analysis of extracts from<br />

HeLa and SW620 cells, untreated or<br />

treated with Human Interferon-γ (hIFN-γ)<br />

#8901 (100 ng/ml, 72 hr), using<br />

#13726 (upper) and GAPDH (D16H11)<br />

XP ® Rabbit mAb #5174 (lower).<br />

2<br />

Lanes<br />

1. HeLa<br />

2. SW620<br />

UCHL1 is expressed in many cell lines<br />

and human non-small cell lung carcinoma.<br />

A<br />

kDa<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

50<br />

40<br />

30<br />

1 2 3 4 5 6 7 8 9 10 11 12 13 14<br />

Sumoylation and Neddylation<br />

Small ubiquitin-related modifier 1, 2, and 3 (SUMO-1, -2, and -3) and NEDD8 are members of the<br />

ubiquitin-like protein family. SUMO and NEDD8 can be covalently attached to proteins (termed<br />

sumoylation and neddylation, respectively) in a manner analogous to ubiquitination using an E1, E2, E3<br />

conjugation system. Unlike ubiquitination, however, sumoylation and neddylation of substrate proteins<br />

does not typically result in degradation. Instead, the SUMO and NEDD modifications affect subcellular<br />

localization, protein function, or protein-protein interactions.<br />

Sumoylation substrates include RanGAP, PML, p53, IκB-α, topoisomerase II, and APP. Sumoylation has<br />

numerous cellular effects including nuclear trafficking, regulation of transcriptional activity, and protein<br />

stability. SUMO modifications can be removed (desumoylation) by proteases such as SENP3, which<br />

catalyzes the release of SUMO2 and SUMO3 monomers from sumoylated substrates.<br />

Neddylation is the covalent attachment of NEDD8 via its C-terminal glycine residue to a lysine residue<br />

within the target protein. In contrast to the ubiquitin pathway, only a handful of neddylation substrates<br />

are known to date. NEDD8 and ubiquitin modifications are closely connected in that neddylation is an<br />

important regulator of E3 ubiquitin ligase activity. For example, neddylation of cullin proteins activates<br />

the SCF (Skp1-Cullin-F-box) E3 ubiquitin ligase complex by promoting complex formation and enhancing<br />

the recruitment of the E2-ubiquitin intermediate. Other neddylation substrates include p53, Mdm2,<br />

RPL11, and VHL.<br />

Proteasome<br />

The 26S proteasome is a highly abundant proteolytic complex involved in the degradation of ubiquitinated<br />

substrate proteins. It consists largely of two sub-complexes, the 20S catalytic core particle (CP)<br />

and the 19S/PA700 regulatory particle (RP) that can cap either end of the CP. The CP consists of two<br />

stacked heteroheptameric β-rings (β1-7) that contain three catalytic β-subunits and are flanked on either<br />

side by two heteroheptameric α-rings (α1-7). The RP includes a base and a lid, each having multiple<br />

subunits. The base, in part, is composed of a heterohexameric ring of ATPase subunits belonging<br />

to the AAA (ATPases Associated with diverse cellular Activities) family. The ATPase subunits function to<br />

unfold the substrate and open the gate formed by the α-subunits, thus exposing the unfolded substrate<br />

to the catalytic β-subunits. The lid consists of ubiquitin receptors and DUBs that function in recruitment<br />

of ubiquitinated substrates and modification of ubiquitin chain topology. Other modulators of<br />

proteasome activity, such as PA28/11S REG, can also bind to the end of the 20S CP and activate it.<br />

Proteasome activity can be inhibited by the chemical modulator bortezomib.<br />

Constitutively expressed core particle subunits PSMB5, PSMB7, and PSMB6 provide chymotrypsin-like,<br />

trypsin-like, and caspase-like activities, respectively. In immune cells involved in antigen presentation,<br />

these subunits are replaced by highly homologous, induced β-subunits PSMB8, PSMB9, and PSMB10<br />

to form the immunoproteasome. The immunoproteasome functions in degradation of proteins into fragments<br />

of the correct size for presentation on MHC class I molecules.<br />

UCHL1<br />

GAPDH<br />

B<br />

Select Reviews<br />

Amm, I., Sommer, T., and Wolf, D.H. (2014) Biochim. Biophys. Acta. 1843, 182−196. • Bhattacharyya, S., Yu, H., and Mim,<br />

C. (2014) Nat. Rev. Mol. Cell Biol. 15, 122−133. • Rabut, G. and Peter, M. (2008) EMBO Rep. 9, 969−976. • Raule, M.,<br />

Cerruti, F., and Cascio, P. (2014) Biochim. Biophys. Acta. 1843, 1942−1947. • Rieser, E., Cordier, S.M., and Walczak, H.<br />

(2013) Trends Biochem. Sci. 38, 94−102. • Ruggiano, A., Foresti, O., and Carvalho, P. (2014) J. Cell Biol. 204, 869−879.<br />

• Schulman, B.A. and Harper, J.W. (2009) Nat. Rev. Mol. Cell Biol. 10, 319−331. • Singhal, S., Taylor, M.C., and Baker, R.T.<br />

(2008) BMC Biochem. 9, S3. • Sriramachandran, A.M. and Dohmen, R.J. (2014) Biochim. Biophys. Acta. 1843, 75−85. •<br />

Zhao, Y., Brickner, J.R., and Majid, M.C. (2014) Trends Cell Biol. 24, 426−434.<br />

Commonly Studied Ubiquitin Targets<br />

Target M P S<br />

ADRM1<br />

• • •<br />

AMFR<br />

•<br />

APC1<br />

•<br />

APC2<br />

• •<br />

APC3<br />

• • •<br />

APC11<br />

• •<br />

BAP1<br />

•<br />

Phospho-BAP1 (Ser592)<br />

•<br />

β-Trcp<br />

•<br />

CAND1<br />

• • •<br />

c-Cbl<br />

• •<br />

Phospho-c-Cbl (Tyr700)<br />

•<br />

Phospho-c-Cbl (Tyr731)<br />

•<br />

Phospho-c-Cbl (Tyr774)<br />

•<br />

Cbl-b<br />

• • •<br />

UBC3<br />

•<br />

CHIP<br />

•<br />

COPS5<br />

• •<br />

CUL1<br />

•<br />

CUL3<br />

•<br />

CUL4A<br />

•<br />

CYLD<br />

• •<br />

Phospho-CYLD (Ser418)<br />

•<br />

DDB-1<br />

• •<br />

DDB-2<br />

•<br />

E2-25K/Hip2<br />

• •<br />

HAUSP<br />

• •<br />

HECTH9<br />

•<br />

ISG15<br />

• • •<br />

ITCH<br />

•<br />

KEAP1<br />

• • •<br />

KLHL12<br />

• •<br />

MIB1<br />

•<br />

NAE1/APPBP1<br />

•<br />

NEDD4<br />

• •<br />

NEDD4L<br />

•<br />

•<br />

Phospho-NEDD4L<br />

(Ser342)<br />

Target M P S<br />

Phospho-NEDD4L<br />

(Ser448) •<br />

NEDD8<br />

• •<br />

NPL4<br />

•<br />

OTUB1<br />

•<br />

OTULIN<br />

•<br />

PA28α<br />

• •<br />

PA28β<br />

•<br />

PA28γ<br />

•<br />

PSMA2<br />

• •<br />

PSMA3<br />

• •<br />

PSMA5<br />

•<br />

PSMA6<br />

•<br />

PSMB5<br />

• •<br />

PSMB6<br />

•<br />

PSMB7<br />

• •<br />

PSMB8/LMP7<br />

•<br />

PSMC3/TBP1<br />

•<br />

PSMC5/TRIP1<br />

•<br />

PSMD10/Gankyrin<br />

•<br />

PSMD11<br />

•<br />

PSMD14<br />

• •<br />

Rad23B<br />

•<br />

RBX1<br />

• •<br />

RCHY1<br />

•<br />

S5a/PSMD4<br />

• •<br />

SENP1<br />

•<br />

SENP3<br />

•<br />

Sharpin<br />

•<br />

Skp1<br />

• • •<br />

Skp2<br />

• • •<br />

SPINK3<br />

•<br />

STAMBP<br />

•<br />

SUMO-1<br />

• •<br />

SUMO-2/3<br />

•<br />

SYVN1<br />

• •<br />

TRIAD1<br />

• •<br />

TRIM25<br />

•<br />

Target M P S<br />

TRIM27<br />

•<br />

UBA2<br />

• • •<br />

UBC3B<br />

•<br />

Ubc9<br />

• •<br />

Ubc12<br />

• •<br />

Ubc13<br />

•<br />

UbcH5C<br />

•<br />

UBE1a<br />

•<br />

UBE1a/b<br />

•<br />

UBE1L2/UBA6<br />

•<br />

UBE2C<br />

•<br />

UBE2L3<br />

• •<br />

UBE2N<br />

•<br />

UBE2S<br />

• •<br />

UBE2T<br />

• •<br />

UBE3A<br />

•<br />

Ubiquitin<br />

• •<br />

• •<br />

K48-linkage Specific<br />

Polyubiquitin<br />

K63-linkage Specific<br />

Polyubiquitin<br />

UBLE1A/SAE1<br />

•<br />

•<br />

UBR5<br />

•<br />

UCHL1<br />

• • •<br />

UCHL3<br />

• • •<br />

USP1<br />

• •<br />

USP2<br />

•<br />

USP4<br />

•<br />

USP8<br />

• •<br />

USP9X<br />

• •<br />

USP10<br />

• • •<br />

USP13<br />

•<br />

USP14<br />

• • •<br />

USP18<br />

•<br />

VCP<br />

• •<br />

VHL<br />

•<br />

VPRBP<br />

•<br />

These protein targets represent key<br />

nodes within ubiquitin signaling<br />

pathways and are commonly studied in<br />

ubiquitin research. Primary antibodies,<br />

antibody conjugates, and antibody<br />

sampler kits containing these targets<br />

are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

S SignalSilence ® siRNA<br />

7<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for NEDD4 have been<br />

cited over 7 times in high-impact, peerreviewed<br />

publications from the global<br />

research community.<br />

Select Citations:<br />

Sun, Y. et al. (2014) Histone deacetylase<br />

5 blocks neuroblastoma cell differentiation<br />

by interacting with N-Myc.<br />

Oncogene 33, 2987−2994.<br />

Liu, P.Y. et al. (2013) The histone<br />

deacetylase SIRT2 stabilizes Myc<br />

oncoproteins. Cell Death Differ. 20,<br />

503−514.<br />

Adler, J.J. et al. (2013) Amot130<br />

adapts atrophin-1 interacting protein<br />

4 to inhibit yes-associated protein<br />

signaling and cell growth. J. Biol.<br />

Chem. 288, 15181−15193.<br />

Xia, P. et al. (2013) WASH inhibits<br />

autophagy through suppression of<br />

Beclin 1 ubiquitination. EMBO J. 32,<br />

2685−2696.<br />

Fu, J. et al. (2013) The short isoform<br />

of the ubiquitin ligase NEDD4L is a<br />

CREB target gene in hepatocytes.<br />

PLoS One 8, e78522.<br />

Andersen, M.N. et al. (2013) A<br />

phosphoinositide 3-kinase (PI3K)-<br />

serum- and glucocorticoid-inducible<br />

kinase 1 (SGK1) pathway promotes<br />

Kv7.1 channel surface expression by<br />

inhibiting Nedd4-2 protein. J. Biol.<br />

Chem. 288, 36841−36854.<br />

Liao, C.K. et al. (2013) Lipopolysaccharide<br />

induces degradation of<br />

connexin43 in rat astrocytes via the<br />

ubiquitin-proteasome proteolytic<br />

pathway. PLoS One 8, e79350.<br />

122 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstubiquitin 123


Section I: Research Areas<br />

chapter 04: Cell Biology<br />

Ubiquitin/Proteasome<br />

E1<br />

Ub<br />

DUBs<br />

Ub<br />

Ub<br />

Ub<br />

ATP<br />

AMP<br />

Ub Ub<br />

Ub<br />

Ub<br />

ATP<br />

ADP<br />

E1<br />

Ub Ub<br />

Ub<br />

Ub<br />

Ub<br />

Peptides<br />

Ub Ub<br />

Ub<br />

Ub<br />

E2<br />

DUBs<br />

E1<br />

Ub Ub<br />

Ub<br />

Ub<br />

E2<br />

Ub<br />

Ub Ub<br />

Ub<br />

Ub<br />

26S<br />

Proteasome<br />

Protein<br />

Substrate<br />

E3<br />

Ub Ub<br />

Ub<br />

Ub<br />

E2<br />

E3<br />

Ub<br />

Ub Ub<br />

Ub<br />

Ub<br />

Ub<br />

Mono-Ub<br />

Multiple<br />

Cycles<br />

Ub<br />

Ub<br />

Multi-Mono-Ub<br />

Endocytosis<br />

DNA Repair<br />

Protein Localization<br />

Trafficking<br />

Ub Ub<br />

Ub<br />

Ub<br />

Ub Ub<br />

Ub<br />

Ub<br />

K48 Poly-Ub K11 Poly-Ub K6 Poly-Ub K27 Poly-Ub K29 Poly-Ub K33 Poly-Ub K63 Poly-Ub Linear Poly-Ub<br />

19S<br />

20S<br />

19S<br />

DNA Damage<br />

Response<br />

TCR Signaling<br />

Lysomal<br />

Degradation<br />

Mitochondrial<br />

Maintenance and<br />

Mitophagy<br />

Activating NF-κB<br />

Signaling<br />

The ubiquitin proteasome pathway, conserved from yeast to mammals, is required for the targeted degradation of most shortlived<br />

proteins in the eukaryotic cell. Targets include cell cycle regulatory proteins, whose timely destruction is vital for controlled<br />

cell division, as well as proteins unable to fold properly within the endoplasmic reticulum.<br />

Ubiquitin modification is an ATP-dependent process carried out by three classes of enzymes. A “ubiquitin activating enzyme” (E1)<br />

forms a thio-ester bond with ubiquitin, a highly conserved 76-amino acid protein. This reaction allows subsequent binding of<br />

ubiquitin to a “ubiquitin conjugating enzyme” (E2), followed by the formation of an isopeptide bond between the carboxy-terminus<br />

of ubiquitin and a lysine residue on the substrate protein. The latter reaction requires a “ubiquitin ligase” (E3). E3 ligases can<br />

be single- or multi-subunit enzymes. In some cases, the ubiquitin-binding and substrate binding domains reside on separate<br />

polypeptides brought together by adaptor proteins or cullins. Numerous E3 ligases provide specificity in that each can modify only<br />

a subset of substrate proteins. Further specificity is achieved by post-translational modification of substrate proteins, including,<br />

but not limited to, phosphorylation.<br />

Effects of monoubiquitination include a role in endocytosis and DNA damage, as well as changes in subcellular protein localization<br />

and trafficking. However, multiple ubiquitination cycles resulting in a polyubiquitin chain are required for targeting a protein<br />

to the proteasome for degradation. The multisubunit 26S proteasome recognizes, unfolds, and degrades polyubiquitinated<br />

substrates into small peptides. The reaction occurs within the cylindrical core of the proteasome complex, and peptide bond<br />

hydrolysis employs a core threonine residue as the catalytic nucleophile. Polyubiquitin chains are also indicated in diverse cellular<br />

processes including DNA damage response, mitochondrial maintenance and mitophagy, lysosomal degradation, T Cell Receptor<br />

signaling, and NF-κB signaling.<br />

Ubiquitinating enzymes (UBEs) catalyze protein ubiquitination, a reversible process countered by deubiquitinating enzyme (DUB)<br />

action. Five DUB subfamilies are recognized, including the USP, UCH, OTU, MJD, and JAMM enzymes. In humans, there are three<br />

proteasomal DUBs: PSMD14 (POH1/RPN11), UCH37 (UCH-L5), and Ubiquitin-Specific Protease 14, which is also known as the<br />

60 kDa subunit of tRNA-guanine transglycosylase (USP14/TGT60 kDa).<br />

Select Reviews:<br />

Amm, I., Sommer, T., and Wolf, D.H. (2014) Biochim. Biophys. Acta. 1843, 182–196. • Budhidarmo, R., Nakatani, Y., and<br />

Day, C.L. (2012) Trends Biochem. Sci. 37, 58–65. • Burrows, J.F. and Johnston, J.A. (2012) Front. Biosci. 17, 1184–1200.<br />

• Campello, S., Strappazzon, F., and Cecconi, F. (2014) Biochim. Biophys. Acta. 1837, 451–460. • Corn, J.E. and Vucic, D.<br />

(2014) Nat. Struc. Mol. Biol. 21, 297–300. • Hammond-Martel, I., Yu, H., Affar, el B. (2012) Cell Signal. 24, 410–421. • Hildebrand,<br />

J.M., Yi, Z., Buchta, C.M., Poovassery, J., Stunz, L.L., and Bishop, G.A. (2011) Immunol. Rev. 244, 55–74. • Hurley,<br />

J.H. and Schulman, B.A. (2014) Cell 157, 300–311. • Ruggiano, A., Foresti, O., and Carvalho, P. (2014) J. Cell Biol. 204,<br />

869–879. • Schaefer, A., Nethe, M., and Hordijk, P.L. (2012) Biochem. J. 442, 13–25. • Tokunaga, F. (2013) J. Biochem.<br />

154, 313–323. • Weissman, A.M., Shabek, N., and Ciechanover, A. (2011) Nat. Rev. Mol. Cell Biol. 12, 605–620. • Zhao, Y.,<br />

Brickner, J.R., Majid, M.C., and Mosammaparast, N. (2014) Trends Cell Biol. 24, 426–434.<br />

Ligase Substrate Function PMID<br />

AMFR KAI1 AMFR is also known as gp78. AMFR is an integral ER membrane protein and<br />

functions in ER-associated degradation (ERAD). AMFR has been found to<br />

promote tumor metastasis through ubiquitination of the metastasis suppressor,<br />

KAI1.<br />

18037895<br />

APC/CDC20 Cyclin B,<br />

Securin<br />

APC/Cdh1<br />

CDC20, Cyclin<br />

B, Cyclin A,<br />

Aurora A, Skp2,<br />

Claspin<br />

The anaphase promoting complex/cyclosome (APC/C) is a multiprotein<br />

complex with E3 ligase activity that regulates cell cycle progression through<br />

degradation of cyclins and other mitotic proteins. APC is found in a complex<br />

with CDC20, CDC27, SPATC1, and TUBG1.<br />

The anaphase promoting complex/cyclosome (APC/C) is a multiprotein<br />

complex with E3 ligase activity that regulates cell cycle progression through<br />

degradation of cyclins and other mitotic proteins. The APC/C-Cdh1 dimeric<br />

complex is activated during anaphase and telophase, and remains active until<br />

onset of the next S phase.<br />

ARIH1 4EHP ARIH1 is an E3 ubiquitin ligase that may regulate protein translation by targeting<br />

eIF4E2 for ubiquitination and degradation by the proteasome.<br />

BIRC2 Smac, TRAF2 BIRC2 is an apoptotic suppressor that prevents caspase activation by forming<br />

a complex with TNF receptor associated factors 1 and 2 (TRAF1 and TRAF2),<br />

which is then recruited to the tumor necrosis factor receptor 2 (TNFR2).<br />

BIRC3 Caspase 3<br />

and 7, Smac,<br />

TRAF1<br />

BIRC4 Caspase 3,<br />

Smac, MEKK2<br />

BIRC3 is an apoptotic suppressor that prevents caspase activation by forming<br />

a complex with TNF receptor associated factors 1 and 2 (TRAF1 and TRAF2),<br />

which is then recruited to the tumor necrosis factor receptor 2 (TNFR2).<br />

BIRC4 is an apoptotic suppressor that prevents caspase activation by forming<br />

a complex with TNF receptor associated factors 1 and 2 (TRAF1 and TRAF2),<br />

which is then recruited to the tumor necrosis factor receptor 2 (TNFR2). BIRC4<br />

is also known as XIAP.<br />

BIRC7 Smac BIRC7 is an E3 ubiquitin ligase with anti-apoptotic activity. BIRC7 supports<br />

cell survival by targeting Smac for ubiquitination and degradation by the<br />

proteasome.<br />

Bmi1 H2A K119 Bmi1 is a component of the polycomb group multiprotein PRC1- like (PcG<br />

PRC1) complex. Bmi1 is required for stimulating PcG PRC1 ubiquitin-protein<br />

ligase activity.<br />

BRCA1<br />

ER-a, Rpb8,<br />

CtIP, FANCD2<br />

BRCA1 is an E3 ubiquitin ligase that maintains genomic stability by repairing<br />

DNA damage. Research studies have shown that mutations of this gene have<br />

been linked to breast cancer.<br />

C6orf157 Cyclin B C6orf157 is also known as H10BH. C6orf157 is an E3 ubiquitin ligase that has<br />

been shown to ubiquitinate cyclin B.<br />

Cbl<br />

Cbl-b and c-Cbl are members of the Cbl family of adaptor proteins that are<br />

highly expressed in hematopoietic cells. Cbl proteins possess E3 ubiquitin<br />

ligase activity that downregulates numerous signaling proteins and RTKs in several<br />

pathways such as EGFR, T cell and B cell receptors, and integrin receptors.<br />

Cbl proteins play an important role in T cell receptor signaling pathways.<br />

CBLL1 CDH1 CBLL1 is also known as Hakai. CBLL1 is an E3 ubiquitin ligase that ubiquitinates<br />

the phosphorylated form of E-Cadherin, causing its degradation and loss<br />

of cell-cell adhesions.<br />

CHFR PLK1, Aurora A CHFR is an E3 ubiquitin ligase that functions as a mitotic stress checkpoint<br />

protein that delays entry into mitosis in response to stress. CHFR has been<br />

shown to ubiquitinate and degrade the kinases PLK1 and Aurora A.<br />

CHIP<br />

CUL3/<br />

BACURD<br />

CUL3/HIB/<br />

SPOP<br />

HSP70/90,<br />

iNOS, Runx1,<br />

LRRK2<br />

RhoA<br />

Ci/Gli<br />

CHIP is an E3 ubiquitin ligase that acts as a co-chaperone protein and interacts<br />

with several heat shock proteins, including HSP70 and HSP90, as well as<br />

the nonheat shock proteins iNOS, Runx1, and LRRK2.<br />

CUL3/BACURD is a ubiquitin ligase complex composed of CUL3 and the BTB<br />

domain adaptor BACURD. CUL3/BACURD controls actin cytoskeleton structure<br />

and cell movement by promoting ubiquitination and degradation of small<br />

GTPase RhoA.<br />

CUL3/HIB/SPOP is an E3 ubiquitin ligase complex composed of Cullin3,<br />

Hedgehog-induced MATH and BTB domain-containing protein (HIB), and SPOP.<br />

CUL3/HIB/SPOP targets the Hedgehog pathway transcription factor (Ci)/Gli for<br />

ubiquitination and degradation by the proteasome.<br />

CUL3/KEAP1 Nrf2, IKKβ CUL3/KEAP1 is part of an E3 ubiquitin ligase complex composed of RBX1,<br />

CULlin3 and the substrate recognition component, KEAP1. CUL3/KEAP1<br />

targets Nrf2, a transcription factor that regulates antioxidant genes in response<br />

to oxidative stress for ubiquitination and degradation by the proteasome. In<br />

addition, CUL1/KEAP1 E3 ligase downregulates NF-κB signaling by targeting<br />

IKKβ ubiquitination.<br />

CUL3/<br />

MEL-26<br />

mei-1<br />

CUL3/MEL-26 is an E3 ubiquitin ligase complex composed of Cullin3 and the<br />

substrate recognition component, MEL-26. MEL-26 targets mei-1 for ubiquitination<br />

and subsequent proteasomal degradation.<br />

17609108,<br />

12070128<br />

10548110,<br />

11562349,<br />

15014503,<br />

19477924<br />

14623119<br />

12525502,<br />

18434593<br />

10862606,<br />

12525502,<br />

15468071<br />

11447297,<br />

12121969,<br />

18761086<br />

16729033<br />

18650381<br />

17392432,<br />

17283126,<br />

16818604,<br />

11239454<br />

15749827<br />

18759930,<br />

9797470<br />

11836526<br />

14562038,<br />

19326084<br />

19913553,<br />

19362296,<br />

19524548,<br />

19536328<br />

19782033<br />

16740475<br />

12682069,<br />

19818716<br />

13679922<br />

Ubiquitin Ligases<br />

This table provides a list of E3 ubiquitin<br />

ligases, along with their substrates<br />

(when known), and corresponding<br />

references. This table was generated<br />

using PhosphoSitePlus ® , Cell Signaling<br />

Technology’s protein modification<br />

resource.<br />

© 2004–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Wenyi Wei, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA for reviewing this diagram.<br />

124 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables 125


Section I: Research Areas<br />

chapter 04: Cell Biology<br />

Ligase Substrate Function PMID<br />

CUL3/Ctb9/<br />

KLHDC5<br />

p60/katanin CUL3/Ctb9/KLHDC5 is an E3 ubiquitin ligase complex required for efficient<br />

p60/katanin removal through promoting ubiquitination of p60/katanin to allow<br />

normal mitotic progression in mammalian cells.<br />

19261606<br />

CUL3/KLHL3 WNK1, WNK4 CUL3/KLHL3 is an E3 ubiquitin ligase complex composed of Cullin3 and kelchlike<br />

3 (KLHL3) protein. WNK4 is a known target of CUL3/KLHL3-mediated<br />

ubiquitination and errors in that process are a common mechanism of human<br />

hereditary hypertension. Furthermore, mutations in KLHL3 and Cullin3 were<br />

identified to cause the human hypertensive disease pseudohypoaldosteronism<br />

type II (PHAII).<br />

CUL3/KLHL8 Rapsyn CUL3/KLHL8, an E3 ubiquitin ligase complex, controls rapsyn stability through<br />

polyubiquitination, which is required for clustering of nicotinic acetylcholine<br />

receptors (nAChRs) at the neuromuscular junction.<br />

CUL3/<br />

KLHL12<br />

CUL3/<br />

KLHL9/<br />

KLHL13<br />

CUL3/<br />

KLHL20<br />

CUL3/<br />

KLHL22<br />

CUL3/<br />

KLHL25<br />

CUL3/SPOP<br />

Dsh<br />

Aurora B<br />

PML, DAPK<br />

PLK1<br />

4E-BP1<br />

Gli2/Gli3, Daxx,<br />

SRC-3, AR,<br />

PTEN<br />

CUL4/CDT2 Cdt1, p21,<br />

Set8, CHK1<br />

CUL4/DDB1/<br />

Cereblon<br />

IKZF1, IKZF3<br />

CUL4/COP1 c-Jun, p53,<br />

ETV1, ETV4,<br />

ETV5<br />

CUL3/KLHL12 E3 ubiquitin ligase targets Dishevelled for poly-ubiquitination<br />

and degradation, thus negatively regulating the Wnt/β-catenin pathway.<br />

CUL3/KLHL9/KLHL13 E3 ubiquitin ligase controls the dynamic behavior of<br />

mitotic chromosomes through ubiquitination of Aurora B, thereby coordinating<br />

mitotic progression and completion of cytokinesis.<br />

CUL3/KLHL20 E3 ubiquitin ligase mediates hypoxia-induced PML proteasomal<br />

degradation via activation by HIF-1. The CUL3/KLHL20 complex also controls<br />

interferon responses by promoting DAPK polyubiquitination and proteasomal<br />

degradation.<br />

CUL/KLHL22 regulates localization of PLK1 on the kinetochore thereby controlling<br />

spindle assembly checkpoint (SAC) activation. Ubiquitination of PLK1<br />

signals degradation-independent removal from kinetochores and fulfillment of<br />

SAC leading to mitotic progression.<br />

CUL3/KLHL25 E3 ubiquitin ligase complex targets hypophosphorylated 4E-BP1<br />

for degradation. Regulation of 4E-BP1 protein levels by CUL3/KLHL25 ubiquitination<br />

induces homeostatic control over the mRNA binding protein, eIF4E, for<br />

which 4E-BP1 acts as a repressor protein.<br />

The CUL3/SPOP E3 ubiquitin ligase displays both tumor suppressor and<br />

oncogenic functions in several types of human tissue. CUL3/SPOP regulates<br />

the proteolysis of the oncogene SRC-3, where underexpression of CUL3/<br />

SPOP leads to overexpression of SRC-3 in prostate cancer. Inversely, SPOP, a<br />

direct transcriptional target of HIFs in some tissues, is overexpressed in 85%<br />

of kidney cancers. The opposing roles of SPOP protein in prostate and kidney<br />

cancers may result from degradation of different substrates, such as AR in<br />

prostate cancers and PTEN in kidney cancer.<br />

CUL4/CDT2 is an E3 ubiquitin ligase complex composed of DCX (DDB1-CUL4-<br />

X-box) and the substrate recognition component, CUL4/CDT2. CUL4/CDT2<br />

regulates cell cycle progression into S phase by targeting CDT1 and SPD1 for<br />

ubiquitination and degradation by the proteasome.<br />

Cereblon forms an E3 ubiquitin ligase complex with CUL4 and DDB1 to regulate<br />

limb development. Lenalidomide-bound Cereblon targets IKZF1 and IKZF3<br />

for ubiquitination and degradation, helping to prevent B cell malignancies.<br />

CUL4/COP1 is an E3 ubiquitin ligase that mediates ubiquitination and subsequent<br />

proteasomal degradation of target proteins. COP1 targets the oncoprotein<br />

c-Jun, transcription factor ETV1 and may target the tumor suppressor p53<br />

for ubiquitination and degradation.<br />

CUL4/DDB2 XPC, H3, H4 CUL4/DDB2 is an E3 ubiquitin ligase complex composed of DCX (DDB1-CUL4-<br />

ROC1) and the substrate recognition component, CUL4/DDB2. CUL4/DDB2<br />

may target histone H2A, histone H3, and histone H4 at sites of UV-induced<br />

DNA damage to induce ubiquitination and degradation by the proteasome.<br />

CUL4/FBW5 TSC2 CUL4/FBW5 is an E3 ubiquitin ligase complex composed of DCX (DDB1-CUL4-<br />

ROC1) and the substrate recognition component, CUL4/FBW5. CUL4/FBW5<br />

regulates TSC2 protein stability and TSC complex turnover.<br />

CUL4/β-TrCP REDD1 CUL4/β-TrCP E3 ligase complex contains DCX (DDB1-CUL4-ROC1) and β-TrCP.<br />

This complex targets REDD1 for ubiquitination and subsequent proteasomal<br />

degradation to re-activate the mTOR signaling pathway as cells recover from<br />

hypoxic stress.<br />

CUL4/RBBP7 p150 CUL4/RBBP7 is an E3 ubiquitin ligase complex composed of DCX (DDB1-<br />

CUL4-ROC1) and the substrate recognition component, RBBP7. CUL4/RBBP7<br />

targets p150 for ubiquitination and degradation.<br />

CUL4/DDB1/<br />

TRCP4A<br />

CUL4/VPRBP TET<br />

N-Myc, C-Myc<br />

CUL4/TRCP4A is an E3 ubiquitin ligase complex composed of DDB1-CUL4 and<br />

the substrate recognition component, TRCP4A. CUL4/DDB1/TRCP4A targets<br />

Myc for ubiquitination and degradation.<br />

CUL4/VPRBP is an E3 complex consisting of CRL4 (DDB1-CUL4-RBX1), and<br />

VPRBP (DCAF1). CRL4/VPRBP is essential for regulating mammalian oocyte<br />

survival and reprogramming via activation of TET methylcytosine dioxygenases.<br />

23387299,<br />

23453970<br />

19158078<br />

16547521<br />

17543862<br />

21840486,<br />

20389280<br />

23455478<br />

22578813<br />

16524876,<br />

19684112,<br />

21577200,<br />

24508459,<br />

24656772<br />

16949367,<br />

18794347,<br />

20932471,<br />

23109433<br />

20223979,<br />

24292623<br />

12615916,<br />

16931761,<br />

21572435,<br />

20062082,<br />

21572435<br />

15882621,<br />

16678110<br />

18381890<br />

19557001<br />

21228219<br />

20551172<br />

24357321<br />

Ligase Substrate Function PMID<br />

CUL5/SOCS1 Dab1 CUL5/SOCS1 is part of an SCF-like ECS (Elongin BC-CUL2/5-SOCS-box 17974915<br />

protein) E3 ubiquitin ligase complex. CUL5/SOCS1 targets components of the<br />

Jak/Stat pathway as well as Dab1, a regulator of cortical development, for<br />

ubiquitination and degradation by the proteasome.<br />

CUL5/SOCS4 EGFR CUL5/SOCS4 is part of an SCF-like ECS (Elongin BC-CUL2/5-SOCS-box protein)<br />

17997974<br />

E3 ubiquitin ligase complex. SOCS4 may target components of cytokine<br />

signal transduction pathways, such as EGF receptor (EGFR) for ubiquitination<br />

and degradation by the proteasome.<br />

CUL5/Vif APOBEC3G CUL5/Vif is part of an SCF-like ECS (Elongin BC-CUL2/5-SOCS-box protein) E3 15574592<br />

ubiquitin ligase complex. Vif targets APOBEC3G and APOBEC3F for ubiquitination<br />

and degradation by the proteasome. The interaction of Vif with APOBEC3G<br />

also blocks its cytidine deaminase activity in a proteasome-independent<br />

manner,<br />

CUL7/ cyclin D1 CUL7/FBXW8 is an SCF-like E3 ubiquitin ligase complex composed of SKP1, 17205132<br />

FBXW8<br />

CUL7, RBX1, GLMN isoform 1, and the substrate recognition component,<br />

FBXW8.<br />

DZIP3 H2AK119 DZIP3 is an E3 ubiquitin ligase that blocks transcriptional elongation by 12538761<br />

ubiquinating H2A at lysine 119.<br />

E6-AP p53, Dlg E6-AP is also known as UBE3A. E6-AP is a HECT domain E3 ubiquitin ligase 17108031<br />

that interacts with Hepatitis C virus (HCV) core protein and targets it for<br />

degradation. The HCV core protein is central to packaging viral DNA and other<br />

cellular processes. E6-AP also interacts with the E6 protein of the human<br />

papillomavirus types 16 and 18, and targets the p53 tumor-suppressor protein<br />

for degradation.<br />

FANCL FANC D2 FANCL is an ubiquitin ligase protein integral to the DNA repair pathway. 12973351<br />

HACE1<br />

HACE1 is an E3 ubiquitin ligase and tumor suppressor. Research has shown 17694067<br />

that aberrant methylation of HACE1 is frequently found in Wilms’ tumors and<br />

colorectal cancer.<br />

HECTD1<br />

HECTD1 is an ubiquitin E3 ligase required for neural tube closure and normal 17442300<br />

development of the mesenchyme.<br />

HECTD2<br />

HECTD2 is a probable E3 ubiquitin ligase and may act as a susceptibility gene 19214206<br />

for neurodegeneration and prion disease.<br />

HECTD3<br />

HECTD3 is a probable E3 ubiquitin ligase and may play a role in cytoskeletal<br />

regulation, actin remodeling, and vesicle trafficking.<br />

18194665<br />

HECW1<br />

DVL1, mutant<br />

SOD1, p53<br />

HECW1 is also known as NEDL1. HECW1 interacts with p53 and the Wnt<br />

signaling protein DVL1, and may play a role in p53-mediated cell death in<br />

neurons.<br />

HECW2 p73 HECW2 is also known as NEDL2. HECW2 ubiquitinates p73, which is a p53<br />

family member. Ubiquitination of p73 increases protein stability.<br />

HERC2 RNF8 HERC2 belongs to a family of E3 ubiquitin ligases involved in membrane<br />

trafficking events. HERC2 plays a role in the DNA damage response through<br />

interaction with RNF8.<br />

HERC3<br />

HERC3 belongs to a family of E3 ubiquitin ligases involved in membrane trafficking<br />

events. HERC3 interacts with hPLIC-1 and hPLIC-2 and localizes to the<br />

late endosomes and lysosomes.<br />

HERC4<br />

HERC4 belongs to a family of E3 ubiquitin ligases involved in membrane<br />

trafficking events. HERC4 is highly expressed in testis and may play a role in<br />

spermatogenesis.<br />

HERC5<br />

HERC5 belongs to a family of E3 ubiquitin ligases involved in membrane<br />

trafficking events. HERC5 is induced by interferon and other pro-inflammatory<br />

cytokines and plays a role in interferon-induced ISG15 conjugation during the<br />

innate immune response.<br />

HLTF PCNA HLTF is both a helicase and an E3 ubiquitin ligase. HLTF participates in<br />

postreplication repair (PRR) of damaged DNA by polyubiquitination of<br />

chromatin-bound PCNA.<br />

HOIP PKC HOIP is the E3 ubiquitin ligase of the LUBEC (linear ubiquitin chain assembly<br />

complex) which ubiquitinates signaling proteins, targeting them for proteasomal<br />

degradation.<br />

HUWE1<br />

N-Myc, C-Myc,<br />

p53, Mcl-1,<br />

TopBP1<br />

HUWE1 is also known as Mule. HUWE1 is a HECT domain E3 ubiquitin ligase<br />

that regulates degradation of Mcl-1 and therefore regulates DNA damage-induced<br />

apoptosis. HUWE1 also controls neuronal differentiation by destabilizing<br />

N-Myc, and regulates p53-dependent and independent tumor suppression<br />

via ARF.<br />

PhosphoSitePlus®<br />

A comprehensive online protein modification resource. www.cellsignal.com/exploration<br />

14684739,<br />

18223681<br />

12890487<br />

20023648<br />

18535780<br />

17967448<br />

16407192,<br />

16815975<br />

18316726<br />

17069764<br />

15989957<br />

126 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables<br />

127


Section I: Research Areas<br />

chapter 04: Cell Biology<br />

Ligase Substrate Function PMID<br />

HYD CHK2 HYD is also known as EDD or UBR5. HYD is a regulator of the DNA damage 18073532<br />

response and is overexpressed in many forms of cancer.<br />

IBRDC2 p21, Bax IBRDC2 is an E3 ubiquitin ligase involved in the regulation of apoptosis. 12853982<br />

IBRDC2 expression can be induced by p53 and may target apoptosis related<br />

proteins p21 and Bax.<br />

IBRDC3 UCKL-1 IBRDC3 is an E3 ubiquitin ligase involved in the cytolytic activities of hematopoietic<br />

16709802<br />

natural killer cells and T cells.<br />

ITCH MKK4, RIP2,<br />

Foxp3<br />

ITCH plays a role in T cell receptor activation and signaling through ubiquitination<br />

of multiple proteins including MKK4, RIP2, and Foxp3. Loss of ITCH function<br />

leads to an aberrant immune response and T helper cell differentiation.<br />

LNX1 NUMB LNX1 is an E3 ubiquitin ligase that plays a role in cell fate determination during<br />

embryogenesis through regulation of NUMB, the negative regulator of Notch<br />

signaling.<br />

LRSAM1 Tsg101 LRSAM is an E3 ubiquitin ligase that mediates intracellular vesicular trafficking<br />

by monoubiquitination of TSG101.<br />

Mahogunin<br />

Mahogunin is an E3 ubiquitin ligase involved in melanocortin signaling. Loss of<br />

mahogunin function leads to neurodegeneration and loss of pigmentation, and<br />

may be the mechanism of action in prion disease.<br />

19737936,<br />

19592251,<br />

20108139<br />

11782429<br />

15256501<br />

19737927,<br />

19524515<br />

MALIN laforin Malin, also known as NHLRC1, is an E3 ubiquitin ligase that promotes the 15930137<br />

ubiquitination and proteasomal degradation of misfolded proteins.<br />

MARCH-I HLA-DRβ MARCH1 is an E3 ubiquitin ligase found on antigen presenting cells (APCs). 19880452<br />

MARCH1 ubiquitinates MHC class II proteins and downregulates their cell<br />

surface expression.<br />

MARCH-II<br />

MARCH-II is a member of the MARCH family of E3 ubiquitin ligases. It associates<br />

15689499<br />

with syntaxin6 in the endosomes and helps to regulate vesicle trafficking.<br />

MARCH-III<br />

MARCH-III is a member of the MARCH family of E3 ubiquitin ligases. MARCH- 16428329<br />

III associates with syntaxin6 in the endosomes and helps to regulate vesicle<br />

trafficking.<br />

MARCH-IV MHC class I MARCH-IV is a member of the MARCH family of E3 ubiquitin ligases. MARCH- 14722266<br />

IV ubiquitinates MHC class I proteins and downregulates their cell surface<br />

expression.<br />

MARCH-V DRP1 MARCH-V is a member of the MARCH family of E3 ubiquitin ligases. March-V 16936636<br />

is located in the mitochondria and aids in the control of mitochondrial morphology.<br />

MARCH-VI<br />

MARCH-VI is also known as TEB4 and is a member of the MARCH family of E3 16373356<br />

ubiquitin ligases. It localizes to the endoplasmic reticulum and participates in<br />

ER-associated protein degradation.<br />

MARCH-VII gp190 MARCH-VII is also known as axotrophin. MARCH-VII was originally identified as 19901269<br />

a neural stem cell gene, but has since been shown to play a role in LIF signaling<br />

in T lymphocytes through degradation of the LIF receptor subunit, gp190.<br />

MARCH-VIII<br />

B7-2, MHC<br />

class II<br />

MARCH-VIII is also known as c-MIR. MARCH-VIII causes the ubiquitination/<br />

degradation of B7-2, which is a co-stimulatory molecule for antigen presentation.<br />

MARCH-VIII has also been shown to ubiquitinate MHC class II proteins.<br />

MARCH-IX ICAM-1, MHC-I MARCH-IX is a member of the MARCH family of E3 ubiquitin ligases. MARCH-<br />

IX mediates ubiquitination of transmembrane proteins, marking them for<br />

endocytosis and sorting to lysosomes via multivesicular bodies.<br />

MARCH-X<br />

MARCH-X is also known as RNF190. MARCH-X is a member of the MARCH<br />

family of E3 ubiquitin ligases. MARCH-X may be involved in spermiogenesis.<br />

MARCH-XI CD4 MARCH-XI is a member of the MARCH family of E3 ubiquitin ligases. MARCH-<br />

IX mediates ubiquitination of CD4, marking it for endocytosis and sorting to<br />

lysosomes via multivesicular bodies.<br />

MDM2 p53 MDM2, an E3 ubiquitin ligase for p53, plays a central role in regulation of<br />

the stability of p53. Akt-mediated phosphorylation of MDM2 at Ser166 and<br />

Ser186 increases its interaction with p300, allowing MDM2-mediated ubiquitination<br />

and degradation of p53.<br />

MEKK1 c-Jun, Erk MEKK1 is a well known protein kinase of the STE11 family. MEKK1 phosphorylates<br />

and activates MKK4/7, which in turn activates JNK1/2/3. MEKK1<br />

contains a RING finger domain and exhibits E3 ubiquitin ligase activity toward<br />

c-Jun and Erk.<br />

MGRN1 Tsg101 MGRN1 is an E3 ubiquitin ligase that mediates intracellular vesicular trafficking<br />

by monoubiquitination of TSG101.<br />

MIB1 Delta, Jagged Mindbomb homolog 1 (MIB1) is an E3 ligase that facilitates the ubiquitination<br />

and subsequent endocytosis of the Notch ligands, Delta and Jagged.<br />

MIB2 Delta, Jagged Mindbomb 2 (MIB2) is an E3 ligase that positively regulates Notch Signaling.<br />

MIB2 has been shown to play a role in myotube differentiation and muscle<br />

stability. MIB2 ubiquitinates NMDAR subunits to help regulate synaptic plasticity<br />

in neurons.<br />

MID1 PP2A Mid1, also known as Midline-1, is an E3 ubiquitin ligase that may target<br />

protein phosphatase 2 for ubiquitination and proteasomal degradation.<br />

16785530<br />

17174307,<br />

14722266<br />

21937444<br />

17604280<br />

9153395<br />

12049732,<br />

17101801<br />

17229889<br />

16000382<br />

15824097,<br />

18216171,<br />

17962190<br />

11685209<br />

Ligase Substrate Function PMID<br />

MKRN1 hTERT, p53, MKRN1 is an E3 ubiquitin ligase that regulates both anti- and pro-apoptotic 15805468<br />

CDKN1A, FLIP1 functions.<br />

MycBP2 Fbxo45, TSC2 MycBP2 is an E3 ubiquitin ligase also known as PAM. MycBP2 associates with<br />

Fbxo45 to play a role in neuronal development. MycBP2 also regulates the<br />

mTOR pathway through ubiquitination of TSC2.<br />

NEDD4<br />

NEDD4 is an E3 ubiquitin ligase highly expressed in the early mouse embryonic<br />

central nervous system. NEDD4 downregulates both neuronal voltagegated<br />

Na+ channels (NaVs) and epithelial Na+ channels (ENaCs) in response<br />

to increased intracellular Na+ concentrations.<br />

NEDD4L Smad2, PTEN NEDD4L is an E3 ubiquitin ligase highly expressed in the early mouse embryonic<br />

central nervous system. NEDD4L has been shown to negatively regulate<br />

TGF-β signaling by targeting Smad2 for degradation.<br />

NEURL Jagged 1, Delta NEURL is an E3 ubiquitin ligase involved in Notch signaling and neurological<br />

determination of cell fate.<br />

OSTM1 Gai3 OSTM1 is an E3 ubiquitin ligase localized to the cell membrane that<br />

regulates membrane associated G-proteins by ubiquitination and<br />

proteasomal degradation.<br />

PARC<br />

Parkin<br />

Pael-R,<br />

CDC-rel,<br />

PLC-g1,MFN1<br />

PARC is a cullin family member that acts as a p53-binding cytoplasmic<br />

anchor protein and is part of an atypical cullin-RING- based E3 ubiquitin ligase<br />

complex.<br />

Parkin is an E3 ubiquitin ligase that has been shown to be a key regulator of<br />

the autophagy pathway. Mutations in Parkin can lead to Parkinson’s Disease.<br />

PCGF1 H2A, K119 PCGF1 is a component of the polycomb group multiprotein PRC1- like (PcG<br />

PRC1) complex. PCGF1 is required for PcG PRC1 mediated monoubiquitination<br />

of H2A Lys119, which is central to the histone code and gene regulation.<br />

PELI1 TRIP, IRAK PELI1 is an E3 ubiquitin ligase that plays a role in Toll-like Receptor (TLR3 and<br />

TLR4) signaling to NF-κB via the TRIP adaptor protein. PELI1 has also been<br />

shown to ubiquitinate IRAK.<br />

PEX10 Pex5 PEX10 is localized to peroxisome membranes and has been associated with<br />

several peroxisomal biogenesis disorders.<br />

PJA1 ELF PJA1 is also known as PRAJA. PJA1 plays a role in downregulating TGF-β<br />

signaling in gastric cancer via ubiquitination of the Smad4 adaptor protein ELF.<br />

PJA2<br />

PJA2 is an E3 ubiquitin ligase found in neuronal synapses. The exact role and<br />

substrates of PJA2 are unclear.<br />

RAD18 PCNA RAD18 is an E3 ubiquitin ligase involved in post-replication repair of UVdamaged<br />

DNA.<br />

RBCK1<br />

SOCS6, PKC,<br />

TAB2/3<br />

RBCK1 is an E3 ligase that acts as an iron sensor by promoting the ubiquitination<br />

of oxidized IREB2 in the presence of high iron and oxygen. RBCK1 is a<br />

component of the LUBAC (linear ubiquitin chain assembly complex).<br />

RCHY1 p27 Kip1, p53 RCHY1, also known as Pirh2, is an E3 ubiquitin ligase that contributes to the<br />

regulation of the cell cycle. RCHY1 is primarily associated with the ubiquitination<br />

and proteasomal degradation of tetrameric p53.<br />

RFFL p53 RFFL is also known as CARP2 and is an E3 ubiquitin ligase that inhibits endosome<br />

recycling. RFFL also degrades p53 through stabilization of MDM2.<br />

RFWD2 MTA1, p53,<br />

FoxO1<br />

RFWD2 is also known as COP1. RFWD2 is an E3 ubiquitin ligase that ubiquitinates<br />

several proteins involved in the DNA damage response and apoptosis<br />

including MTA1, p53, and FoxO1.<br />

Rictor SGK1 Rictor interacts with CULlin1-Rbx1 to form an E3 ubiquitin ligase complex and<br />

promotes ubiquitination and degradation of SGK1.<br />

RING1 H2A, K119 RING1, also known as RNF1, is an E3 ubiquitin ligase of the polycomb group<br />

multiprotein PRC1-like (PcG PRC1) complex. RING1 is required for PcG PRC1<br />

mediated monoubiquitination of H2A Lys119, which is central to the histone<br />

code and gene regulation.<br />

RNF2 H2A, K119,<br />

Geminin<br />

RNF2, also known as Ring2, is an E3 ubiquitin ligase of the polycomb group<br />

multiprotein PRC1-like (PcG PRC1) complex. RNF2 is required for PcG PRC<br />

mediated monoubiquitination of H2A Lys119, which is central to the histone<br />

code and gene regulation.<br />

RNF5 JAMP, paxillin RNF5 is also known as RMA5. RNF5 plays a role in ER-associated degradation<br />

of misfolded proteins and ER stress response through ubiquitination of JAMP.<br />

RNF5 also plays a role in cell motility and has been shown to ubiquitinate<br />

paxillin.<br />

RNF6<br />

LIM1, Androgen<br />

receptor<br />

RNF6 is an E3 ubiquitin ligase involved in the regulation of cell motility and<br />

differentiation. RNF6 targets LIMK for ubiquitination and degradation, inhibiting<br />

cytoskeleton stability.<br />

RNF8 H2A, H2AX RNF8 is a RING domain E3 ubiquitin ligase that plays a role in the repair<br />

of damaged chromosomes. RNF8 ubiquitinates Histone H2A and H2A.X at<br />

double-strand breaks (DSBs) which recruits 53BP1 and BRCA1 repair proteins.<br />

19398581,<br />

18308511<br />

9618557,<br />

9792722<br />

19917253,<br />

17218260<br />

17003037,<br />

11696324<br />

12826607<br />

12526791<br />

20074049,<br />

18671761,<br />

17553932,<br />

16672220<br />

18460542<br />

19734906,<br />

17675297<br />

15283676<br />

16096365<br />

12036302<br />

17720710,<br />

18245774<br />

17449468,<br />

16643902,<br />

12654245,<br />

18006823,<br />

18344599<br />

15229288,<br />

18382127<br />

19805145,<br />

16931761,<br />

18815134<br />

20832730<br />

16359901<br />

17157253,<br />

15509584<br />

19269966,<br />

12861019<br />

16204183<br />

18001824<br />

128 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables 129


Section I: Research Areas<br />

chapter 04: Cell Biology<br />

Ligase Substrate Function PMID<br />

RNF11 Smurf2 RNF11 is a required component of a ubiquitin-editing protein complex involved 14562029<br />

in modifying cellular inflammatory response to LPS and TNF signaling.<br />

RNF12 CLIM, Ldb1,<br />

Ldb2<br />

RNF12, also known as RLIM, is an E3 ubiquitin ligase. RNF12 is involved in<br />

telomere regulation and X chromosome inactivation.<br />

12874135,<br />

11882901<br />

RNF19 SOD1 RNF19 is also known as Dorfin. Accumulation and aggregation of mutant 19610091<br />

SOD1 leads to ALS disease. RNF19 ubiquitinates mutant SOD1 protein, causing<br />

a decrease in neurotoxicity.<br />

RNF20 Histone H2B RNF20 is also known as BRE1. RNF20 is an E3 ubiquitin ligase that monoubiquitinates<br />

18832071<br />

Histone H2B. H2B ubiquitination is associated with areas of<br />

active transcription.<br />

RNF34 Caspase-8, -10 RNF34 is also known as RFI. RNF34 inhibits death receptor mediated apoptosis 16596200<br />

through ubiquitination/degradation of caspase-8 and -10.<br />

RNF40 Histone H2B RNF40 is also known as BRE1-B. RNF40 forms a protein complex with RNF20<br />

resulting in the ubiquitination of Histone H2B. H2B ubiquitination is associated<br />

with areas of active transcription.<br />

16307923<br />

RNF41<br />

RNF111<br />

ErbB3, BIRC6,<br />

Parkin<br />

Smad, SnoN,<br />

c-Ski<br />

RNF41 is an E3 ubiquitin ligase that has been implicated in the regulation of<br />

hematopoietic progenitor cell differentiation.<br />

RNF111 is an E3 ubiquitin ligase that participates in mesoderm patterning<br />

by promoting the ubiquitination and proteasomal degradation of downstream<br />

Smads.<br />

RNF123 CDKN1B RNF123 is an E3 ubiquitin ligase that functions as part of the KPC complex.<br />

RNF123 aids in cell cycle regulation by targeting CDKN1B for ubiquitination<br />

and proteasomal degradation during G1.<br />

RNF125<br />

RNF125 is also known as TRAC-1. RNF125 has been shown to positively<br />

regulate T cell activation.<br />

RNF128<br />

RNF128 is also known as GRAIL. RNF128 promotes T cell anergy and may<br />

play a role in actin cytoskeletal organization in T cell/APC interactions.<br />

RNF135 RIG-1 RNF135 is an E3 ubiquitin ligase involved in viral innate immunity. RNF135<br />

targets the cytoplasmic viral nucleic acid receptor RIG-1 for ubiquitination and<br />

degradation by the proteasome.<br />

RNF138 TCF/LEF RNF138 is also known as NARF. RNF138 is associated with Nemo-like Kinase<br />

(NLK) and suppresses Wnt/β-Catenin signaling through ubiquitination/degradation<br />

of TCF/LEF.<br />

RNF167<br />

TSSC5<br />

(SLC22A18)<br />

RNF167 may act as an E3 ubiquitin ligase involved in the regulation of kidney<br />

transporter function.<br />

RNF168 H2A, H2A.X RNF168 is an E3 ubiquitin ligase that helps protect genome integrity by<br />

working together with RNF8 to ubiquitinate Histone H2A and H2A.X at DNA<br />

double-strand breaks (DSB).<br />

RNF180 Zic2 RNF180 is an E3 ubiquitin ligase involved in neurological development.<br />

RNF180 targets the ZIC2 transcription factor for polyubiquitination and degradation<br />

by the proteasome.<br />

RNF182 ATP6VOC RNF 182 is an E3 ubiquitin ligase that targets ATP6V0C, a component of<br />

vacuolar ATPase, for polyubiquitination and degradation by the proteasome.<br />

RNF190<br />

see MARCH-X<br />

SCF/β-TrCP<br />

IκBα, Wee1,<br />

Cdc25A,<br />

β-Catenin<br />

SCF/β-TrCP is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, β-TrCP (also known as<br />

BTRC). SCF/β-TrCP mediates the ubiquitination of proteins involved in cell cycle<br />

progression, signal transduction, and transcription. SCF/β-TrCP also regulates<br />

the stability of β-catenin and participates in Wnt signaling.<br />

SCF/FBXW2 GCMa SCF/FBXW2 is an E3 ubiquitin ligase complex composed of SCF (SKP1-<br />

CUL1-F-box protein) and the substrate recognition component, FBXW2. SCF/<br />

FBXW2 promotes ubiquitination of GCMa which is important for trophoblast<br />

cell differentiation.<br />

SCF/FBXW5 SASS6, Eps8 SCF/FBXW5 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXW5. SCF/FBXW5<br />

mediates the ubiquitination of SASS6, preventing centriole duplication.<br />

SCF/FBXW7 Cyclin-E, c-<br />

Myc, c-Jun<br />

SCF/FBXW8<br />

IRS-1, TBC1D3,<br />

Cyclin D1,<br />

IGFBP2, HPK1<br />

SCF/FBXW7 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXW7. SCF/FBXW7<br />

mediates the ubiquitination of proteins involved in cell cycle progression, signal<br />

transduction, and transcription. Target proteins for SCF/FBXW7 include the<br />

phosphorylated forms of c-Myc, Cyclin E, Notch intracellular domain (NICD),<br />

and c-Jun. Research has found that defects in FBXW7 may be a cause of many<br />

types of human cancers including T-ALL, colon cancer and breast cancer.<br />

SCF/FBXW8 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXW8. SCF/FBXW8<br />

plays critical roles in various cellular processes such as cell cycle progression,<br />

cell differentiation, development, and growth factor signaling pathway.<br />

14765125,<br />

12411582,<br />

18541373<br />

18451154,<br />

14657019,<br />

17591695<br />

15531880<br />

17990982<br />

19833735<br />

19017631<br />

16714285<br />

16314844<br />

19203579<br />

18363970<br />

18298843<br />

10230406,<br />

15070733,<br />

14603323,<br />

10339577<br />

15640526<br />

18381890,<br />

23314863<br />

11533444,<br />

15150404,<br />

16023596<br />

23142081,<br />

23029530,<br />

17205132,<br />

24362026<br />

Ligase Substrate Function PMID<br />

SCF/<br />

FBXW10<br />

CBX1, CBX5 SCF/FBXW10 is an E3 ubiquitin ligase complex composed of SCF (SKP1-<br />

CUL1-F-box protein) and the substrate recognition component, FBXW10. SCF/<br />

FBXW10 contributes to gene expression by degradation of heterochromatin<br />

components CBX5 and CBX1.<br />

20498703<br />

SCF/<br />

FBXW15<br />

SCF/Skp2/<br />

FBXL1<br />

SCF/FBXL2<br />

HBO1<br />

p27, p21,<br />

FoxO1<br />

Aurora B, p85b,<br />

APP, Cyclin D2<br />

SCF/FBXW15 is an E3 ubiquitin ligase complex composed of SCF (SKP1- 23319590<br />

CUL1-F-box protein) and the substrate recognition component, FBXW15. SCF/<br />

FBXW15 controls DNA replication via degradation of the origin recognition<br />

complex protein HBO1.<br />

SCF/Skp2 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-Fbox<br />

protein) and the substrate recognition component, Skp2. SCF/Skp2 medi-<br />

10559916<br />

15668399,<br />

ates the ubiquitination of proteins involved in cell cycle progression (specifically<br />

the G1/S transition), signal transduction and transcription. Target proteins for<br />

SCF/Skp2 include the phosphorylated forms of p27Kip1, p21Waf1/Cip1, and<br />

FoxO1.<br />

SCF/FBXL2 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXL2. SCF/FBXL2 is<br />

involved in cell cycle regulation, growth factor signaling and synapse formation.<br />

SCF/FBXL3 CRY1, CRY2 SCF/FBXL3 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXL3. SCF/FBXL3<br />

mediates circadian clock function by ubiquitination and subsequent degradation<br />

of CRY1 and CRY2.<br />

SCF/FBXL4<br />

SCF/FBXL5<br />

KDM4A/JM-<br />

JD2A<br />

IRP2,<br />

p150(Glued)<br />

SCF/FBXL4 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXL4. SCF/FBXL4<br />

regulates chromatin structure through degradation of the lysine demethylase<br />

KDM4A/JMJD2A.<br />

SCF/FBXL5 is an ubiquitin ligase complex also known as SCF (SKP1-cullin-Fbox).<br />

FBXL5 is an F-box protein that functions as an iron sensor by promoting<br />

the ubiquitination and subsequent degradation of IREB2/IRP2 under high iron<br />

and oxygen conditions.<br />

SCF/FBXL7 Aurora A SCF/FBXL7 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXL7. SCF/FBXL7<br />

colocalizes with and promotes degradation of Aurora A during mitosis leading<br />

to impaired cell proliferation.<br />

SCF/FBXL10<br />

p15, c-Fos,<br />

Ink4a, RipK3<br />

SCF/FBXL10 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXL10. SCF/FBXL10<br />

plays critical roles in cell proliferation, senescence and stem cell self-renewal.<br />

SCF/FBXL11 p65 FBXL11 is a lysine demethylase that regulates NF-κB signaling pathway through<br />

p65 demethylation. A substrate for ubiquitination has yet to be identified.<br />

SCF/FBXL12<br />

SCF/FBXL14<br />

SCF/FBXL15<br />

SCF/FBXL19<br />

Ku80,<br />

Calmodulin<br />

kinase I<br />

SNAIL1, SLUG,<br />

Mkp3<br />

Timeless,<br />

SMURF1,<br />

SMURF2<br />

ST2L, Rac1,<br />

RhoA<br />

SCF/FBXL12 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXL12. SCF/FBXL12<br />

is involved in regulating cell cycle progression and DNA damage response.<br />

SCF/FBXL14 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component,FBXL14. SCF/FBXL14<br />

is associated with metastasis and cancer development.<br />

SCF/FBXL15 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXL15. FBXL15 targets<br />

negative regulators of the BMP signaling pathway, including SMURF1 and<br />

SMURF2, for ubiquitination and subsequent proteasomal degradation. FBXL15<br />

is required for dorsal/ventral pattern formation and bone mass maintenance.<br />

SCF/FBXL15 also targets the Drosophila circadian clock protein timeless.<br />

SCF/FBXL19 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXL19. SCF/FBXL19<br />

is an important regulator of cell proliferation and migration, cytoskeleton<br />

reorganization, and pulmonary inflammation.<br />

SCF/FBXL20 RIM1 SCF/FBXL20 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXL20. FBXL20 is<br />

localized to the synapse and its regulation of RIM1 by ubiquitination may play a<br />

role in neural transmission.<br />

SCF/FBXL21 CRY SCF/FBXL21 is an E3 ubiquitin ligase complex composed of SCF (SKP1-<br />

CUL1-F-box protein) and the substrate recognition component, FBXL21. SCF/<br />

FBXL21 maintains circadian clock rhythms through controlling CRY turnover<br />

cooperatively with SCF/FBXL3.<br />

PhosphoSitePlus®<br />

A comprehensive online protein modification resource. www.cellsignal.com/exploration<br />

23928698,<br />

23604317,<br />

22399757,<br />

22323446<br />

17463251,<br />

17463252,<br />

17462724<br />

21757720<br />

19762597,<br />

19762596<br />

22306998<br />

18836456,<br />

21252908,<br />

21540074,<br />

22825849<br />

20080798<br />

23324393,<br />

23707388<br />

19955572,<br />

16887825,<br />

22410791<br />

16794082,<br />

21572392<br />

22660580,<br />

23512198,<br />

23871831<br />

17803915<br />

23452856,<br />

23452855,<br />

18953409<br />

130 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables<br />

131


Section I: Research Areas<br />

chapter 04: Cell Biology<br />

Ligase Substrate Function PMID<br />

SCF/FBXL22 ACTN/FLNC SCF/FBXL22 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXL22. FBXL22<br />

is exclusively expressed in cardiomyocytes, and promotes ubiquitination and<br />

degradation of sarcomeric proteins, α-actinin-2 (ACTN) and Filamin C (FLNC).<br />

22972877<br />

SCF/FBXO1 CP110, RRM2 SCF/FBXO1 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXO1. FBXO1<br />

targets CP110 for ubiquitination and subsequent proteasomal degradation during<br />

cell cycle G2 phase, thereby inhibiting centrosome reduplication.<br />

SCF/FBXO2 Pre-integrin<br />

β-1, CFTR,<br />

Connexin 26,<br />

NR1, SHPS-1,<br />

UL9<br />

SCF/FBXO3 HIPK2, p300,<br />

Fbxl2<br />

SCF/FBXO4<br />

SCF/FBXO6<br />

SCF/FBXO7<br />

TERF1, Cyclin<br />

D1<br />

Chk1, ATRN,<br />

TCRa, TFRC<br />

BIRC2, DL-<br />

GAP5, CD43<br />

SCF/FBXO2 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXO2 (NFB42).<br />

FBXO2 targets misfolded glycoproteins for ubiquitination and proteasomal<br />

degradation by recognition of sugar chains in the endoplasmic reticulumassociated<br />

degradation (ERAD) pathway.<br />

SCF/FBXO3 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXO3. FBXO3<br />

targets HIPK2 and p300 for ubiquitination and rapid degradation by the<br />

proteasome. The inclusion of PML in a complex with SCF/FBXO3, HIPK2, and<br />

p300 delays degradation of HIPK2 and allows synergistic activation of p53/<br />

TP53-dependent transactivation.<br />

SCF/FBXO4 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXO4. FBXO4 may<br />

play a role in telomere homeostasis by recognition of TERF1 and promotion of<br />

its ubiquitination together with UBE2D1.<br />

SCF/FBXO6 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXO6. FBXO6<br />

targets misfolded glycoproteins for ubiquitination and proteasomal degradation<br />

by recognition of sugar chains in the endoplasmic reticulum-associated<br />

degradation (ERAD) pathway. FBXO6 also targets the kinase Chk1, a cell cycle<br />

regulator involved in entry into mitosis.<br />

SCF/FBXO7 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXO7. SCF/FBXO7<br />

targets BIRC2 (cIAP1), an inhibitor of apoptosis, and DLGAP5, a cell cycle<br />

regulator, for ubiquitination and proteasomal degradation.<br />

SCF/FBXO8 Arf6 SCF/FBXO8 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXO8. FBXO8<br />

mediates the ubiquitination of Arf6. SCF/FBXO8 may function as a guanine<br />

nucleotide exchange factor (GEF) that activates ARF G proteins. Loss of FBXO8<br />

correlates with poor survival in hepatocellular carcinoma.<br />

SCF/FBXO9 Tel2, Tti1 SCF/FBXO9 is an E3 ubiquitin ligase complex composed of SCF (SKP1-<br />

CUL1-F-box protein) and the substrate recognition component, FBXO9. Three<br />

isoforms of the human protein are produced by alternative splicing, and it is<br />

linked to promoting survival in multiple myeloma.<br />

SCF/FBXO10 Bcl2 SCF/FBXO10 is an E3 ubiquitin ligase complex composed of SCF (SKP1-<br />

CUL1-F-box protein) and the substrate recognition component, FBXO10. SCF/<br />

FBXO10 induces apoptosis in B-cells through degrading Bcl2.<br />

SCF/FBXO11 Cdt2, Bcl6, p53 SCF/FBXO11 is an E3 ubiquitin ligase complex composed of SCF (SKP1-<br />

CUL1-F-box protein) and the substrate recognition component, FBXO11.<br />

SCF/FBXO11 promotes the neddylation of p53 to suppress its transcriptional<br />

activity, and crosstalks with the cullin 4-RING ubiquitin ligase, CRL4-Cdt2,<br />

to control cell cycle progression. Mutations and deletions in FBXO11 lead to<br />

BCL6 stabilization and B-cell lymphoma. FBXO11 is also regarded as a haploinsufficient<br />

tumor suppressor gene.<br />

SCF/FBXO15<br />

P-glycoprotein/<br />

ABCB1<br />

SCF/FBXO15 is an E3 ubiquitin ligase complex composed of SCF (SKP1-<br />

CUL1-F-box protein) and the substrate recognition component, FBXO15. SCF/<br />

FBXO15 activity may affect anticancer drug resistance through controlling the<br />

expression of P-glycoprotein (P-gp)/ABCB1 on cancer cell surfaces.<br />

20596027,<br />

22632967<br />

12140560,<br />

15809437,<br />

14701835,<br />

17494702<br />

18809579,<br />

23542741,<br />

18809579<br />

17081987,<br />

19645770,<br />

17081987<br />

19716789,<br />

22268729,<br />

12939278<br />

16510124,<br />

21652635<br />

18094045<br />

23263282<br />

23431138<br />

23478445,<br />

23478441,<br />

23892434,<br />

22113614,<br />

17098746<br />

23465077<br />

SCF/FBXO17 Arf1 SCF/FBXO17 is an E3 ubiquitin ligase complex composed of SCF (SKP1- 19836238<br />

CUL1-F-box protein) and the substrate recognition component, FBXO17. In<br />

fission yeast, SCF/FBXO17 binds the Arf1 transcription factor and promotes<br />

Arf degradation to suppress stress-related gene expression. Stress-induced Arf<br />

phosphorylation dissociates Arf interaction with FBXO17.<br />

SCF/FBXO22 KDM4A SCF/FBXO22 is an E3 ubiquitin ligase complex composed of SCF (SKP1-<br />

CUL1-F-box protein) and the substrate recognition component, FBXO22. SCF/<br />

FBXO22 is involved in transcriptional control through degradation of lysine<br />

demethylase KDM4A.<br />

21768309<br />

SCF/FBXO25<br />

NKX2.55, Isl1,<br />

Hand1, Elk-1<br />

SCF/FBXO25 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXO25. FBXO25<br />

is a cardiac-specific F-box protein. SCF/FBXO25 maintains cardiac protein<br />

homeostasis through degrading NKX2.5, Isl1, and Hand1, and suppresses<br />

mitogenic response by downregulating Elk-1.<br />

21596019,<br />

23940030<br />

Ligase Substrate Function PMID<br />

SCF/FBXO31 Cyclin D1,<br />

Par6c<br />

SCF/FBXO31 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXO31 (FBXO14).<br />

SCF/FBXO31 targets phosphorylated cyclin D1 for ubiquitination and degradation<br />

by the proteasome, resulting in G1 cell cycle arrest.<br />

19412162<br />

SCF/FBXO32<br />

DUSP1, eIF3-f,<br />

MyoD, BK-β(1),<br />

FOXO1<br />

SCF/FBXO32 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXO32 (MAFbx).<br />

FBXO32 is a cardiac-specific F-box protein. SCF/FBXO32 maintains cardiac<br />

protein homeostasis. FBXO32 promoter is highly methylated in pediatric softtissue<br />

sarcoma.<br />

SCF/FBXO33 YB-1 SCF/FBXO33 is an E3 ubiquitin ligase complex composed of SCF (SKP1-<br />

CUL1-F-box protein) and the substrate recognition component, FBXO33.<br />

SCF/FBXO33 is involved in gene expression and protein translation through<br />

degradation of Y-box protein YB-1.<br />

SCF/FBXO40 IRS-1 SCF/FBXO40 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXO40. FBXO40 is<br />

a skeletal muscle-specific F-box protein. SCF/FBXO40 controls muscle size by<br />

regulating IRS-1 protein stability.<br />

SCF/FBXO42 p53 SCF/FBXO42 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXO42. FBXO42<br />

targets p53/TP53 for ubiquitination and degradation by the proteasome.<br />

SCF/FBXO44 BRCA1 SCF/FBXO44 is an E3 ubiquitin ligase complex composed of SCF (SKP1-<br />

CUL1-F-box protein) and the substrate recognition component, FBXO44. SCF/<br />

FBXO44 activity may contribute to breast cancer development through targeting<br />

BRCA1 for proteasomal degradation.<br />

SCF/FBXO45 UNC13A, p73 SCF/FBXO45 is an E3 ubiquitin ligase complex composed of SCF (SKP1-CUL1-<br />

F-box protein) and the substrate recognition component, FBXO45. SCF/FBXO45<br />

aids in the regulation of neurotransmission at mature neurons by targeting<br />

UNC13A for ubiquitin dependent degradation by the proteasome. FBXO45 also<br />

targets the apoptotic protein p73 for ubiquitination and degradation.<br />

SHPRH PCNA SHPRH is an E3 ubiquitin ligase that plays a role in DNA replication through<br />

ubiquitination of PCNA. PCNA ubiquitination prevents genomic instability from<br />

stalled replication forks after DNA damage.<br />

SIAH1<br />

β-catenin, Bim,<br />

TRB3<br />

SIAH1 is an E3 ubiquitin ligase that plays a role in inhibition of Wnt signaling<br />

through ubiquitination of β-catenin. SIAH1 has also been shown to promote<br />

apoptosis through upregulation of Bim, and to ubiquitinate the signaling adaptor<br />

protein TRB3.<br />

SIAH2 HIPK2, PHD1/3 SIAH2 is an E3 ubiquitin ligase that plays a role in hypoxia through ubiquitination<br />

and degradation of HIPK2. SIAH2 also ubiquitinates PHD1/3, which<br />

regulates levels of HIF-1α in response to hypoxia.<br />

SMURF1<br />

Smad1/5,<br />

RhoA, MEKK2<br />

SMURF1 is an E3 ubiquitin ligase that interacts with BMP pathway Smad<br />

effectors, leading to Smad protein ubiquitination and degradation. Smurf1<br />

negatively regulates osteoblast differentiation and bone formation in vivo.<br />

SMURF2 Smads, Mad2 SMURF2 is an E3 ubiquitin ligase that interacts with Smads from both the BMP<br />

and TGF-β pathways. SMURF2 also regulates the mitotic spindle checkpoint<br />

through ubiquitination of Mad2.<br />

SYVN1<br />

ERAD, Pael-<br />

R,p53, IRE-1<br />

SYVN1 is an E3 ubiquitin ligase involved in the ER-associated degradation<br />

(ERAD) pathway. SYVN1 targets misfolded proteins and appropriately folded<br />

short-lived proteins for ubiquitination and degradation by the proteasome.<br />

TOPORS p53, NKX3.1 TOPORS is an E3 ubiquitin ligase and a SUMO ligase. TOPORS ubiquitinates<br />

and sumoylates p53, which regulates p53 stability. TOPORS has also been<br />

shown to ubiquitinate the tumor suppressor NKX3.1.<br />

TRAF2<br />

Rip1, other<br />

TRAFs<br />

TRAF2 is a weak E3 ubiquitin ligase that acts as a component of several<br />

ubiquitination complexes. TRAF2 ligase activity is activated in the presence<br />

of cytoplasmic sphingosine-1-phosphate. TRAF2 is a major regulator of the<br />

apoptosis and cell survival machinery.<br />

TRAF6 NEMO, Akt1 TRAF6 is an E3 ubiquitin ligase that functions as an adaptor protein in IL-1R,<br />

CD40, and TLR signaling. TRAF6 promotes NF-κB signaling through K63<br />

polyubiquitination of IKK, resulting in IKK activation. TRAF6 has also been<br />

shown to ubiquitinate Akt1, causing its translocation to the cell membrane.<br />

TRAF7<br />

TRAF7 is an E3 ubiquitin ligase and SUMO ligase that functions as an adaptor<br />

protein in TNF Receptor and TLR signaling. TRAF7 has been shown to be capable<br />

of self-ubiquitination and plays a role in apoptosis via MEKK3-mediated<br />

activation of NF-κB.<br />

TRIAD3 TLRs, RIP1 Triad 3 is an E3 ubiquitin ligase found in peripheral blood leukocytes of the immune<br />

system that regulates antiviral and cytokine induced cellular responses.<br />

TRIM8 SOCS-1 TRIM8 is an E3 ubiquitin ligase that regulates cytokine induced signal<br />

transduction by targeting SOCS1 for ubiquitination and degradation by the<br />

proteasome.<br />

22586590,<br />

21454680,<br />

19073596,<br />

24213577<br />

16797541<br />

22033112<br />

19509332<br />

23086937<br />

19581926<br />

18719106<br />

16413921,<br />

19775288,<br />

18276110<br />

19043406,<br />

15210114<br />

10458166,<br />

15820682<br />

11158580,<br />

18852296<br />

14593114,<br />

17059562,<br />

17170702,<br />

18369366<br />

19473992,<br />

18077445<br />

11909853,<br />

15175328<br />

19713527,<br />

11057907<br />

15001576<br />

15107846,<br />

16968706<br />

12163497<br />

132 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables 133


Section I: Research Areas<br />

chapter 04: Cell Biology<br />

Ligase Substrate Function PMID<br />

TRIM11 Humanin,<br />

ARC105, Pax6<br />

TRIM11 is an E3 ubiquitin ligase that may promote the degradation of insoluble<br />

ubiquitinated proteins. TRIMM11 may also aid in anti-viral cellular functions.<br />

12670303,<br />

16904669,<br />

18628401<br />

TRIM13<br />

TRIM13 is an E3 ubiquitin ligase that targets membrane and secretory proteins 17314412<br />

for ubiquitination and proteasomal degradation in the endoplasmic reticulumassociated<br />

degradation (ERAD) pathway.<br />

TRIM21 IgG1 HC, IRF3 TRIM21 is an E3 ubiquitin ligase involved in intracellular antibody-mediated<br />

degradation of viral components by the proteasome.<br />

TRIM25 RIG-1 TRIM25 is an E3 ubiquitin ligase involved in viral innate immunity. TRIM25<br />

targets the cytoplasmic viral nucleic acid receptor RIG-1 for ubiquitination and<br />

degradation by the proteasome.<br />

TRIM32 actin, piasy TRIM32 is an E3 ubiquitin ligase involved in viral lysosome related vesicle<br />

trafficking. TRIM32 targets DTNBP1 for ubiquitination and degradation by the<br />

proteasome. TRIM32 may also mediate the activity of HIV Tat proteins.<br />

TRIM33 Smad4 TRIM33 is an E3 ubiquitin ligase involved in the regulation of the TGF-b/ BMP<br />

signaling pathway. TRIM33 targets SMAD4 for ubiquitination, nuclear exclusion,<br />

and proteasomal degradation.<br />

TRIM41 PKC TRIM41 is an E3 ubiquitin ligase that targets protein kinase C for ubiquitination<br />

and proteasomal degradation.<br />

18022694,<br />

18641315<br />

12075357<br />

14578165,<br />

16243356<br />

15820681<br />

17893151<br />

TRIM54 TRIM54 is an E3 ubiquitin ligase that may target and stabilize microtubules. 15967462<br />

TRIM55<br />

TRIM55 is an E3 ubiquitin ligase that may regulate gene expression and 15967462<br />

protein turnover in muscle cells<br />

TRIM63 Troponin I, TRIM63 is also known as Murf-1. TRIM63 is a muscle-specific E3 ubiquitin ligase 19506036<br />

MyBP-C, whose expression is upregulated during muscle atrophy. TRIM63 has been shown<br />

to ubiquitinate several important muscle proteins including troponin I, MyBP-C, and<br />

MyLC1/2.<br />

MyLC1/2<br />

TRIM63 is also known as Murf-1. TRIM63 is a muscle-specific E3 ubiquitin 19506036<br />

ligase whose expression is upregulated during muscle atrophy. TRIM63 has<br />

been shown to ubiquitinate several important muscle proteins including<br />

troponin I, MyBP-C, and MyLC1/2.<br />

UBE3B<br />

UBE3B is an E3 ubiquitin ligase identified through sequence analysis. The 12837265<br />

specific substrates and cellular function of UBE3B is currently unknown.<br />

UBE3C<br />

UBE3C is an E3 ubiquitin ligase also known as KIAA10. UBE3C is highly expressed<br />

in muscle and may interact with the transcriptional regulator TIP120B.<br />

12692129<br />

UBR1<br />

UBR1 is an E3 ubiquitin ligase responsible for proteasomal degradation of<br />

misfolded cytoplasmic proteins. UBR1 has also been shown to be a ubiquitin<br />

ligase of the N-end rule proteolytic pathway, which regulates degradation of<br />

short-lived proteins.<br />

UBR2 Histone H2A UBR2 is an E3 ubiquitin ligase that has been shown to ubiquitinate histone<br />

H2A, resulting in transcriptional silencing. UBR2 is also part of the N-end rule<br />

proteolytic pathway.<br />

19041308,<br />

17962019<br />

20080676,<br />

19008229<br />

UHRF1 Histone H3 UHRF1 is an epigenetic regulator that is also a putative E3 ubiquitin ligase. 14993289<br />

UHRF2 PCNP UHRF2 is also known as NIRF. UHRF2 is a nuclear protein that may regulate<br />

cell cycle progression through association with Chk2. UHRF2 also ubiquitinates<br />

PCNP and has been shown to play a role in degradation of nuclear aggregates<br />

containing polyglutamine repeats.<br />

VHL HIF-1α VHL is the substrate recognition component of the ECV (Elongin B/C, CULlen-2,<br />

VHL) E3 ubiquitin ligase complex responsible for degradation of the<br />

transcription factor HIF-1α. Ubiquitination and degradation of HIF-1α takes<br />

place only during periods of normoxia, but not during hypoxia, thereby playing<br />

a central role in the regulation of gene expression by oxygen.<br />

VPS18 SNK VPS18 is an E3 ubiquitin ligase that regulates intracellular vesicle trafficking.<br />

VPS18 may also regulate the POLO-like kinase SNK during the cell cycle.<br />

WWP1 ErbB4 WWP1 is an E3 ubiquitin ligase commonly found to be overexpressed in breast<br />

cancer. WWP1 has been shown to ubiquitinate and degrade ErbB4. Interestingly,<br />

the WWP1 homolog in C. elegans was found to increase life expectancy<br />

in response to dietary restriction.<br />

WWP2 oct-4, PTEN WWP2 is an E3 ubiquitin ligase that has been shown to ubiquitinate/degrade<br />

the stem cell pluripotency factor Oct-4. WWP2 also ubiquitinates the transcription<br />

factor EGR2 to inhibit activation-induced T cell death.<br />

ZNRF1<br />

ZNRF1 is an E3 ubiquitin ligase highly expressed in neuronal cells. ZNRF1 is<br />

found in synaptic vesicle membranes and may regulate neuronal transmissions<br />

and plasticity.<br />

PhosphoSitePlus®<br />

A comprehensive online protein modification resource. www.cellsignal.com/cstpsp<br />

15178429,<br />

14741369,<br />

19218238<br />

11292862<br />

16203730<br />

19561640,<br />

19553937<br />

19274063,<br />

19651900,<br />

21532586<br />

14561866<br />

DUB Substrate Function PMID<br />

ATXN3<br />

ATXN3L<br />

BRCC36<br />

COPS5<br />

COPS6<br />

RAD23A,<br />

RAD23B,<br />

STUB1/CHIP<br />

FAM175A/<br />

Abraxas<br />

TP53, MIF,<br />

JUN, UCHL1,<br />

ESR1,<br />

RanBP9<br />

TP53, MIF,<br />

c-Jun,<br />

UCHL1<br />

ATXN3 is a transcriptional regulation deubiquitination enzyme that preferentially 17696782<br />

displays ubiquitin isopeptidase activity toward long, four or greater, ubiquitin chains.<br />

ATXN3-like is a deubiquitination enzyme that displays ubiquitin isopeptidase 21118805<br />

activity toward K48- and K63-linked chains.<br />

BRCC36 (BRCC3) is a deubiquitination enzyme that preferentially displays ubiquitin 14636569,<br />

isopeptidase activity toward K63-linked chains. BRCC36 targets K63-linked ubiquitin<br />

chains on H2A and H2X at the site of DNA double strand breaks as a component<br />

16707425<br />

of the BRCA complex.<br />

COPS5 (CSN5) is the protease subunit of the COP9 signalosome complex (CSN), a<br />

key regulator of the ubiquitin conjugation pathway. COPS5 is essential for the CSN<br />

isopeptidase activity responsible for deneddylation of cullin-RING E3 ubiquitin ligase<br />

complexes.<br />

COPS6 is a component of the COP9 signalosome complex (CSN) which is involved<br />

in several cellular and developmental processes. The CSN complex regulates the<br />

ubiquitin (Ubl) conjugation pathway through the deneddylation of the cullin subunits<br />

of SCF-type E3 ligase complexes. This decreases the Ubl ligase activity of SCF-type<br />

complexes such as SCF, CSA, or DDB2.<br />

9535219,<br />

11285227,<br />

23926111<br />

11337588,<br />

12732143,<br />

12628923<br />

USP17L2 CDC25A USP17L2 is a deubiquitination enzyme that preferentially displays ubiquitin 14699124,<br />

isopeptidase activity toward CDC25A, preventing CDC25A degradation and allowing 20228808<br />

cell cycle progression.<br />

eIF3F Notch eIF3F deubiquitinates activated Notch1 promoting its nuclear import, thereby acting 21124883<br />

as a positive regulator of Notch signaling.<br />

eIF3H<br />

eIF3H is involved in various stps of the initiation of protein synthesis as a component 16766523<br />

of the eukaryotic translation initiation factor 3 (eIF-3) complex.<br />

JOSD1<br />

JOSD1 is a deubiquitination enzyme that displays low ubiquitin isopeptidase activity 21118805<br />

in vitro.<br />

JOSD2<br />

MPND<br />

MYSM1<br />

H2A,<br />

E4BP4,<br />

GFI1<br />

JOSD2 is a deubiquitination enzyme that displays ubiquitin isopeptidase activity<br />

toward K63-linked chains, and to a lesser extent K48-linked chains.<br />

MPND is an MPN domain and JAMM motif-containing protein with predicted<br />

ubiquitin isopeptidase activity.<br />

MYSM1 is a deubiquitinating enzyme that acts as a transcriptional co-activator by<br />

directing preferential ubiquitin isopeptidase activity toward monoubiquinated H2A in<br />

hyperacetylated nucleosomes.<br />

OTU1 VCP OTU1, also known as YOD1, is a deubiquitination enzyme that displays ubiquitin<br />

isopeptidase activity toward K48- and K63-linked polyubiquitin or di-ubiquitin<br />

chains. OTU1 is a part of the endoplasmic reticulum-associated degradation (ERAD)<br />

pathway for misfolded lumenal proteins.<br />

OTUB1<br />

RNF128,<br />

UbcH5,<br />

Smad2/3,<br />

c-IAP<br />

OTUB1 is a deubiquitination enzyme that preferentially displays ubiquitin isopeptidase<br />

activity toward polyubiquitinated K48-linked chains. OTUB1 regulates protein<br />

turnover by preventing degradation and also plays a unique role in the regulation<br />

of T cell anergy. Furthermore, OTUB1 regulates p53 stability and activity via noncanonical<br />

inhibition of the MDM2 cognate Ub-conjugating enzyme (E2) UbcH5.<br />

OTUB1 also inhibits the ubiquitination of phospho-SMAD2/3 by binding to and<br />

inhibiting the E2 ubiquitin-conjugating enzymes independent of its catalytic activity.<br />

OTUB1 regulates NF-κB and MAPK signaling pathways as well as TNF-dependent<br />

cell death by modulating c-IAP1 stability.<br />

OTUB2 RNF-8 OTUB2 is a deubiquitination enzyme that displays ubiquitin isopeptidase activity<br />

toward K48- and K63-linked chains. OTUB2 regulates protein turnover by preventing<br />

degradation. OTUB2 fine-tunes the speed of DSB-induced ubiquitination.<br />

OTUD1<br />

OTUD1 is a member of the deubiquitinating enzyme ovarian tumor domain (OTU)<br />

superfamily.<br />

OTUD3<br />

OTUD3 is a member of the deubiquitinating enzyme ovarian tumor domain (OTU)<br />

superfamily.<br />

OTUD4 XPC OTUD4 is a member of the deubiquitinating enzyme ovarian tumor domain (OTU)<br />

superfamily.<br />

OTUD5<br />

OTUD6A<br />

OTUD6B<br />

OTUD7A/<br />

Cezanne 2<br />

OTUD7B/<br />

Cezanne<br />

OTUD7C/<br />

A20<br />

TRAF3,<br />

p53<br />

TRAF6,<br />

TRAF3<br />

NAF1,<br />

TAX1BP1,<br />

TRAF2<br />

OTUD5 is a deubiquitination enzyme that displays ubiquitin isopeptidase activity<br />

toward K48- and K63-linked chains. OTUD5 negatively regulates type I interferon<br />

(IFN) production by deubiquitination of TRAF3.<br />

OTUD6A is a member of the deubiquitinating enzyme ovarian tumor domain (OTU)<br />

superfamily.<br />

OTUD6B is a member of the deubiquitinating enzyme ovarian tumor domain (OTU)<br />

superfamily.<br />

OTUD7A is a deubiquitination enzyme that displays ubiquitin isopeptidase activity<br />

toward K48- and K63-linked chains.<br />

OTUD7B is a deubiquitination enzyme that displays ubiquitin isopeptidase activity<br />

toward K48- and K63-linked chains. OTUD7B negatively regulates NF-κB.<br />

OTUD7C is a ubiquitination-editing enzyme that displays ubiquitin isopeptidase activity<br />

toward K63-linked chains and ubiquitination of K48-linked chains. OTUD7C is<br />

an essential regulator of inflammatory signaling pathways in the lymphoid system.<br />

17696782,<br />

21118805<br />

17707232,<br />

24062447,<br />

24014243<br />

19818707<br />

12704427,<br />

14661020,<br />

24403071,<br />

24071738,<br />

23524849,<br />

22124327<br />

12704427,<br />

18954305,<br />

24560272<br />

17991829<br />

17991829<br />

17991829,<br />

24366067<br />

17991829,<br />

24143256<br />

17991829<br />

17991829<br />

12682062<br />

11463333,<br />

23334419<br />

9882303,<br />

14748687<br />

Deubiquitinase<br />

Table<br />

This table provides a list of<br />

deubiquitinases (DUBs), along with<br />

their substrates (when known) and<br />

corresponding references.<br />

134 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables<br />

135


Section I: Research Areas<br />

chapter 04: Cell Biology<br />

DUB Substrate Function PMID<br />

OTULIN Met-1<br />

polyubiquitin<br />

OTULIN disassembles Met1-Ub which is important for reducing Met1-Ub accumulation<br />

after stimulation of nucleotide-oligomerization domain-containing protein 2<br />

(NOD2). Depletion of OTULIN alters signaling downstream of NOD2.<br />

23806334<br />

POH1<br />

PRPF8<br />

SNRP116,<br />

WDR57/<br />

SPF38<br />

POH1 is the metalloprotease deubiquitination enzyme component of the 26S<br />

proteasome that displays ubiquitin isopeptidase activity toward K63-linked chains.<br />

PRPF8 is a member of the deubiquitinating enzyme metalloprotease JAMM domain<br />

superfamily. PRPF8 is known to be a central component of the spliceosome, while<br />

PRPF8 ubiquitin isopeptidase activity is controversial.<br />

PSMD7 TRIM5 PSMD7 is a regulatory subunit of the 26S proteasome, and is involved in the ATPdependent<br />

degradation of ubiquitinated proteins.<br />

STAMBP<br />

STAMB-<br />

PL1<br />

CXCR4,<br />

EGFR, ErbB2<br />

STAM-binding protein (STAMBP or AMSH) is an endosomal deubiquitination enzyme<br />

that preferentially displays ubiquitin isopeptidase activity toward K63-linked chains.<br />

STAM-binding protein-like 1 (STAMBPL1 or AMSHLP) is a deubiquitination enzyme<br />

that preferentially displays ubiquitin isopeptidase activity toward K63-linked chains.<br />

TRABID TRAF6, APC TRABID is a deubiquitination enzyme that preferentially displays ubiquitin isopeptidase<br />

activity toward K63-linked chains. TRABID acts as a positive regulator of the<br />

Wnt signaling pathway by deubiquitinating APC protein, a Wnt signaling pathway<br />

negative regulator.<br />

UCHL1<br />

UCHL2/<br />

BAP1<br />

COPS5,<br />

CHT, NCAM,<br />

β-Catenin<br />

BRCA1,<br />

HCFC1<br />

UCHL1 is a member of the ubiquitin C-terminal hydrolase (UCH) deubiquitinase<br />

superfamily. UCHL1 functions as a ubiquitin hydrolase involved in the processing of<br />

both ubiquitin precursors and ubiquitinated substrates, generating free monomeric<br />

Ub. UCHL1 may play a role in regulating cholinergic function through CHT ubiquitination<br />

and degradation.<br />

UCHL1/Bap1 is a member of the ubiquitin C-terminal hydrolase (UCH) deubiquitinase<br />

superfamily. UCHL1/ Bap1 is a BRCA1-associated, nuclear localized ubiquitin<br />

hydrolase that suppresses cell growth.<br />

136 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

9374539,<br />

19214193<br />

2139226,<br />

8702566<br />

22078707,<br />

7755639<br />

18758443,<br />

20159979,<br />

22800866<br />

18758443<br />

18281465,<br />

21834987<br />

9790970,<br />

24525247,<br />

23061666,<br />

22641175<br />

9528852<br />

UCHL3 ENAC UCHL3 is a member of the ubiquitin C-terminal hydrolase (UCH) deubiquitinase<br />

superfamily. UCHL3 functions as a ubiquitin hydrolase involved in the processing of<br />

both ubiquitin precursors and ubiquitinated substrates, generating free monomeric<br />

Ub. UCHL3 shows dual specificity toward both ubiquitin (Ub) and NEDD8, a Ub-like<br />

molecule.<br />

2530630<br />

UCHL5<br />

USP1<br />

USP2<br />

TGF-β<br />

Receptor I<br />

FANCD2,<br />

PCNA,<br />

PHLPP1<br />

CCND1,<br />

PER1<br />

USP3 H2A, Rig-I ,<br />

H2A, γH2A.X<br />

USP4<br />

USP5/<br />

ISOT<br />

ADORA2A,<br />

RB1,<br />

Rig-I, RIP1,<br />

TRAF2,<br />

TRAF6,<br />

PDK1<br />

p53,<br />

TRIML1<br />

UCHL5 is a member of the ubiquitin C-terminal hydrolase (UCH) deubiquitinase<br />

superfamily. UCHL5 is the deubiquitination enzyme component of the 19S regulatory<br />

subunit of the 26S proteasome that displays ubiquitin isopeptidase activity<br />

toward K48-linked chains.<br />

USP1 is a member of the ubiquitin-specific processing protease (USP/USB) deubiquitinase<br />

superfamily. USP1 is a negative regulator of DNA repair machinery.<br />

USP2 is a member of the ubiquitin-specific processing protease (USP/USB) deubiquitinase<br />

superfamily. USP2 is characterized by its C19 peptidase activity, which is<br />

involved in ubiquitin recycling and in the disassembly of various forms of polymeric<br />

ubiquitin and ubiquitin-like protein complexes. USP2 is also a core component of<br />

circadian rhythm machinery.<br />

USP3 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP3 deubiquitinates monoubiquitinated histone H2A<br />

and H2B. USP3 is required for proper progression through S phase and subsequent<br />

mitotic entry.<br />

USP4 is a member of the ubiquitin-specific processing protease (USP/USB) deubiquitinase<br />

superfamily. USP4 is a proto-oncogene that deubiquitinates target proteins<br />

such as the receptor ADORA2A and TRIM21 and plays a role in the regulation of<br />

quality control in the ER.<br />

USP5 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP5 preferentially cleaves branched and K48-linked<br />

polymers. USP5 binds linear and K63-linked polyubiquitin with a lower affinity.<br />

Knock-down of USP5 causes the accumulation of p53/TP53 and an increase in<br />

p53/TP53 transcriptional activity.<br />

USP6 NF-κB USP6 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP6 exhibits an ATP-dependent C-terminal isopeptidase<br />

activity.<br />

USP7/<br />

HAUSP<br />

FOXO4,<br />

PTEN p53,<br />

MDM2,<br />

Tip60, MCM,<br />

FoxP3 ,<br />

UBE2E1,<br />

NF-κB,<br />

Aurora A<br />

USP7, also known as herpes virus-associated ubiquitin-specific protease (Hausp),<br />

is a member of the ubiquitin-specific processing protease (USP/USB) deubiquitinase<br />

superfamily. USP7 deubiquitinates target proteins such as FoxO4, p53/TP53,<br />

MDM2, PTEN and DAXX. USP7 is Involved in cell proliferation during early embryonic<br />

development and also plays a role in the regulation of early adipogenesis.<br />

16906146,<br />

18922472,<br />

23500140<br />

15694335,<br />

16531995,<br />

22426999<br />

17290220,<br />

19917254,<br />

19838211,<br />

23213472<br />

17980597,<br />

24366338,<br />

24196443<br />

7784062,<br />

16316627,<br />

23388719,<br />

23313255,<br />

22029577,<br />

22347420<br />

19098288<br />

20418905,<br />

22081069<br />

11923872,<br />

14506283,<br />

15053880,<br />

23775119,<br />

24190967,<br />

23973222,<br />

23603909,<br />

23267096,<br />

23348568<br />

DUB Substrate Function PMID<br />

USP8/<br />

UBPY<br />

USP9X<br />

EPS15,<br />

CLOCK,<br />

HIF-1α, Smo<br />

SMAD4,<br />

MARK4,<br />

NUAK1,<br />

BIRC5/<br />

survivin,<br />

Smurf1,<br />

Mcl-1, ERG,<br />

Bcl10, PEX5<br />

USP8 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP8 is an essential growth-regulated enzyme that is<br />

indispensible for cell proliferation and survival. USP8 regulates endosomal ubiquitin<br />

dynamics, cargo sorting, membrane traffic at early endosomes, and maintenance<br />

of EGFR stability. In normoxia, USP8 maintains HIF1α and HIF transcriptional output<br />

which is essential for endosome-mediated ciliogenesis.<br />

USP9X is an X-linked member of the ubiquitin-specific processing protease<br />

(USP/USB) deubiquitinase superfamily. USP9X hydrolyzes both ‘Lys-29’- and<br />

‘Lys-33’-linked polyubiquitin chains. USP9X functions to regulate cell-cell contact<br />

interactions, TGF-β/BMP signaling, chromosome alignment and segregation, and<br />

specifically deubiquitinates monoubiquitinated Smad4. Moreover, USP9X is a<br />

mTORC1 and mTORC2 binding partner that negatively regulates mTOR activity and<br />

skeletal muscle differentiation.<br />

USP9Y SMAD4 USP9Y is a Y-linked member of the ubiquitin-specific processing protease (USP/<br />

USB) deubiquitinase superfamily required for sperm production. USP9Y functions to<br />

regulate TGF-β/BMP signaling, and specifically deubiquitinates monoubiquitinated<br />

Smad4.<br />

USP10 G3BP, p53/<br />

TP53, SNX3,<br />

NEMO, SirT6<br />

USP11<br />

USP12<br />

USP13/<br />

ISOT3<br />

USP14<br />

USP15<br />

BRCA2,<br />

CHUK/IKKA,<br />

RANBP9/<br />

RANBPM ,<br />

PML, ALK5<br />

WDR48,<br />

PHLPP1,<br />

Notch<br />

PTEN, Stat1,<br />

Ubl4A<br />

FANCC,<br />

CXCR4,<br />

ERN1, REST,<br />

I-κB<br />

E6, ubH2B,<br />

TRIM25,<br />

KEAP1,<br />

REST, BRAP,<br />

TβR-I<br />

USP10 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP10 functions as an essential regulator of p53/TP53<br />

stability following DNA damage.<br />

USP11 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP11 aids in the regulation of pathways leading to<br />

NF-κB activation and also DNA repair after double-stranded DNA breaks. Depletion<br />

of USP11 causes inhibition of TGFβ-induced epithelial to mesenchymal transition.<br />

USP12 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP12 requires interaction with WDR48 for high<br />

deubiquitinase activity. WDR48, in complex with deubiquitinase USP12, suppresses<br />

Akt-dependent cell survival signaling by stabilizing PHLPP1. USP12 and its activator<br />

UAF1 deubiquitinate nonactivated Notch.<br />

USP13 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP13 is part of an autophagy regulatory loop involving<br />

the deubiquitination of USP10 that leads to regulation of p53 stability. USP13 is a<br />

tumor suppressing protein that functions through deubiquitylation and stabilization<br />

of PTEN. USP13 also modulates STAT1 and plays a role in host defense against<br />

viral infection.<br />

USP14 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP14 is one of three proteasome-associated deubiquitinases,<br />

along with POH and UCHL5. USP14 is thought to antagonize substrate<br />

degradation as a part of the proteasome.<br />

USP15 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP15 preferentially cleaves K48-linked polymers.<br />

USP15 deubiquitination protects APC and human papillomavirus type 16 protein E6<br />

target proteins against proteasomal degradation. USP15 is a critical regulator of the<br />

TRIM25- and RIG-I-mediated antiviral immune response. USP15 also regulates the<br />

TGF-β pathway and is a key factor in glioblastoma pathogenesis.<br />

USP16 H2A USP16 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP16 acts as a transcriptional co-activator by<br />

specifically targeting H2A for deubiquitination. USP16 deubiquitination of H2A<br />

is also required for entry into mitosis.<br />

USP17<br />

RIG-I, MDA-<br />

5, SDS3<br />

USP17 regulates virus-induced type I IFN signaling through deubiquitination of<br />

RIG-I and MDA5. USP17 specifically deubiquitinates Lys-63-linked ubiquitin chains<br />

from SDS3.<br />

USP18 TAK1, TAB1 USP18 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP18 catalyzes the removal of ISG15, an interferonregulated<br />

ubiquitin-like protein, which maintains the critical cellular balance of<br />

ISG15-conjugated proteins important for normal development and brain function.<br />

USP18 deubiquitinates the TAK1/TAB1 complex thus inhibiting NF-κB and NFAT<br />

activation during Th17 differentiation.<br />

USP19 RNF123 USP19 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP19 deubiquitinates target proteins involved in<br />

cell proliferation, myogenesis, regulation of hypoxia, and modulation of the ERAD<br />

protein degradation pathway.<br />

USP20<br />

VHL, DIO2,<br />

HIF1α<br />

USP20 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP20 cleaves K48- and K63-linked chains. USP20<br />

deubiquitinates β2-adrenergic receptor (ADRB2) as well as target proteins involved<br />

in thyroid hormone regulation and regulation of hypoxia.<br />

9628861,<br />

16520378,<br />

17711858,<br />

23154984,<br />

24378640,<br />

22253573<br />

16322459,<br />

18254724,<br />

19135894,<br />

23184937,<br />

23097624,<br />

24591637,<br />

23690623,<br />

22371489<br />

19246359<br />

11439350,<br />

18632802,<br />

19398555,<br />

24270572,<br />

24332849<br />

15314155,<br />

17897950,<br />

18408009,<br />

24487962,<br />

22773947<br />

19075014,<br />

24145035,<br />

22778262<br />

9841226,<br />

24270891,<br />

23940278,<br />

24424410<br />

18162577,<br />

19135427,<br />

19106094,<br />

23754622,<br />

23615914<br />

16005295,<br />

19576224,<br />

24526689,<br />

24399297,<br />

23727018,<br />

23708518,<br />

23105109,<br />

22344298<br />

10077596,<br />

17914355<br />

20368735,<br />

21239494<br />

10777664,<br />

23825189<br />

19465887<br />

12056827,<br />

12865408,<br />

15776016<br />

www.cellsignal.com/csttables 137


Section I: Research Areas<br />

chapter 04: Cell Biology<br />

DUB Substrate Function PMID<br />

USP21<br />

USP22<br />

H2A, RIG-I,<br />

GATA-3<br />

ATXN7L3,<br />

NFATc2<br />

USP21 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP21 is also known as USP23. USP21 acts as a<br />

transcriptional co-activator by specifically targeting H2A for deubiquitination.<br />

USP21 is capable of removing the ubiquitin-like NEDD8 from NEDD8 conjugates.<br />

USP21 acts as a negative regulator in antiviral responses by binding and deubiquitinating<br />

RIG-I. USP21 also stabilizes the transcription factor GATA-3 by mediating its<br />

deubiquitination.<br />

USP22 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP22 deubiquitinates histones H2A and H2B as a<br />

component of the histone acetylation (HAT) complex SAGA. USP22 deubiquitinates<br />

specific targets required for transcription, nuclear receptor-mediated transactivation,<br />

and cell cycle progression. USP22 positively regulates NFATc2 through its<br />

deubiquitinase activity and promotes IL2 expression.<br />

10799498,<br />

24493797,<br />

23395819<br />

18206972,<br />

18206973,<br />

18469533,<br />

24561192<br />

USP23 H2A See USP21 10799498<br />

USP24 DDB-2 USP24 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. Mutations of the USP24 gene may correlate with risk<br />

of Parkinson’s disease. USP24 interacts with and regulates stability of the DNA<br />

damage specific protein, DDB-2.<br />

USP25<br />

ACTA1,<br />

MYBPC1,<br />

TRAF3,<br />

TRAF5,<br />

TRAF6<br />

USP25 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP25 cleaves both K48- and K63-linked chains.<br />

The USP25 muscle-specific isoform may have a role in the regulation of muscular<br />

differentiation and function. USP25 targets TRAF5 and TRAF6 for deubiquitination<br />

and thus negatively regulates IL-17-mediated signaling and inflammation.<br />

USP26 AR USP26 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP26 regulates the androgen receptor signaling<br />

pathway by targeting the androgen receptor for deubiquitination.<br />

USP27X<br />

USP27X is an X-linked member of the ubiquitin-specific processing protease (USP/<br />

USB) deubiquitinase superfamily.<br />

USP28<br />

P53bp1,<br />

Chk2, LSD1<br />

USP28 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP28 can bind to and deubiquitinate several target<br />

proteins in the DNA damage pathway, resulting in their stability, including p53BP1<br />

and Chk2. USP28 also plays an important role in Myc related signaling by binding<br />

through FBW7α to Myc. USP28 stabilizes LSD1 via deubiquitination, and USP28<br />

overexpression has been linked to the upregulation of LSD1 upregulation in multiple<br />

cancer cell lines and breast tumor samples.<br />

USP29 Claspin USP29 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP29 helps regulate the ATR-Chk1 pathway and the<br />

control of DNA replication via Claspin deubiquitination.<br />

USP30<br />

USP30 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP30 may participate in the maintenance of mitochondrial<br />

morphology.<br />

USP31<br />

USP31 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily.<br />

USP32<br />

USP32 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP32 is highly expressed in breast cancer cell lines<br />

and may be involved in tumorigenesis.<br />

USP33<br />

USP34<br />

USP35<br />

USP36<br />

USP37<br />

USP38<br />

ADRB2,<br />

CP110,<br />

RalB<br />

AXIN1,<br />

AXIN2<br />

RNA Polymerase<br />

I<br />

FZR1/CDH1,<br />

PLZF/RARα<br />

USP33 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP33 is involved in cellular migration, β2-adrenergic<br />

receptor/ADRB2 recycling, and G protein-coupled receptor (GPCR) signaling. In<br />

addition, USP33 regulates centrosome biogenesis by deubiquitinating CP110.<br />

USP33 also regulates the autophagy and innate immune response of RalB through<br />

deubiquitination of RalB.<br />

USP34 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP34 acts as an activator of the Wnt signaling<br />

pathway downstream of the β-catenin destruction complex by deubiquitinating and<br />

stabilizing AXIN1 and AXIN2.<br />

USP35 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily.<br />

USP36 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP36 may play a role in the maintenance of stem<br />

cells and regulation of cellular differentiation. Furthermore, USP36 regulates rRNA<br />

production through control of RNA Polymerase I stability.<br />

USP37 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP37 antagonizes the anaphase-promoting complex<br />

(APC/C) during G1/S transition by mediating deubiquitination of cyclin A (CCNA1 and<br />

CCNA2), thereby promoting S phase entry. In addition, USP37 is involved in acute<br />

promyelocytic leukemia (APL) through regulating the stability of the oncogenic fusion<br />

protein PLZF/RARα.<br />

USP38 is a member of the ubiquitin-specific processing protease (USP/USB) deubiquitinase<br />

superfamily. USP38 is expressed in skeletal muscle and adrenal gland.<br />

16917932,<br />

23159851<br />

10612803,<br />

11597335,<br />

16501887,<br />

23674823,<br />

23042150<br />

20501646<br />

12838346<br />

17558397,<br />

16901780,<br />

24075993<br />

10958632,<br />

24632611<br />

18287522<br />

14715245<br />

12604796,<br />

20549504<br />

12865408,<br />

23486064,<br />

24056301<br />

21383061<br />

14715245<br />

22622177,<br />

22902402<br />

21596315,<br />

23208507<br />

19615732<br />

DUB Substrate Function PMID<br />

USP39<br />

USP39 is a member of the ubiquitin-specific processing protease (USP/USB) 11350945<br />

deubiquitinase superfamily. USP39 may play a role in mRNA splicing as a<br />

competitor of ubiquitin C-terminal hydrolases (UCHs).<br />

USP40<br />

USP40 may be a nonprotease homologue of the ubiquitin-specific processing 16917932<br />

protease (USP/USB) superfamily.<br />

USP41<br />

USP41 is a member of the ubiquitin-specific processing protease (USP/USB) 14715245<br />

deubiquitinase superfamily.<br />

USP42<br />

USP42 is a member of the ubiquitin-specific processing protease (USP/USB) 14715245<br />

deubiquitinase superfamily. USP42 may play a role in spermatogenesis.<br />

USP43<br />

USP43 is a member of the ubiquitin-specific processing protease (USP/USB) 14715245<br />

deubiquitinase superfamily.<br />

USP44 Cdc20,<br />

Histone H2B<br />

USP44 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP44 regulates the cell cycle by deubiquitination of<br />

17443180,<br />

22681888<br />

CDC20, leading to stabilization of the MAD2L1-CDC20-APC/C ternary complex and<br />

avoidance of premature anaphase entry. USP44 modulates H2B ubiquitylation thus<br />

regulating stem cell differentiation.<br />

USP45<br />

USP45 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily.<br />

14715245<br />

USP46<br />

USP47<br />

USP48<br />

GAD1/<br />

GAD67,<br />

PHLPP<br />

POLB,<br />

CDC25A,<br />

katanin-p60<br />

TRAF2,<br />

RELA, NHE3<br />

USP46 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP46 requires interaction with WDR48 for high<br />

deubiquitinase activity. USP46 may act by mediating the deubiquitination of<br />

GAD1/GAD67. USP46 also regulates Akt signaling in colon cancer through<br />

control of PHLPP activation.<br />

USP47 is a member of the ubiquitin-specific processing protease (USP/USB) deubiquitinase<br />

superfamily. USP47 regulates base excision repair by deubiquitinating<br />

monoubiquitinated DNA polymerase β (POLB). USP47 may also regulate cell growth<br />

and survival by targeting CDC25A. USP47 promotes axonal growth of cultured<br />

rat hippocampal neurons through specifically deubiquitinating and stabilizing<br />

katanin-p60.<br />

USP48 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP48 may be involved in the regulation of NF-κB<br />

activation by the TNF receptor superfamily via its interactions with RelA and TRAF2.<br />

USP48 can also prevent NHE3 degradation by deubiquitination and thus helps<br />

regulate blood pressure and sodium balance.<br />

19075014,<br />

22391563<br />

19966869,<br />

23904609<br />

16214042,<br />

24308971<br />

USP49 Histone H2B USP49 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP49 specifically regulates Histone H2B by deubiquitination<br />

14715245,<br />

23824326<br />

and is required for efficient cotranscriptional splicing of exons.<br />

USP50<br />

USP50 is a nonprotease homologue of the ubiquitin-specific processing protease 14715245<br />

(USP/USB) superfamily.<br />

USP51<br />

USP51 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily.<br />

14715245<br />

USP52 PAN3 USP52 is a member of the ubiquitin-specific processing protease (USP/USB)<br />

deubiquitinase superfamily. USP52 is a member of the Pan nuclease complex,<br />

which regulates mRNA stability.<br />

USP53<br />

USP53 is a nonprotease homologue of the ubiquitin-specific processing protease<br />

(USP/USB) superfamily.<br />

USP54<br />

USP54 is a nonprotease homologue of the ubiquitin-specific processing protease<br />

(USP/USB) superfamily.<br />

USPL1<br />

USPL1 is a nonprotease homologue of the ubiquitin-specific processing protease<br />

(USP/USB) superfamily.<br />

USPL2/<br />

CYLD<br />

NF-κB,<br />

HDAC6,<br />

RIP1<br />

CYLD deubiquitinase regulates inflammation and cell proliferation by down regulating<br />

NF-κB signaling through removal of ubiquitin chains from several NF-κB pathway<br />

proteins. CYLD is a negative regulator of proximal events in Wnt/β-catenin signaling<br />

and is a critical regulator of natural killer T cell development. In selenite-treated<br />

colorectal cancer cells, CYLD regulates RIP1 deubiquitination and triggers apoptosis.<br />

VCPIP1 VCP VCPIP1 (valosin containing protein p97/p47 complex-interacting protein) is a<br />

member of the deubiquitinating enzyme ovarian tumor domain (OTU) superfamily.<br />

VCPIP1 is necessary for VCP-mediated reassembly of Golgi stacks after mitosis.<br />

14583602,<br />

16284618<br />

14715245<br />

14715245<br />

12917689,<br />

12917690,<br />

24577083<br />

15037600<br />

138 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables 139


05<br />

Section I: Research Areas<br />

Cellular Metabolism<br />

Energy is indispensible for all forms of life, and glucose is the primary energy source for most cells of<br />

the body. Glucose metabolism is central to cell proliferation, growth, survival, and tumor progression.<br />

Insulin Signaling and Glucose Transport<br />

The maintenance of glucose homeostasis is an essential physiological process that is regulated<br />

by hormones. An elevation in blood glucose levels during feeding stimulates insulin release from<br />

pancreatic β cells through a glucose-sensing pathway. Insulin stimulates glucose uptake from blood<br />

into skeletal muscle, cardiac muscle, and adipose tissue through a signaling cascade mediated by the<br />

insulin receptor (IR). Insulin binding to the IR results in activation of the insulin receptor substrate (IRS)<br />

proteins and subsequent signaling to the PI3K/Akt and Erk1/2 pathways, resulting in translocation of<br />

Glut4 from vesicles to the plasma membrane, glucose uptake, cell proliferation, and survival. Glut2,<br />

on the other hand, mediates glucose transport across liver cell membrane where glucose is converted<br />

to glycogen. When blood glucose levels are high, insulin suppresses glucose production in the liver.<br />

When blood glucose levels are low, liver cells degrade glycogen to glucose and/or synthesize glucose<br />

from noncarbohydrate substrates to release it into the blood. Aberrant insulin signaling can result in<br />

diabetes, obesity, atherosclerosis, and neurodegenerative disease.<br />

The insulin-like growth factor-I (IGF-I) receptor is similar in structure to the IR and can exist either as a<br />

homodimer or as a heterodimer with the IR. IGF-I receptor uses a similar signaling cascade as the IR to<br />

promote cell proliferation and survival.<br />

AMPK Signaling<br />

During conditions of glucose deprivation when cellular ATP levels fall, the serine/threonine kinase<br />

AMPK becomes active. AMPK is an energy sensor that is activated by an elevated AMP:ADP/ATP ratio<br />

due to cellular and environmental stress, such as low glucose, heat shock, hypoxia, or ischemia. AMPK<br />

activation positively regulates signaling pathways that replenish cellular ATP supplies. For example,<br />

activation of AMPK enhances both the transcription and translocation of Glut4, resulting in an increase<br />

in insulin-stimulated glucose uptake. It also stimulates catabolic processes such as fatty acid oxidation<br />

and glycolysis via inhibition of ACC and activation of PFK2. AMPK also activates ULK1 to trigger<br />

autophagy, which recycles cellular components for energy production. AMPK negatively regulates<br />

several proteins central to ATP-consuming processes such as mTORC2, glycogen synthase, SREBP-1,<br />

and TSC2, resulting in the downregulation or inhibition of gluconeogenesis and of glycogen, lipid, and<br />

protein synthesis.<br />

ACC, a key enzyme in the biosynthesis and<br />

oxidation of fatty acids, is commonly phosphorylated<br />

at Ser79 in breast carcinoma.<br />

Phospho-Acetyl-CoA Carboxylase (Ser79) (D7D11) Rabbit mAb #11818:<br />

IHC analysis of paraffin-embedded human breast carcinoma using #11818.<br />

chapter 05: cellular metabolism<br />

Oligomycin treatment<br />

results in phosphorylation<br />

of AMPKα at<br />

Thr172.<br />

kDa<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

– +<br />

Phospho-<br />

AMPKα<br />

(Thr172)<br />

AMPKα<br />

Oligomycin<br />

Phospho-AMPKα (Thr172) (40H9)<br />

Rabbit mAb #2535: WB analysis of<br />

extracts from C2C12 cells, untreated<br />

or oligomycin-treated (0.5 µM), using<br />

#2535 (upper) or AMPKα Antibody<br />

#2532 (lower).<br />

Growth Factors<br />

for IGF-I/II Signaling<br />

#8917 Human Insulin-like Growth<br />

Factor I (hIGF-I)<br />

#5238 Human Insulin-like Growth<br />

Factor II (hIGF-II)<br />

#9897 Mouse Insulin-like Growth<br />

Factor I (mIGF-I)<br />

Insulin expression in rat pancreas<br />

Stimulation of cells with IGF-I or insulin<br />

results in phosphorylation of IGF-I receptor<br />

β at Tyr1135/1136 and insulin receptor β at<br />

Tyr1150/1151.<br />

Phospho-IGF-I-Receptor β (Tyr1135/1136)/Insulin Receptor β (Tyr1150/1151)<br />

(19H7) Rabbit mAb #3024: WB analysis of extracts from untreated and IGF-treated<br />

HeLa cells and untreated and insulin-treated H-4-II-E cells using #3024.<br />

Insulin (C27C9) Rabbit mAb #3014: Confocal IF analysis of rat pancreas<br />

using #3014 (green). Blue pseudocolor = DRAQ5 ® #4084 (fluorescent DNA dye).<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

HeLa<br />

H-4-II-E<br />

Phospho-IGF-I<br />

Receptor β/Insulin<br />

Receptor β<br />

Cancer Metabolism<br />

The role of metabolism in cancer has been a focus of extensive research. In the 1920s, Otto Warburg<br />

observed that tumor cells produce energy by undergoing high rates of glycolysis and lactate fermentation<br />

in the cytosol in the presence of oxygen, a condition usually found only in the oxygen-deprived<br />

state. In contrast, normal cells typically have lower rates of glycolysis followed by oxidation of pyruvate<br />

in mitochondria. This cancer-associated phenomenon was later termed the Warburg Effect. Several<br />

signaling pathways such as Akt, Erk1/2, and AMPK converge on the key glycolytic enzymes phosphofructokinase<br />

(PFK) and pyruvate kinase M2 isoform (PKM2) to regulate ATP synthesis. The transcription<br />

factors c-Myc and p53 regulate glutamine metabolism, lipid metabolism, and the pentose phosphate<br />

pathway, creating the cellular components (nucleotides, lipids, proteins, Krebs Cycle intermediates)<br />

necessary for supporting and sustaining rapid tumor cell growth.<br />

Hexokinase II, an enzyme that catalyzes the first<br />

step in glycolysis, is expressed in lung carcinoma.<br />

Hexokinase II (C64G5) Rabbit mAb #2867: IHC analysis of paraffin-embedded human<br />

lung carcinoma using #2867.<br />

Glycolytic enzyme<br />

PKM2 is expressed in<br />

cancer cells.<br />

PKM2 (D78A4) XP ® Rabbit mAb<br />

#4053: IHC analysis of paraffin-embedded<br />

human lung carcinoma using #4053.<br />

20<br />

– + – –<br />

– – – +<br />

IGF<br />

Insulin<br />

Chemical Modulators<br />

for AMPK Signaling<br />

Insulin stimulation results in phosphorylation<br />

of AS160 at Thr642, a key event in insulinstimulated<br />

glucose transport.<br />

Phospho-AS160 (Thr642) (D27E6) Rabbit mAb #8881: WB analysis of extracts<br />

from serum starved HeLa cells, untreated (-) or insulin-treated (150 nM, 15 min), using<br />

#8881 (upper) or AS160 (C69A7) Rabbit mAb #2670 (lower). The phospho-specificity<br />

of the antibody is verified by λ phosphatase treatment.<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

– +<br />

– –<br />

– + Insulin<br />

+ + λ phosphatase<br />

Phospho-AS160<br />

(Thr642)<br />

AS160<br />

Select Reviews<br />

Burkewitz, K., Zhang, Y., and Mair, W.B. (2014) Cell Metab. 20, 10−25. • Guo, S. (2014) J. Endocrinol. 220, T1−T23. • Iqbal,<br />

M.A., Gupta, V., Gopinath, P., et al. (2014) FEBS Lett. 588, 2685–2692. • Metallo, C.M. and Vander Heiden, M.G. (2013) Mol.<br />

Cell 49, 388−398. • Mouchiroud, L., Eichner, L.J., Shaw, R.J., et al. (2014) Cell Metab. 20, 26−40. • Richter, E.A. and<br />

Hargreaves, M. (2013) Physiol. Rev. 93, 993−1017. • Ruderman, N.B., Carling, D., Prentki, M., et al. (2013) J. Clin. Invest.<br />

123, 2764−2772. • Yang, W. and Lu, Z. (2013) Cancer Lett. 339, 153−158. • Zhang, C., Liu, J., Liang, Y., et al. (2013) Nat.<br />

Commun. 4, 2935.<br />

#9944 AICAR<br />

AMP analog that<br />

activates AMPK<br />

#9996 Oligomycin<br />

ATP synthase inhibitor; inhibits<br />

oxidative phosphorylation;<br />

Activates AMPK<br />

140 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstcellularmetabolism 141


Section I: Research Areas<br />

These protein targets represent key<br />

nodes within cellular metabolism signaling<br />

pathways and are commonly studied<br />

in cellular metabolism research. Primary<br />

antibodies, antibody conjugates, and<br />

antibody sampler kits containing these<br />

targets are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

E PathScan ® ELISA Kits<br />

S SignalSilence ® siRNA<br />

C Antibody Conjugate<br />

Cytoplasmic protein<br />

IDH1 catalyzes the<br />

oxidative decarboxylation<br />

of isocitrate to<br />

α-ketoglutarate and<br />

functions as a tumor<br />

suppressor.<br />

IDH1 (D2H1) Rabbit mAb #8137:<br />

Confocal IF analysis of 293T cells using<br />

#8137 (green). Blue pseudocolor =<br />

DRAQ5 ® #4084 (fluorescent DNA dye).<br />

Commonly Studied Cellular Metabolism Targets Target M P E S C<br />

Phospho-IGF-I Receptor β (Tyr980) •<br />

Target M P E S C Target M P E S C<br />

Phospho-IGF-I Receptor β (Tyr1131)/ • •<br />

4F2hc/CD98<br />

•<br />

C1QBP<br />

• •<br />

Insulin Receptor β (Tyr1146)<br />

ABCC4<br />

• •<br />

CA2<br />

•<br />

Phospho-IGF-I Receptor β (Tyr1135) •<br />

ABCG2<br />

•<br />

CA9<br />

•<br />

Phospho-IGF-I Receptor β • •<br />

ACAD9<br />

•<br />

CA12<br />

•<br />

(Tyr1135/1136)/Insulin Receptor β<br />

ACAD10<br />

• CAD<br />

•<br />

(Tyr1150/1151)<br />

ACAT2<br />

•<br />

Phospho-CAD (Ser1859) •<br />

Phospho-IGF-I Receptor β (Tyr1316) •<br />

Acetyl-CoA Carboxylase • • • • Catalase<br />

•<br />

IGFBP-2<br />

•<br />

Phospho-Acetyl-CoA Carboxylase • • • • CLIC4<br />

•<br />

IGFBP-3<br />

•<br />

(Ser79)<br />

COX IV • • •<br />

Insulin Receptor β • • •<br />

Acetyl-CoA Carboxylase 1 • • CPT1A<br />

•<br />

Phospho-Insulin Receptor β (Tyr1146) •<br />

Acetyl-CoA Carboxylase 2 •<br />

CRABP1<br />

•<br />

Phospho-Insulin Receptor β •<br />

AceCS1<br />

•<br />

(Tyr1150/1151)<br />

CCTα<br />

• •<br />

ACO2<br />

• •<br />

Phospho-Insulin Receptor β (Tyr1345)<br />

CTR1/SLC31A1<br />

•<br />

• •<br />

ACSL1<br />

• •<br />

Phospho-Insulin Receptor β (Tyr1361)<br />

CYP11A1<br />

•<br />

•<br />

ADH1<br />

•<br />

Insulin<br />

CYP3A4<br />

•<br />

• • •<br />

Adiponectin<br />

•<br />

C-Peptide<br />

DHCR24/Seladin-1 •<br />

• •<br />

AKR1C2<br />

•<br />

IRAP<br />

DLST<br />

• •<br />

• •<br />

ALDH1A1<br />

•<br />

IRS-1<br />

DPYD<br />

•<br />

• • • •<br />

Aldolase A • • •<br />

Phospho-IRS-1 (panTyr)<br />

Enolase-1<br />

• •<br />

•<br />

AMACR<br />

•<br />

Phospho-IRS-1 (Ser302)<br />

Enolase-2<br />

• •<br />

• • •<br />

AMPKα • • • •<br />

Phospho-IRS-1 (Ser307)<br />

ENPP1<br />

• •<br />

• •<br />

Phospho-AMPKα (Thr172) • • • •<br />

Phospho-IRS-1 (Ser318)<br />

FAAH1<br />

• •<br />

•<br />

AMPK-α1<br />

•<br />

Phospho-IRS-1 (Ser332/Ser336)<br />

FABP1<br />

•<br />

•<br />

Phospho-AMPKα1 (Ser485) • •<br />

Phospho-IRS-1 (Ser612)<br />

FABP4<br />

• •<br />

• • •<br />

Phospho-AMPKα1 (Ser485)/AMPKα2 •<br />

Phospho-IRS-1 (Ser636/Ser639)<br />

Fatty Acid Synthase • • • •<br />

•<br />

(Ser491)<br />

Phospho-IRS-1 (Ser789)<br />

FTH1<br />

•<br />

•<br />

AMPK-α2 • •<br />

Phospho-IRS-1 (Tyr895)<br />

Fetuin A<br />

•<br />

•<br />

AMPK-β1<br />

• •<br />

Phospho-IRS-1 (Ser1101)<br />

FHIT<br />

•<br />

•<br />

Phospho-AMPK-β1 (Ser108) •<br />

Phospho-IRS-1 (Tyr1222)<br />

Phospho-FHIT (Tyr114) •<br />

•<br />

Phospho-AMPK-β1 (Ser182) •<br />

IRS-2<br />

FoxA2/HNF3β<br />

• •<br />

• •<br />

AMPK-β1/β2<br />

•<br />

Phospho-IRS-2 (Tyr)<br />

FoxC2<br />

•<br />

•<br />

AMPK-β2<br />

•<br />

LAT1<br />

Fumarase<br />

•<br />

•<br />

AMPK-γ1<br />

•<br />

LDH-A<br />

Glucose-6-Phosphate Dehydrogenase • •<br />

• •<br />

AMPK-γ2<br />

•<br />

LDH-A/LDH-C<br />

GAPDH • •<br />

•<br />

AMPK-γ3<br />

•<br />

Phospho-LDH-A (Tyr10)<br />

GATA-2<br />

•<br />

•<br />

Amylase<br />

• •<br />

Lipin 1<br />

GFAT1<br />

• •<br />

•<br />

ANT2/SLC25A5<br />

•<br />

LKB1<br />

GFAT2<br />

•<br />

•<br />

AQP2<br />

•<br />

Phospho-LKB1 (Thr189)<br />

GLDC<br />

•<br />

•<br />

Arginase-1<br />

•<br />

Phospho-LKB1 (Ser334)<br />

Glucagon • • •<br />

•<br />

ARK5<br />

•<br />

Phospho-LKB1 (Ser428)<br />

Glut4<br />

• •<br />

•<br />

AS160<br />

• •<br />

LXR-β<br />

Glycogen Synthase • •<br />

•<br />

Phospho-AS160 (Ser318) •<br />

Malic Enzyme (Mitochondrial)<br />

Phospho-Glycogen Synthase (Ser641) •<br />

•<br />

Phospho-AS160 (Ser588) •<br />

MDH2<br />

GPX1<br />

• •<br />

• •<br />

Phospho-AS160 (Thr642) • •<br />

MDR1/ABCB1<br />

GUCY1A2<br />

•<br />

•<br />

ASCT2<br />

• •<br />

Mitofusin-1<br />

hERG1α<br />

•<br />

•<br />

ATF-4<br />

•<br />

Mitofusin-2<br />

Hexokinase I<br />

• •<br />

•<br />

ATGL<br />

• •<br />

MO25α/CAB39<br />

Hexokinase II<br />

• •<br />

•<br />

ATP Citrate Lyase<br />

• •<br />

MRP1/ABCC1<br />

HMOX2/HO-2<br />

•<br />

•<br />

Phospho-ATP Citrate Lyase (Ser455) •<br />

MRP2<br />

HNF-1α<br />

•<br />

• •<br />

ATPIF1<br />

• •<br />

MRP3/ABCC3<br />

HNF4α<br />

• •<br />

•<br />

BCAT1<br />

•<br />

MRP4/ABCC4<br />

HO-1<br />

• •<br />

•<br />

BCAT2<br />

•<br />

MTAP<br />

HSL<br />

•<br />

• •<br />

C/EBP-α<br />

• •<br />

NME1/NDKA<br />

Phospho-HSL (Ser563) •<br />

• •<br />

Phospho-C/EBPα (Ser21) •<br />

NPC1L1<br />

Phospho-HSL (Ser565) •<br />

•<br />

Phospho-C/EBPα (Thr222/226) •<br />

NQO1<br />

Phospho-HSL (Ser660) •<br />

•<br />

C/EBP-β<br />

•<br />

NRF1<br />

IDH1<br />

• •<br />

•<br />

Phospho-C/EBPβ (Ser105) •<br />

OGDH<br />

IDH2<br />

•<br />

•<br />

Phospho-C/EBPβ (Thr235) •<br />

O-GlcNAc<br />

IGF-I Receptor<br />

•<br />

• •<br />

C/EBP-δ<br />

•<br />

OGT<br />

IGF-I Receptor β • • • •<br />

•<br />

PANK2<br />

•<br />

142 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

Target M P E S C<br />

PANK4<br />

•<br />

PASK<br />

•<br />

PCK1<br />

•<br />

PCK2<br />

• •<br />

Pyruvate Dehydrogenase • •<br />

PDHK1<br />

•<br />

Pdx1<br />

•<br />

Perilipin<br />

• •<br />

PFKFB2<br />

•<br />

Phospho-PFKFB2 (Ser483) •<br />

PFKFB3<br />

•<br />

PFKL<br />

•<br />

PFKP<br />

• •<br />

PGAM1<br />

•<br />

PGC1α<br />

•<br />

PGD<br />

•<br />

PGRMC1<br />

• •<br />

PHGDH<br />

•<br />

PiT1/SLC20A1<br />

•<br />

PKM1<br />

•<br />

PKM1/2<br />

• •<br />

PKM2 • • •<br />

Phospho-PKM2 (Tyr105) •<br />

PLA2G1B<br />

•<br />

Prdx1<br />

•<br />

PTP1B<br />

•<br />

SCAP<br />

•<br />

SCD1<br />

• •<br />

SDHA<br />

• •<br />

SGLT1<br />

•<br />

SHMT1<br />

•<br />

SHMT2<br />

•<br />

SIK2<br />

•<br />

SLC4A4/NBC1<br />

•<br />

SLC7A11/xCT<br />

•<br />

SNARK/NUAK2<br />

•<br />

SOD2<br />

•<br />

Phospho-SREBP-1c (Ser372) •<br />

StAR<br />

•<br />

Succinyl-CoA Synthetase • •<br />

Synip<br />

•<br />

TBC1D1<br />

•<br />

Phospho-TBC1D1 (Thr590) •<br />

Phospho-TBC1D1 (Ser660) •<br />

Phospho-TBC1D1 (Ser700) •<br />

Thioredoxin 1<br />

• •<br />

Thioredoxin 2<br />

•<br />

Thymidine Kinase 1<br />

•<br />

Thymidylate Synthase • •<br />

TORC1/CRTC1<br />

• •<br />

Phospho-TORC1/CRTC1 (Ser151) •<br />

TORC2/CRTC2<br />

• •<br />

TORC3/CRTC3<br />

• •<br />

Transketolase<br />

•<br />

TRXR1<br />

•<br />

Tug<br />

•<br />

TRXR2/TXNRD2<br />

•<br />

Tyrosinase<br />

•<br />

UGT<br />

•<br />

VMS1<br />

•<br />

chapter 05: cellular metabolism<br />

220<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for Phospho-AMPK<br />

(Thr172) have been cited over 220 times<br />

in high-impact, peer-reviewed publications<br />

from the global research community.<br />

Select Citations:<br />

Murthy, A. et al. (2014) A Crohn’s<br />

disease variant in Atg16l1 enhances<br />

its degradation by caspase 3. Nature<br />

506, 456−462.<br />

Sakamaki, J. et al. ((2014) Role of the<br />

SIK2-p35-PJA2 complex in pancreatic<br />

beta-cell functional compensation.<br />

Nature Cell Biol. 16, 234−244.<br />

Liu, P.P. et al. (2014) Metabolic<br />

regulation of cancer cell side population<br />

by glucose through activation of<br />

the Akt pathway. Cell Death Differ. 21,<br />

124−135.<br />

Takayama, H. et al. (2014) Metformin<br />

suppresses expression of the selenoprotein<br />

P gene via an AMP-activated<br />

kinase (AMPK)/FoxO3a pathway in<br />

H4IIEC3 hepatocytes. J. Biol. Chem.<br />

289, 335−345.<br />

Wu, Y. et al. (2014) Phosphorylation of<br />

p53 by TAF1 inactivates p53-dependent<br />

transcription in the DNA damage<br />

response. Mol. Cell 53, 63−74.<br />

Wang, L. et al. (2014) Pten deletion<br />

in RIP-Cre neurons protects against<br />

type 2 diabetes by activating the<br />

anti-inflammatory reflex. Nat. Med.<br />

20, 484−492.<br />

Jang, Y.H. et al. (2014) Phospholipase<br />

D-mediated autophagic regulation is<br />

a potential target for cancer therapy.<br />

Cell Death Differ. 21, 533−546.<br />

Thompson, A.M. et al. (2014)<br />

Resveratrol induces vascular smooth<br />

muscle cell differentiation through<br />

stimulation of SirT1 and AMPK. PLoS<br />

One 9, e85495.<br />

Cheng, H. et al. (2014) A genetic<br />

mouse model of invasive endometrial<br />

cancer driven by concurrent loss of<br />

Pten and Lkb1 Is highly responsive<br />

to mTOR inhibition. Cancer Res. 74,<br />

15−23.<br />

Demir, U. et al. (2014) Metformin<br />

anti-tumor effect via disruption of the<br />

MID1 translational regulator complex<br />

and AR downregulation in prostate<br />

cancer cells. BMC Cancer 14, 52.<br />

Wang, W. et al. (2014) Human T-cell<br />

leukemia virus type 1 Tax-deregulated<br />

autophagy pathway and c-FLIP<br />

expression contribute to resistance<br />

against death receptor-mediated<br />

apoptosis. J. Virol. 88, 2786−2798.<br />

Spruiell, K. et al. (2014) Role of<br />

pregnane X receptor in obesity and<br />

glucose homeostasis in male mice.<br />

J. Biol. Chem. 289, 3244−3261.<br />

www.cellsignal.com/cstcellularmetabolism 143


Section I: Research Areas<br />

Insulin Receptor Signaling<br />

Glucose<br />

GLUT4<br />

PDK1<br />

Bad<br />

SGK<br />

Apoptosis<br />

ENaC<br />

Sodium<br />

Transport<br />

Cytoplasm<br />

Nucleus<br />

GLUT4<br />

Translocation<br />

GLUT4<br />

vesicle<br />

GLUT4<br />

Exocytosis<br />

PP1<br />

mTORC1<br />

GβL Raptor<br />

mTOR<br />

DEPTOR<br />

FoxO3<br />

SNARE<br />

Complex<br />

Synip<br />

TC10<br />

CIP4/2<br />

GSK-3<br />

SGK<br />

Crkll<br />

PP2A<br />

14-3-3<br />

AS160<br />

TBC1D1<br />

14-3-3<br />

GS<br />

Glycogen<br />

Synthesis<br />

Transcription<br />

CAP<br />

C3G<br />

Rac1<br />

ATP-citrate<br />

lyase<br />

FoxO4<br />

Flotillin<br />

Cav<br />

SREBP<br />

Fatty Acid<br />

Synthesis<br />

mTORC2<br />

Sin1 PRR5<br />

Rictor GβL<br />

mTOR<br />

DEPTOR<br />

Cbl<br />

APS<br />

EHD1<br />

EHBP1<br />

PKCλ/ζ<br />

Lipin1<br />

Akt2<br />

FoxO1<br />

PDK1<br />

PTEN<br />

SHIP<br />

PRAS40<br />

Apoptosis,<br />

Autophagy,<br />

Glucose and<br />

Lipid Metabolism<br />

4E-BP1<br />

PIP 3<br />

Akt<br />

TSC2<br />

TSC1<br />

Rheb<br />

p85<br />

PI3K<br />

p110<br />

144 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

Gab1<br />

SHP-2<br />

IRS<br />

Shc<br />

GRB2<br />

IRS-1<br />

eIF4E<br />

HSL<br />

Protein Synthesis,<br />

Growth, and SREBP-1<br />

Proliferation<br />

Lipolysis<br />

Lipin1<br />

Akt2<br />

mTORC2<br />

mTORC1<br />

Fatty Acid<br />

and Cholesterol<br />

Synthesis<br />

PKCλ/ζ<br />

LKB1<br />

AMPK<br />

SREBP<br />

p70<br />

S6K<br />

Erk1/2<br />

LXRα<br />

PDE3B<br />

[cAMP]<br />

PKA<br />

USF<br />

Insulin Receptor<br />

PKCθ<br />

FFA<br />

NO<br />

SIK2<br />

SOCS3<br />

Nck<br />

Crkll<br />

Fyn<br />

IKK<br />

ROS<br />

CBP/p300<br />

PTP1B<br />

iNOS<br />

Jnk<br />

Torc2<br />

Disruption of<br />

CBP/Torc2/CREB<br />

Complex<br />

Gluconeogenesis<br />

TNFR1<br />

TNF<br />

GRB10<br />

Degradation<br />

SOS<br />

Ras<br />

c-Raf<br />

MEK1/2<br />

Erk1/2<br />

Erk1/2<br />

Growth<br />

Insulin is the major hormone controlling critical energy functions such as glucose and lipid metabolism. Insulin activates the insulin receptor tyrosine kinase (IR), which<br />

phosphorylates and recruits different substrate adaptors such as the IRS family of proteins. Tyrosine phosphorylated IRS then displays binding sites for numerous signaling<br />

partners. Among them, PI3K has a major role in insulin function, mainly via the activation of the Akt/PKB and the PKCζ cascades. Activated Akt induces glycogen synthesis<br />

through inhibition of GSK-3; protein synthesis via mTOR and downstream elements; and cell survival through inhibition of several pro-apoptotic agents (Bad, FoxO transcription<br />

factors, GSK-3, and MST1). Akt phosphorylates and directly inhibits FoxO transcription factors, which also regulate metabolism and autophagy. Inversely, AMPK is known to<br />

directly regulate FoxO3 and activate transcriptional activity. Insulin signaling also has growth and mitogenic effects, which are mostly mediated by the Akt cascade as well as<br />

by activation of the Ras/MAPK pathway. The insulin signaling pathway inhibits autophagy via the ULK1 kinase, which is inhibited by Akt and mTORC1, and activated by AMPK.<br />

Insulin stimulates glucose uptake in muscle and adipocytes via translocation of GLUT4 vesicles to the plasma membrane. GLUT4 translocation involves the PI3K/Akt pathway<br />

and IR-mediated phosphorylation of CAP, and formation of the CAP:CBL:CRKII complex. In addition, insulin signaling inhibits gluconeogenesis in the liver, through disruption<br />

of CREB/CBP/mTORC2 binding. Insulin signaling induces fatty acid and cholesterol synthesis via the regulation of SREBP transcription factors. Insulin signaling also promotes<br />

fatty acid synthesis through activation of USF1 and LXR. A negative feedback signal emanating from Akt/PKB, PKCζ, p70 S6K, and the MAPK cascades results in serine<br />

phosphorylation and inactivation of IRS signaling.<br />

Select Reviews:<br />

Altarejos, J.Y. and Montminy, M. (2011) Nat. Rev. Mol. Cell Biol. 12, 141–151. • Cheng, Z., Tseng, Y., and White, M.F. (2010) Trends Endocrinol. Metab. 21, 589–958.<br />

• Fritsche, L., Weigert, C., Häring, H.U., and Lehmann, R. (2008) Curr. Med. Chem. 15, 1316–1329. • Guo, S. (2014) J. Endocrinol. 220,T1–T23. • Rowland, A.F.,<br />

Fazakerley, D.J., and James, D.E. (2011) Traffic 12, 672–681. • Siddle, K. (2011) J. Mol. Endocrinol. 47, R1–R10. • Shao, W. and Espenshade, P.J. (2012) Cell Metab.<br />

16, 414–419. • Wong, R.H. and Sul, H.S. (2010) Curr. Opin. Pharmacol. 10, 684–691.<br />

© 2003–2015 Cell Signaling Technology, Inc. • We would like to thank Ashley Webb and Prof. Anne Brunet Stanford University, Palo Alto, CA for reviewing this diagram.<br />

Warburg Effect<br />

Pentose<br />

Phosphate<br />

Shunt<br />

NADPH<br />

Ribulose-5P<br />

Nucleotide<br />

Synthesis<br />

NADPH<br />

Folate<br />

Metabolism<br />

6-P-<br />

NADP<br />

Gluconate<br />

Glycine<br />

NADP<br />

Macropinocytosis and<br />

other scavenging pathways<br />

Malic Enzyme<br />

Akt<br />

Amino Acids<br />

and Lipids<br />

Serine<br />

Malate<br />

Hexokinase<br />

NADP<br />

PHGDH<br />

OMM Stability<br />

Inhibition Cytochrome C Release<br />

Inhibition of Apoptosis<br />

Bax<br />

NADPH<br />

NADPH<br />

6-P-<br />

Gluconolactone<br />

p53<br />

ANT<br />

VDAC<br />

VDAC<br />

Lactate<br />

ANT<br />

TIGAR<br />

HIF-1α<br />

c-Myc<br />

NADP<br />

Malate<br />

Glucose<br />

Transporters<br />

Hexokinase<br />

Glucose-6-P<br />

Fructose-6-P<br />

PFK<br />

Fructose<br />

Bisphosphate<br />

3-Phosphoglycerate<br />

p53<br />

Glycolysis<br />

Glucose<br />

PEP<br />

PKM2<br />

Pyruvate<br />

IDH2<br />

Ras<br />

Citrate<br />

UCP2<br />

Ras<br />

LDHA<br />

Pyruvate PDHK<br />

Dehydrogenase<br />

Pyruvate<br />

Acetyl-CoA<br />

Oxaloacetate<br />

Krebs<br />

Cycle<br />

α-Ketoglutarate<br />

AMPK<br />

c-Myc<br />

c-Myc<br />

ULK1<br />

NADP<br />

Glutaminase<br />

NADPH<br />

Glutamine Transporters<br />

Glutamine<br />

Glutaminolysis<br />

LKB1<br />

Protein Synthesis<br />

chapter 05: cellular metabolism<br />

Growth Factors<br />

PI3K<br />

Akt<br />

mTOR<br />

Autophagy<br />

β-Oxidation<br />

Citrate<br />

ACL<br />

Acetyl-CoA<br />

IDH1 ACC<br />

Epigenetic<br />

Regulation<br />

Ras<br />

Amino Acids<br />

SREBP<br />

HIF-1α<br />

Cell Proliferation<br />

MEK1/2<br />

Erk1/2<br />

c-Myc<br />

Fatty Acid Oxidation<br />

Fatty Acids<br />

Acetate<br />

ACSS2<br />

AMPK<br />

Fatty Acid/<br />

Lipid Synthesis<br />

Cancer cells rely on a variety of metabolic fuels, and the specific nutrients used are impacted by both the genetic and environmental context of the cancer cell.<br />

Most mammalian cells use glucose as a fuel source. Glucose is metabolized by glycolysis in a multistep set of reactions resulting in the creation of pyruvate. In typical cells<br />

under normal oxygen levels, much of this pyruvate enters the mitochondria where it is oxidized by the Krebs Cycle to generate ATP to meet the cell’s energy demands. However,<br />

in cancer cells or other highly proliferative cell types, much of the pyruvate from glycolysis is directed away from the mitochondria to create lactate through the action of<br />

lactate dehydrogenase (LDH/LDHA)—a process typically reserved for the low oxygen state. Lactate production in the presence of oxygen is termed “aerobic glycolysis” or the<br />

Warburg Effect.<br />

Cancer cells frequently use glutamine as another fuel source, which enters the mitochondria and can be used to replenish Krebs Cycle intermediates or to produce more pyruvate<br />

through the action of malic enzyme. Highly proliferative cells need to produce excess lipid, nucleotide, and amino acids for the creation of new biomass. Excess glucose is<br />

diverted through the pentose phosphate shunt (PPS) and serine/glycine biosynthesis pathway to create nucleotides. Fatty acids are critical for new membrane production and<br />

are synthesized from citrate in the cytosol by ATP-citrate lyase (ACL) to generate acetyl-CoA. Acetate can also be a source of carbon for acetyl-CoA production when available.<br />

De novo lipid synthesis requires NADPH reducing equivalents, which can be generated through the actions of malic enzyme, IDH1, and also from multiple steps within the PPS<br />

pathway and serine/glycine metabolism. These reducing equivalents are also part of the defense against the increased levels of reactive oxygen species that are characteristic<br />

of cancer cells. There is also evidence that some cancer cells can scavenge extracellular protein, amino acids, and lipids. Macropinocytosis, a process that allows bulk uptake<br />

of extracellular material that can be delivered to the lysosome, is one way the cells can catabolize extracellular material and provide nutrients for cell metabolism. These<br />

nutrients can generate ATP or NADPH, or contribute directly to biomass.<br />

Several signaling pathways contribute to the Warburg Effect and other metabolic phenotypes of cancer cells. Growth factor stimulation results in signaling through RTKs to<br />

activate PI3K/Akt and Ras. Akt promotes glucose transporter activity and stimulates glycolysis through activation of several glycolytic enzymes including hexokinase and phosphofructokinase<br />

(PFK). Akt phosphorylation of apoptotic proteins such as Bax makes cancer cells resistant to apoptosis and helps stabilize the outer mitochondrial membrane<br />

(OMM) by promoting attachment of mitochondrial hexokinase (mtHK) to the VDAC channel complex. RTK signaling to c-Myc results in transcriptional activation of numerous<br />

genes involved in glycolysis and lactate production. The p53 oncogene transactivates TP-53-induced Glycolysis and Apoptosis Regulator (TIGAR) and results in increased<br />

NADPH production by PPS. Signals impacting levels of hypoxia inducible factor (HIF) can increase expression of enzymes such as LDHA to promote lactate production, as<br />

well as pyruvate dehydrogenase kinase to inhibit the action of pyruvate dehydrogenase and limit entry of pyruvate into the Krebs Cycle. There is also increasing evidence that<br />

availability of metabolic substrates can influence gene expression by affecting epigenetic marks on histones and DNA.<br />

Select Reviews:<br />

Dang, C.V., Le, A., and Gao, P. (2009) Clin. Cancer Res. 15, 6479–6483. • Gogvadze, V., Zhivotovsky, B., and Orrenius, S. (2010) Mol. Aspects Med. 31, 60–74. •<br />

Hensley, C.T., Wasti, A.T., and DeBerardinis, R.J. (2013) J. Clin. Invest. 123, 3678–3684. • Kaelin, W.G. Jr. and McKnight, S.L. (2013) Cell 153, 56–69. • Lunt, S.Y. and<br />

Vander Heiden, M.G. (2011) Annu. Rev. Cell Dev. Biol. 27, 441–464. • Samudio, I., Fiegl, M., and Andreeff, M. (2009) Cancer Res. 69, 2163–2166. • Tennant, D.A.,<br />

Durán, R.V., and Gottlieb, E. (2010) Nat. Rev. Cancer 10, 267–277. • White, E. (2013) Genes Dev. 27, 2065–2071. • Vander Heiden, M.G., Cantley, L.C., and Thompson,<br />

C.B. (2009) Science 324, 1029–1033.<br />

© 2010–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Matthew G. Vander Heiden, Massachusetts Institute of Technology, Cambridge, MA for reviewing this diagram.<br />

www.cellsignal.com/cstpathways 145


Section I: Research Areas<br />

chapter 05: cellular metabolism<br />

AMPK Signaling<br />

Acute Regulation of Metabolism<br />

Glucose<br />

Glut4<br />

Glut<br />

Trafficking<br />

Glycogen<br />

Synthesis<br />

Glycolysis<br />

GS<br />

PFKFB3<br />

Sterol/Isoprenoid<br />

Synthesis<br />

Fatty Acid<br />

Oxidation<br />

Glut1<br />

Leptin<br />

Glut4<br />

vesicle<br />

HMG-CoA<br />

Reductase<br />

CPT1<br />

ATGL<br />

Lipolysis<br />

Glut1<br />

vesicle<br />

Glut<br />

Translocation<br />

HSL<br />

Adiponectin<br />

Receptor<br />

Malonyl<br />

CoA<br />

Tau<br />

CLIP170<br />

Microtubules<br />

Cytoskeletal Signaling<br />

TXNIP<br />

TBC1D1<br />

α-Adrenergic<br />

Receptor<br />

PLCβ Gα q<br />

[cAMP]<br />

AMP + ADP<br />

ATP<br />

PKA<br />

LKB1<br />

β γ<br />

AMPKα<br />

p300<br />

SirT1<br />

[Ca 2+ ]<br />

CaMKK<br />

ACC<br />

Myosin<br />

Light<br />

Chain2<br />

Actin<br />

Dynamics<br />

Brain<br />

MYPT1<br />

MBS85<br />

CRY1<br />

STRAD<br />

MO25α<br />

HNF4α<br />

HDAC4<br />

HDAC5<br />

HDAC7 TORC2/<br />

CRTC2<br />

FoxO<br />

Low Glucose,<br />

Hypoxia, Ischemia,<br />

Heat Shock<br />

AICAR<br />

Exercise<br />

Histamine<br />

Thrombin<br />

CREB<br />

PGC1α<br />

Gluconeogenesis<br />

Ras<br />

FIP200 ATG13<br />

SREBP-1<br />

B-Raf<br />

Erk<br />

p90RSK<br />

ULK1<br />

Other RTKs<br />

PI3K<br />

Akt<br />

TSC2<br />

TSC1<br />

Raptor<br />

mTOR GβL<br />

4E-BP1<br />

p70<br />

S6K<br />

Autophagy<br />

Beclin<br />

PI3K Atg14<br />

Class III<br />

Fatty Acid<br />

Synthase<br />

Lipogenesis<br />

Transcriptional Control of Metabolism<br />

Insulin<br />

Receptor<br />

AMP-activated protein kinase (AMPK) plays a key role as a master regulator of cellular energy homeostasis. The kinase is activated in response to stresses that deplete<br />

cellular ATP supplies such as low glucose, hypoxia, ischemia, and heat shock. It exists as a heterotrimeric complex composed of a catalytic α subunit and regulatory β and<br />

γ subunits. Binding of AMP to the γ subunit allosterically activates the complex, making it a more attractive substrate for phosphorylation on Thr172 in the activation loop of<br />

the α subunit by its major upstream AMPK kinase, LKB1. AMPK can also be directly phosphorylated on Thr172 by CaMKK2 in response to changes in intracellular calcium as<br />

occurs following stimulation by metabolic hormones including adiponectin and leptin.<br />

As a cellular energy sensor responding to low ATP levels, AMPK activation positively regulates signaling pathways that replenish cellular ATP supplies, including fatty acid<br />

oxidation and autophagy. AMPK negatively regulates ATP-consuming biosynthetic processes including gluconeogenesis, lipid and protein synthesis. AMPK accomplishes<br />

this through direct phosphorylation of a number of enzymes directly involved in these processes as well as through transcriptional control of metabolism by phosphorylating<br />

transcription factors, co-activators, and co-repressors.<br />

Due to its role as a central regulator of both lipid and glucose metabolism, AMPK is considered to be a potential therapeutic target for the treatment of type II diabetes mellitus,<br />

obesity, and cancer. AMPK has also been implicated in a number of species as a critical modulator of aging through its interactions with mTOR and sirtuins.<br />

Select Reviews:<br />

Cantó, C. and Auwerx, J. (2010) Cell. Mol. Life Sci. 67, 3407–3423. • Carling, D., Mayer, F.V., and Sanders, M.J., Gamblin SJ (2011) Nat. Chem. Biol. 512–518. • Hardie,<br />

D.G. (2011) Genes Dev. 25, 1895–1908. • Hardie, D.G., Ross, F.A., and Hawley, S.A. (2012) Chem. Biol. 19, 1222–1236. • Mihaylova, M.M. and Shaw, R.J. (2011) Nat.<br />

Cell Biol. 13, 1016–1023. • Steinberg, G.R. and Kemp, B.E. (2009) Physiol. Rev. 89, 1025–1078. • Zhang, B.B., Zhou, G., and Li, C. (2009) Cell Metab. 9, 407–416.<br />

Insulin<br />

Rheb<br />

Protein Metabolism and Autophagy<br />

Substrate<br />

ACC1<br />

AMPKA<br />

Isoform<br />

AMPKA1<br />

AMPKA2<br />

Published Data Human<br />

Organism Site Site Sequence (+/-7) PMID<br />

human S80 S80 LHIRssMsGLHLVkQ 17276402<br />

19176702<br />

18303014<br />

15371448<br />

AMPKA2 mouse S79 S80 FHMRSSMsGLHLVKQ 15866171<br />

AMPKA1 rat<br />

AMPKA2<br />

S79<br />

S1200<br />

S1215<br />

S80<br />

S1201<br />

S1216<br />

FHMRSsMsGLHLVKQ<br />

IPTLNRMsFASNLNH<br />

YGMTHVAsVSDVLLD<br />

12015362<br />

7915280<br />

1688796<br />

2900138<br />

1967580<br />

Substrate Function and<br />

Effect of Phosphorylation<br />

Acetyl-CoA carboxylase (ACC) catalyzes<br />

the carboxylation of acetyl-CoA to malonyl-<br />

CoA in the biosynthesis and oxidation<br />

of fatty acids. Phosphorylation by AMPK<br />

inhibits the enzymatic activity of ACC.<br />

ACC2 AMPKA1 human S222 S222 PTMRPSMsGLHLVKR 17276402 Acetyl-CoA carboxylase (ACC) catalyzes<br />

the carboxylation of acetyl-CoA to malonyl-<br />

CoA in the biosynthesis and oxidation<br />

of fatty acids. Phosphorylation by AMPK<br />

inhibits the enzymatic activity of ACC.<br />

AMPKA1 AMPKA1 human S360<br />

S486<br />

S494<br />

S496<br />

T183<br />

T388<br />

AMPKA1 rat S496<br />

T183<br />

T269<br />

AMPKB1 AMPKA1 human S24<br />

S108<br />

S174<br />

S177<br />

T80<br />

T158<br />

AMPKA1 rat S24<br />

S25<br />

S96<br />

S101<br />

S108<br />

S182<br />

S360<br />

S486<br />

S494<br />

S496<br />

T183<br />

T388<br />

S496<br />

T183<br />

T269<br />

S24<br />

S108<br />

S174<br />

S177<br />

T80<br />

T158<br />

S24<br />

S25<br />

S96<br />

S101<br />

S108<br />

S182<br />

LATsPPDsFLDDHHL<br />

DEItEAksGtAtPQR<br />

GtAtPQRsGsVsNYR<br />

AtPQRsGsVsNYRSC<br />

SDGEFLRtsCGsPNy<br />

EtPRARHtLDELNPQ<br />

AtPQRsGsVsNYRSC<br />

SDGEFLRtsCGsPNy<br />

VDPMKRAtIKDIREH<br />

HKtPRRDssGGTKDG<br />

sKLPLTRsHNNFVAI<br />

MVDSQKCsDVsELss<br />

SQKCsDVsELsssPP<br />

APAQARPtVFRWTGG<br />

NIIQVKKtDFEVFDA<br />

HKTPRRDssGGTKDG<br />

KTPRRDssGGTKDGD<br />

KEVYLSGsFNNWsKL<br />

SGsFNNWsKLPLTRs<br />

sKLPLTRsQNNFVAI<br />

DVSELSSsPPGPYHQ<br />

19376078 AMPK is a heterotrimeric complex<br />

composed of a catalytic α subunit and<br />

regulatory β and γ subunits, each of which<br />

is encoded by two or three distinct genes<br />

(α1, 2; β1, 2; γ1, 2, 3). The kinase is activated<br />

by an elevated AMP/ATP ratio due<br />

17728241<br />

17023420<br />

16340011<br />

9305909<br />

to cellular and environmental stress, such<br />

as heat shock, hypoxia, and ischemia. Accumulating<br />

evidence indicates that AMPK<br />

not only regulates the metabolism of fatty<br />

acids and glycogen, but also modulates<br />

protein synthesis and cell growth through<br />

EF2 and TSC2/mTOR pathways, as well<br />

as blood flow via eNOS/nNOS. AMPKA1<br />

phosphorylation is required for AMPK<br />

activation.<br />

19376078 AMPK is a heterotrimeric complex<br />

composed of a catalytic α subunit and<br />

regulatory β and γ subunits, each of which<br />

is encoded by two or three distinct genes<br />

(α1, 2; β1, 2; γ1, 2, 3). The kinase is activated<br />

by an elevated AMP/ATP ratio due<br />

9305909<br />

12764152<br />

9305909<br />

to cellular and environmental stress, such<br />

as heat shock, hypoxia, and ischemia. Accumulating<br />

evidence indicates that AMPK<br />

not only regulates the metabolism of fatty<br />

acids and glycogen, but also modulates<br />

protein synthesis and cell growth through<br />

EF2 and TSC2/mTOR pathways, as well as<br />

blood flow via eNOS/nNOS. The β1 subunit<br />

is post-translationally modified by multisite<br />

phosphorylation to regulate AMPK<br />

activation and localization.<br />

AS160 AMPKA2 mouse S711 S704 PSLHTSFsAPSFTAP 19923418 AS160 is a Rab GTPase-activating protein<br />

that regulates insulin-stimulated Glut4<br />

trafficking. Phosphorylation of AS160<br />

by AMPK is involved in the regulation of<br />

contraction-stimulated Glut4 translocation.<br />

CFTR AMPKA1 human S737<br />

S768<br />

S737<br />

S768<br />

EPLERRLsLVPDSEQ<br />

LQARRRQsVLNLMTH<br />

19095655<br />

19419994<br />

CFTR is a plasma membrane cyclic<br />

AMP activated chloride channel that is<br />

expressed in the epithelial cells of the lung<br />

and several other organs. CFTR channels<br />

are kept closed by AMPK mediated phosphorylation<br />

in non-stimulated epithelium.<br />

ChREBP AMPKA1 rat S568 S556 TLLRPPEsPDAVPEI 11724780 Carbohydrate-responsive element-binding<br />

protein (ChREBP) is a transcriptional<br />

repressor that regulates cellular energy homeostasis.<br />

AMPK phosphorylation inhibits<br />

the ability of CHREBP to bind DNA.<br />

CK1-E AMPKA1 human S389 S389 RGAPANVsssDLtGR 17525164 CK1-E (Casein Kinase I epsilon) is a<br />

member of a family of protein kinases<br />

implicated in multiple processes including<br />

DNA repair, cell morphology, and Wnt<br />

signaling. Multiple inhibitory autophosphorylation<br />

sites have been identified near<br />

the C-terminus of CK1-E, including an<br />

AMPK site that results in increased CK1-E<br />

activity.<br />

AMPK<br />

Substrates<br />

The table provides a list of substrates<br />

for AMPK, along with corresponding<br />

phosphorylation sites and references.<br />

This table was generated using<br />

PhosphoSitePlus ® , Cell Signaling<br />

Technology’s protein modification<br />

resource. See page 250 for more<br />

information on PhosphoSitePlus ® .<br />

© 2006–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Reuben Shaw, The Salk Institute for Biological Studies, La Jolla, CA, for reviewing this diagram.<br />

146 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables 147


Section I: Research Areas<br />

chapter 05: cellular metabolism<br />

Substrate<br />

AMPKA<br />

Isoform<br />

Published Data Human<br />

Organism Site Site Sequence (+/-7) PMID<br />

Substrate Function and<br />

Effect of Phosphorylation<br />

CRY1 AMPKA1 mouse S71 S71 ANLRKLNsRLFVIRG 19833968 CRY1 is a member of the DNA photolyase<br />

class-1 family that acts as a regulator of<br />

the circadian clock. CRY1 is regulated in<br />

rhythmic fashion by AMPK phosphorylation-induced<br />

degradation.<br />

eEF2K AMPKA1 human S398 S398 DSLPSsPsSATPHSQ 14709557 Eukaryotic elongation factor 2 kinase<br />

(eEF2k) phosphorylates and inactivates<br />

eEF2, resulting in the inhibition of peptidechain<br />

elongation. AMPK phosphorylation of<br />

eEF2K increases its ability to phosphorylate<br />

eEF2.<br />

eNOS AMPKA1 cow S1179 S1177 TSRIRtQsFSLQERH 12107173 Endothelial nitric-oxide synthase (eNOS)<br />

AMPKA1 human S633<br />

S1177<br />

AMPKA1 rat T494<br />

S1176<br />

S633,<br />

S1177<br />

T495,<br />

S1177<br />

WRRKRKEssNTDSAG<br />

TsRIRtQsFsLQERQ<br />

TGITRKKtFKEVANA<br />

TSRIRTQsFsLQERQ<br />

12791703<br />

17276402<br />

20479254<br />

10025949<br />

catalyzes the production of nitric oxide<br />

(NO), a key regulator of blood pressure,<br />

vascular remodeling, and angiogenesis.<br />

eNOS is activated by AMPK phosphorylating<br />

Ser1177 in response to various stimuli.<br />

GABBR1 AMPKA1 rat S948 S918 ELRHQLQsRQQLRSR 17224405 The metabotropic GABA(B) receptor is<br />

coupled to G proteins that modulate slow<br />

inhibitory synaptic transmission. Functional<br />

GABA(B) receptors form heterodimers<br />

of GABA(B)R1 and GABA(B)R2 where<br />

GABA(B)R1 binds the GABA ligand and<br />

GABA(B)R2 is the primary G protein contact<br />

site. AMPK mediated phosphorylation<br />

of GABA receptors increases activity as<br />

part of a neuroprotective mechanism.<br />

GABBR2 AMPKA1 rat S783 S784 VTSVNQAsTSRLEGL 17224405 The metabotropic GABA(B) receptor is<br />

coupled to G proteins that modulate slow<br />

inhibitory synaptic transmission. Functional<br />

GABA(B) receptors form heterodimers<br />

of GABA(B)R1 and GABA(B)R2 where<br />

GABA(B)R1 binds the GABA ligand and<br />

GABA(B)R2 is the primary G protein contact<br />

site. AMPK mediated phosphorylation<br />

of GABA receptors increases activity as<br />

part of a neuroprotective mechanism.<br />

GBF1 AMPKA1 human T1337 T1337 GKIHRsAtDADVVNs 18063581 Golgi-specific brefeldin A resistance factor<br />

1 promotes guanine nucleotide exchange<br />

in the Golgi apparatus. GBF1 phosphorylation<br />

by AMPK occurs in response to low<br />

glucose, resulting in Golgi disassembly<br />

and lowered intracellular levels of ATP.<br />

GFAT AMPKA1 human S261 S261 CNLsRVDsttCLFPV 17941647<br />

19170765<br />

GFAT, glutamine:fructose-6-phosphate<br />

aminotransferase 1, is the rate-limiting<br />

enzyme of the hexosamine biosynthesis<br />

pathway generating the building blocks for<br />

protein and lipid glycosylation. GFAT activity<br />

is regulated by AMPK phosphorylation.<br />

GYS1 AMPKA1 rabbit S8 S8 MPLSRTLsVSsLPGL 2567185 Glycogen synthase 1 (GYS1) is a key<br />

enzyme in the regulation of glycogen<br />

synthesis in muscle. AMPK mediated phosphorylation<br />

leads to inactivation of GYS1.<br />

H2B AMPKA1 human S37 S37 RKRsRkEsysIyVyk 20647423 The nucleosome, made up of four core<br />

histone proteins (H2A, H2B, H3, and H4),<br />

is the primary building block of chromatin.<br />

In response to metabolic stress, AMPK is<br />

recruited to responsive genes and phosphorylates<br />

histone H2B at S37, activating<br />

transcription.<br />

HAS2 AMPKA2 human T110 T110 LQSVKRLtYPGIKVV 21228273 Hyaluronan synthase 2 (HAS2) regulates<br />

the synthesis of hyaluronan (HA), an extracellular<br />

matrix protein involved in cell<br />

motility, proliferation, tumorigenesis, and<br />

inflammation. HAS2 phosphorylation by<br />

AMPK results in a loss of HAS2 enzymatic<br />

activity and impaired HA regulated functions.<br />

HDAC5<br />

AMPKA1,<br />

AMPKA2<br />

human S259,<br />

S498<br />

S259<br />

S498<br />

FPLRKTAsEPNLKVR<br />

RPLSRtQssPLPQsP<br />

18184930 Histone deacetylase 5 (HDAC5) acts as a<br />

repressor of transcription by removing histone<br />

tail acetylations, promoting a closed<br />

chromatin configuration. AMPK mediated<br />

phosphorylation inhibits the repression<br />

activity of HDAC5.<br />

Substrate<br />

AMPKA<br />

Isoform<br />

Published Data Human<br />

Organism Site Site Sequence (+/-7) PMID<br />

Substrate Function and<br />

Effect of Phosphorylation<br />

HNF4α AMPKA1 human S313 S313 GkIkRLRsQVQVsLE 12740371 Hepatocyte nuclear factor 4α (HNF4α) is<br />

a transcription factor that belongs to the<br />

steroid hormone receptor superfamily and<br />

regulates lipid homeostasis in the liver.<br />

AMPK phosphorylation of HNF4α inhibits<br />

dimer formation and DNA binding, resulting<br />

in increased protein degradation.<br />

HSL AMPKA1 human S855 S855 EPMRRsVsEAALAQP 16188906,<br />

2537200<br />

IKKβ AMPKA2 human S177<br />

S181<br />

S177<br />

S181<br />

AKELDQGsLCtsFVG<br />

DQGsLCtsFVGTLQy<br />

HSL (hormone-sensitive lipase) catalyzes<br />

the hydrolysis of triacylglycerol, the ratelimiting<br />

step in lipolysis. AMPK phosphorylation<br />

of HSL reduces HSL phosphorylation<br />

by PKA and inhibits HSL activity.<br />

21673972 The NF-κB/Rel transcription factors are<br />

present in the cytosol in an inactive state,<br />

in a complex with the inhibitory IκB proteins.<br />

IκB kinase (IKK) complex containing<br />

the IKKβ catalytic subunit targets IκB for<br />

proteasomal degradation. Activation of IKK<br />

depends upon AMPK phosphorylation of<br />

the activation loop of IKKβ.<br />

IRS1 AMPKA1 mouse S789 S794 QHLRLSSsSGRLRYT 11598104 Insulin receptor substrate 1 (IRS1) is one<br />

of the major substrates of the insulin<br />

receptor kinase. Insulin signaling pathway<br />

activity is increased by AMPK phosphorylation<br />

of IRS1.<br />

KCNMA1 AMPKA1 mouse S722 S722 GRSERDCsCMSGRVR 21209098 Calcium-activated potassium channel<br />

subunit a-1 (KCNMA1) is a K+ channel<br />

activated by membrane depolarization,<br />

increased cytosolic Ca2+, and cytosolic<br />

Mg2+. KCNMA1 regulates several<br />

membrane polarization activities, as well<br />

as acting as an oxygen mediator under<br />

hypoxic conditions. KCNMA1 is inhibited<br />

by AMPK phosphorylation in cell types that<br />

do not monitor oxygen levels.<br />

Kir6.2 AMPKA1 rat S385 S385 AKPKFSIsPDSLS__ 19357830 ATP-sensitive inward rectifier potassium<br />

channel 11 (Kir6.2) is a G protein mediated<br />

receptor that allows K+ to flow into<br />

the cell. The Kir6.2 channel is closed by<br />

AMPK mediated phosphorylation to allow<br />

insulin secretion in pancreatic beta cells.<br />

KLC1 AMPKA1 human S521 S521 ENMEkRRsREsLNVD 20074060 Kinesin light chain 1 (KLC1), also known as<br />

KNS2, is a motor protein that associates<br />

with microtubule components of the<br />

cytoskeleton. The intracellular trafficking<br />

of organelles may be regulated by AMPK<br />

mediated phosphorylation of KLC1.<br />

KLC2 AMPKA1 human S545<br />

S582<br />

S545<br />

S582<br />

GSLRRsGsFGKLRDA<br />

PRMKRAssLNFLNKs<br />

21725060 Kinesin light chain 2 (KLC2) is a motor<br />

protein that associates with microtubule<br />

components of the cytoskeleton. The<br />

intracellular trafficking of organelles<br />

may be regulated by AMPK mediated<br />

phosphorylation of KLC2.<br />

KPNA2 AMPKA1 human S105 S105 QAARKLLsREkQPPI 15342649 Importin subunit a-2 (KPNA2) is an adaptor<br />

subunit of the Importin nuclear protein<br />

import receptor. KPNA2 phosphorylation<br />

by AMPK is required for Importin nuclear<br />

import mediation activity.<br />

Kv2.1<br />

AMPKA1<br />

AMPKA2<br />

rat<br />

S444<br />

S541<br />

S444,<br />

S541<br />

ERAKRNGsIVsMNMK<br />

SKMAKTQsQPILNTK<br />

22006306 Potassium voltage-gated channel subfamily<br />

B member 1 (Kv2.1) mediates voltage<br />

dependent flow of K+ across membranes.<br />

Kv2.1 mediated action potential frequency<br />

is modulated under stress conditions via<br />

phosphorylation by AMPK.<br />

mTOR AMPKA1 mouse T2446 T2446 NKRsRtRtDsysAGQ 14970221 The mammalian target of rapamycin<br />

(mTOR) is a Ser/Thr protein kinase that<br />

functions as an ATP and amino acid<br />

sensor to balance nutrient availability and<br />

cell growth. AMPK phosphorylates mTOR<br />

in response to nutrient deprivation and<br />

inhibits mTOR response to growth factor<br />

phosphorylation.<br />

148 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables 149


Section I: Research Areas<br />

chapter 05: cellular metabolism<br />

AMPKA Published Data Human<br />

Substrate Function and<br />

Substrate Isoform Organism Site Site Sequence (+/-7) PMID Effect of Phosphorylation<br />

NKCC2 AMPKA1 human S130 S130 GPKVNRPsLLEIHEQ 19176702 NKCC2 is an electroneutral cation chloridecoupled<br />

efflux cotransporter that regulates<br />

AMPKA1 mouse S126 S130 GPKVNRPsLLEIHEQ 17341212<br />

cell volume and maintains cellular homeostasis<br />

in response to osmotic and oxidative<br />

stress. NKCC2 chloride efflux activity<br />

is inhibited by AMPK phosphorylation,<br />

thereby increasing intracellular chloride<br />

concentration in the kidney.<br />

p27Kip1 AMPKA1 human T198 T198 PGLRRRQt______ 17237771 p27 Kip1 is a member of the Cip/Kip family<br />

of cyclin-dependent kinase inhibitors<br />

AMPKA1 mouse S83 S83 WQEVERGsLPEFyYR 17237771<br />

T170 T170 QNKRANRtEENVSDG 18701472 that enforces the G1 restriction point via<br />

T197 T198 KPGLRRQt______ 20146253 its inhibitory binding to CDK2/cyclin E and<br />

other CDK/cyclin complexes. p27Kip1<br />

stability is increased by AMPK mediated<br />

phosphorylation, resulting in increased<br />

survival under stress conditions.<br />

p300 AMPKA1 human S89 S89 SELLRSGsSPNLNMG 11518699 The transcriptional coactivator p300 associates<br />

with transcriptional regulators and<br />

signaling molecules, integrating multiple<br />

signal transduction pathways with the<br />

transcriptional machinery. AMPK mediated<br />

phosphorylation represses p300 activity<br />

by disrupting the association of p300 with<br />

nuclear receptors.<br />

p53 AMPKA1 human S20<br />

T18<br />

150 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

S20<br />

T18<br />

PLsQEtFsDLWKLLP<br />

EPPLsQEtFsDLWKL<br />

AMPKA2 mouse S15 S15 IsLELPLsQEtFsGL 15866171<br />

17339337 The p53 tumor suppressor protein plays<br />

a major role in cellular response to DNA<br />

damage and other genomic aberrations.<br />

Activation of p53 can lead to either cell<br />

cycle arrest and DNA repair or apoptosis.<br />

DNA damage induces phosphorylation of<br />

p53 and leads to a reduced interaction<br />

between p53 and its negative regulator,<br />

the oncoprotein MDM2.<br />

PFKFB2 AMPKA1 human S466 S466 PVRMRRNsFtPLSSS 12853467 Phosphofructokinase (PFK) catalyzes the<br />

phosphorylation of fructose-6-phosphate<br />

in glycolysis. PFKFB2 initiated glycolysis is<br />

activated by AMPK phosphorylation.<br />

PFKFB3 AMPKA1 human S461 S461 NPLMRRNsVtPLAsP 12065600,<br />

15896703<br />

PGC-1α AMPKA2 mouse T177<br />

S538<br />

T178<br />

S539<br />

NHTHRIRtNPAIVkT<br />

SLFDVSPsCSSFNSP<br />

Phosphofructokinase (PFK) catalyzes the<br />

phosphorylation of fructose-6-phosphate<br />

in glycolysis. PFKFB2 initiated glycolysis is<br />

activated by AMPK phosphorylation.<br />

17609368 PGC-1α interacts with a diverse array of<br />

transcription factors to regulate adaptive<br />

thermogenesis, energy metabolism,<br />

glucose uptake, gluconeogenesis, insulin<br />

secretion, and mitochondrial biogenesis.<br />

PGC-1α activity in skeletal muscle is<br />

induced by AMPK-mediated phosphorylation.<br />

PLD1 AMPKA2 human S505 S505 GSVKRVTsGPsLGSL 20231899 Phosphatidylcholine-specific phospholipase<br />

D (PLD) hydrolyzes phosphatidylcholine<br />

(PC) to produce choline and<br />

phosphatidic acid (PA). PA is the precursor<br />

of the second messenger, diacylglycerol<br />

(DAG). PLD1 is activated by AMPK<br />

phosphorylation, leading to an increase<br />

in glucose uptake in muscle under stress<br />

deprivation conditions.<br />

PPP1R3C AMPKA1 human S33<br />

S293<br />

PPP2R5C AMPKA1 human S298<br />

S336<br />

S33<br />

S293<br />

S298<br />

S336<br />

MRLCLAHsPPVKSFL<br />

LESTIFGsPRLASGL<br />

KYWPKTHsPKEVMFL<br />

RQLAKCVsSPHFQVA<br />

19171932 Protein phosphatase 1 is a serine/<br />

threonine phosphatase holoenzyme<br />

composed of a catalytic subunit and an<br />

inhibitory regulatory subunit. PPP1R3C is a<br />

regulatory subunit that confers specificity<br />

for increasing glycogen synthesis. AMPK<br />

targets PPP1R3C for phosphorylation and<br />

proteasomal degradation, which inhibits<br />

glycogen synthesis.<br />

19366811 Protein phosphatase 2 is a tripartite<br />

serine/threonine phosphatase holoenzyme<br />

composed of a catalytic subunit, a<br />

structural subunit and a regulatory<br />

subunit. AMPK phosphorylates the regulatory<br />

subunit PPP2R5C, which results in<br />

dephosphorylation of the catalytic subunit<br />

and increased PP2A activity.<br />

AMPKA Published Data<br />

Substrate Isoform<br />

Raf1 AMPKA1 human S259<br />

S621<br />

Human<br />

Organism Site Site Sequence (+/-7) PMID<br />

S259<br />

S621<br />

sQRQRststPNVHMV<br />

PKINRsAsEPsLHRA<br />

Substrate Function and<br />

Effect of Phosphorylation<br />

9091312 Raf-1 (c-Raf) is recruited by GTP-bound<br />

Ras to activate the MEK-MAP kinase<br />

pathway. Inhibitory 14-3-3 protein binding<br />

sites on c-Raf can be phosphorylated by<br />

AMPK.<br />

raptor AMPKA1 human S792 S792 DKMRRASsYSsLNsL 18439900 The regulatory associated protein of mTOR<br />

(Raptor) was identified as an mTOR binding<br />

partner that mediates mTOR signaling<br />

to downstream targets. AMPK phosphorylation<br />

of raptor is essential for inhibition<br />

of the raptor-containing mTOR complex 1<br />

(mTORC1) and induces cell cycle arrest<br />

when cells are stressed for energy.<br />

Rb AMPKA1 mouse S804 S811 IYIsPLKsPyKIsEG 19217427 The retinoblastoma tumor suppressor<br />

protein, Rb, regulates cell proliferation by<br />

controlling progression through the restriction<br />

point within the G1-phase of the cell<br />

cycle. AMPK regulation of brain development<br />

is achieved by modulating control of<br />

the cell cycle via phosphorylation of Rb.<br />

smMLCK AMPKA1 chicken S1749 S1760 RAIGRLSsMAMISGM 18426792 Smooth muscle myosin light chain kinase<br />

(smMLCK) is activated by high Ca 2+ induced<br />

calcium/calmodulin. Smooth muscle<br />

contraction is activated smMLCK mediated<br />

phosphorylation of myosin light chains.<br />

Smooth muscle contraction is attenuated<br />

by AMPK phosphorylation and inactivation<br />

of smMLCK.<br />

TBC1D1 AMPKA1 human S237<br />

T596<br />

S237<br />

T596<br />

RPMRKSFsQPGLRsL<br />

AFRRRANtLsHFPIE<br />

17995453 TBC1D1 is a Rab GTPase activating protein<br />

involved in vesicle trafficking in response<br />

to insulin. AMPK acts in association with<br />

insulin and growth factor signaling to activate<br />

TBC1D1 mediated vesicle regulation.<br />

TIF-IA AMPKA1 human S635 S635 DTHFRsPsSSVGsPP 19815529 RNA polymerase I-specific transcription<br />

initiation factor RRN3 (TIF-IA) is required<br />

for RNA polymerase I initiation. Transcription<br />

of rRNA is inhibited during times of<br />

stress by AMPK phosphorylation inhibition<br />

of TIF-IA.<br />

TORC2 AMPKA1 mouse S171 S171 SALNRtssDsALHTs 16148943 Torc2 (transducer of regulated CREB activity<br />

2) functions as a CREB co-activator and<br />

is implicated in mediating the effects of<br />

hormone and glucose sensing pathways.<br />

Torc2 is phosphorylated by AMPK at<br />

Ser171 and becomes sequestered in the<br />

cytoplasm, inhibiting hepatic gluconeogenesis.<br />

TSC2 AMPKA1 rat S1389<br />

T1271<br />

S1387,<br />

T1271<br />

QPLsKSSsSPELQTL<br />

PTLPRSNtVASFSSL<br />

16959574<br />

14651849<br />

Tuberin is a product of the TSC2 tumor<br />

suppressor gene and an important regulator<br />

of cell proliferation and tumor development.<br />

AMPK phosphorylates tuberin during<br />

periods of energy starvation, enhancing<br />

tuberin activity and resulting in increased<br />

translation.<br />

ULK1 AMPKA1 human S638 S638 FDFPKtPssQNLLAL 21383122 ULK1 is a serine/threonine kinase involved<br />

AMPKA1 mouse S467 S467, sAIRRsGsttPLGFG 21205641 in axon growth, endocytosis of critical<br />

S555 S556 GLGCRLHsAPNLSDF<br />

growth factors such as NGF, and can act<br />

as a convergence point for multiple signals<br />

that control autophagy. AMPK, activated<br />

during low nutrient conditions, directly<br />

phosphorylates ULK1 at multiple sites to<br />

promote autophagy.<br />

VASP AMPKA1 human T278 T278 LARRRKAtQVGEktP 17082196 VASP belongs to the Ena/VASP family of<br />

adaptor proteins linking the cytoskeletal<br />

system to the signal transduction pathways<br />

and that it functions in cytoskeletal<br />

organization, fibroblast migration, platelet<br />

activation, and axon guidance. AMPK<br />

phosphorylation of VASP leads to specific<br />

cytoskeletal rearrangements.<br />

ZNF692 AMPKA1 human S470 S470 VAAHRSKsHPALLLA 17097062 ZNF692, also known as AREBP, is a zinc<br />

finger transcription factor involved in the<br />

expression of gluconeogenesis genes.<br />

ZNF692 DNA binding ability is abrogated<br />

by AMPK mediated phosphorylation during<br />

times of metabolic stress.<br />

www.cellsignal.com/csttables 151


06<br />

Section I: Research Areas<br />

ESC Signaling<br />

ESCs can be identified using markers specific<br />

to the pluripotency transcription factors, the<br />

cell surface glycoproteins SSEA3/4, and the<br />

TRA-1-60 and TRA-1-81 antigens.<br />

Development and Differentiation<br />

Embryogenesis is a complex and highly ordered program of proliferation and directed differentiation,<br />

during which a fertilized egg undergoes transformation from an undifferentiated single-cell entity<br />

(zygote) into a multicellular organism composed of numerous, functionally-distinct cell types. This<br />

transformation is under the control of an intricate array of epigenetic, genetic, and biochemical signaling<br />

cascades that collectively influence the fate and behavior of individual cells in a developmentally<br />

appropriate context.<br />

Embryonic Stem Cells<br />

ESCs<br />

Embryonic stem cells (ESCs) are derived from the inner cell mass of a cleavage-stage embryo, or blastocyst.<br />

ESCs exhibit a suite of distinct properties that make them an exceptionally powerful research<br />

tool in developmental biology and a source of significant therapeutic potential in regenerative medicine.<br />

These features include (1) pluripotency, defined as the ability to differentiate into any cell lineage of the<br />

body; and (2) the capacity for self-renewal, which allows them to be propagated indefinitely in culture.<br />

Distinct cell signaling pathways are responsible for endowing and maintaining these properties in<br />

ESCs. In humans, foremost among these are the BMP/TGF-β signaling pathway, which signals through<br />

SMAD proteins, and the FGF signaling pathway, which activates MAPK and Akt pathways. The Wnt signaling<br />

pathway has also been implicated in the maintenance of pluripotency, through a noncanonical<br />

Wnt signaling mechanism that serves to maintain a balance between the transcriptional activator TCF1<br />

and the repressor TCF3. Likewise, the Hippo signaling pathway also plays an important role in ESC<br />

biology. Down-regulation of the transcriptional co-activator YAP, a hallmark of Hippo pathway activation,<br />

results in a loss of ESC pluripotency, whereas over-expression of YAP prevents ESC differentiation,<br />

even in culture conditions that otherwise promote differentiation. The role of NOTCH signaling in ESC<br />

biology is somewhat less clear. Human (but not mouse) ESCs have an active Notch signaling pathway,<br />

and Notch inactivation results in reduced human ESC proliferation in culture, suggesting that Notch signaling<br />

may be required for ESC proliferation. This mechanism, however, does not appear to be widely<br />

conserved among mammals.<br />

membrane<br />

cytoplasm<br />

FGF2<br />

IGF<br />

LRP<br />

Wnt<br />

TGF-β/<br />

Activin/Nodal<br />

BMP4<br />

Cell-Cell<br />

Contact<br />

iPSCs<br />

Induced pluripotent stem cells (iPSCs) are pluripotent, ESC-like cells that can be experimentally derived<br />

from differentiated cells by forced expression of a defined set of reprogramming factors, the best known<br />

of which are Oct-4, Sox2, KLF4, and c-Myc (commonly known as the Yamanaka factors, after Shinya<br />

Yamanaka, the scientist who first generated iPSCs in 2006 [Takahashi, K. and Yamanaka, S. (2006)<br />

Cell 126, 663–676.]. In vitro reprogrammed iPSCs exhibit a gene expression signature similar to that<br />

of ESCs and, like their embryonic counterparts, are the subject of intense investigation due to their<br />

enormous potential for use in regenerative medicine, drug screening, and basic developmental biology<br />

research. Alternatively, iPSCs may be generated by autologous somatic cell nuclear transfer (SCNT).<br />

There is considerable interest in this approach, as SCNT-derived pluripotent cells exhibit an epigenetic<br />

signature that more closely resembles ESCs than do iPSCs generated by in vitro reprogramming.<br />

A B C<br />

D E F<br />

Transcription Factors and Development<br />

Transcription factors play an instrumental role in both pluripotency and cell fate specification (differentiation)<br />

by regulating the global expression patterns of developmentally important genes. In ESCs<br />

and iPSCs, pluripotency is induced and maintained by the expression of three key transcription factors:<br />

Oct-4, Sox2, and Nanog. These transcription factors promote a pluripotency gene expression signature,<br />

regulate their own expression through an auto-regulatory loop, and serve as important biochemical<br />

or molecular markers of pluripotency. Research studies are revealing a complex network of additional<br />

transcription factors that function to maintain pluripotency, through transcriptional regulation of the core<br />

pluripotency factor network.<br />

chapter 06: Development and differentiation<br />

Expression of<br />

pluripotency markers<br />

in human iPSCs<br />

StemLight Pluripotency Antibody<br />

Kit #9656: Projected confocal z-stack<br />

of human iPS cells using TRA-1-60(S)<br />

(TRA-1-60(S)) Mouse mAb (A), TRA-1-<br />

81 (TRA-1-81) Mouse mAb (B), SSEA4<br />

(MC813) Mouse mAb (C), Oct-4A<br />

(C30A3) Rabbit mAb (D), Sox2 (D6D9)<br />

XP ® Rabbit mAb (E) and Nanog (D73G4)<br />

XP ® Rabbit mAb (F). Actin filaments<br />

were labeled with DY-554 phalloidin<br />

(red). Blue pseudocolor = DRAQ5 ®<br />

#4084 (fluorescent DNA dye).<br />

nucleus<br />

PI3K<br />

Akt<br />

Erk1/2<br />

β-catenin Smad2/3 Smad1/5/9<br />

152 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

Oct-4<br />

Sox2<br />

Oct-4 Sox2 FoxD3<br />

Oct-4 Sox2 Nanog<br />

Oct-4,<br />

Sox2,<br />

FoxD3,<br />

FGF4<br />

Nanog<br />

gene<br />

expression<br />

YAP<br />

Pluripotency<br />

and Self Renewal<br />

hESC Markers:<br />

Oct-4<br />

Nanog<br />

Sox2<br />

SSEA 3/4<br />

TRA-1-60<br />

TRA-1-81<br />

Oct-4, Sox2, and Nanog pluripotency factors regulate<br />

their expression through an auto-regulatory network.<br />

SimpleChIP ® Stem Cell Master Regulator Assay Kit #8980:<br />

Chromatin IPs were performed with cross-linked chromatin<br />

from 4 x 10 6 NCCIT cells and 10 μl of Nanog, Oct-4, and Sox2<br />

antibodies or 2 μl of Normal Rabbit IgG, using SimpleChIP ®<br />

Enzymatic Chromatin IP Kit (Magnetic Beads) #9003. The<br />

enriched DNA was quantified by real-time PCR using human<br />

Nanog promoter primers, SimpleChIP ® Human Oct-4 Promoter<br />

Primers #4641, SimpleChIP ® Human Sox2 Promoter Primers<br />

#4649, and SimpleChIP ® Human α Satellite Repeat Primers<br />

#4486. The amount of immunoprecipitated DNA in each sample<br />

is represented as a percent of the total input chromatin.<br />

% of total input chromatin<br />

3.0<br />

2.5<br />

2.0<br />

1.5<br />

1.0<br />

0.5<br />

0<br />

Nanog Oct-4 Sox2 α Satellite<br />

Generation of Induced Pluripotent Stem Cells<br />

Download a free copy of ‘Generation of Induced Pluripotent Stem Cells’, a white paper written by Alison Becker<br />

from <strong>CST</strong> in collaboration with Ryan M. Walsh and Konrad Hochedlinger of the Harvard Stem Cell Institute.<br />

www.cellsignal.com/cstliterature<br />

Nanog (D73G4) XP ® Rabbit mAb<br />

(ChIP Formulated) #5232<br />

Oct-4A (C30A3C1) Rabbit mAb<br />

(ChIP Formulated) #5677<br />

Sox2 (D6D9) XP ® Rabbit mAb<br />

(ChIP Formulated) #5024<br />

Normal Rabbit IgG #2729<br />

www.cellsignal.com/cstdevelopment 153


Section I: Research Areas<br />

chapter 06: Development and differentiation<br />

GATA-6, a zinc finger<br />

protein that regulates<br />

endoderm development,<br />

is expressed in colon<br />

carcinoma (KM12) but<br />

not ovarian carcinoma<br />

(SK-OV-3) cells.<br />

Events<br />

Sox2 (Alexa Fluor ® 647 Conjugate)<br />

Pluripotency marker Sox2 is expressed<br />

in embryonic carcinoma NTERA-2 cells.<br />

Sox2 (D6D9) XP ® Rabbit mAb (Alexa Fluor ® 647 Conjugate) #5067:<br />

Flow cytometric analysis of HeLa cells (blue) and NTERA-2 cells (green)<br />

using #5067.<br />

While much of modern stem cell research has focused on the isolation or generation of pluripotent<br />

stem cells, more recent efforts have been directed towards understanding and exploiting the process<br />

of transdifferentiation, or direct reprogramming. Transdifferentiation describes a process whereby a<br />

terminally differentiated cell is converted into a functionally distinct cell type, without first proceeding<br />

through a pluripotent phase of development. Transdifferentiation is considered a promising avenue<br />

of exploration in regenerative medicine, as transdifferentiated cells could provide for direct in situ<br />

replacement of damaged cells without the need for in vitro differentiation, as required by iPSCs. Transdifferentiation<br />

can be experimentally induced by the forced expression of a combination of transcription<br />

factors, unique for each cell type. For example, mouse cardiac fibroblasts can be transdifferentiated<br />

into cardiomyocytes by forced expression of GATA-4, MEF2C, and Tbx5 (Qian, L. et al. (2012) Nature<br />

485, 593–598.).<br />

Miwi is expressed in<br />

mouse testes and is a<br />

marker for germ cells.<br />

Several additional transcription factor families regulate differentiation along lineage-specific pathways.<br />

These include members of the zinc finger, homeobox, forkhead box (Fox), helix-loop-helix (bHLH), T<br />

box, Paired box (Pax), and Sox protein families. Members within each group are classified by the presence<br />

of unique DNA binding domains, which bind to specific promoter regions to regulate expression<br />

of genes necessary for each stage of development. One example are the GATA proteins, an extremely<br />

large, highly conserved family of zinc finger proteins, the members of which play diverse and critically<br />

important roles throughout development. Similarly, homeobox transcription factors, the product of Hox<br />

genes, contain a helix-turn-helix homeodomain and are critical for regulating the transcription of genes<br />

that specify development along the anterior-posterior body axis.<br />

Germ cell marker DDX4 is expressed in mouse testes but not mouse brain.<br />

DDX4 (D10C5) Rabbit mAb #8761:<br />

Confocal IF analysis of mouse testes<br />

(left) and mouse brain (right) using<br />

#8761 (green). Actin filaments were<br />

labeled with DY-554 phalloidin (red).<br />

Blue pseudocolor = DRAQ5 ® #4084<br />

(fluorescent DNA dye).<br />

Miwi (D92B7) XP ® Rabbit mAb #6915:<br />

Confocal IF analysis of mouse testes<br />

using #6915 (green). Actin filaments<br />

were labeled with DY-554 phalloidin (red).<br />

Blue pseudocolor = DRAQ5 ® #4084<br />

(fluorescent DNA dye).<br />

GATA-6 (D61E4) XP ® Rabbit mAb<br />

#5851: Confocal IF analysis of KM12 (top)<br />

and SK-OV-3 (bottom) cells using #5851<br />

(green). Actin filaments were labeled with<br />

DY-554 phalloidin (red).<br />

Stem Cell Differentiation and Transdifferentiation<br />

Pluripotent stem/progenitor cells, including ESCs and iPSCs, can be induced to develop into lineagespecific<br />

progenitor cells, representing each of the three primary germ layers established during<br />

gastrulation: ectoderm, mesoderm, and endoderm. Further differentiation then proceeds progressively<br />

along the respective lineage-specific pathways, culminating in terminal differentiation and yielding a<br />

cell with a lineage-specific functional phenotype. Cells that have differentiated into a specific lineage<br />

may be identified using lineage markers—antigens with a spatially or temporally restricted pattern of<br />

expression that can be used to identify cells within specific lineage pathways.<br />

Neurofilament-L (C28E10) Rabbit<br />

mAb #2837 and β3-Tubulin (TU-20)<br />

Mouse mAb #4466: Confocal IF<br />

analysis of neuroepithelial clusters<br />

differentiated from human iPS cells,<br />

showing multiple neurite extensions,<br />

using #2837 (red) and #4466 (green).<br />

Blue pseudocolor = DRAQ5 ® #4084<br />

(fluorescent DNA dye).<br />

Neurofilament-L<br />

and β3-Tubulin<br />

are markers for<br />

neuronal cells and<br />

antibodies for these<br />

proteins label cells<br />

in neuroepithelial<br />

clusters.<br />

Induced Pluripotency (iPS)<br />

Embryonic Stem Cell<br />

Primordial Germ Cell<br />

Ectoderm<br />

Mesoderm<br />

Endoderm<br />

Development and Differentiation Signaling<br />

Development along each lineage is regulated by several signaling pathways that control cell division,<br />

growth, and differentiation, including BMP/TGF-β, Notch, Wnt/β-catenin, Hedgehog, and Hippo pathways.<br />

Each of these pathways is regulated by a complex array of genetic, epigenetic, and exogenous<br />

signaling factors that serve to guide cell fate and behavior during development and differentiation.<br />

Additional details of signaling nodes within each of these pathways can be found in the pathway<br />

diagrams to follow.<br />

β-catenin is<br />

abundantly expressed<br />

in the mammalian gut,<br />

where it regulates<br />

epithelial cell adhesion.<br />

Neural Stem Cell<br />

Mesenchymal<br />

Stem Cell<br />

Neural Crest Glial<br />

Neuron<br />

Adipocyte, Hematopoietic Hepatocyle Pancreatic Cell<br />

Progenitor Progenitor Myocyte, Osteocyte Stem Cell<br />

Astrocyte Oligodendrocyte Neuron<br />

Hemangioblast<br />

© 2002–2015 Cell Signaling Technology, Inc.<br />

Endodermal<br />

Progenitor<br />

Wnt/β-catenin Signaling<br />

The widely conserved Wnt/β-Catenin pathway regulates stem cell pluripotency and cell fate decisions<br />

during development. Wnt signaling is triggered by binding of the Wnt ligand to Frizzled receptors in<br />

complex with the co-receptor LRP5/6, initiating a signaling cascade that results in stabilization and<br />

nuclear translocation of the transcriptional co-regulator, β-catenin. Nuclear β-catenin functions as a<br />

transcriptional co-activator, promoting the transcription of genes that regulate proliferation and differentiation.<br />

β-catenin also plays a highly important role in the planar-cell-polarity pathway, regulating<br />

cell-cell contact via adherens junctions.<br />

β-Catenin (D10A8) XP ® Rabbit mAb<br />

#8480: Confocal IF analysis of mouse<br />

colon using #8480 (green). Actin filaments<br />

were labeled with DY-554 phalloidin (red).<br />

Blue pseudocolor = DRAQ5 ® #4084<br />

(fluorescent DNA dye).<br />

154 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstdevelopment 155


Section I: Research Areas<br />

chapter 06: Development and differentiation<br />

YAP localizes to<br />

the nucleus in low<br />

confluence cells, but<br />

is translocated to the<br />

cytoplasm during<br />

high confluence<br />

(contact inhibition).<br />

Hippo Signaling<br />

Hippo signaling is an evolutionarily conserved pathway that plays a critical role in development by<br />

regulating cell proliferation, apoptosis, and stem cell self-renewal. Activation of the Hippo pathway<br />

(e.g., by cell-cell contact) suppresses cell proliferation by initiating a signaling cascade that results in<br />

phosphorylation, cytoplasmic retention, and proteasomal degradation of the transcriptional co-activators<br />

YAP and TAZ, thereby preventing the transcription of genes that promote proliferation.<br />

A B C<br />

Notch Signaling<br />

The Notch pathway is a contact-dependent signaling cascade mediated by the Notch receptor on the<br />

receiving cell and the Delta-like and Jagged ligands on the signal-sending cell. Receptor-ligand binding<br />

results in a series of cleavages to the Notch receptor, culminating in release of the Notch intracellular<br />

domain (NICD) by γ-secretase. The NICD translocates to the nucleus where it interacts with RBPSUH<br />

and Mastermind-like (MAML) proteins to regulate transcription of target genes that mediate cell fate<br />

decisions in neuronal, cardiac, immune, and endocrine development.<br />

Hedgehog Signaling<br />

The evolutionarily conserved Hedgehog (Hh) pathway plays a critical role in a time and positiondependent<br />

fashion during development. Hedgehog ligands (Sonic, Desert, and Indian Hedgehog) act as<br />

classic morphogens, whose concentration gradient promotes specific developmental outcomes at distinct<br />

concentration thresholds. Hedgehog signaling occurs when a hedgehog family ligand binds to the<br />

receptor Patched (PTCH), releasing Smoothened (SMO) from suppression and allowing it to translocate to<br />

primary cilium. Once there, SMO signals through G proteins, ultimately leading to nuclear translocation<br />

of GLI zinc finger transcription factors. Hedgehog signaling is instrumental early in embryogenesis,<br />

orchestrating neural plate patterning and ventral polarity of the neural tube, and anterior-posterior axis<br />

patterning (e.g. during limb formation). Defects in Hh signaling are the most common cause of midline<br />

birth defects, resulting in disorders such as holoprosencephaly, which is characterized by incomplete<br />

separation of the forebrain hemispheres and their derivatives. The Hedgehog pathway has also been<br />

shown to play a role in maintenance of adult stem cells, particularly neural progenitors and hematopoietic<br />

stem cells.<br />

YAP (D8H1X) XP ® Rabbit mAb #14074: Confocal IF analysis of low confluence MCF 10A cells (A), high confluence MCF 10A (B), and<br />

YAP-negative RL-7 cells (C) using #14074 (green). Blue pseudocolor in lower images = DRAQ5 ® #4084 (fluorescent DNA dye). Increased<br />

nuclear localization of YAP protein is seen in low confluence (proliferating) cells.<br />

TGF-β/BMP Signaling<br />

Transforming growth factor-β (TGF-β) superfamily signaling plays a critical role in the regulation of cell<br />

proliferation, differentiation, developmental patterning and morphogenesis, and disease pathogenesis.<br />

In the canonical TGF-β signaling pathway, ligand binding to one of the three types of TGF-β receptors<br />

results in receptor activation and phosphorylation of the Smad2/3 or Smad1/5/9 effector proteins.<br />

These phosphorylated Smads dimerize with the co-activating Smad4 and translocate to the nucleus,<br />

where they stimulate transcription of target genes. TGF-β receptors can also signal through noncanonical<br />

(Smad-independent) pathways that promote cytoskeletal rearrangement and adhesion through<br />

activation of RhoA, Rac/cdc42, and PI3K pathways.<br />

BMP2 treatment results in phosphorylation<br />

of Smad1/5 at Ser463/465.<br />

Phospho-Smad1 (Ser463/465)/Smad5 (Ser463/465)/Smad9 (Ser465/467)<br />

(D5B10) Rabbit mAb #13820: WB analysis of extracts from Hep G2 or MEF cells,<br />

untreated (-) or treated with Human BMP2 #4697 (50 ng/ml, 30 min; +), using<br />

#13820 (upper) and Smad1 (D59D7) XP ® Rabbit mAb #6944 (lower).<br />

Lanes<br />

1. Hep G2<br />

2. MEF<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

1 2<br />

Phospho-Smad1<br />

(Ser463/465)/<br />

Smad5 (Ser463/465)/<br />

Smad9 (Ser465/467)<br />

Smad1<br />

Development and Cancer<br />

Ectopic activation or dysregulation of embryonic signaling pathways is recognized as an important<br />

contributor to tumorigenesis in many contexts. For example, constitutive activation of the Hedgehog<br />

pathway by mutations in PTCH or SMO are strongly linked to basal cell carcinoma and medulloblastoma.<br />

Likewise, mutations in components of the Wnt signaling pathway, which can lead to increased stability<br />

and transcriptional activity of β-catenin, are directly associated with colorectal, gastric, hepatocellular,<br />

ovarian, breast, and prostrate cancers. In Hippo signaling, the central pathway mediators, YAP and<br />

TAZ, function as oncogenes, and amplification of YAP has been associated with a variety of tumors<br />

including hepatocellular, lung, colorectal, breast, and liver carcinomas. The TGF-β signaling pathway<br />

is recognized as a potentially important driver of cancer metastasis, through its ability to regulate the<br />

process of epithelial-mesenchymal transition (EMT).<br />

Cancer stem cells (CSC) are a rare sub-population of tumorigenic cells found within tumors that (like<br />

normal stem cells) are multipotent and self-renewing. These cells posses tumor-initiating potential and<br />

can differentiate into all the other cell types that compose the bulk of the tumor. Cell surface markers<br />

can distinguish CSCs from differentiated cancer cells in many tumor types and be used to purify subpopulations<br />

of CSCs. These include CD133, CD44, CD24, ALDH1, and EpCAM, among others.<br />

Select Reviews<br />

Addis, R.C. and Epstein, J.A. (2013) Nat. Med. 19, 829−836. • Ader, M. and Tanaka, E.M. (2014) Curr. Opin. Cell Biol. 31C,<br />

23−28. • Gieseck, R.L. 3rd, Colquhoun, J., and Hannan, N.R. (2014) Biochim. Biophys. Acta. Jun 2 [Epub ahead of print] •<br />

Ingham, P.W. and McMahon, A.P. (2001) Genes and Dev. 15, 3059−3087. • Isobe, K.I., Cheng, Z., Nishio, N. et al. (2014) Nat.<br />

Biotechnol. 31, 411−421. • Itoh, F., Watabe, T., and Miyazono, K. (2014) Semin. Cell Dev. Biol. 32C, 98−106. • Jamieson,<br />

C., Sharma, M., and Henderson, B.R. (2014) Semin. Cancer Biol. 27C, 20−29. • Jiang, J. and Hui, C-C. (2008) Developmental<br />

Cell 15, 801−812. • Jopling, C., Boue, S., and Izpisua Belmonte, J.C. (2011) Nat. Rev. Mol. Cell Biol. 12, 79−89. • Kreso,<br />

A. and Dickemail, J.E. (2014) Cell Stem Cell 14, 275–291. • Lien, W.H. and Fuchs, E. (2014) Genes Dev. 28, 1517−1532. •<br />

Ntziachristos, P., Lim, J.S., Sage, J., et al. (2014) Cancer Cell 25, 318−334. • Robbins, D.J., Fei, D.L., and Riobo, N.A. (2012)<br />

Sci. Signal. 5, re6. • Sánchez, A.A. and Yamanaka, S. (2014) Cell 157, 110−119. • Sarkar, A. and Hochedlinger, K. (2013)<br />

Cell Stem Cell 12, 15−30. • Torres-Padilla, M.E. and Chambers, I. (2014) Development 141, 2173−2181. • Varelas, X.<br />

(2014) Development 141, 1614−1626.<br />

Elevated levels of<br />

β-catenin are found in<br />

colon carcinoma.<br />

β-Catenin (D10A8) XP ® Rabbit mAb<br />

#8480: IHC analysis of paraffin-embedded<br />

human colon carcinoma using #8480.<br />

Amplification of YAP is<br />

associated with breast<br />

adenocarcinoma.<br />

20<br />

– +<br />

– +<br />

Human BMP2<br />

Cancer Research<br />

Please visit our website to learn more about the scientific tools and educational resources we have online for cancer<br />

signaling and proteomic analysis, including discussion of key disease drivers. www.cellsignal.com/cancerguide<br />

YAP (D8H1X) XP ® Rabbit mAb #14074:<br />

IHC analysis of paraffin-embedded human<br />

breast adenocarcinoma using #14074.<br />

156 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstdevelopment<br />

157


Section I: Research Areas<br />

chapter 06: Development and differentiation<br />

These protein targets represent key<br />

nodes within development and differentiation<br />

signaling pathways and are<br />

commonly studied in development and<br />

stem cell research. Primary antibodies,<br />

antibody conjugates, and antibody<br />

sampler kits containing these targets<br />

are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

C Antibody Conjugate<br />

Commonly Studied Development and Differentiation Targets<br />

Target M P C<br />

ACVR1<br />

AFP<br />

•<br />

• • •<br />

AGR2 •<br />

ALPP •<br />

Angiopoietin-2 •<br />

APC<br />

•<br />

Axin1 •<br />

Axin2 • •<br />

Phospho-BCL9L<br />

(Ser915)<br />

•<br />

Bcl-11B •<br />

BMP4 •<br />

BMP7<br />

•<br />

BMPR2<br />

•<br />

Brachyury •<br />

CACYBP • •<br />

•<br />

β-Catenin (Aminoterminal<br />

Antigen)<br />

β-Catenin (Carboxyterminal<br />

Antigen)<br />

β-Catenin<br />

Phospho-β-Catenin<br />

(Ser33/37)<br />

Phospho-β-Catenin<br />

(Ser33/37/Thr41)<br />

Non-phospho (Active)<br />

β-Catenin (Ser33/37/<br />

Thr41)<br />

Phospho-β-Catenin<br />

(Thr41/Ser45)<br />

Phospho-β-Catenin<br />

(Ser45)<br />

Phospho-β-Catenin<br />

(Ser552)<br />

Phospho-β-Catenin<br />

(Ser675)<br />

•<br />

• • •<br />

•<br />

•<br />

• • •<br />

• •<br />

•<br />

•<br />

• •<br />

Acetyl-β-Catenin (Lys49) • •<br />

CDCP1 •<br />

•<br />

Phospho-CDCP1<br />

(Tyr707)<br />

Phospho-CDCP1<br />

(Tyr734)<br />

Phospho-CDCP1<br />

(Tyr743)<br />

Phospho-CDCP1<br />

(Tyr806)<br />

CDX2<br />

CEACAM1<br />

CK1<br />

Cripto<br />

CtBP1<br />

CtBP2<br />

DAX1<br />

DAZL<br />

DDX4<br />

DKK1<br />

DKK2<br />

DLK1<br />

DLL1<br />

DLL3<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

• •<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

Target M P C<br />

DLL4<br />

•<br />

Deltex-2<br />

•<br />

Dvl2<br />

• •<br />

Dvl3<br />

•<br />

Endoglin • •<br />

EOMES<br />

•<br />

E-Ras •<br />

Acidic FGF •<br />

Basic FGF •<br />

FIH<br />

•<br />

FLI1<br />

•<br />

Flightless-I •<br />

FoxC1 • •<br />

FoxD3 •<br />

FoxK1<br />

•<br />

FoxK2<br />

•<br />

FoxP1 • •<br />

FoxP2 •<br />

Fragilis<br />

•<br />

Frizzled5 • •<br />

Frizzled6 •<br />

GATA-1 • • •<br />

GATA-3 • •<br />

GATA-6 • •<br />

GCNF/NR6A1 •<br />

GFI1b •<br />

GLI1 • • •<br />

GLI2<br />

•<br />

Gremlin<br />

•<br />

Heregulin •<br />

HES1 •<br />

HIF-1β/ARNT • •<br />

HIF-1α •<br />

Hydroxy-HIF-1α (Pro564) •<br />

HIF-2α •<br />

Id2<br />

•<br />

ID3 • •<br />

Jagged1 •<br />

Jagged2 •<br />

KIBRA<br />

•<br />

KIF3A •<br />

KIF3B<br />

•<br />

KLF4 • •<br />

LEF1 • •<br />

Lefty1 •<br />

LIMD1<br />

•<br />

LIN28A • • •<br />

LIN28B • •<br />

Lmx1B •<br />

LRP5 •<br />

LRP6 •<br />

Phospho-LRP6<br />

(Ser1490) •<br />

MAML1 • •<br />

MAML2 • •<br />

MESD2 •<br />

Mic-1 • •<br />

Target M P C<br />

MIS-R2<br />

•<br />

MITF •<br />

MOB1 • •<br />

Phospho-MOB1 (Thr12) •<br />

Phospho-MOB1 (Thr35) •<br />

Msx1<br />

•<br />

c-Myb •<br />

MyoD1 • •<br />

NAC1<br />

•<br />

Naked1 • •<br />

Naked2 •<br />

Nanog • • •<br />

NDRG1 • •<br />

Phospho-NDRG1<br />

(Ser330) • •<br />

Phospho-NDRG1<br />

(Thr346) • • •<br />

NDRG2<br />

•<br />

NDRG3<br />

•<br />

NDRG4 •<br />

NKX2.2<br />

•<br />

NKX2.5 •<br />

Notch1 •<br />

Cleaved Notch1<br />

(Val1754) • •<br />

Notch2 •<br />

Notch3 • •<br />

Notch4 •<br />

Nucleostemin •<br />

Numb • •<br />

Phospho-Numb (Ser276) • •<br />

Oct-1 • •<br />

Oct-4 • • •<br />

OTX2 •<br />

PAX2<br />

•<br />

PAX3<br />

•<br />

PAX5 • •<br />

PAX8<br />

•<br />

PAX9 •<br />

PDAP1<br />

•<br />

PHD-2/Egln1 • •<br />

PTCH1 •<br />

PTCH2<br />

•<br />

PTPN14<br />

•<br />

PTPN18 •<br />

RAIG1 • •<br />

RBPSUH • •<br />

RUNX2 •<br />

RUNX3/AML2 •<br />

Sall4 • •<br />

Sara<br />

•<br />

SAV1 • •<br />

Scribble<br />

•<br />

Phospho-Scribble •<br />

SCF<br />

•<br />

SFRP1 • •<br />

Shh<br />

• •<br />

Target M P C<br />

Shh/Ihh<br />

•<br />

SIX1<br />

•<br />

Slug<br />

•<br />

Smad1 • •<br />

Phospho-Smad1<br />

(Ser206) • •<br />

Phospho-Smad1<br />

(Ser463/465)/Smad5<br />

(Ser463/465)/Smad9 • • •<br />

(Ser426/428)<br />

Phospho-Smad1/5<br />

(Ser463/465) • •<br />

Smad2 •<br />

•<br />

Phospho-Smad2<br />

(Ser245/250/255)<br />

Phospho-Smad2<br />

(Ser465/467)<br />

Smad2/3<br />

Phospho-Smad2<br />

(Ser465/467)/Smad3<br />

(Ser423/425)<br />

Smad3<br />

Phospho-Smad3<br />

(Ser423/425)<br />

Smad4<br />

Smad5<br />

Smurf1<br />

Smurf2<br />

• • •<br />

• • •<br />

• •<br />

• •<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

Select Citations:<br />

Palla, A.R. et al. (2014) Reprogramming activity of<br />

NANOGP8, a NANOG family member widely expressed in<br />

cancer. Oncogene 33, 2513–2519.<br />

Zhou, W. et al. (2014) Snail contributes to the maintenance<br />

of stem cell-like phenotype cells in human pancreatic<br />

cancer. PLoS One 9, e87409.<br />

Ono, T. et al. (2014) A single-cell and feeder-free culture system<br />

for monkey embryonic stem cells. PLoS One 9, e88346.<br />

Yan, X. et al. (2014) 5-azacytidine improves the osteogenic<br />

differentiation potential of aged human adipose-derived<br />

mesenchymal stem cells by DNA demethylation. PLoS One<br />

9, e90846.<br />

Han, H. et al. (2013) MBNL proteins repress ES-cellspecific<br />

alternative splicing and reprogramming. Nature 498,<br />

241–245.<br />

Hamalainen, R.H. et al. (2013) Tissue- and cell-type-specific<br />

manifestations of heteroplasmic mtDNA 3243A>G mutation<br />

in human induced pluripotent stem cell-derived disease<br />

model. Proc. Natl. Acad. Sci. USA 110, 3622–3630.<br />

Zhang, J. et al. (2013) NANOG modulates stemness in human<br />

colorectal cancer. Oncogene 32, 4397–4405.<br />

Jinesh, G.G. et al. (2013) Blebbishields, the emergency<br />

program for cancer stem cells: sphere formation and tumorigenesis<br />

after apoptosis. Cell Death Differ. 20, 382–395.<br />

Kregel, S. et al. (2013) Sox2 is an androgen receptorrepressed<br />

gene that promotes castration-resistant prostate<br />

cancer. PLoS One 8, e53701.<br />

Demir, K. et al. (2013) RAB8B is required for activity and<br />

caveolar endocytosis of LRP6. Cell Rep. 4, 1224–1234.<br />

Target M P C<br />

Snail •<br />

SnoN<br />

•<br />

Sox1<br />

•<br />

Sox2 • • •<br />

SPARC • •<br />

SSEA1 •<br />

SSEA4 •<br />

SUFU • •<br />

TACE • •<br />

TAZ<br />

•<br />

TAZ/YAP •<br />

TCF1 • •<br />

TCF3 •<br />

TCF4 •<br />

TCF8 •<br />

TCF12/HEB •<br />

TEAD1 • •<br />

Pan-TEAD •<br />

TEAD2<br />

•<br />

TEAD3<br />

•<br />

TFF1/pS2 •<br />

Pro-TGF-α •<br />

TGF-β • •<br />

TGF-β Receptor I •<br />

TGF-β Receptor II •<br />

Target M P C<br />

TGF-β Receptor III • •<br />

Thap11/Ronin •<br />

TLE1/2/3/4 •<br />

TRA-1-60 •<br />

TRA-1-81 •<br />

TRA-2-54 (Alkaline<br />

phosphatase) •<br />

TRIB2 •<br />

TRIM33 •<br />

Thyroid Transcription<br />

Factor 1 (TTF-1) •<br />

Phospho-TTF1 (Ser327) •<br />

UTF1<br />

•<br />

VEGF-B<br />

•<br />

VEGF-C<br />

•<br />

WBP2<br />

•<br />

WIF1 • •<br />

Wnt3a • •<br />

Wnt5a<br />

•<br />

Wnt5a/b •<br />

WTX •<br />

YAP<br />

• •<br />

Phospho-YAP (Ser127) • •<br />

Phospho-YAP (Ser397) •<br />

ZFX<br />

•<br />

Garg, N. et al. (2013) microRNA-17-92 cluster is a<br />

direct Nanog target and controls neural stem cell through<br />

Trp53inp1. EMBO J. 32, 2819–2832.<br />

Bhatia, S. et al. (2013) Demarcation of stable subpopulations<br />

within the pluripotent hESC compartment. PLoS One<br />

8, e57276<br />

Vaira, V. et al. (2013) Regulation of lung cancer metastasis<br />

by Klf4-Numb-like signaling. Cancer Res. 73, 2695–2705.<br />

Tanaka, A. et al. (2013) Efficient and reproducible myogenic<br />

differentiation from human iPS cells: prospects for modeling<br />

Miyoshi Myopathy in vitro. PLoS One 8, e61540.<br />

Wang, T. et al. (2013) Subtelomeric hotspots of aberrant<br />

5-hydroxymethylcytosine-mediated epigenetic modifications<br />

during reprogramming to pluripotency. Nat. Cell Biol. 15,<br />

700–711.<br />

Romorini, L. et al. (2013) Effect of antibiotics against Mycoplasma<br />

sp. on human embryonic stem cells undifferentiated<br />

status, pluripotency, cell viability and growth. PLoS One 8,<br />

e70267.<br />

Abad, M. et al. (2013) Reprogramming in vivo produces<br />

teratomas and iPS cells with totipotency features. Nature<br />

502, 340–345.<br />

Hasmim, M. et al. (2013) Cutting edge: Hypoxia-induced<br />

Nanog favors the intratumoral infiltration of regulatory T<br />

cells and macrophages via direct regulation of TGF-beta1. J.<br />

Immunol. 191, 5802–5806.<br />

Abhold, E.L. et al. (2012) EGFR kinase promotes acquisition<br />

of stem cell-like properties: a potential therapeutic target in<br />

head and neck squamous cell carcinoma stem cells. PLoS<br />

One 7, e32459.<br />

24<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for Nanog have been<br />

cited over 24 times in high-impact,<br />

peer-reviewed publications from the<br />

global research community.<br />

158 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstdevelopment 159


Section I: Research Areas<br />

chapter 06: Development and differentiation<br />

Wnt/b-Catenin Signaling<br />

Notch Signaling<br />

OFF-State<br />

Cadherin<br />

β-catenin<br />

α-catenin<br />

p120<br />

DKKs<br />

LRP5/6<br />

SFRP<br />

Frizzled<br />

ON-State<br />

Porc WIF<br />

WLS<br />

SFRP<br />

Wnt<br />

LRP5/6<br />

CK1 GSK-3<br />

Axin<br />

Frizzled<br />

Dvl<br />

Naked<br />

R-spondins<br />

LGR5/6<br />

RNF43<br />

ZNRF3<br />

Signal<br />

Sending<br />

Cell<br />

Inactive<br />

Ligand<br />

Delta<br />

Jagged<br />

Active<br />

Ligand<br />

Neur<br />

or Mib<br />

ub<br />

ub<br />

Delta<br />

Jagged<br />

Endosome<br />

ub<br />

Ligand<br />

Repositioning<br />

Epsin<br />

ub<br />

Ligand-receptor<br />

Internalization<br />

Skp<br />

Proteasomal<br />

Degradation<br />

β-TrCP<br />

ub β-catenin<br />

Tankyrase<br />

PP2A<br />

Axin<br />

APC GSK-3<br />

WTX<br />

β-catenin<br />

CK1<br />

CYLD<br />

β-catenin<br />

Rac1<br />

ub<br />

Dvl<br />

PAR-1<br />

ub TAB2<br />

TAK1<br />

TAB1<br />

NLK<br />

Extracellular<br />

Space<br />

Signal<br />

Receiving<br />

Cell<br />

Notch<br />

Fringe<br />

Deltex<br />

NEDD4<br />

Golgi<br />

ub<br />

Notch<br />

Recycling<br />

Activated<br />

Receptor<br />

Endosome<br />

NUMB<br />

α-Adaptin<br />

ADAM/<br />

TACE<br />

S2 Cleavage<br />

TM<br />

NICD<br />

S3 Cleavage<br />

FBX7<br />

Nicastrin<br />

Presenilin<br />

APH-1<br />

PEN-2<br />

γ-Secretase<br />

Complex<br />

Ubiquitin<br />

Degradation<br />

Cytoplasm<br />

Nucleus<br />

OFF<br />

HDAC<br />

Groucho<br />

TCF3<br />

HDAC<br />

Groucho β-catenin<br />

Hesx1<br />

FoxO<br />

Pygo BCL9<br />

Brg1<br />

β-catenin<br />

CBP<br />

LEF1/TCF1<br />

FoxO<br />

ICAT<br />

β-catenin<br />

Chibby<br />

NLK<br />

TAZ<br />

Target Genes:<br />

Myc, Cyclin D1,<br />

TCF-1, PPAR-δ,<br />

MMP-7, Axin-2,<br />

CD44, etc.<br />

Pit1<br />

Snail1<br />

Migration<br />

Adhesion<br />

S1 Cleavage Furin<br />

ER<br />

Cytoplasm<br />

Nucleus<br />

O-Fut<br />

CtBP1 CIR HDAC<br />

KDM5A<br />

SMRT SHARP<br />

CSL SKIP<br />

Inactive<br />

Lysosomal<br />

Degradation<br />

KDM5A<br />

HAT<br />

MAML<br />

SKIP<br />

CSL<br />

Active<br />

Notch Target Genes<br />

HES Family<br />

Myc<br />

p21<br />

Cyclin D3<br />

The conserved Wnt/β-Catenin pathway regulates stem cell pluripotency and cell fate decisions during development. This developmental cascade integrates signals from other<br />

pathways, including retinoic acid, FGF, TGF-β, and BMP, within different cell types and tissues. The Wnt ligand is a secreted glycoprotein that binds to Frizzled receptors, leading<br />

to the formation of a larger cell surface complex with LRP5/6. Frizzleds are ubiquitinated by ZNRF3 and RNF43, whose activity is inhibited by R-spondin binding to LGR5/6.<br />

In this manner R-spondins increase sensitivity of cells to the Wnt ligand. Activation of the Wnt receptor complex triggers displacement of the multifunctional kinase GSK-3β<br />

from a regulatory APC/Axin/GSK-3β-complex. In the absence of Wnt-signal (Off-state), β-catenin, an integral E-cadherin cell-cell adhesion adaptor protein and transcriptional<br />

co-regulator, is targeted by coordinated phosphorylation by CK1 and the APC/Axin/GSK-3β-complex leading to its ubiquitination and proteasomal degradation through the<br />

β-TrCP/Skp pathway. In the presence of Wnt ligand (On-state), the co-receptor LRP5/6 is brought in complex with Wnt-bound Frizzled. This leads to activation of Dishevelled<br />

(Dvl) by sequential phosphorylation, poly-ubiquitination, and polymerization, which displaces GSK-3β from APC/Axin through an unclear mechanism that may involve substrate<br />

trapping and/ or endosome sequestration. Stablized β-catenin is translocated to the nucleus via Rac1 and other factors, where it binds to LEF/TCF transcription factors, displacing<br />

co-repressors and recruiting additional co-activators to Wnt target genes. Additionally, β-catenin cooperates with several other transcription factors to regulate specific<br />

targets. Importantly, researchers have found β-catenin point mutations in human tumors that prevent GSK-3β phosphorylation and thus lead to its aberrant accumulation.<br />

E-cadherin, APC, R-spondin and Axin mutations have also been documented in tumor samples, underscoring the deregulation of this pathway in cancer. Wnt signaling has<br />

also been shown to promote nuclear accumulation of other transcriptional regulator implicated in cancer, such as TAZ and Snail1. Furthermore, GSK-3β is involved in glycogen<br />

metabolism and other signaling pathways, which has made its inhibition relevant to diabetes and neurodegenerative disorders.<br />

Select Reviews:<br />

Angers, S. and Moon, R.T. (2009) Nat. Rev. Mol. Cell Biol. 10, 468–477. • Cadigan, K.M. and Waterman, M.L. (2012) Cold Spring Harb. Perspect. Biol. 4, a007906. •<br />

Clevers, H. and Nusse, R. (2012) Cell 149, 1192–1205. • de Lau, W., Peng, W.C., Gros, P. and Clevers, H. (2014) Genes Dev. 28, 305–316. • Fearon, E.R. (2009) Cancer<br />

Cell 16, 366–368. • MacDonald, B.T., Tamai, K. and He, X. (2009) Dev. Cell 17, 9–26. • Metcalfe, C. and Bienz, M. (2011) J. Cell. Sci. 124, 3537–3544. • Niehrs, C.<br />

(2012) Nat. Rev. Mol. Cell Biol. 13, 767–779. • Nusse, R. (2010) The Wnt Homepage. • Petersen, C.P. and Reddien, P.W. (2009) Cell 139, 1056–1068. • Sokol, S.Y.<br />

(2011) Development 138, 4341–4350. • van Amerongen, R. and Nusse, R. (2009) Development 136, 3205–3214. • Valenta, T., Hausmann, G., and Basler, K. (2012)<br />

EMBO J. 31, 2714–2736.<br />

© 2003–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Kenneth Cadigan, University of Michigan, Ann Arbor, MI, for reviewing this diagram.<br />

160 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

Notch signaling is an evolutionarily conserved pathway in multicellular organisms that regulates cell-fate determination during development and maintains adult tissue homeostasis.<br />

The Notch pathway mediates juxtacrine cellular signaling wherein both the signal sending and receiving cells are affected through ligand-receptor crosstalk by which<br />

an array of cell fate decisions in neuronal, cardiac, immune, and endocrine development are regulated. Notch receptors are single-pass transmembrane proteins composed<br />

of functional extracellular (NECD), transmembrane (TM), and intracellular (NICD) domains. Notch receptors are processed in the ER and Golgi within the signal-receiving cell<br />

through cleavage and glycosylation, generating a Ca 2+ -stabilized heterodimer composed of NECD noncovalently attached to the TM-NICD inserted in the membrane (S1<br />

cleavage). The processed receptor is then endosome-transported to the plasma membrane to enable ligand binding in a manner regulated by Deltex and inhibited by NUMB.<br />

In mammalian signal-sending cells, members of the Delta-like (DLL1, DLL3, DLL4) and the Jagged (JAG1, JAG2) families serve as ligands for Notch signaling receptors.<br />

Upon ligand binding, the NECD is cleaved away (S2 cleavage) from the TM-NICD domain by TACE (TNF-α ADAM metalloprotease converting enzyme). The NECD remains<br />

bound to the ligand and this complex undergoes endocytosis/recycling within the signal-sending cell in a manner dependent on ubiquitination by Mib. In the signal-receiving<br />

cell, γ-secretase (also involved in Alzheimer’s disease) releases the NICD from the TM (S3 cleavage), which allows for nuclear translocation where it associates with the CSL<br />

(CBF1/Su(H)/Lag-1) transcription factor complex, resulting in subsequent activation of the canonical Notch target genes: Myc, p21, and the HES-family members. The Notch<br />

signaling pathway has spurred interest for pharmacological intervention due to its connection to human disease. Importantly, researchers have found Notch receptor activating<br />

mutations leading to nuclear accumulation of NICD are common in adult T cell acute lymphoblastic leukemia and lymphoma. In addition, loss-of-function Notch receptor and<br />

ligand mutations are implicated in several disorders, including Alagille syndrome and CADASIL, an autosomal dominant form of cerebral arteriopathy.<br />

Select Reviews:<br />

Ables, J.L., Breunig, J.J., Eisch, A.J., Rakic, P. (2011) Nat. Rev. Neurosci. 12, 269–283. • Andersson, E.R., Lendahl, U. (2014) Nat. Rev. Drug Discov. 13, 357–378. • Aster,<br />

J.C., Blacklow, S.C., Pear, W.S. (2011) J. Pathol. 223, 262–273. • Bai, G., Pfaff, S.L. (2011) Neuron 72, 9–21. • de la Pompa, J.L., Epstein, J.A. (2012) Dev. Cell 22, 244–<br />

254. • Ntziachristos, P., Lim, J.S., Sage, J., Aifantis, I. (2014) Cancer Cell. 24. • Ranganathan, P., Weaver, K.L., Capobianco, A.J. (2011) Nat. Rev. Cancer 11, 338–351.<br />

• Weinmaster, G., Fischer, J.A. (2011) Dev. Cell 21, 134–144. • Yuan, J.S., Kousis, P.C., Suliman, S., Visan, I., Guidos, C.J. (2010) Annu. Rev. Immunol. 28, 343–365.<br />

© 2006–2015 Cell Signaling Technology, Inc. • We would like to thank Dr. Hans Widlund, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

www.cellsignal.com/cstpathways 161


Section I: Research Areas<br />

chapter 06: Development and differentiation<br />

Hippo Signaling<br />

Adherens Junction Tight Junction<br />

Crb<br />

ZO-2<br />

YAP/TAZ<br />

AMOT<br />

YAP/TAZ<br />

AMOT<br />

β-TrCP<br />

FRMD<br />

Merlin<br />

KIBRA<br />

SIK1-3<br />

α-catenin<br />

Mst1/2<br />

LKB1<br />

14-3-3 SAV1<br />

MARK4<br />

YAP<br />

14-3-3<br />

YAP<br />

RASSF<br />

Ajuba<br />

CK1<br />

Merlin<br />

CD44<br />

<br />

FAT4<br />

<br />

LATS1/2<br />

YAP/TAZ<br />

<br />

TAOK-1-3<br />

MOB1<br />

GPCR Signaling<br />

PP2A<br />

F-actin<br />

ITCH<br />

PP1<br />

Gα 12/13<br />

ASPP2<br />

Gα q/11<br />

Gα i/o<br />

AMOT<br />

Gα s<br />

Mechanical Signaling<br />

Rho<br />

Hedgehog Signaling<br />

PKA<br />

SUFU<br />

GLI 1/2/3<br />

SUFU<br />

β−TrCP<br />

CK1<br />

GLI 1/2/3<br />

GSK-3β<br />

Proteasome<br />

Repressor GLI 3-R<br />

no transcription of<br />

target genes<br />

Cytoplasm<br />

KIF7<br />

Primary<br />

Cilium<br />

GLI 1/2/3<br />

Microtubules<br />

SMO<br />

GLI 1/2 Degradation<br />

Off-State<br />

Skn<br />

PTCH1<br />

GLI 3-R<br />

SCUBE2<br />

CDON/BOC<br />

Nucleus<br />

Hh<br />

HhN<br />

DISP1<br />

PTCH2<br />

Primary<br />

Cilium<br />

ER/Golgi<br />

HhN<br />

Microtubules<br />

SUFU<br />

GLI 1/2/3<br />

GLI 1/2-Act<br />

Activator<br />

KCTD11<br />

Cyclin D, Cyclin E,<br />

Myc, GLI1, PTCH1,<br />

PTCH2, Hhip1<br />

Cytoplasm<br />

KIF7<br />

Hh Secreting Cell<br />

Gα i<br />

β-Arrestin<br />

SMO<br />

On-State<br />

PTCH1<br />

GLI 1/2-Act<br />

Mammalian<br />

Ligands:<br />

Ihh<br />

Dhh<br />

Shh<br />

CDON/BOC<br />

Nucleus<br />

Hhip1 GAS1<br />

The evolutionarily conserved Hedgehog (Hh) pathway is essential for normal embryonic development and plays critical roles in adult tissue maintenance, renewal and regeneration.<br />

Secreted Hh proteins act in a concentration- and time-dependent manner to initiate a series of cellular responses that range from survival and proliferation to cell fate<br />

specification and differentiation.<br />

HhN<br />

VGL4<br />

TEAD1-4<br />

WBP2<br />

YAP/TAZ<br />

TEAD1-4<br />

YAP/TAZ<br />

Smad<br />

YAP/TAZ<br />

p73<br />

YAP/TAZ<br />

RUNX<br />

Nucleus<br />

Transcription<br />

Cytoplasm<br />

Hippo signaling is an evolutionarily conserved pathway that controls organ size by regulating cell proliferation, apoptosis, and stem cell self renewal. In addition, dysregulation<br />

of the Hippo pathway contributes to cancer development. Core to the Hippo pathway is a kinase cascade, wherein Mst1/2 (ortholog of Drosophila Hippo) kinases and SAV1<br />

form a complex to phosphorylate and activate LATS1/2. LATS1/2 kinases in turn phosphorylate and inhibit the transcription co-activators YAP and TAZ, two major downstream<br />

effectors of the Hippo pathway. When dephosphorylated, YAP/TAZ translocate into the nucleus and interact with TEAD1-4 and other transcription factors to induce expression<br />

of genes that promote cell proliferation and inhibit apoptosis. The Hippo pathway is involved in cell contact inhibition, and its activity is regulated at multiple levels: Mst1/2 and<br />

LATS1/2 are regulated by upstream molecules such as Merlin, KIBRA, RASSFs, and Ajuba; 14-3-3, α-catenin, AMOT, and ZO-2 retain YAP/TAZ in the cytoplasm, adherens<br />

junctions, or tight junctions by binding; Mst1/2 and YAP/TAZ phosphorylation and activity are modulated by phosphatases; Lats1/2 and YAP/TAZ stability are regulated by<br />

protein ubiquitination; and LATS1/2 activity is also regulated by the cytoskeleton. Despite extensive study of the Hippo pathway in the past decade, the exact nature of extracellular<br />

signals and membrane receptors regulating the Hippo pathway remains elusive.<br />

Select Reviews:<br />

Badouel, C. and McNeill, H. (2011) Cell 145, 484–484. • Genevet, A. and Tapon, N. (2011) Biochem. J. 436, 213–224. • O’Hayre, M., Degese, M.S., and Gutkind, J.S.<br />

(2014) Curr. Opin. Cell Biol. 27, 126–135. • Pan, D. (2010) Dev. Cell 19, 491–505. • Sudol, M. and Harvey, K.F. (2010) Trends Biochem. Sci. 35, 627–633. • Yu, F.X.<br />

and Guan, K.L. (2013) Genes Dev. 27, 355–371. • Zhao, B., Li, L., Lei, Q., and Guan, K.L. (2010) Genes Dev. 24, 862–874. • Zhao, B., Tumaneng, K., and Guan, K.L.<br />

(2011) Nat. Cell Biol. 13, 877–883.<br />

Proper levels of Hh signaling require the regulated production, processing, secretion and trafficking of Hh ligands– in mammals this includes Sonic (Shh), Indian (Ihh) and<br />

Desert (Dhh). All Hh ligands are synthesized as precursor proteins that undergo autocatalytic cleavage and concomitant cholesterol modification at the carboxy terminus<br />

and palmitoylation at the amino terminus, resulting in a secreted, dually-lipidated protein. Hh ligands are released from the cell surface through the combined actions of<br />

Dispatched and Scube2, and subsequently trafficked over multiple cells through interactions with the cell surface proteins LRP2 and the Glypican family of heparan sulfate<br />

proteoglycans (GPC1-6).<br />

Hh proteins initiate signaling through binding to the canonical receptor Patched (PTCH1) and to the co-receptors GAS1, CDON and BOC. Hh binding to PTCH1 results in derepression<br />

of the GPCR-like protein Smoothened (SMO) that results in SMO accumulation in cilia and phosphorylation of its cytoplasmic tail. SMO mediates downstream signal<br />

transduction that includes dissociation of GLI proteins (the transcriptional effectors of the Hh pathway) from kinesin-family protein, Kif7, and the key intracellular Hh pathway<br />

regulator SUFU.<br />

GLI proteins also traffic through cilia and in the absence of Hh signaling are sequestered by SUFU and Kif7, allowing for GLI phosphorylation by PKA, GSK3β and CK1, and<br />

subsequent processing into transcriptional repressors (through cleavage of the carboxy-terminus) or targeting for degradation (mediated by the E3 ubiquitin ligase β-TrCP). In<br />

response to activation of Hh signaling, GLI proteins are differentially phopshorylated and processed into transcriptional activators that induce expression of Hh target genes,<br />

many of which are components of the pathway (e.g. PTCH1 and GLI1). Feedback mechanisms include the induction of Hh pathway antagonists (PTCH1, PTCH2 and Hhip1)<br />

that interfere with Hh ligand function, and GLI protein degradation mediated by the E3 ubiquitin ligase adaptor protein, SPOP.<br />

In addition to vital roles during normal embryonic development and adult tissue homeostasis, aberrant Hh signaling is responsible for the initiation of a growing number of<br />

cancers including, classically, basal cell carcinoma, medulloblastoma, and rhabdomyosarcoma; more recently overactive Hh signaling has been implicated in pancreatic,<br />

lung, prostate, ovarian, and breast cancer. Thus, understanding the mechanisms that control Hh pathway activity will inform the development of novel therapeutics to treat a<br />

growing number of Hh-driven pathologies.<br />

Select Reviews:<br />

Beachy, P.A., Hymowitz, S.G., Lazarus, R.A., Leahy, D.J., and Siebold, C. (2010) Genes Dev. 24, 2001–2012. • Eaton, S. (2008) Nat. Rev. Mol. Cell Biol. 9, 437–445. •<br />

Hui, C.C. and Angers, S. (2011) Annu. Rev. Cell Dev. Biol. 27, 513–537. • Ingham, P.W., Nakano, Y., and Seger, C. (2011) Rev. Genet. 12, 393–406. • Ng, J.M. and<br />

Curran, T. (2011) Nat. Rev. Cancer 11, 493–501. • Wilson, C.W. and Chuang, P.T. (2010) Development 137, 2079–2094. • Teglund, S., and Toftgard, R. (2010) Biochim.<br />

Biophys. Acta. 1805, 181–208. • Briscoe, J., and Therond, P.P. (2013) Nat. Rev. Mol. Cell Biol. 14, 416–429. • Goetz, S.C., and Anderson, K.V. (2010) Nat. Rev. Genet.<br />

11, 331–344. • Falkenstein, K.N., and Vokes, S.A. (2014) Semin. Cell Dev. Biol. 33, 73–80.<br />

© 2010–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Kun-Liang Guan, University of California, San Diego, CA for reviewing this diagram.<br />

162 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

© 2006–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Hans Widlund, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

www.cellsignal.com/cstpathways 163


Section I: Research Areas<br />

chapter 06: Development and differentiation<br />

TGF-b Signaling<br />

ESC Pluripotency Differentiation<br />

RhoA<br />

mDia ROCK<br />

MLC LIMK<br />

Cofilin<br />

PAK<br />

Rac/<br />

Cdc42<br />

c-Abl<br />

TGF-β Ligands:<br />

TGF-βs, Activins, Nodals<br />

Par6<br />

PKC<br />

PI3K<br />

Akt<br />

mTOR<br />

Type II<br />

Receptor<br />

PP2A<br />

Type I<br />

Receptor<br />

Shc<br />

SARA<br />

Smad2/3<br />

TRAF4/6<br />

Smurf2 Smad7<br />

JNK<br />

GRB2<br />

USP4/11/15<br />

TAK1<br />

p38<br />

NF-κB<br />

SOS<br />

TMEPAI<br />

Smad7<br />

Smad2/3<br />

Smurf2<br />

Smad2/3 Smad4<br />

Smad2/3<br />

Ras<br />

Erk1/2<br />

Smad4<br />

Smurf1<br />

Type I<br />

Receptor<br />

Smad1/5/9<br />

Smad1/5/9<br />

Smad1/5/9 Smad4<br />

Smad1/5/9<br />

BMP Ligands:<br />

BMP-2, -4, -7, MIS<br />

Smurf1<br />

Smad6/7<br />

Type II<br />

Receptor<br />

LIMK<br />

Cofilin<br />

p38<br />

TAK1<br />

JNK<br />

IGF-IR<br />

PI3K<br />

Akt<br />

FGFR<br />

Ras/<br />

Raf<br />

MEK1/2<br />

Erk1/2<br />

LRP<br />

Wnt<br />

Frizzled<br />

Gα q/o<br />

PP2A<br />

APC<br />

Axin GSK-3<br />

β-catenin<br />

β-catenin<br />

TGF-β/Activin/Nodal<br />

SARA<br />

Smad2/3<br />

Smad2/3<br />

Smad2/3 Smad4<br />

Smad2/3<br />

Smad4<br />

BMP<br />

Smad1/5/9<br />

Smad1/5/9<br />

γ−Secretase<br />

Smad1/5/9 Smad4<br />

Smad1/5/9<br />

Notch<br />

NICD<br />

Actin Polymerization<br />

Stress Fibers<br />

4E-<br />

BP1/2<br />

Cell Adhesion<br />

p70 S6K<br />

Actin Polymerization<br />

Stress Fibers<br />

Cytoplasm<br />

Nucleus<br />

Erk1/2<br />

TCFs<br />

β-catenin<br />

Smad2/3<br />

Smad1/5/9<br />

NICD<br />

Cytoplasm<br />

Nucleus<br />

Smad4 CBP<br />

Smad2/3<br />

Smad2/3 TF<br />

Smad4 CBP<br />

Smad1/5/9<br />

Smad1/5/9 TF<br />

Gene Expression<br />

Oct-4 Sox2 Nanog<br />

Gene Expression<br />

Gene Expression<br />

Polycomb<br />

Transcription Factors:<br />

AP-1 homeodomain<br />

bZIP Sp1<br />

RUNX nuclear receptors<br />

Fox IRF-7<br />

bHLH<br />

Corepressors:<br />

c-Ski/SnoN<br />

Evi1<br />

TGIF<br />

SIP1<br />

Tob (BMP only)<br />

HDAC4/5<br />

Coactivators:<br />

CBP/p300<br />

SMIF<br />

MSG1<br />

ARC105<br />

Pluripotency and Self Renewal<br />

hESC markers:<br />

Oct-4 SSEA3/4<br />

Sox2 TRA-1-60<br />

Nanog TRA-1-81<br />

Differentiation<br />

Endoderm<br />

Mesoderm<br />

Ectoderm<br />

Primordial Germ Cells<br />

Transforming growth factor-β (TGF-β) superfamily signaling plays a critical role in the regulation of cell growth, differentiation, and development in a wide range of biological<br />

systems. In general, signaling is initiated with ligand-induced oligomerization of serine/threonine receptor kinases and phosphorylation of the cytoplasmic signaling molecules<br />

Smad2 and Smad3 for the TGF-β/activin pathway, or Smad1/5/9 for the bone morphogenetic protein (BMP) pathway. Carboxy-terminal phosphorylation of Smads by activated<br />

receptors results in their partnering with the common signaling transducer Smad4, and translocation to the nucleus. Activated Smads regulate diverse biological effects by<br />

partnering with transcription factors resulting in cell-state specific modulation of transcription. Inhibitory Smads, i.e. Smad6 and Smad7 antagonize activation of R-Smads.<br />

The expression of inhibitory Smads (I-Smads) 6 and 7 is induced by both activin/TGF-β and BMP signaling as part of a negative feedback loop. The stability of TGF-β family<br />

receptors and/or Smads are regulated by Smurf E3 ubiquitin ligases and USP4/11/15 deubiquitinases. TGF-β/activin and BMP pathways are modulated by MAPK signaling<br />

at a number of levels. Moreover, in certain contexts, TGF-β signaling can also affect Smad-independent pathways, including Erk, SAPK/JNK, and p38 MAPK pathways. Rho<br />

GTPase (RhoA) activates downstream target proteins, such as mDia and ROCK, to prompt rearrangement of the cytoskeletal elements associated with cell spreading, cell<br />

growth regulation, and cytokinesis. Cdc42/Rac regulates cell adhesion through downstream effector kinases PAK, PKC, and c-Abl following TGF-β activation.<br />

Select Reviews:<br />

Horbelt, D., Denkis, A., and Knaus, P. (2012) Int. J. Biochem. Cell Biol. 44, 469–474. • Ikushima, H. and Miyazono, K. (2010) Nat. Rev. Cancer 10, 415–424. • Kitisin, K.,<br />

Saha, T., Blake, T., et al. (2007) Sci. STKE 399, cm1. • Meulmeester, E. and ten Dijke, P. (2011) J. Pathol. 223, 205–218. • Schmierer, B. and Hill, C.S. (2007) Nat. Rev.<br />

Mol. Cell Biol. 8, 970–982. • Xiao, Y.T., Xiang, L.X., and Shao, J.Z. (2007) Biochem. Biophys. Res. Commun. 362, 550–553. • Zhang, L., Zhou, F., de Vinuesa, A.G., et al.<br />

(2013) Mol. Cell 51, 559–572.<br />

Two distinguishing characteristics of embryonic stem cells (ESCs) are pluripotency and the ability to self-renew. These traits, which allow ESCs to grow into any cell type in<br />

the adult body and divide continuously in the undifferentiated state, are regulated by a number of cell signaling pathways. In human ESCs (hESCs), the predominant signaling<br />

pathways involved in pluripotency and self-renewal are TGF-β, which signals through Smad2/3/4, and FGFR, which activates the MAPK and Akt pathways. The Wnt pathway<br />

also promotes pluripotency, although this may occur through a non-canonical mechanism involving a balance between the transcriptional activator, TCF1, and the repressor,<br />

TCF3. Signaling through these pathways supports the pluripotent state, which relies predominantly upon three key transcription factors: Oct-4, Sox2, and Nanog. These<br />

transcription factors activate gene expression of ESC-specific genes, regulate their own expression, suppress genes involved in differentiation, and also serve as hESCs markers.<br />

Other markers used to identify hESCs are the cell surface glycolipid SSEA3/4, and glycoproteins TRA-1-60 and TRA-1-81. In vitro, hESCs can be coaxed into derivatives of the<br />

three primary germ layers, endoderm, mesoderm, or ectoderm, as well as primordial germ cell-like cells. One of the primary signaling pathways responsible for this process is<br />

the BMP pathway, which uses Smad1/5/9 to promote differentiation by both inhibiting expression of Nanog, as well as activating the expression of differentiation-specific genes.<br />

Notch also plays a role in differentiation through the notch intracellular domain (NICD). As differentiation continues, cells from each primary germ layer further differentiate along<br />

lineage-specific pathways.<br />

Select Reviews:<br />

Bilic, J. and Izpisua Belmonte, J.C. (2012) Stem Cells 30, 33–41. • Dalton, S. (2013) Curr. Opin. Cell Biol. 25, 241–246. • Guenther, M.G. (2011) Epigenomics 3, 323–343.<br />

• Jaenisch, R. and Young, R. (2008) Cell 132, 567–582. • Ng, H.H. and Surani, M.A. (2011) Nat. Cell Biol. 13, 490–496. • Pan, G. and Thomson, J.A. (2007) Cell Res.<br />

17, 42–49. • Welham, M.J., Kingham, E., Sanchez-Ripoll, Y., Kumpfmueller, B., Storm, M., and Bone, H. (2011) Biochem. Soc. Trans. 39, 674–678. • Young, R.A. (2011)<br />

Cell 144, 940–954.<br />

© 2003–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Peter ten Dijke, Leiden University Medical Center, Leiden, The Netherlands, for reviewing this diagram.<br />

164 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

© 2009–2015 Cell Signaling Technology, Inc. • We would like to thank Justin Brumbaugh and Prof. Konrad Hochedlinger,<br />

HHMI and MGH Cancer Center, Center for Regenerative Medicine, Harvard University, Cambridge, MA, for reviewing this diagram.<br />

www.cellsignal.com/cstpathways 165


Section I: Research Areas<br />

Molecular rendering of<br />

tumor vascularization<br />

Angiogenesis<br />

Angiogenesis is the formation of new blood vessels from pre-existing larger blood vessels. It is required<br />

during embryogenesis, and defects in or interference with this process have severe consequences<br />

for embryonic development. In adult organisms, angiogenesis plays a key role in normal physiological<br />

processes such as wound healing and inflammation, and in mammalian placental development.<br />

The process of angiogenesis can also be hijacked by tumors, which require a supply of oxygen and<br />

nutrients to support the high metabolic rate of tumor cells.<br />

VEGF Receptor Signaling<br />

Vascular endothelial growth factor receptors (VEGFR) are a receptor tyrosine kinase family composed of<br />

seven extracellular immunoglobulin (Ig)-like domains, a transmembrane region, and a cytoplasmic tail<br />

containing an active kinase domain. VEGFR1 plays an important role in EC function and normal vascular<br />

development, as well as in hematopoietic function. VEGFR2 is a major receptor for VEGF-induced signaling<br />

in endothelial cells. Upon ligand binding, VEGFR2 undergoes autophosphorylation and becomes<br />

activated. Signaling from VEGFR2 is necessary for the execution of VEGF-stimulated proliferation,<br />

chemotaxis, and sprouting, as well as survival of cultured ECs in vitro and in vivo. VEGFR3 expression is<br />

largely restricted to adult lymphatic endothelium and is thought to control lymphangiogenesis.<br />

VEGFR phosphorylation<br />

at tyrosine residues<br />

activates downstream<br />

signaling cascades that<br />

support angiogenesis.<br />

Tyr801<br />

Tyr951<br />

Tyr966<br />

Tyr1008<br />

Tyr1054<br />

Tyr1059<br />

Tyr1175<br />

Tyr1214<br />

VEGF<br />

lg-like<br />

domains<br />

P<br />

Akt<br />

P TSAd<br />

P<br />

P PLCγ<br />

P<br />

Cbl<br />

P<br />

P PLCγ Cbl<br />

P Shb<br />

P Grb2 Gab1<br />

SHP2<br />

Src<br />

PI3K<br />

PLCγ<br />

PI3K<br />

Src<br />

chapter 06: Development and differentiation<br />

Vascular permeability,<br />

cell migration<br />

Endothelial cell migration<br />

Differentiation, tubulogenesis<br />

Cell proliferation, angiogenesis<br />

Focal adhesion formation,<br />

cell migration<br />

Cell migration, lamellipodia<br />

formation, capillary formation<br />

Kinase domain<br />

Autophosphorylation<br />

Angiogenesis Signaling<br />

When angiogenesis is stimulated, pro-angiogenic growth factors such as VEGF, PDGF, FGF, and TGF<br />

are released. These growth factors bind their cognate receptors on endothelial cells (EC) within preexisting<br />

vessels. This triggers a signaling cascade that activates several signaling pathways such as<br />

PI3K/Akt, Erk1/2, Smad, and Notch and results in EC proliferation and migration. New blood vessel<br />

formation occurs as ECs use matrix metalloproteases (MMPs) and integrins to digest extracellular<br />

matrix and migrate into new territory where they lengthen and form tubes.<br />

VEGF treatment results in phosphorylation<br />

of VEGFR-2 at Tyr1175.<br />

PathScan ® Phospho-VEGFR-2 (Tyr1175) Sandwich ELISA Kit #7335:<br />

Treatment of HUVEC with VEGF stimulates phosphorylation of VEGFR-2 at<br />

Tyr1175, detected by #7335, but does not affect the level of total VEGFR-2<br />

protein detected by PathScan ® Total VEGFR-2 Sandwich ELISA kit #7340.<br />

Absorbance at 450 nm is shown in the top figure, while the corresponding<br />

western blot using Phospho-VEGFR-2 (Tyr1175) Rabbit mAb #2478 (right<br />

panel) or VEGFR-2 Rabbit mAb (7340-55B11) (left panel), is shown in the<br />

bottom figure.<br />

Untreated<br />

VEGF-treated<br />

Absorbance 450nm<br />

3.5<br />

3.0<br />

2.5<br />

2.0<br />

1.5<br />

1.0<br />

0.5<br />

0<br />

kDa<br />

200<br />

140<br />

Total VEGFR-2<br />

Phospho-VEGFR-2<br />

(Tyr1175)<br />

– + – + VEGF<br />

TPA induces expression of MMP-9, a protease that digests<br />

extracellular matrix and is associated with tumor angiogenesis.<br />

MMP-9 (D6O3H) XP ® Rabbit mAb #13667:<br />

IHC analysis of paraffin-embedded human breast<br />

carcinoma (A) using #13667. WB analysis of<br />

concentrated, serum-free cultured medium from<br />

U-2 OS cells, untreated (-) or treated with TPA<br />

#4174 (200 nM, 48 hr; +) (B), using #13667.<br />

A<br />

B<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

– +<br />

MMP-9<br />

TPA<br />

Neuropilin-2<br />

In addition to neuronal guidance, the single pass, transmembrane glycoprotein neuropilin-2<br />

is a co-receptor for VEGFR2 and VEGFR3, and plays a role in the development of vascular<br />

and lymphatic systems through binding VEGF 165 and VEGF145.<br />

Neuropilin-2 is expressed in the<br />

developing mouse embryo.<br />

Neuropilin-2 (D39A5) XP ® Rabbit mAb #3366: Confocal<br />

IF analysis of E14.5 mouse embryo using #3366 (green).<br />

Blue pseudocolor = DRAQ5 ® #4084 (fluorescent DNA dye).<br />

<strong>CST</strong> Technical<br />

Support<br />

When you contact <strong>CST</strong> for technical<br />

support, you can be confident you<br />

will be working with a colleague<br />

you can trust, striving to provide the<br />

highest quality products and services<br />

to you, our customer. Please see our<br />

Technical Support resource page<br />

online for contact information.<br />

www.cellsignal.com/cstsupport<br />

166 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstangiogenesis<br />

167


Section I: Research Areas<br />

chapter 06: Development and differentiation<br />

Elevated PDGF<br />

Receptor β expression<br />

is found within stromal<br />

cells of many forms<br />

of cancer.<br />

PDGF Receptor β (28E1) Rabbit mAb<br />

#3169: IHC analysis of paraffin-embedded<br />

human colon carcinoma using #3169.<br />

CD73 expression on<br />

tumor cells promotes<br />

angiogenesis.<br />

NT5E/CD73 (D7F9A) Rabbit mAb<br />

#13160: IHC analysis of paraffin-embedded<br />

human lung carcinoma using #13160.<br />

Pericyte Signaling<br />

Pericytes are support cells that provide structural stability for newly formed blood vessels, promote EC<br />

survival, and regulate vasoconstriction and dilation. This is done through a reciprocal signaling mechanism<br />

in which PDGF-BB secreted into the matrix by ECs acts as a ligand for PDGFR-β located on the<br />

pericyte membrane. In return, pericytes produce and secrete VEGF that signals through the endothelial<br />

VEGF receptor.<br />

Tumor Signaling<br />

Tumor angiogenesis occurs when cancer cells stimulate new blood vessel growth in order to bring<br />

oxygen and nutrients to a tumor. As a tumor grows in size, diffusion is no longer sufficient to oxygenate<br />

the cells at the center of the mass, creating a hypoxic environment. Hypoxia stabilizes levels of HIF-1α,<br />

a transcription factor that responds to changing oxygen levels. Under hypoxic conditions, HIF-1α binds<br />

HIF-1β to activate transcription of angiogenesis-promoting genes. Cancer cells also secrete a variety<br />

of growth factors and cytokines that stimulate classical angiogenic signaling pathways, extracellular<br />

matrix remodeling, and an inflammatory response that leads to new blood vessel formation.<br />

Under normoxia conditions, HIF-1α is<br />

hydroxylated at Pro564, marking it for degradation.<br />

PathScan ® Hydroxy-HIF-1α (Pro564) Sandwich ELISA Kit #13201:<br />

Treatment of HeLa cells with the hydroxylase inhibitor dimethyloxaloylglycine<br />

(DMOG) results in decreased hydroxylation of HIF-1α, as detected by<br />

#13201, but does not affect the level of total HIF-1α detected by PathScan ®<br />

Total HIF-1α Sandwich ELISA Kit #13127. Absorbance at 450 nm is shown<br />

in the top figures, while corresponding western blots using a total HIF-1α<br />

antibody (left) and Hydroxy-HIF-1α (Pro564) (D43B5) XP ® Rabbit mAb #3434<br />

(right) are shown in the bottom figures. Treatment of HeLa cells with the<br />

proteasome inhibitor MG-132 #2194 stabilizes HIF-1α protein.<br />

Untreated<br />

MG-132-treated<br />

MG-132-treated,<br />

DMOG-treated<br />

Total HIF-1α<br />

Hydroxy-HIF-1α<br />

(Pro564)<br />

Angiogenesis in Cancer<br />

Cancer cells secrete a variety of growth factors and cytokines that stimulate classical angiogenesis<br />

signaling pathways and extracellular matrix remodeling. Cytokines can also induce an inflammatory<br />

response that initiates angiogenesis and consequent vascularization of the tumor. Sprouting angiogenesis<br />

is the first step in this neovascularization process. In response to stimuli released by tumor cells,<br />

a single EC migrates toward the angiogenic factors and proliferates to form sprouts of ECs surrounding<br />

a lumen that is connected to the mother blood vessel. Tumors may also be vascularized by intussusceptive<br />

angiogenesis. This process proceeds much more quickly than sprouting and is essentially the<br />

splitting of an existing blood vessel into two new vessels. The ECs, pericytes, and basement membrane<br />

associated with a tumor exhibit abnormalities leading to tortuous, poorly organized, and leaky vasculature.<br />

However, this sub-optimal structure is often sufficient to supply the tumor with oxygen, nutrients,<br />

and soluble factors that help it survive and expand. It is through this vascular system that some tumor<br />

cells are able to escape and travel through the circulatory system to new locations. Such metastatic<br />

colonization is a key factor in the fatal outcomes of many types of cancer, and the density of tumor<br />

angiogenesis has been linked to tumor metastasis and patient survival rates. Accordingly, angiogenesis<br />

is under investigation as an important prognostic indicator in cancer, and angiogenesis inhibitors are<br />

attractive as candidate therapeutics.<br />

Absorbance 450nm<br />

3.5<br />

3.0<br />

2.5<br />

2.0<br />

1.5<br />

1.0<br />

0.5<br />

0<br />

– + + – + + MG-132<br />

– – + – – + DMOG<br />

Angiogenesis Resources<br />

Please visit our website for additional resources and products relating to the study of Angiogenesis.<br />

www.cellsignal.com/cstangiogenesis<br />

In ECs, VE-Cadherin signaling, expression, and localization<br />

correlate with vascular permeability and tumor angiogenesis.<br />

VE-Cadherin (D87F2) XP ® Rabbit<br />

mAb #2500: Confocal IF analysis of<br />

HUVEC (VE-Cadherin positive; left) and<br />

HeLa cells (VE-Cadherin negative; right)<br />

using #2500 (green). Actin filaments<br />

have been labeled with DY-554 phalloidin<br />

(red). Blue pseudocolor = DRAQ5 ®<br />

#4084 (fluorescent DNA dye).<br />

FGFR1 binds acidic FGF and basic FGF, both potent promoters of angiogenesis.<br />

A<br />

B<br />

Events<br />

60<br />

50<br />

40<br />

60<br />

50<br />

40<br />

30<br />

FGF Receptor 1<br />

FGF Receptor 1 (D8E4) XP ® Rabbit mAb #9740: IHC analysis of paraffin-embedded human breast carcinoma (A) using<br />

#9740. Flow cytometric analysis of A-204 cells (B) using #9740 (blue) compared to concentration matched Rabbit (DA1E)<br />

mAb IgG XP ® Isotype Control #3900 (red). WB analysis of extracts from A-204 (FGFR1 positive), KG-1a (FGFR1 oncogenic<br />

partner-FGFR1 fusion), A172 (FGFR1 low expression), and HT-29 (FGFR1 negative) cells (C) using #9740 (upper) and β-Actin<br />

(D6A8) Rabbit mAb #8457 (lower).<br />

Angiogenesis Modulators<br />

Select Reviews<br />

Benazzi, C., Al-Dissi, A., Chau, C.H., et al. (2014) Scientific World Journal 2014, 919570. • Claesson-Welsh, L. (2003)<br />

Biochem. Soc. Trans. 31, 20–24. • Ferrara, N., Gerber, H.P., and LeCouter, J. (2003) Nat. Med. 9, 699–676. • Jain, R.K.<br />

(2005) Science 307, 58–62. • Karkkainen, M.J. and Petrova, T.V. (2000) Oncogene 19, 5598–5605. • Koch, S., Tugues, S.,<br />

Li, X., Gualandi, L., and Claesson-Welsh, L. (2011) Biochem. J. 437, 169–183. • Lu, X. and Kang, Y. (2010) Clin. Cancer Res.<br />

16, 5928–5935. • Makanya, A.N., Hluchchuk, R., and Djonov, V.G. (2009) Angiogenesis 12, 113–123. • Meyer, M., Clauss,<br />

M., Lepple-Wienhues, A., et al. (1999) EMBO J. 18, 363–374. • Patel-Hett, S. and D’Amore, P.A. (2011) Int. J. Dev. Biol. 55,<br />

353–363. • Rahimi, N., Dayanir, V., and Lashkari, K. (2000) J. Biol. Chem. 275, 16986–16992. • Ribatti, D., Nico, B., and<br />

Crivellato, E. (2011) Int. J. Dev. Biol. 55, 261–268. • Robinson, C.J. and Stringer, S.E. (2001) J. Cell Sci. 114, 853–865. •<br />

Saharinen, P., Eklund, L., Pulkki, K., et al. (2011) Trends Mol. Med. 17, 347–362. • Sakurai, T. and Kudo, M. (2011) Oncology<br />

81 Suppl 1, 24–29. • Senger, D.R. and Davis, G.E. (2011) Cold Spring Harb. Perspect Biol. 3, a005090. • Weis, S.M. and<br />

Cheresh, D.A. (2011) Nat. Med. 17, 1359–1370.<br />

C<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

1 2 3 4<br />

Promoters of Angiogenesis<br />

Inhibitors of Angiogenesis<br />

Acidic fibroblast growth factor (aFGF) #5234<br />

ADAMTS1<br />

Angiogenin<br />

Angiostatin<br />

Basic fibroblast growth factor (bFGF) #8910<br />

Endostatin<br />

Epidermal growth factor (EGF) #8916 Interferons (alpha) #8927<br />

Granulocyte colony-stimulating factor (GM-CSF) #8922 Platelet factor 4<br />

Hepatocyte growth factor (HGF)<br />

Prolactin 16 kDa fragment<br />

Interleukin 8 (IL-8) #8921<br />

Thrombospondin<br />

Placental growth factor (PGF)<br />

Tissue inhibitor of metalloproteinase-1 (TIMP-1)<br />

Platelet-derived endothelial growth factor (PEGF)<br />

Tissue inhibitor of metalloproteinase-2 (TIMP-2)<br />

Transforming growth factor alpha (TGF-α) #5495<br />

Tissue inhibitor of metalloproteinase-3 (TIMP-3)<br />

Tumor necrosis factor alpha (TNF-α) #8902<br />

Vascular endothelial growth factor (VEGF) #8908, #8065<br />

FGFR1<br />

FOP-<br />

FGFR1<br />

β-Actin<br />

Lanes<br />

1. A-204<br />

2. KG-1a<br />

3. A172<br />

4. HT-29<br />

168 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstangiogenesis<br />

169


Section I: Research Areas<br />

These protein targets represent key<br />

nodes within angiogenesis signaling<br />

pathways and are commonly studied<br />

in angiogenesis research. Primary<br />

antibodies, antibody conjugates, and<br />

antibody sampler kits containing these<br />

targets are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

E PathScan ® ELISA Kits<br />

S SignalSilence ® siRNA<br />

C Antibody Conjugate<br />

163<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for VEGF Receptor 2<br />

have been cited over 163 times in highimpact,<br />

peer-reviewed publications from<br />

the global research community.<br />

Commonly Studied Angiogenesis Targets<br />

Target M P E S C<br />

ADAMTS1<br />

•<br />

Angiopoietin-2<br />

•<br />

CA9<br />

•<br />

CBP<br />

• •<br />

Acetyl-CBP (Lys1535)/p300 (Lys1499) •<br />

CD31 (PECAM-1)<br />

•<br />

Cripto<br />

• •<br />

CYR61<br />

•<br />

DLL4<br />

•<br />

eNOS<br />

• •<br />

EphA2<br />

• •<br />

Phospho-EphA2 (Tyr594) •<br />

EphB1<br />

•<br />

Acidic FGF<br />

•<br />

FGF Receptor 1 • • • •<br />

FGF Receptor 2<br />

• • • •<br />

FGF Receptor 3<br />

• •<br />

FGF Receptor 4 • • •<br />

FIH<br />

•<br />

Gremlin<br />

•<br />

HIF-1α<br />

• •<br />

Hydroxy-HIF-1α (Pro564) • •<br />

HIF-1β/ARNT<br />

• •<br />

HO-1<br />

• •<br />

Integrin α6<br />

•<br />

Integrin αV<br />

•<br />

Integrin β1<br />

• •<br />

Integrin β3<br />

• •<br />

Integrin β5<br />

• •<br />

Jagged1<br />

•<br />

Maspin<br />

•<br />

MMP-2<br />

• •<br />

MMP-7<br />

•<br />

MMP-9<br />

• •<br />

NDRG1 • • •<br />

Select Citations:<br />

Whiteus, C. et al. (2014) Perturbed neural activity disrupts<br />

cerebral angiogenesis during a postnatal critical period.<br />

Nature 505, 407−411.<br />

Behjati, S. et al. (2014) Recurrent PTPRB and PLCG1 mutations<br />

in angiosarcoma. Nat. Genet. 46, 376−379.<br />

Chen, P.Y., Qin, L., Zhuang, Z.W., Tellides, G., Lax, I.,<br />

Schlessinger, J., Simons, M. (2014) The docking protein<br />

FRS2alpha is a critical regulator of VEGF receptors signaling.<br />

Proc. Natl. Acad. Sci. USA 111, 5514−5519.<br />

Ria, R. et al. (2014) HIF-1alpha of bone marrow endothelial<br />

cells implies relapse and drug resistance in patients with<br />

multiple myeloma and may act as a therapeutic target. Clin.<br />

Cancer Res. 20, 847−858.<br />

Blosser, W. et al. (2014) A method to assess target gene involvement<br />

in angiogenesis in vitro and in vivo using lentiviral<br />

vectors expressing shRNA. PLoS One 9, e96036<br />

Riquelme, E. et al. (2014) VEGF/VEGFR-2 Upregulates EZH2<br />

Expression in Lung Adenocarcinoma Cells and EZH2 Depletion<br />

Enhances the Response to Platinum-Based and VEGFR-<br />

2-Targeted Therapy. Clin. Cancer Res. 20, 3849−3861.<br />

Yang, Y. et al. (2013) GAB2 induces tumor angiogenesis in<br />

NRAS-driven melanoma. Oncogene 32, 3627−3637.<br />

Nakayama, M. et al. (2013) Spatial regulation of VEGF receptor<br />

endocytosis in angiogenesis. Nat. Cell Biol. 15, 249−260.<br />

Target M P E S C<br />

NDRG2<br />

•<br />

NDRG3<br />

•<br />

NDRG4<br />

•<br />

Neuropilin-2<br />

•<br />

Notch1 • •<br />

Cleaved Notch1 (Val1744) • • •<br />

Notch2<br />

•<br />

Notch3<br />

• •<br />

Notch4<br />

•<br />

NT5E/CD73<br />

• •<br />

PDGF Receptor α • • • •<br />

Phospho-PDGF Receptor α (Tyr754) •<br />

PDGF Receptor β • • •<br />

Phospho-PDGF Receptor β (Tyr740) •<br />

Phospho-PDGF Receptor β (Tyr771) •<br />

Phospho-PDGF Receptor β (Tyr1009) •<br />

PHD-2/Egln1<br />

• •<br />

RECK<br />

•<br />

Ron<br />

•<br />

Phospho-Ron (panTyr)<br />

•<br />

Spry1<br />

• •<br />

Tenascin C<br />

•<br />

TGF-β Receptor I<br />

•<br />

TGF-β Receptor III<br />

• •<br />

Tie2<br />

•<br />

TIMP1<br />

•<br />

TIMP2<br />

•<br />

TIMP3<br />

•<br />

uPAR<br />

• •<br />

VE-Cadherin<br />

• •<br />

VEGF Receptor 1<br />

•<br />

VEGF Receptor 2 • • • •<br />

VEGF Receptor 3<br />

• •<br />

VHL<br />

•<br />

Planas-Paz, L. et al. (2012) Mechanoinduction of lymph<br />

vessel expansion. EMBO J. 31, 788−804.<br />

Fang, L. et al. (2013) Control of angiogenesis by AIBPmediated<br />

cholesterol efflux. Nature 498, 118−122.<br />

Harris, N.C. et al. (2013) The propeptides of VEGF-D<br />

determine heparin binding, receptor heterodimerization, and<br />

effects on tumor biology. J. Biol. Chem. 288, 8176−8186.<br />

Mujahid, S. et al. (2013) MiR-221 and miR-130a regulate<br />

lung airway and vascular development. PLoS One 8,<br />

e55911.<br />

Singh, N.K. et al. (2013) Both Kdr and Flt1 play a vital role in<br />

hypoxia-induced Src-PLD1-PKCgamma-cPLA(2) activation<br />

and retinal neovascularization. Blood 121, 1911−1923.<br />

Gourlaouen, M. et al. (2013) Essential role for endocytosis<br />

in the growth factor-stimulated activation of ERK1/2 in<br />

endothelial cells. J. Biol. Chem. 288, 7467−7480.<br />

Breitbach, C.J. et al. (2013) Oncolytic vaccinia virus disrupts<br />

tumor-associated vasculature in humans. Cancer Res. 73,<br />

1265−1275.<br />

Tang, J.R. et al. (2013) The NF-kappaB inhibitory proteins<br />

IkappaBalpha and IkappaBbeta mediate disparate responses<br />

to inflammation in fetal pulmonary endothelial cells.<br />

J. Immunol. 190, 2913−2923.<br />

170 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

Angiogenesis Signaling in Tumor Neovascularization<br />

Endothelial<br />

Cell<br />

Key<br />

Angiopoietin 1<br />

Angiopoietin 2<br />

bFGF<br />

Ephrin<br />

PDGF<br />

SLIT<br />

VEGF<br />

Tie2<br />

EPH<br />

ROBO3<br />

ROBO1/2<br />

Pericyte<br />

Integrins<br />

MMPs<br />

Basement<br />

Membrane<br />

Tip Cell<br />

FGFR<br />

Integrins<br />

VEGFR2<br />

Akt<br />

4E-<br />

BP1<br />

Tie2<br />

Neuropilin<br />

elF4E1<br />

• Growth Factors<br />

• Cytokines<br />

• ECM Proteases<br />

Tumor Cell<br />

chapter 06: Development and differentiation<br />

PHDs<br />

Erk1/2<br />

HIF-1α<br />

Precursor<br />

Endothelial Cell<br />

OH<br />

HRE<br />

OH<br />

HIF-1α<br />

HIF-1α<br />

CBP/p300<br />

HIF-1β<br />

Normoxia<br />

HIF-1β<br />

Hypoxia<br />

Target Genes<br />

Platelet<br />

MMPs<br />

Nucleus<br />

PDGFR<br />

• Growth Factors<br />

• Cytokines<br />

• ECM Proteases<br />

Ets2<br />

HIF-1α<br />

Stat3<br />

Target Genes<br />

Tumor Associated<br />

Macrophage (TAM)<br />

Angiogenesis, the formation of new blood vessels from pre-existing blood vessels, plays a key role in tumorigenesis. When a small dormant tumor initiates angiogenesis,<br />

referred to as the ‘angiogenic switch’, it secretes factors that induce sprouting and chemotaxis of endothelial cells (ECs) towards the tumor mass. Within the hypoxic environment<br />

of the inner tumor mass the transcription factor Hypoxia-Inducible-Factor-1-α (HIF-1α) is stabilized and activates the expression of multiple genes contributing to the angiogenic<br />

process. HIF-1α induced proteins include Vascular Endothelial Growth Factor (VEGF) and Basic Fibroblast Growth Factor (bFGF), which promote vascular permeability and EC<br />

growth, respectively. Other secreted factors, such as PDGF, angiopoietin 1 and angiopoietin 2 facilitate chemotaxis, while ephrins guide newly formed blood vessels through<br />

maintenance of cell-cell separation. Other HIF-1α induced gene products include matrix metalloproteinases (MMPs) that breakdown the extracellular matrix to facilitate EC<br />

migration and release associated growth factors. Certain integrins such as αVβ3 found on the surface of angiogenic ECs help the sprouting ECs adhere to the provisional Extracellular<br />

Matrix (ECM), migrate and survive. Factors secreted into the microenvironment surrounding the tumor activate tumor-associated macrophages (TAMs), that subsequently<br />

produce angiogenic factors, such as VEGF and MMPs, further promoting angiogenesis. Pericytes function as support cells enveloping the basolateral surface of ECs and regulate<br />

vasoconstriction and dilation under normal physiologic conditions. During the process of tumor angiogenesis sprouting vessels lack pericytes, which are later recruited by<br />

ECs to provide structural support that indirectly promotes tumor survival. For example, PDGF secreted by ECs acts as a ligand for PDGF receptor located on the pericyte<br />

membrane, causing pericytes to produce and secrete VEGF that signals through the endothelial VEGF receptor.<br />

Select Reviews:<br />

Folkman, J. (2007) Nat. Rev. Drug Disc. 6, 273–286. • Guo, C., Buranych, A., Sarkar, D., et al. (2013) Vasc. Cell. 5, 20. • Keith, B., Johnson, R.S., and Simon, M.C.<br />

(2012) Nat. Rev. Cancer 12, 9–22. • Klagsbrun, M. and Eichmann, A. (2005) Cytokine and Growth Factor Rev. 16, 535–548. • Raza, A., Franklin, M.J., and Dudek, A.Z.<br />

(2010) J. Hematol. 85, 593–598. • Sakurai, T. and Kudo, M. (2011) Oncology 81 Suppl. 1, 24–29. • Senger, D.R. and Davis, G.E. (2011) Cold Spring Harb. Perspect. Biol.<br />

3, a005090. • van Hinsbergh, V.W. and Koolwijk, P. (2008) Cardiovasc. Res. 78, 203–212.<br />

© 2008–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Diane Bielenberg, Harvard Medical School, Children’s Hospital, Boston, MA, for reviewing this diagram.<br />

www.cellsignal.com/cstpathways 171


07<br />

Section I: Research Areas<br />

Syk is expressed<br />

in B cells and<br />

other cell lines.<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

1 2 3 4<br />

Syk<br />

Syk (D3Z1E) XP ® Rabbit mAb #13198:<br />

WB analysis of extracts from various cell<br />

lines using #13198.<br />

Lanes<br />

1. SW620<br />

2. SR<br />

3. A20<br />

4. YB2/0<br />

Immunology and Inflammation<br />

B Cell and T Cell Receptor Signaling and Adaptive Immunity<br />

B and T lymphocytes mediate the humoral and cell-mediated immune responses, respectively, which<br />

make up the adaptive arm of the immune system. B cells mature in the bone marrow and differentiate<br />

into antibody-secreting plasma cells. In contrast, T cells are thymus-derived and, as effector cells,<br />

orchestrate cell-mediated immunity.<br />

The B cell receptor (BCR) is composed of a membrane-bound antibody (immunoglobulin or Ig) flanked<br />

by Igα/Igβ (CD79A/CD79B) heterodimers. When membrane Ig binds antigen, the CD79 heterodimer<br />

transduces signals through its cytoplasmic immunoreceptor tyrosine-based activation motif (ITAM)<br />

domains. The T cell receptor (TCR) consists of a membrane-bound αβ heterodimer (TCRαβ), four CD3<br />

chains (two CD3ε, one CD3γ, one CD3δ), and a ζ−chain homodimer. The TCRαβ dimer recognizes<br />

antigenic peptides, while the associated signaling chains transduce signals with their cytoplasmic ITAM<br />

domains. Thus, the lymphocyte antigen receptors use similar models of membrane-bound antigen<br />

receptors linked to signal-transducing accessory chains.<br />

Signaling through the BCR and TCR involves activation of a number of Src family tyrosine kinases (Blk,<br />

Fyn, and Lyn in B cells and Fyn and Lck in T cells), which are responsible for phosphorylation of the<br />

receptor-associated ITAM motifs. Phosphorylated ITAMs act as docking sites for Syk family tyrosine<br />

kinases (Syk in B cells and Zap-70 in T cells). Activated Syk kinases amplify signals through phosphorylation<br />

of downstream adaptor proteins, thereby initiating a cascade of intracellular signaling molecules.<br />

In addition to mediating cell activation, lymphocyte receptor signaling drives B and T cell development,<br />

differentiation, proliferation, and survival.<br />

Events<br />

Zap-70 is expressed in T cells.<br />

Zap-70 (D1C10E) XP ® Rabbit mAb (Alexa Fluor ® 488 Conjugate) #9473: Flow<br />

cytometric analysis of Ramos (B cells; blue) and Jurkat (T cells; green) cells using #9473.<br />

Jak/Stat Signaling<br />

The Jak/Stat signaling pathway is utilized by a large number of cytokines, growth factors, and hormones<br />

upon binding to their specific receptors. Receptor-mediated tyrosine phosphorylation of Jak family<br />

members triggers phosphorylation of Stat proteins, resulting in their nuclear translocation, binding<br />

to specific DNA elements, and subsequent activation of transcription. The remarkable range and<br />

specificity of responses regulated by the Stats is determined, in part, by the tissue-specific expression<br />

of different cytokine receptors, Jaks, and Stats, as well as by the combinatorial coupling of various<br />

Stat members to different receptors. Stat1 is activated in response to a large number of ligands and is<br />

essential for responsiveness to IFN-α and IFN-γ. Stat3 is constitutively activated in a number of human<br />

tumors and possesses both oncogenic potential and antiapoptotic activities. Stat4 has been most extensively<br />

investigated as a mediator of IL-12 responses. Stat5 is activated in response to a wide variety<br />

of ligands including IL-2, GM-CSF, growth hormone, and prolactin.<br />

Cytokine stimulation results in<br />

phosphorylation of Stat3 at Tyr705.<br />

Phospho-Stat3 (Tyr705) (D3A7) XP ® Rabbit mAb (Alexa Fluor ®<br />

488 Conjugate) #4323: Flow cytometric analysis of Jurkat cells,<br />

untreated (blue) or IFN-α treated (green), using #4323 compared to<br />

isotype control antibody (red).<br />

Growth factor<br />

stimulation results<br />

in phosphorylation<br />

of Stat5 at Tyr694.<br />

A<br />

Events<br />

Phospho-Stat3 (Tyr705)<br />

(Alexa Fluor ® 488 Conjugate)<br />

B<br />

chapter 07: immunology and inflammation<br />

Phospho-Stat5 (Tyr694) (D47E7)<br />

XP ® Rabbit mAb #4322: Confocal<br />

IF analysis of A-431 cells, treated<br />

with Human Epidermal Growth Factor<br />

(hEGF) #8916 (A) or untreated (B),<br />

using #4322 (green) and Pan-Keratin<br />

(C11) Mouse mAb #4545 (red).<br />

Zap-70<br />

TLR Signaling and Innate Immunity<br />

The innate arm of the immune system consists of a host of immune cells and resistance mechanisms<br />

that act as the first line of defense against invading pathogens. The toll-like receptors (TLRs) are a family<br />

of evolutionarily conserved pattern recognition receptors (PRRs) that recognize the pathogen-associated<br />

molecular patterns (PAMPs) found in microbial pathogens. TLR1, 2, 4, 5, and 6 are expressed at the<br />

cell surface, while TLR3, 7, 8, and 9 have been shown to localize to intracellular vesicles. Activation<br />

of TLRs through ligand binding triggers a signaling cascade involving a variety of intracellular signaling<br />

adaptors including MyD88, IRAKs, and TRAF6. TLR signaling leads to the activation of the MAP kinase,<br />

NF-κB, and IRF signaling pathways, which mediate inflammation through the production of inflammatory<br />

cytokines, type I IFN, chemokines, and antimicrobial peptides. TLR signaling in innate immune cells, particularly<br />

dendritic cells, leads to their activation and subsequent induction of adaptive immune responses.<br />

TLR2 expression in mouse macrophages and dendritic cells<br />

Toll-like Receptor 2 (E1J2W) Rabbit mAb (Mouse Specific) #13744: WB analysis<br />

of extracts from Raw 264.7 cells, mouse bone marrow-derived macrophages (BMDM),<br />

and mouse bone marrow-derived dendritic cells (BMDC) using #13744.<br />

kDa 1 2 3<br />

200<br />

172 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

140<br />

100<br />

80<br />

60<br />

50<br />

TLR2<br />

Lanes<br />

1. Raw 264.7<br />

2. BMDM<br />

3. BMDC<br />

NF-κB Signaling<br />

Transcription factors of the nuclear factor κB (NF-κB)/Rel family play a pivotal role in inflammatory and<br />

immune responses. There are five family members in mammals: RelA, c-Rel, RelB, NF-κB1 (p105/p50),<br />

and NF-κB2 (p100/p52). Both p105 and p100 are co-translationally processed by the proteasome to<br />

produce p50 and p52, respectively. Rel proteins bind p50 and p52 to form dimeric complexes that<br />

bind DNA and regulate transcription. In unstimulated cells, NF-κB is sequestered in the cytoplasm by<br />

IκB inhibitory proteins. NF-κB-activating agents can induce the phosphorylation of IκB proteins, targeting<br />

them for rapid degradation through the ubiquitin-proteasome pathway and releasing NF-κB to enter<br />

the nucleus where it regulates gene expression. NIK and IKKα (IKK1) regulate the phosphorylation and<br />

processing of NF-κB2 (p100) to produce p52, which translocates to the nucleus.<br />

NF-κB1 p105/p50 associates with promoters for<br />

IκBα and IL-8, but not with α satellite repeat element.<br />

NF-κB1 p105/p50 (D4P4D) Rabbit mAb #13586: Chromatin IPs were<br />

performed with cross-linked chromatin from 4 x 10 6 HeLa cells treated<br />

with Human Tumor Necrosis Factor-α (hTNF-α) #8902 (30 ng/ml, 1 hr)<br />

and either 10 μl of #13586 or 2 μl of Normal Rabbit IgG #2729 using<br />

SimpleChIP ® Enzymatic Chromatin IP Kit (Magnetic Beads) #9003. The<br />

enriched DNA was quantified by real-time PCR using SimpleChIP ® Human<br />

IκBα Promoter Primers #5552, human IL-8 promoter primers, and<br />

SimpleChIP ® Human α Satellite Repeat Primers #4486. The amount of<br />

immunoprecipitated DNA in each sample is represented as a percent of<br />

the total input chromatin.<br />

NF-κB1 p105/p50 (D4P4D)<br />

Rabbit mAb #13586<br />

Normal Rabbit<br />

IgG #2729<br />

% of total input chromatin<br />

4.0<br />

3.5<br />

3.0<br />

2.5<br />

2.0<br />

1.5<br />

1.0<br />

0.5<br />

0<br />

IκB-α<br />

IL-8<br />

α Satellite<br />

TNF-α treatment<br />

results in translocation<br />

of NF-κB p65 (RelA) to<br />

the nucleus.<br />

NF-κB p65 (D14E12) XP ® Rabbit<br />

mAb #8242: Confocal IF analysis of<br />

HT-1080 cells, untreated (top) or treated<br />

with hTNF-α #8902 (20 ng/ml, 20 min)<br />

(bottom), using #8242 (green). Actin<br />

filaments were labeled with DY-554<br />

phalloidin (red). Blue pseudocolor =<br />

DRAQ5 ® #4084 (fluorescent DNA dye).<br />

www.cellsignal.com/cstimmunology 173


Section I: Research Areas<br />

chapter 07: immunology and inflammation<br />

PD-L1 is expressed<br />

in lung carcinoma.<br />

PD-L1 (E1L3N ® ) XP ® Rabbit mAb<br />

#13684: IHC analysis of paraffin-embedded<br />

human lung carcinoma using #13684.<br />

These protein targets represent<br />

key nodes within immunology and<br />

inflammation signaling pathways and<br />

are commonly studied in immunology<br />

and inflammation research. Primary<br />

antibodies, antibody conjugates, and<br />

antibody sampler kits containing these<br />

targets are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

E PathScan ® ELISA Kits<br />

S SignalSilence ® siRNA<br />

C Antibody Conjugate<br />

Immune Checkpoints<br />

Activation of T lymphocytes by antigen-presenting cells (APCs) requires engagement of the T cell<br />

receptor with MHC class I or II molecules and co-stimulatory signals generated from CD28 (on T cells)<br />

binding to CD80 or CD86 (on APCs). However, under certain circumstances, such as maintaining<br />

self-tolerance or preventing collateral tissue damage, T cell engagement is coupled with inhibitory<br />

signals that repress T cell activation and response, known as immune checkpoints. Immune checkpoint<br />

proteins such as PD-1 and CTLA-4, which are commonly upregulated in infitrating T cells, bind their<br />

corresponding ligands, PD-L1 and CD80/86 respectively, which are upregulated in cancer cells as a<br />

means to evade immune detection and downregulate T cell response. Activating antitumor immunity<br />

through the blockade of immune checkpoint proteins has become a promising therapeutic strategy for<br />

the treatment of cancer.<br />

Select Reviews<br />

Borroto, A., Abia, D., and Alarcón, B. (2014) Immunol. Lett. 161, 113−117. • Burger, J.A. and Chiorazzi, N. (2013) Trends Immunol.<br />

34, 592−601. • Dorritie, K.A., Redner, R.L., and Johnson, D.E. (2014) Adv. Biol. Regul. 56, 30–44. • Fu, G., Rybakin, V.,<br />

Brzostek, J., et al. (2014) Trends Immunol. 35, 311−318. • Gasparini, C., Celeghini, C., Monasta, L., et al. (2014) Cell Mol. Life<br />

Sci. 71, 2083−2102. • Gerondakis, S., Fulford, T.S., Messina, N.L., et al. (2014) Nat. Immunol. 15, 15−25. • Harwood, N.E. and<br />

Batista, F.D. (2010) Annu. Rev. Immunol. 28, 185–210. • Kawakami, Y., Yaguchi, T., Park, J.H., et al. (2013) Front. Oncol. 3, 136.<br />

• Maddaly, R., Pai, G., Balaji, S., et al. (2010) FEBS Lett. 584, 4883–4894. • Mifsud, E.J., Tan, A.C., and Jackson, D.C. (2014)<br />

Front. Immunol. 5, 79. • Pardoll, D.M. (2012) Nat. Rev. Cancer 12, 252–264. • Reuven, E.M., Fink, A., and Shai, Y. (2014)<br />

Biochim. Biophys. Acta. 1838, 1586−1593. • Vanneman, M. and Dranoff, G. (2012) Nat. Rev. Cancer 12, 237−251.<br />

Commonly Studied Immunology and Inflammation Targets<br />

Target M P E S C<br />

A20/TNFAIP3 • •<br />

ABIN-1 • •<br />

ADAP •<br />

AID •<br />

AIM2 • •<br />

Aiolos •<br />

AML1 • •<br />

Phospho-AML1<br />

(Ser249)<br />

•<br />

β2-microglobulin • • •<br />

BACH2 •<br />

BAFF •<br />

Basigin/EMMPRIN • •<br />

BATF •<br />

BCL6 • •<br />

Bcl10 •<br />

Blimp-1/PRDI-BF1 •<br />

Blk •<br />

BLNK •<br />

Phospho-BLNK<br />

(Tyr96) •<br />

Btk • •<br />

•<br />

Phospho-Btk<br />

(Ser180)<br />

Phospho-Btk<br />

(Tyr223)<br />

CARD9<br />

CARD11<br />

Phospho-CARD11<br />

(Ser652)<br />

CBFβ<br />

CCR2<br />

CD3ε<br />

CD4<br />

CD8<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

Target M P E S C<br />

CD9 •<br />

CD10 •<br />

CD13 •<br />

CD19 •<br />

Phospho-CD19<br />

(Tyr531) •<br />

CD31 (PECAM-1) •<br />

CD34 •<br />

CD44 • • • •<br />

CD45 •<br />

CD46 •<br />

CD79A • •<br />

Phospho-CD79A<br />

(Tyr182) •<br />

CD82 • •<br />

CIITA •<br />

CISH •<br />

CrkL •<br />

•<br />

Phospho-CrkL<br />

(Tyr207)<br />

Cytokine Receptor<br />

Common β-Chain<br />

Cox1<br />

•<br />

• •<br />

Cox2 • • • •<br />

Cyclophilin A •<br />

DAP12 •<br />

DC-SIGN<br />

Dectin-1<br />

E2A<br />

ERC1<br />

ERC1α<br />

ETO<br />

Evi-1<br />

Fgr<br />

FoxP3<br />

• •<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

Target M P E S C<br />

Fyn • •<br />

Galectin-1/LGALS1 • •<br />

Galectin-3/LGALS3 •<br />

GIMAP5 •<br />

GP130 •<br />

GRK6 •<br />

Helios •<br />

HPK1 •<br />

HS1 • •<br />

Phospho-HS1<br />

(Tyr397) • • •<br />

IFI16 •<br />

IFIT1 •<br />

IFITM1 •<br />

IFITM2 •<br />

IFN-α •<br />

IFN-γ • •<br />

IGBP1 •<br />

Ikaros • •<br />

IκBα • • • •<br />

Phospho-IκBα<br />

(Ser32) • • •<br />

Phospho-IκBα<br />

(Ser32/Ser36)<br />

IκBα (Aminoterminal<br />

Antigen) • •<br />

IκBα (Carboxyterminal<br />

Antigen)<br />

IκBβ<br />

Phospho-IκBε<br />

(Ser18/22)<br />

IκBζ<br />

IKKα<br />

Phospho-IKKα<br />

(Ser176)/IKKβ<br />

(Ser177)<br />

•<br />

• •<br />

•<br />

•<br />

• • • •<br />

•<br />

174 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

•<br />

Target M P E S C<br />

Phospho-IKKα/β<br />

(Ser176/180) • •<br />

IKKβ • • •<br />

•<br />

Phospho-IKKβ<br />

(Ser177/181)<br />

IKKγ<br />

Phospho-IKKγ<br />

(Ser376)<br />

IKKε<br />

Phospho-IKKε<br />

(Ser172)<br />

IL-1β<br />

IL-1RA<br />

IL-2<br />

IL-2Rα<br />

IL-2Rβ<br />

Mouse IL-3<br />

Neutralizing<br />

Human IL-4<br />

Neutralizing<br />

IL-4<br />

IL-6<br />

IL10<br />

IL-17A<br />

Human IL-17A<br />

Neutralizing<br />

IL17R<br />

IDO<br />

• •<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

IRAK1 • • • •<br />

•<br />

Phospho-IRAK1<br />

(Thr209)<br />

Phospho-IRAK1<br />

(Thr387)<br />

IRAK2<br />

IRAK4<br />

Phospho-IRAK4<br />

•<br />

•<br />

•<br />

(Thr345/Ser346) • •<br />

IRAK-M •<br />

IRF-1 • •<br />

IRF-2 •<br />

IRF-3 • •<br />

Phospho-IRF-3<br />

(Ser396) •<br />

IRF-4 • •<br />

IRF-5 • •<br />

IRF-6 •<br />

IRF-7 • • •<br />

•<br />

Phospho-IRF-7<br />

(Ser471/472)<br />

Phospho-IRF-7<br />

(Ser477)<br />

IRF-8<br />

Itk<br />

Jak1<br />

Phospho-Jak1<br />

(Tyr1022/1023)<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

Jak2 • • •<br />

•<br />

Phospho-Jak2<br />

(Tyr221)<br />

Phospho-Jak2<br />

(Tyr1007)<br />

Phospho-Jak2<br />

(Tyr1007/1008)<br />

•<br />

• •<br />

Target M P E S C<br />

Phospho-Jak2<br />

(Tyr1008) • •<br />

Jak3 • •<br />

Phospho-Jak3<br />

(Tyr980/Tyr981) •<br />

Langerin<br />

LAT<br />

•<br />

•<br />

•<br />

Phospho-LAT<br />

(Tyr171)<br />

Phospho-LAT<br />

(Tyr191)<br />

Lck<br />

Phospho-Lck<br />

(Tyr505)<br />

LGP2<br />

LITAF<br />

5-Lipoxygenase<br />

Phospho-5-Lipoxygenase<br />

(Ser271)<br />

Phospho-5-Lipoxygenase<br />

(Ser663)<br />

LRF/Pokemon<br />

Lsp1<br />

Lyn<br />

Phospho-Lyn<br />

(Tyr507)<br />

MALT1<br />

Mannose Receptor<br />

MAVS<br />

MCP-1 mouse<br />

MDA-5<br />

MEIS1/2<br />

• •<br />

• •<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

Miz-1 • •<br />

MNDA • •<br />

MyD88 • •<br />

Myeloperoxidase •<br />

NALP1 •<br />

NDP52 • •<br />

NFAT1 • •<br />

NFAT2 •<br />

NFAT3 •<br />

NFAT4 •<br />

NF-κB p65 • • • •<br />

•<br />

Phospho-NF-κB<br />

p65 (Ser468)<br />

Phospho-NF-κB<br />

p65 (Ser536) • • • •<br />

Acetyl-NF-κB p65<br />

(Lys310)<br />

Methyl-NF-κB p65<br />

(Lys310)<br />

NF-κB p105<br />

Phospho-NF-κB<br />

p105 (Ser932)<br />

NF-κB p105/p50<br />

• •<br />

NF-κB2 p100/p52 • •<br />

Phospho-NF-κB2<br />

p100/p52<br />

(Ser866/Ser870)<br />

NIK<br />

NLRC4<br />

NLRP3<br />

NLRX1<br />

Nod1<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

Target M P E S C<br />

NOS (pan) •<br />

iNOS • • •<br />

NTAL/LAB • •<br />

Phospho-p40phox<br />

(Thr154) •<br />

p47phox • •<br />

p67phox •<br />

Pbx1 •<br />

PD-L1 •<br />

PIAS1 •<br />

PIAS3 • • •<br />

PIAS4 •<br />

Pim-1 • •<br />

Pim-2 •<br />

Pim-3 •<br />

Pirin •<br />

Prolactin Receptor •<br />

PTPN22 •<br />

PU.1 • • •<br />

RAG1 •<br />

RAGE •<br />

RAGE 1 • •<br />

RANK •<br />

RANK Ligand •<br />

RANTES •<br />

c-Rel • • •<br />

RelB • •<br />

Phospho-RelB<br />

(Ser552) • • •<br />

Rig-I • •<br />

RIP • •<br />

RIP2 • •<br />

Phospho-RIP2<br />

(Ser176) • •<br />

RIP3 • •<br />

RIP4 •<br />

SAMHD1 •<br />

SARM1 •<br />

SDF1 • •<br />

SH2D1A • •<br />

SHIP1 • •<br />

Phospho-SHIP1<br />

(Tyr1020) •<br />

SHIP2 • •<br />

•<br />

Phospho-SHIP2<br />

(Tyr986/Tyr987)<br />

Phospho-SHIP2<br />

(Tyr1135)<br />

SHP-1<br />

Phospho-SHP-1<br />

(Tyr564)<br />

SINTBAD<br />

SLP76<br />

Phospho-SLP76<br />

(Ser376)<br />

SOCS1<br />

SOCS2<br />

SOCS3<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

Stat1 • • •<br />

Phospho-Stat1<br />

(Tyr701) • • •<br />

285<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for Phospho-Stat3<br />

(Tyr705) have been cited over 285 times<br />

in high-impact, peer-reviewed publications<br />

from the global research community.<br />

Select Citations:<br />

Mauer, J. et al. (2014) Signaling by<br />

IL-6 promotes alternative activation<br />

of macrophages to limit endotoxemia<br />

and obesity-associated resistance to<br />

insulin. Nat. Immunol. 15, 423−430.<br />

Kothari, P. et al. (2014) IL-<br />

6-mediated induction of matrix<br />

metalloproteinase-9 is modulated<br />

by JAK-dependent IL-10 expression<br />

in macrophages. J. Immunol. 192,<br />

349−357.<br />

Zhou, C. et al. (2014) PTTG acts as<br />

a STAT3 target gene for colorectal<br />

cancer cell growth and motility.<br />

Oncogene 33, 851−861.<br />

Kryczek, I. et al. (2014) IL-22(+)<br />

CD4(+) T cells promote colorectal<br />

cancer stemness via STAT3 transcription<br />

factor activation and induction<br />

of the methyltransferase DOT1L.<br />

Immunity 40, 772−784.<br />

Mayeur, C. et al. (2014) The type I<br />

BMP receptor Alk3 is required for the<br />

induction of hepatic hepcidin gene<br />

expression by interleukin-6. Blood<br />

123, 2261−2268.<br />

Becker, T.M. et al. (2014) Mutant B-<br />

RAF-Mcl-1 survival signaling depends<br />

on the STAT3 transcription factor.<br />

Oncogene 33, 1158−1166.<br />

Xiang, M. et al. (2014) STAT3 induction<br />

of miR-146b forms a feedback<br />

loop to inhibit the NF-kappaB to IL-6<br />

signaling axis and STAT3-driven cancer<br />

phenotypes. Sci. Signal. 7, ra11.<br />

Wang, Y. et al. (2014) GdX/UBL4A<br />

specifically stabilizes the TC45/<br />

STAT3 association and promotes<br />

dephosphorylation of STAT3 to<br />

repress tumorigenesis. Mol. Cell 53,<br />

752−765.<br />

Schmidt, J.W. et al. (2014) Stat5<br />

regulates the phosphatidylinositol<br />

3-kinase/Akt1 pathway during<br />

mammary gland development and<br />

tumorigenesis. Mol. Cell Biol. 34,<br />

1363−1377.<br />

Nagashima, H. et al. (2014) The<br />

adaptor TRAF5 limits the differentiation<br />

of inflammatory CD4(+) T cells<br />

by antagonizing signaling via the<br />

receptor for IL-6. Nat. Immunol. 15,<br />

449−456.<br />

www.cellsignal.com/cstimmunology 175


Section I: Research Areas<br />

chapter 07: immunology and inflammation<br />

Jak and Cytokine<br />

Receptor Mutants<br />

This table lists Jak and cytokine receptor<br />

mutations found in various cancers,<br />

along with corresponding publications.<br />

Target M P E S C<br />

Phospho-Stat1<br />

(Ser727) • •<br />

Stat2 •<br />

Phospho-Stat2<br />

(Tyr690) •<br />

Stat3<br />

• • • • •<br />

Phospho-Stat3<br />

(Tyr705) • • • •<br />

Phospho-Stat3<br />

(Ser727)<br />

Acetyl-Stat3<br />

(Lys685)<br />

Stat3α<br />

Stat4<br />

• • •<br />

•<br />

• •<br />

•<br />

Phospho-Stat4<br />

(Tyr693) • • •<br />

Stat5 • • • •<br />

Phospho-Stat5<br />

(Tyr694) • • • •<br />

Stat5a<br />

Stat6<br />

•<br />

• • • •<br />

Phospho-Stat6<br />

(Tyr641) • • • •<br />

STING<br />

Syk<br />

•<br />

• •<br />

•<br />

Phospho-Syk<br />

(panTyr)<br />

Target M P E S C<br />

Phospho-Syk<br />

(Tyr323) •<br />

Phospho-Syk<br />

(Tyr525/526) • • • •<br />

TAL1 •<br />

TAP1 •<br />

TAP2 •<br />

T-bet/TBX21<br />

(V365) • •<br />

TBK1/NAK • •<br />

Phospho-TBK1/<br />

NAK (Ser172) • •<br />

Tec •<br />

THEMIS •<br />

ThPOK •<br />

TIRAP •<br />

Toll-like Receptor 1 •<br />

Toll-like Receptor 2 • •<br />

Toll-like Receptor 3 •<br />

Toll-like Receptor 4 •<br />

Toll-like Receptor 6 •<br />

Toll-like Receptor 7 • •<br />

Toll-like Receptor 8 •<br />

Toll-like Receptor 9 • •<br />

•<br />

•<br />

Human TNF-α<br />

Neutralizing<br />

Target M P E S C<br />

Mouse TNF-α<br />

Neutralizing •<br />

TNF-α • • • •<br />

TNF-R1 • •<br />

Tollip •<br />

Phospho-TPOR<br />

(Tyr626) •<br />

TREX1 •<br />

TRIF •<br />

TWEAK •<br />

TWEAK Receptor/<br />

Fn14 •<br />

Tyk2 • • •<br />

Phospho-Tyk2<br />

(Tyr1054/1055) •<br />

VCAM1 •<br />

Yes •<br />

ZAP70 • • •<br />

•<br />

Phospho-ZAP70<br />

(Tyr319)<br />

Phospho-Zap-70<br />

(Tyr319)/Syk<br />

(Tyr352)<br />

Phospho-ZAP70<br />

(Tyr493)<br />

• • •<br />

•<br />

Jak Mutants Cytokine Receptor Disease References<br />

Jak2 V617F EpoR, TpoR (MPL), G-CSFR Myeloproliferative neoplasms 1–5<br />

(MNPs), PV, ET, PMF<br />

Jak2 K539L, exon 12 mutants EpoR MNP: PV 6<br />

Jak2 T875N Undetermined AML (AMKL) 7<br />

Jak3 A572V Undetermined AML (AMKL) (cell lines) 8<br />

Jak1 V658F, Jak1 A634D, R879H, R724S IL2R, IL9R, other undetermined T-ALL 9,10<br />

Jak1 R683G/S Jak2 DIREED TLSPR Pediatric and Down syndrome ALL 11–15<br />

Jak2 V617I, Jak2 R564Q, Jak2 S755R/ R938Q TpoR (MPL) Hereditary thrombocytosis 16–18<br />

Receptor Mutants Cytokine Receptor Disease References<br />

TpoR W515L/K/A Jak2 MPNs: ET, PMF 19–21<br />

TpoR S505N 22<br />

TpoR S487A 23<br />

TLSPR F232S / TLSPR translocations Jak2 R683 mutants Pediatric and Down syndrome ALL 13, 24–26<br />

References: (1) James, C. et al. (2005) Nature 434, 1144–1148. • (2) Baxter, E.J. et al. (2005) Lancet 365, 1054–1061. •<br />

(3) Kralovics, R. et al. (2005) N. Engl. J. Med. 352, 1779–1790. • (4) Levine, R.L. et al. (2005) Cancer Cell 7, 387–397. • (5)<br />

Vainchenker, W. et al. (2008) Semin. Cell Dev. Biol. 19, 385–393. • (6) Scott, L.M. et al. (2007) N. Engl. J. Med. 356, 459–468.<br />

• (7) Mercher, T. et al. (2006) Blood 108, 2770–2779. • (8) Walters, D.K. et al. (2006) Cancer Cell 10, 65–75. • (9) Flex, E. et<br />

al. (2008) J. Exp. Med. 205, 751–758. • (10) Jeong, E.G. et al. (2008) Clin. Cancer Res. 14, 3716–3721. • (11) Malinge, S.<br />

et al. (2007) Blood 109, 2202–2204. • (12) Mullighan, C.G. et al. (2009) Proc. Natl. Acad. Sci. USA 106, 9414–9418. • (13)<br />

Mullighan, C.G. et al. (2009) Nat. Genet. 41, 1243–1246. • (14) Bercovich, D., et al. (2008) Lancet 372, 1484–1492. • (15)<br />

Kearney, L. et al. (2009) Blood 113, 646–648. • (16) Mead, A.J. et al. (2013) Blood 121, 4156–4165. • (17) Etheridge, S.L.<br />

et al. (2014) Blood 123, 1059–1068. • (18) Marty, C. et al. (2014) Blood 123, 1372–1383. • (19) Pikman, Y. et al. (2006)<br />

PLoS Med. 3, e270. • (20) Pardanani, A.D. et al. (2006) Blood 108, 3472–3476. • (21) Pecquet, C. et al. (2010) Blood 115,<br />

1037–1048. • (22) Ding, J. et al. (2004) Blood 103, 4198–4200. • (23) Malinge, S. et al. (2008) Blood 112, 4220–4226.<br />

• (24) Russell, L.J. et al. (2009) Blood 114, 2688–2698. • (25) Chapiro, E. et al. (2010) Leukemia 24, 642–645. • (26)<br />

Hertzberg, L. et al. (2010) Blood 115, 1006–1017.<br />

Ligand Receptor Jak-Kinase Other Tyrosine Kinases Stat Family Members<br />

IL-6 IL-6Rα+gp130 Jak1,2, Tyk2 Hck Stat1, Stat3<br />

IL-11 IL-11R+gp130 Jak1,2, Tyk2 Src, Yes Stat3<br />

CNTF, CT-1, LIF, OSM CNTFR+gp130, CT-1R+gp130,<br />

LIFR+gp130, OSMR+gp130<br />

Jak1,2, Tyk2 Src family Predominant: Stat3<br />

Secondary: Stat1,5<br />

G-CSF G-CSFR Jak2, Tyk2 Lyn Stat3<br />

IL-12 (p40+p35) IL-12Rβ1+IL-12Rβ2 Jak2, Tyk2 Lck Stat4<br />

Leptin LeptinR Jak2 Not determined Stat3,5,6<br />

IL-3 IL-3Rα+βc Jak2 Fyn, Hck, Lyn Stat3,5,6<br />

IL-5 IL-5R+βc Jak2 Btk Stat3,5,6<br />

GM-CSF GM-CSFR+βc Jak2 Hck, Lyn Stat3,5<br />

Angiotensin GPCR Jak2, Tyk2 Stat1,2,3<br />

Serotonin GPCR Jak2 Stat3<br />

α-Thrombin GPCR Jak2 Stat1,3<br />

Chemokines CXCR4 Jak2,3<br />

IL-2 IL-2Rα+IL-2Rb+γc Jak1,2,3 Fyn, Hck, Lck, Syk, Tec Stat3,5<br />

IL-4 IL-4Rα+γcR or IL-4Rα+IL-13Rα1 Jak1,3 Lck, Tec Stat6<br />

IL-7 IL-7R+γc Jak1,3 Lyn Stat3,5<br />

IL-9 IL-9R+γc Jak1,3 Not determined Stat1,3,5<br />

IL-13 IL-13Rα1+ IL-4Rα Jak1,2, Tyk2 Ctk Stat6<br />

IL-15 IL-15Rα+IL-2Rβ+γc Jak1,3 Lck Stat3,5<br />

IL-19 IL-20Rα+IL-20Rβ Jak1, Stat3<br />

IL-20 IL-20Rα+IL-20Rβ, IL-22R+IL-20Rβ Jak1, Stat3<br />

IL-21 IL-21R+γc Jak1,3 Stat1,3,5<br />

IL-22 IL-22R+IL-10Rβ Jak1, Tyk2 Stat1,3,5<br />

IL-23 (p40+p19) IL-12Rβ1+IL-23R Jak2 Tyk2 Stat4<br />

IL-24 same as IL-20 Jak1, Stat3<br />

IL-26 IL-20Rα+IL-10Rβ Jak1, Tyk2 Stat3<br />

IL-27 (EBI3+p28) gp130+WSX1 Jak1,2, Tyk2 Stat1,2,3,4,5<br />

IL-28A, IL-28B, IL-29 IL-28R+IL-10Rβ Jak1, Tyk2 Stat1,2,3,4,5<br />

IL-31 IL-31Rα+OSMR Jak1,2, Tyk2 Stat1,3,5<br />

IL-35 (p35+EBI3) gp130+WSX1 Jak1,2, Tyk2 Stat1,3,5<br />

GH GHR Jak2 Src family Stat3,5 (mainly Stat5a)<br />

Tpo TpoR (c-Mpl) Jak2, Tyk2 Lyn Stat1,3,5<br />

Epo, Pro EpoR, ProlactinR Jak2 Src Family Stat5 (mainly Stat5a)<br />

Interferon (IFNα/β) IFNAR1+IFNAR2 Jak1, Tyk2 Lck Predominant: Stat1,2<br />

Secondary: Stat3,4,5<br />

IFN-γ IFN-gR1+IFN-γR2 Jak1, Jak2 Hck, Lyn Stat1<br />

IL-10 IL-10Rα+ IL-10Rβ Jak1, Tyk2 Not determined Stat1,3,5<br />

TLSP TLSPR and IL-7R Jak1, possibly<br />

Jak2<br />

Not determined Stat3,5<br />

EGF EGFR Jak1 EGFR, Src Stat1,3,5<br />

PDGF PDGFR Jak1,2 PDGFR, Src Stat1,3,5<br />

© 2002–2014 Cell Signaling Technology, Inc.<br />

We would like to thank Prof. Stefan Constantinescu, Ludwig Institute for Cancer Research, Brussels, Belgium for contributing to this table.<br />

Jak/Stat<br />

Utilization<br />

This table lists the combinatorial use<br />

of tyrosine kinases and Stat proteins<br />

in cytokine/growth factor signaling.<br />

Protein Kinases: Introduction<br />

Please visit our website to learn more about Protein Kinases. www.cellsignal.com/cstkinases<br />

176 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/csttables<br />

177


CRAC Channel<br />

Orai1<br />

Section I: Research Areas<br />

chapter 07: immunology and inflammation<br />

B Cell Receptor Signaling<br />

BCR<br />

Internalization<br />

p38<br />

JNK<br />

BCR<br />

Ag<br />

Gab<br />

Shc<br />

GRB2<br />

Lyn CD22 SHP-1<br />

SHP-2<br />

p85 p110<br />

PI3K PIP 3<br />

FcγRIIB1<br />

Lipid<br />

Lyn<br />

Raft Ezrin CD19<br />

Aggregation<br />

Syk<br />

SHIP<br />

SHP-1<br />

PTEN<br />

CD45<br />

PIR-B<br />

RhoA<br />

Dok-3<br />

clathrin<br />

SHP-2<br />

PI(4,5)P 2<br />

Btk<br />

Ca 2+<br />

Bam32 Cbp/PAG<br />

Cbl<br />

IP3R<br />

Csk<br />

Bam32<br />

STIM1<br />

PLCγ2<br />

Intracellular<br />

IP Ca 2+ 3<br />

Store<br />

Akt<br />

Ca<br />

LAB<br />

2+<br />

CIN85<br />

GRB2<br />

DAG<br />

Vav<br />

SOS<br />

Rac/<br />

PKC<br />

cdc42 CD19 HS1<br />

Ras<br />

PIP 3<br />

GRP<br />

Pyk2<br />

DAG Ras<br />

CARMA1<br />

Ras TAK1<br />

Rac<br />

GAP<br />

Bcl10<br />

RapL<br />

MALT1<br />

c-Raf<br />

Rap<br />

CaM GSK-3 mTOR<br />

Riam<br />

Dok-1<br />

MEKKs<br />

FcγRIIB1<br />

IKK<br />

DAG<br />

Cofilin<br />

CD19 MEK1/2<br />

Calcineurin<br />

MKK3/4/6 MKK4/7<br />

p70<br />

IκB<br />

S6K<br />

PKC Ca 2+ Erk1/2<br />

NF-κB<br />

CD40<br />

IκB<br />

p38<br />

NFAT Protein<br />

JNK1/2<br />

Proteasomal<br />

Synthesis<br />

Degradation<br />

Cytoskeletal<br />

Rearrangements and<br />

Integrin Activation<br />

Cytoplasm<br />

Nucleus<br />

Transmembrane Protein<br />

mIg<br />

α/β α/β<br />

BLNK<br />

GRB2<br />

LAB<br />

Erk1/2<br />

mIg<br />

BCAP<br />

NF-κB<br />

CD19<br />

CaMK<br />

Ca 2+<br />

Glucose<br />

Uptake<br />

Glycolysis<br />

ATP<br />

Generation<br />

Akt<br />

T Cell Receptor Signaling<br />

Ca 2+ Ca 2+ Ca 2+ Clustering<br />

CRAC<br />

Channel<br />

Calcineurin<br />

CaM<br />

NFAT<br />

CaMKIV<br />

CREB<br />

NFAT<br />

LFA-1<br />

Actinin Talin<br />

Calpain<br />

F-Actin<br />

IP 3<br />

IP3R<br />

Intracellular<br />

Ca 2+ Store<br />

SOS<br />

PIP 2<br />

DAG<br />

RasGRP<br />

Ras<br />

Raf<br />

MEK1/2<br />

Erk1/2<br />

GRB2<br />

LAT<br />

GADS<br />

PLCγ1<br />

NCK<br />

Itk<br />

SLP-76<br />

WASP<br />

Rac/cdc42<br />

MEKK1<br />

MKK4/7<br />

JNK<br />

ADAP<br />

Vav<br />

TAK1<br />

HPK1<br />

GADD45α<br />

ζ ζ<br />

Zap-70<br />

Dlgh1<br />

p38 MAPK<br />

α β<br />

c-Cbl<br />

DAG<br />

TAK1<br />

MKK7<br />

δ ε ε γ<br />

JNK2<br />

TCR/CD3<br />

Complex<br />

PDK1<br />

PKCθ<br />

MALT1Bcl10<br />

Carma1<br />

IKKβ<br />

IKKγ<br />

CD4<br />

Lck<br />

IKKα<br />

Rel<br />

IκB<br />

NF-κB<br />

IκB<br />

CD45<br />

GβL<br />

Proteasomal<br />

Degradation<br />

CD28<br />

PI3K<br />

PIP 3<br />

Akt<br />

mTORC1<br />

Raptor<br />

mTOR<br />

DEPTOR<br />

Cell Proliferation<br />

Survival<br />

MEF2C<br />

CREB<br />

ATF-2 Jun Bcl-6 Egr-1 Elk-1 Bcl-xL Bfl-1 Oct-2 Ets-1 NFAT<br />

Transcription<br />

FoxO<br />

Transcription<br />

Growth Arrest,<br />

Apoptosis<br />

Cytoplasm<br />

Nucleus<br />

IL-2<br />

Gene<br />

NFAT Fos<br />

Jun NF-κB Rel<br />

Nuclear<br />

Membrane<br />

The B cell antigen receptor (BCR) is composed of membrane immunoglobulin (mIg) molecules and associated Igα/Igβ (CD79a/CD79b) heterodimers (α/β). The mIg subunits<br />

bind antigen, resulting in receptor aggregation, while the α/β subunits transduce signals to the cell interior. BCR aggregation rapidly activates the Src family kinases Lyn,<br />

Blk, and Fyn as well as the Syk and Btk tyrosine kinases. This initiates the formation of a ‘signalosome’ composed of the BCR, the aforementioned tyrosine kinases, adaptor<br />

proteins such as CD19 and BLNK, and signaling enzymes such as PLCγ2, PI3K, and Vav. Signals emanating from the signalosome activate multiple signaling cascades that<br />

involve kinases, GTPases, and transcription factors. This results in changes in cell metabolism, gene expression, and cytoskeletal organization. The complexity of BCR signaling<br />

permits many distinct outcomes, including survival, tolerance (anergy) or apoptosis, proliferation, and differentiation into antibody-producing cells or memory B cells. The<br />

outcome of the response is determined by the maturation state of the cell, the nature of the antigen, the magnitude and duration of BCR signaling, and signals from other<br />

receptors such as CD40, the IL-21 receptor, and BAFF-R. Many other transmembrane proteins, some of which are receptors, modulate specific elements of BCR signaling.<br />

A few of these, including CD45, CD19, CD22, PIR-B, and FcγRIIB1 (CD32), are indicated here in yellow. The magnitude and duration of BCR signaling are limited by negative<br />

feedback loops including those involving the Lyn/CD22/SHP-1 pathway, the Cbp/Csk pathway, SHIP, Cbl, Dok-1, Dok-3, FcγRIIB1, PIR-B, and internalization of the BCR. In<br />

vivo, B cells are often activated by antigen-presenting cells that capture antigens and display them on their cell surface. Activation of B cells by such membrane-associated<br />

antigens requires BCR-induced cytoskeletal reorganization. Please refer to the diagrams for the PI3K/Akt signaling pathway, the NF-κB signaling pathway, and the regulation<br />

of actin dynamics for more details about these pathways.<br />

Select Reviews:<br />

Dal Porto, J.M., Gauld, S.B., Merrell, K.T., et al. (2004) Mol. Immunol. 41, 599–613. • Goodnow, C.C., Vinuesa, C.G., Randall, K.L., et al. (2010) Nat. Immunol. 11,<br />

681–688. • Harwood, N.E. and Batista, F.D. (2010) Annu. Rev. Immunol. 28, 185–210. • Harwood, N.E. and Batista, F.D. (2008) Immunity 28, 609–619. • Kurosaki, T.,<br />

Shinohara, H., and Baba, Y. (2010) Annu. Rev. Immunol. 28, 21–55. • Szydłowski, M., Jabłońska, E., and Juszczyński, P. (2014) Int. Rev. Immunol. 33, 146–157.<br />

Tumor cells employ multiple defense strategies to evade detection by the immune system. One common strategy, upregulation of immune checkpoint proteins and ligands, takes<br />

advantage of a natural immune mechanism for self-tolerance and prevention of collateral tissue damage. Immune checkpoint proteins, such as PD-1, CTLA-4, and many others,<br />

are located on T cells and engage with their corresponding ligand on tumor cells or dendritic cells, sending inhibitory signals that repress T cell activation or response. One of<br />

the first discovered checkpoint proteins, CTLA-4, plays a role at the stage of T cell priming by binding to the CD28 ligands CD80 or CD86 to prevent co-stimulatory signals<br />

necessary for T cell activation. In contrast, the PD-1/PD-L1 checkpoint acts later in the process, inhibiting anti-tumor immune responses by effector T cells such as CD4 + T<br />

helper 1 (Th1) cells and CD8 + cytotoxic T lymphocytes (CTLs), leading to decreases in IFNγ production and cytolytic activity. Upregulation of PD-L1 expression on the tumor cell<br />

surface is mediated by IFNγR signaling to Stat1, as well as oncogenic signaling through several receptor tyrosine kinases (EGFR, ALK, ROS, HER2, and others) to activate the<br />

MAPK, Akt, and Stat3 pathways.<br />

Cells in the tumor microenvironment can also influence tumor progression. FoxP3 + /CD4 + T regulatory cells (T Regs ) and myeloid-derived suppressor cells (MSCs) secrete immunosuppressive<br />

cytokines IL-10 and TGF-β to inhibit the activity of Th1 cells and CTLs. Natural killer (NK) cells release cytotoxic granules against the tumor cell and secrete<br />

IFNγ, which stimulates surrounding pro-inflammatory M1 macrophages. Pro-tumorigenic M2 macrophages suppress anti-tumor immune responses via production of IL-10<br />

and TGF-β and promote metastasis through release of MMPs. MMPs and TGF-β are also released by surrounding mast cells.<br />

Select Reviews:<br />

Burbach, B.J., Medeiros, R.B., Mueller, K.L., and Shimizu, Y. (2007) Immunol. Rev. 218, 65–81. • Chen, L. and Flies, D.B. (2013) Nat. Rev. Immunol. 13, 227–242. •<br />

Cheng, J., Montecalvo, A., and Kane, L.P. (2011) Immunol. Res. 50, 113–117. • Cronin, S.J. and Penninger, J.M. (2007) Immunol. Rev. 220, 151–168. • Fracchia, K.M.,<br />

Pai, C.Y., and Walsh, C.M. (2013) Front Immunol. 4, 324. • Marsland, B.J. and Kopf, M. (2008) Trends Immunol. 29, 179–185. • Thome, M. (2008) Nat. Rev. Immunol. 8,<br />

495–500.<br />

© 2002–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Michael R. Gold, University of British Columbia, Vancouver, British Columbia, Canada for reviewing this diagram.<br />

178 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

© 2004–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Sankar Ghosh, Columbia University, New York, NY for reviewing this diagram.<br />

www.cellsignal.com/cstpathways 179


Section I: Research Areas<br />

chapter 07: immunology and inflammation<br />

Toll-like Receptor Signaling<br />

UNC93B1<br />

TLR9<br />

TLR9<br />

MyD88<br />

TLR7<br />

LPS<br />

dsRNA or<br />

5'-triphosphate RNA<br />

CpG<br />

ATP<br />

MAVS<br />

Anti-viral<br />

Compounds,<br />

ssRNA<br />

MyD88<br />

CD14<br />

TLR8<br />

MD-2<br />

TLR4<br />

TLR4<br />

TRAM TRIF TIRAP<br />

RIG-I<br />

MyD88<br />

IRF-7<br />

IRF-7<br />

SARM1<br />

Endosome<br />

UNC93B1 UNC93B1<br />

dsRNA<br />

TLR3<br />

TLR3<br />

MyD88<br />

TRAF3<br />

TRIF<br />

SARM1<br />

IRF-3<br />

IRF-3<br />

Uropathogenic<br />

Bacteria,<br />

Profilin<br />

ST2L<br />

TLR11<br />

TLR11<br />

MyD88<br />

TRIAD3A<br />

SOCS1<br />

IKKε<br />

TBK1<br />

TOLLIP<br />

RIP1<br />

Proteasomal<br />

Degradation<br />

Flagellin<br />

TLR5<br />

TLR5<br />

MyD88<br />

IRAK-4<br />

IRAK-1<br />

TRAF6<br />

Ubc13<br />

UEV1A<br />

IKKγ/<br />

NEMO<br />

IKKβ IKKα<br />

MyD88<br />

IRAK-M<br />

IκBα ub<br />

p65/RelA<br />

NF-κB<br />

TIRAP<br />

IRAK-2<br />

A20<br />

ECSIT<br />

Triacyl<br />

Lipopeptide<br />

TLR2<br />

TLR1<br />

TAK1<br />

TAB1/2<br />

MKK 4/7<br />

JNK<br />

MyD88<br />

FADD<br />

MEKK-1<br />

TIRAP<br />

MKK 3/6<br />

p38 MAPK<br />

Diacyl<br />

Lipopeptide<br />

TLR2<br />

TLR6<br />

Casp-8<br />

Apoptosis<br />

Jak/Stat Signaling: IL-6 Receptor Family<br />

p120<br />

ras-GAP<br />

E-Ras<br />

PI3K<br />

Akt<br />

Tumor-like<br />

Properties<br />

of ES Cells<br />

Cytoplasm<br />

Nucleus<br />

Erk<br />

Raf<br />

MEK<br />

Erk<br />

C/EBPβ<br />

NF-κB<br />

AP-1<br />

etc.<br />

Erk<br />

Ras<br />

Erk<br />

Shc<br />

GRB2<br />

TF<br />

TF<br />

Accessory<br />

TF Motif<br />

PI3K<br />

Akt<br />

mTOR<br />

SHP-2<br />

SOCS3<br />

Jak<br />

gp130<br />

Stat3<br />

Stat3<br />

Stat3<br />

Jak<br />

Stat3<br />

ISRE/GAS<br />

Mcl-1<br />

IL-6<br />

Apoptosis<br />

PIAS<br />

SUMO<br />

PTP<br />

SHP-1<br />

Stat<br />

SOCS<br />

Renewal<br />

of ES Cells<br />

Crosstalk<br />

Tumor Cells<br />

EGF<br />

EGFR<br />

Feedback<br />

Inhibition<br />

Stat<br />

Src<br />

CIS, SOCS, Mcl-1, APPs, TIMP-1,<br />

Pim-1, c-Myc, cytokines, TFs, etc.<br />

Cytoplasm<br />

Nucleus<br />

Inflammation, Immune Regulation, Survival, Proliferation<br />

Transcription<br />

Factors<br />

Toll-like receptors (TLRs) recognize distinct pathogen-associated molecular patterns and play a critical role in innate immune responses. They participate in the first line of<br />

defense against invading pathogens and play a significant role in inflammation, immune cell regulation, survival, and proliferation. To date, 11 members of the TLR family<br />

have been identified, of which TLR1, TLR2, TLR4, TLR5, TLR6, and TLR11 are located on the cell surface and TLR3, TLR7, TLR8, and TLR9 are localized to the endosomal/<br />

lysosomal compartment. The activation of the TLR signaling pathway originates from the cytoplasmic Toll/IL-1 receptor (TIR) domain that associates with a TIR domain-containing<br />

adaptor, MyD88. Upon stimulation with ligands, MyD88 recruits IL-1 receptor-associated kinase-4 (IRAK-4) to TLRs through interaction of the death domains of both<br />

molecules. IRAK-1 is activated by phosphorylation and associates with TRAF6, thereby activating the IKK complex and leading to activation of MAP kinases (JNK, p38 MAPK)<br />

and NF-κB. Tollip and IRAK-M interact with IRAK-1 and negatively regulate the TLR-mediated signaling pathways. Additional modes of regulation for these pathways include<br />

TRIF-dependent induction of TRAF6 signaling by RIP1 and negative regulation of TIRAP-mediated downstream signaling by ST2L, TRIAD3A, and SOCS1. Activation of MyD88-<br />

independent pathways occurs via TRIF and TRAF3, leading to recruitment of IKKε/TBK1, phosphorylation of IRF3, and expression of interferon-β. TIR domain containing<br />

adaptors such as TIRAP, TRIF, and TRAM regulate TLR-mediated signaling pathways by providing specificity for individual TLR signaling cascades. TRAF3 plays a critical role in<br />

the regulation of both MyD88-dependent and TRIF-dependent signaling via TRAF3 degradation, which activates MyD88-dependent signaling and suppresses TRIF-dependent<br />

signaling (and vice versa).<br />

Select Reviews:<br />

Barton, G.M. and Kagan, J.C. (2009) Nat. Rev. Immunol. 9, 535–542. • Blasius, A.L. and Beutler, B. (2010) Immunity 32, 305–315. • Kawai, T., and Akira, S. (2010) Nat.<br />

Immunol. 11, 373–384. • Lester, S.N. and Li, K. (2014) J. Mol. Biol. 426, 1246–1264. • Li, X., Jiang, S., and Tapping, R.I. (2010) Cytokine 49, 1–9. • McGettrick, A.F.<br />

and O’Neill, L.A. (2010) Curr. Opin. Immunol. 22, 20–27. • Miggin, S.M. and O’Neill, L.A. (2006) J. Leukoc. Biol. 80, 220–226. • Pasare, C. and Medzhitov, R. (2005)<br />

Adv. Exp. Med. Biol. 560, 11–18. • Reuven, E.M., Fink, A., and Shai, Y. (2014) Biochim. Biophys. Acta. 1838, 1586–1593.<br />

Jaks and Stats are critical components of many cytokine receptor systems; regulating growth, survival, differentiation, and pathogen resistance. An example of these pathways<br />

is shown for the IL-6 (or gp130) family of receptors, which coregulate B cell differentiation, plasmacytogenesis, and the acute phase reaction. Cytokine binding induces<br />

receptor dimerization, activating the associated Jaks, which phosphorylate themselves and the receptor. The phosphorylated sites on the receptor and Jaks serve as docking<br />

sites for the SH2-containing Stats, such as Stat3, and for SH2-containing proteins and adaptors that link the receptor to MAP kinase, PI3K/Akt, and other cellular pathways.<br />

Phosphorylated Stats dimerize and translocate into the nucleus to regulate target gene transcription. Members of the suppressor of cytokine signaling (SOCS) family dampen<br />

receptor signaling via homologous or heterologous feedback regulation. Jaks or Stats can also participate in signaling through other receptor classes, as outlined in the Jak/<br />

Stat Utilization Table. Researchers have found Stat3 and Stat5 to be constitutively activated by tyrosine kinases other than Jaks in several solid tumors.<br />

The Jak/Stat pathway mediates the effects of cytokines, like erythropoietin, thrombopoietin, and G-CSF, which are protein drugs for the treatment of anemia, thrombocytopenia,<br />

and neutropenia, respectively. The pathway also mediates signaling by interferons, which are used as antiviral and antiproliferative agents. Researchers have found that<br />

dysregulated cytokine signaling contributes to cancer. Aberrant IL-6 signaling contributes to the pathogenesis of autoimmune diseases, inflammation, and cancers such as<br />

prostate cancer and multiple myeloma. Jak inhibitors currently are being tested in models of multiple myeloma. Stat3 can act as an oncogene and is constitutively active in<br />

many tumors. Crosstalk between cytokine signaling and EGFR family members is seen in some cancer cells. Research has shown that in glioblastoma cells overexpressing<br />

EGFR, resistance to EGFR kinase inhibitors is induced by Jak2 binding to EGFR via the FERM domain of the former [Sci. Signal. (2013) 6, ra55].<br />

Activating Jak mutations are major molecular events in human hematological malignancies. Researchers have found a unique somatic mutation in the Jak2 pseudokinase<br />

domain (V617F) that commonly occurs in polycythemia vera, essential thrombocythemia, and idiopathic myelofibrosis. This mutation results in the pathologic activation Jak2,<br />

associated with receptors for erythropoietin, thrombopoietin, and G-CSF, which control erythroid, megakaryocytic, and granulocytic proliferation and differentiation. Researchers<br />

have also shown that somatic acquired gain-of-function mutations of Jak1 are found in adult T cell acute lymphoblastic leukemia. Somatic activating mutations in Jak1,<br />

Jak2, and Jak3 have also been identified in pediatric acute lymphoblastic leukemia (ALL). Furthermore, Jak2 mutations have been detected around pseudokinase domain<br />

R683 (R683G or DIREED) in Down syndrome childhood B-ALL and pediatric B-ALL.<br />

Select Reviews:<br />

Beekman, R. and Touw, I.P. (2010) Blood 115, 5131–5136. • Neurath, M.F. and Finotto, S. (2011) Cytokine Growth Factor Rev. 22, 83–89. • Sansone, P. and Bromberg,<br />

J. (2012) J. Clin. Oncol. 30, 1005–1014. • Vainchenker, W. and Constantinescu, S.N. (2013) Oncogene 32, 2601–2613. • Yu, H., Pardoll, D., and Jove, R. (2009) Nat.<br />

Rev. Cancer 9, 798–809.<br />

© 2009–2015 Cell Signaling Technology, Inc. • We would like to thank Dr. Pranoti Mandrekar, University of Massachusetts Medical School, Worcester, MA, for reviewing this diagram.<br />

180 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

© 2002–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Stefan Constantinescu, Ludwig Institute for Cancer Research, Brussels, Belgium for reviewing this diagram.<br />

www.cellsignal.com/cstpathways 181


Section I: Research Areas<br />

chapter 07: immunology and inflammation<br />

NF-κB Signaling<br />

Stress: ROIs,<br />

UV, metals,<br />

ischemia, shear<br />

UV<br />

Ag<br />

BCR<br />

JNK<br />

p38<br />

ub K63-ubiquitin<br />

ub K48-ubiquitin<br />

Cytoplasm<br />

Nucleus<br />

Ag-MHC<br />

TCR<br />

For detailed signaling,<br />

see BCR Pathway.<br />

CK2<br />

RelA/cRel IκBα/ε<br />

NF-κB1<br />

p50<br />

LPS, CpG,<br />

ssRNA, dsRNA<br />

TLRs<br />

IκBζ<br />

Pellino<br />

ub<br />

RelA/cRel IκBα/β/ε<br />

NF-κB1<br />

p50<br />

ub<br />

Bcl-3<br />

NF-κB<br />

p50/52<br />

NF-κB<br />

p50/52<br />

ub<br />

NAP1 NAK<br />

RSK1<br />

CYLD<br />

p65/<br />

RelA<br />

NF-κB<br />

p50/52<br />

ac<br />

IL-1<br />

IL-1R<br />

MyD88<br />

ub IRAK1/4<br />

TRAF2/6<br />

c-IAP1/2<br />

For detailed signaling,<br />

see TLR Pathway.<br />

Ubc13<br />

TRAF6<br />

A20<br />

For detailed signaling, UEV1A<br />

ITCH TAX1BP1<br />

ub<br />

see TCR Pathway.<br />

ub<br />

RNF11<br />

TRAF6<br />

TAB1/2<br />

TAK1<br />

LUBAC<br />

ELKS<br />

HOIL1 HOIP<br />

IKKβ IKKα<br />

SHARPIN<br />

IKKγ/<br />

NEMO<br />

ub<br />

ub<br />

ub<br />

β-TrCP<br />

Nuclear-cytoplasmic<br />

shuttling of nonphosphorylated<br />

forms<br />

Proteasomal<br />

Degradation<br />

IKKα/β/ε<br />

PKCζ<br />

PKA C<br />

p65/<br />

RelA<br />

NF-κB<br />

p50/52<br />

PCAF<br />

CBP/<br />

p300<br />

HDAC<br />

Survival, Proliferation, Inflammation, Immune Regulation<br />

TNFR<br />

TRADD<br />

TRAF2/5<br />

RIP<br />

ub<br />

Tax<br />

ub<br />

p65/<br />

IκBα RelA<br />

NF-κB2<br />

p52<br />

GSK-3β<br />

CK2<br />

SUMO<br />

SUMO<br />

IKKγ/<br />

NEMO<br />

PIASγ<br />

MSK1<br />

ATM<br />

Growth Factors:<br />

BMP, EGF, HGH,<br />

Insulin, NGF, TGF-α<br />

ub<br />

ubc13<br />

Akt<br />

Cot<br />

ub<br />

Genotoxic<br />

Stress<br />

IKKγ/<br />

NEMO<br />

PARP1<br />

GF-Rs<br />

Ras<br />

PDK1<br />

β-TrCP<br />

IKKα<br />

IKKα<br />

PI3K<br />

H3<br />

ub<br />

ub<br />

IKKα<br />

IKKα<br />

TRAF2<br />

c-IAP1/2<br />

IKKα<br />

LT, CD40L,<br />

BAFF/BLys<br />

LTβR,<br />

CD40,<br />

BR3<br />

NIK<br />

NF-κB2<br />

p100<br />

NF-κB2<br />

p52<br />

NF-κB2<br />

p52<br />

TRAF3<br />

IKKα<br />

Proteasomal<br />

Processing<br />

RelB<br />

RelB<br />

RelB<br />

Lymphogenesis, B Cell Maturation<br />

Nuclear factor-κB (NF-κB)/Rel proteins include NF-κB2 p52/p100, NF-κB1 p50/p105, c-Rel, RelA/p65, and RelB. These proteins function as dimeric transcription factors that<br />

regulate the expression of genes influencing a broad range of biological processes including innate and adaptive immunity, inflammation, stress responses, B-cell development,<br />

and lymphoid organogenesis. In the classical (or canonical) pathway, NF-κB/Rel proteins are bound and inhibited by IκB proteins. Proinflammatory cytokines, LPS, growth<br />

factors, and antigen receptors activate an IKK complex (IKKβ, IKKα, and NEMO), which phosphorylates IκB proteins. Phosphorylation of IκB leads to its ubiquitination and<br />

proteasomal degradation, freeing NF-κB/Rel complexes. Active NF-κB/Rel complexes are further activated by post-translational modifications (phosphorylation, acetylation,<br />

glycosylation) and translocate to the nucleus where, either alone or in combination with other transcription factors including AP-1, Ets, and Stat, they induce target gene<br />

expression. In the alternative (or noncanonical) NF-κB pathway, NF-κB2 p100/RelB complexes are inactive in the cytoplasm. Signaling through a subset of receptors, including<br />

LTβR, CD40, and BR3, activates the kinase NIK, which in turn activates IKKα complexes that phosphorylate C-terminal residues in NF-κB2 p100. Phosphorylation of NF-κB2<br />

p100 leads to its ubiquitination and proteasomal processing to NF-κB2 p52. This creates transcriptionally competent NF-κB p52/RelB complexes that translocate to the<br />

nucleus and induce target gene expression. Only a subset of NF-κB agonists and target genes are shown here.<br />

Select Reviews:<br />

Gilmore T.D. (2014) www.nf-kb.org • Hayden M.S. and Ghosh S. (2008) Cell 132, 344–362. • Perkins N.D. (2006) Oncogene 25, 6717–30. • Sun S-C. (2012) Immunol<br />

Rev. 246, 125–140. • Chen J. and Chen Z.J. (2013) Curr. Opin. Immunol. 25, 4–12.<br />

TNF<br />

CYLD<br />

Tumor Immunology<br />

MMPs<br />

VEGF<br />

T cell apoptosis<br />

IL-10<br />

IDO<br />

DC<br />

T Reg<br />

FoxP3<br />

CD4 +<br />

IL-10<br />

TGF-β<br />

IL-35<br />

Th1<br />

T-bet<br />

TNF-α<br />

IL-2<br />

CCL22<br />

MMPs<br />

VEGF<br />

Angiogenesis<br />

IFNγ<br />

T cell<br />

Immune<br />

Checkpoint<br />

MSC<br />

Arginase<br />

IL-10<br />

TGF-β<br />

IFNγ-R<br />

CTL<br />

T-bet<br />

T cell<br />

priming<br />

IL-1β<br />

TNF-α<br />

CD4 +<br />

T cell<br />

Tumor-draining Lymph Node<br />

Proliferation and production<br />

of anti-tumor antibodies<br />

Jak3<br />

Stat3<br />

cytotoxic<br />

granules<br />

Stat1<br />

B cell<br />

IL-2, 4, 5<br />

Tumor-specific<br />

CD8 + T cell<br />

TIM-3<br />

Galectin-9<br />

<br />

B7-H3<br />

Class I antigen<br />

presentation;<br />

IDO; PD-L1<br />

Activation/Response Change<br />

<br />

B7-H4<br />

Genes of cell growth and survival;<br />

PD-L1, VEGF, IL-6, IL-10<br />

PD-1<br />

PD-L1<br />

Nucleus<br />

NF-κB<br />

TCR<br />

MHC<br />

Stat3<br />

Akt<br />

MAPK<br />

DC<br />

M2<br />

Macrophage<br />

IFNγ<br />

cytotoxic<br />

granules<br />

TNF-R<br />

oncogenic<br />

signaling<br />

IL-6<br />

NK<br />

T-bet<br />

Jak<br />

TNF-α<br />

IL-1β<br />

Tumor-promoting<br />

Macrophage<br />

IL-10<br />

TGF-β<br />

M1<br />

Macrophage<br />

IL-6R<br />

Tumor Cell<br />

MMPs<br />

IL-4<br />

CCL22<br />

Inflammation<br />

IL-1β<br />

TNF-α<br />

ALK<br />

ROS1<br />

RTK: EGFR<br />

HER2<br />

etc.<br />

Mast cell<br />

Tumor cells employ multiple defense strategies to evade detection by the immune system. One common strategy, upregulation of immune checkpoint proteins and ligands,<br />

takes advantage of a natural immune mechanism for self-tolerance and prevention of collateral tissue damage. Immune checkpoint receptors, such as PD-1, CTLA-4, and<br />

many others, are located on T cells and engage with their corresponding ligand on tumor cells and dendritic cells, sending inhibitory signals that repress T cell activation<br />

or response. One of the first discovered checkpoint proteins, CTLA-4, plays a role at the stage of T cell priming by binding to the CD28 ligands CD80 or CD86 to prevent<br />

co-stimulatory signals necessary for T cell activation. In contrast, the PD-1/PD-L1 checkpoint acts later in the process, inhibiting anti-tumor immune responses by effector<br />

T cells such as CD4 + T helper 1 (Th1) cells and CD8 + cytotoxic T lymphocytes (CTLs), leading to decreases in IFNγ production and cytolytic activity. Upregulation of PD-L1<br />

expression on the tumor cell surface is mediated by IFNγR signaling to Stat1, as well as oncogenic signaling through several receptor tyrosine kinases (EGFR, ALK, ROS,<br />

HER2, and others) to activate the MAPK, Akt, and Stat3 pathways.<br />

Cells in the tumor microenvironment can also influence tumor progression. FoxP3 + /CD4 + T regulatory cells (Tregs) and myeloid-derived suppressor cells (MDSCs) secrete immunosuppressive<br />

cytokines IL-10 and TGF-β to inhibit the activity of Th1 cells and CTLs. Natural killer (NK) cells release cytotoxic granules against the tumor cell and secrete<br />

IFNγ, which stimulates surrounding pro-inflammatory M1 macrophages. Pro-tumorigenic M2 macrophages suppress anti-tumor immune responses via production of IL-10<br />

and TGF-β and promote metastasis through release of MMPs. MMPs and TGF-β are also released by surrounding mast cells.<br />

Select Reviews:<br />

Pardoll, D.M. (2012) Nat. Rev. Cancer 12, 252–264. • Vanneman, M. and Dranoff, G. (2012) Nat. Rev. Cancer 12, 237–251. • Kawakami, Y., Yaguchi, T., and Park, J.H.,<br />

et al. (2013) Front. Oncol. 3, 136. • Elinav, E., Nowarski, R., Thaiss, C.A., et al. (2013) Nat. Rev. Cancer 13, 759–771. • Mentlik, J.A., Cohen, A.D., and Campbell, K.S.<br />

(2013) Front. Immunol. 4, 481. • Gajewski, T.F., Schreiber, H., and Fu, Y.X. (2013) Nat. Immunol. 14, 1014–1022. • Krstic, J. and Santibanez, J.F. (2014) ScientificWorld-<br />

Journal, 521754.<br />

MMPs<br />

TGF-β<br />

Th2<br />

IL-13<br />

© 2009–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Thomas D. Gilmore, Boston University, Boston, MA, for reviewing this diagram.<br />

182 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

© 2014–2015 Cell Signaling Technology, Inc. • We would like to thank Glenn Dranoff, M.D., Susanne H.C. Baumeister, M.D.,<br />

Karrie Wong, Ph.D., and Girija Goyal, Dana Farber Cancer Institute and Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

www.cellsignal.com/cstpathways 183


08<br />

Section I: Research Areas<br />

Neuronal<br />

Development Markers<br />

Neural progenitor: Nestin, Musashi-1<br />

Astroglial precursor: Notch1<br />

Oligodendrocyte progenitor: A2B5,<br />

PDGFRα<br />

Neuronal differentiation: CEND1<br />

Neuronal stem cell marker: Sox1, Sox2<br />

Neuron: Neurofilament L, Neurofilament<br />

M, Neurofilament H, β3-tubulin,<br />

MAP2, Tau<br />

Oligodendrocytes: CNPase, MAG, MBP<br />

Schwann: Vimentin<br />

Astroglia: GFAP<br />

Neuroscience<br />

Neuroscience is a broad scientific area consisting of cellular and molecular biology, anatomy, physiology,<br />

and development of neurons and the nervous system. The field also includes cognitive neuroscience<br />

and behavioral research.<br />

Neuronal Development<br />

Development of the peripheral and central nervous systems begins early in embryogenesis and can<br />

be tracked throughout its different stages using lineage markers specific to each stage of neuronal<br />

development. Neural stem cells are derived from the ectoderm and differentiate into neural crest cells,<br />

glial progenitor cells, and neuronal progenitor cells. Markers for neural stem cells include Sox1 and<br />

Sox2. The neural crest further differentiates into a diverse array of cell types including neurons, glia,<br />

craniofacial cartilage, and connective tissue, and is sometimes referred to as the fourth primary germ<br />

layer. Neural crest markers include FoxD3 and Notch1. Glial progenitor cells develop into astrocytes,<br />

which provide structural support and help form the blood-brain barrier, and oligodendrocytes, which<br />

form the insulating myelin sheaths that surround axons. Neuronal progenitor cells, which can be identified<br />

using the markers Nestin and Musashi-1, give rise to mature neurons.<br />

Sox1, a marker for neuronal stem cells, is expressed<br />

in 1-day old rat brain but not adult rat brain.<br />

Sox1 Antibody #4194: Confocal IF<br />

analysis of postnatal day 1 (left) and<br />

adult (right) rat brain using #4194 (green)<br />

and Neurofilament-L (DA2) Mouse<br />

mAb #2835 (red). Blue pseudocolor =<br />

DRAQ5 ® #4084 (fluorescent DNA dye).<br />

Synaptic signaling occurs when the signal from one neuron is transmitted across the synaptic cleft<br />

to another neuron through the action of neurotransmitters (NTs). These NTs, such as dopamine,<br />

glutamate, and GABA (γ-aminobutyric acid), are stored in synaptic vesicles in the presynaptic neuron.<br />

Upon receiving an action potential, vesicles containing NT dock, prime, and fuse to the presynaptic<br />

membrane in a highly regulated mechanism through the action of SNARE family (VAMP, syntaxin-1,<br />

SNAP25), chaperone (complexin), and calcium binding proteins (synaptotagmin). Vesicle fusion results<br />

in NT release into the synaptic cleft, where it binds one of several receptors on the postsynaptic<br />

membrane. Receptor families such as the dopamine receptor, which is a GPCR, can signal through<br />

adenylate cyclase to activate PKA and other signaling intermediates to regulate gene expression<br />

through the actions of CREB and other transcription factors. Other NTs bind ion channels such as<br />

NMDAR or AMPAR that regulate flux of Ca 2+ and Na + , thereby perpetuating the action potential through<br />

the postsynaptic neuron. Continual NT release into a synapse and clustering of postsynaptic receptors<br />

can strengthen synaptic signaling over time. Synaptic plasticity, or the ability to modulate the number<br />

of NT receptors at the synapse, is a mechanism involved in many adaptative processes such as stress,<br />

addiction, and learning and memory.<br />

VAMP2, a SNARE<br />

protein expressed in<br />

brain tissue, cell lines,<br />

and primary neurons,<br />

facilitates the docking,<br />

priming, and fusion of<br />

NT-containing vesicles<br />

to the presynaptic<br />

membrane.<br />

A<br />

Neuronal Markers<br />

Markers are proteins with a very specific and well-defined localization that are used to identify or localize<br />

a subset of neurons (i.e. glutamatergic or GABAergic) or cellular compartment (i.e. presynaptic or<br />

postsynaptic compartment).<br />

B<br />

kDa<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

1 2 3 4 5 6<br />

VAMP2<br />

chapter 08: Neuroscience<br />

VAMP2 (D6O1A) Rabbit mAb #13508:<br />

Confocal IF analysis of primary rat cortical<br />

neurons grown for 21 days (A) using<br />

#13508 (green). Blue pseudocolor =<br />

DRAQ5 ® #4084 (fluorescent DNA dye).<br />

WB analysis of extracts from various cell<br />

lines and tissues (B) using #13508.<br />

Lanes<br />

1. mouse brain<br />

2. rat brain<br />

3. human cerebellum<br />

4. OVCAR8<br />

5. SK-N-SH<br />

6. Neuro-2a<br />

Synaptic Signaling<br />

Neurons are the building blocks of extensive neural networks. Transfer of information between neurons<br />

occurs at the synapse, where the neuronal information is converted from electrical action potentials<br />

into neurochemical signals. The synapse comprises a presynaptic active zone, the synaptic cleft, and<br />

the postsynaptic density.<br />

Neuronal Type<br />

Glutamatergic: EAAT1, EAAT2, EAAT3, VGluT1, VGluT2<br />

Dopaminergic: Tyrosine hydroxylase, Parkin<br />

GABAergic: GAD1, GAD2, DARPP-32<br />

Subcellular Compartment<br />

Presynaptic: Synapsin-1, Synaptophysin, SYT1, NSF<br />

Postsynaptic: PSD95, SHANK2<br />

Dentrite: MAP2<br />

Axon: β3-tubulin, Tau<br />

PSD95, a scaffolding<br />

protein within the<br />

postsynaptic density, is<br />

expressed in rat retina.<br />

Synaptic vesicle<br />

Voltage-gated<br />

Ca 2+ channels<br />

Presynaptic<br />

Active Zone<br />

Synapsin-1 (D12G5) XP ® Rabbit mAb<br />

#5297: Confocal IF analysis of mouse<br />

brain (left) or primary rat cortical neurons<br />

grown for 21 days (right) using #5297<br />

(green) and β3-Tubulin (TU-20) Mouse<br />

mAb #4466 (red). Blue pseudocolor =<br />

DRAQ5 ® #4084 (fluorescent DNA dye).<br />

Synapsin-1, a<br />

presynaptic marker,<br />

is expressed in<br />

mouse brain and<br />

primary neurons.<br />

PSD95 (D27E11) XP ® Rabbit mAb<br />

#3450: Confocal IF analysis of rat retina<br />

using #3450 (red) and Neurofilament-L<br />

(DA2) Mouse mAb #2835 (green). Blue<br />

pseudocolor = DRAQ5 ® #4084 (fluorescent<br />

DNA dye).<br />

Neurotransmitter<br />

Neurotransmitter<br />

receptors<br />

Synaptic<br />

Cleft<br />

Post-synaptic<br />

density<br />

MAP2 (D5G1) XP ® Rabbit mAb #8707:<br />

Confocal IF analysis of frozen rat cerebellum<br />

(left) or primary rat cortical neurons<br />

grown for 21 days (right) using #8707<br />

(green). Blue pseudocolor = DRAQ5 ®<br />

#4084 (fluorescent DNA dye).<br />

MAP2, a marker<br />

for dendrites, is<br />

expressed in rat<br />

cerebellum and<br />

primary neurons.<br />

184 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstneuroscience 185


Section I: Research Areas<br />

α-Synuclein is the<br />

main component of<br />

pathogenic Lewy<br />

bodies and Lewy<br />

neurites characteristic<br />

of Parkinson’s<br />

disease.<br />

Rev-Erbα regulates<br />

expression of several<br />

genes, including<br />

circadian regulator<br />

protein BMAL1.<br />

Neurodegeneration<br />

Devastating diseases arise from loss of neuron structure or function and are generally known as neurodegenerative<br />

diseases. One of the most common neurodegenerative diseases worldwide is Alzheimer’s<br />

disease (AD). This condition is characterized by the presence of extracellular amyloid plaques that form<br />

through abnormal APP (amyloid β precursor protein) processing and aggregation of β-amyloid peptides.<br />

AD is also characterized by the formation of neurofibrillary tangles that result from hyperphosphoryation<br />

of the tau protein. Parkinson’s disease, another neurodegenerative disorder, occurs when genetic mutation<br />

or environmental toxins result in misfolded α-synuclein protein that aggregates to form Lewy bodies.<br />

These aggregates alter dopamine signaling, particularly in the nigrostriatal pathway, ultimately leading<br />

to neuronal dysfunction and cell death.<br />

% of total input chromatin<br />

0.007<br />

0.006<br />

0.005<br />

0.004<br />

0.003<br />

0.002<br />

0.001<br />

0<br />

A<br />

BMAL1 NR1D1 α Satellite<br />

β-amyloid fragments aggregate<br />

to form the amyloid plaques<br />

characteristic of Alzheimer’s disease.<br />

β-Amyloid (D54D2) XP ® Rabbit mAb #8243: Confocal IF analysis of paraffin-embedded<br />

human Alzheimer’s brain using #8243 (green) and Tau (Tau46) Mouse mAb #4019 (red).<br />

Blue pseudocolor = DRAQ5 ® #4084 (fluorescent DNA dye).<br />

Circadian Rhythms<br />

Circadian rhythms govern many key physiological processes that fluctuate with a period of approximately<br />

24 hours. These processes include the sleep-wake cycle, glucose, lipid and drug metabolism, heart<br />

rate, hormone secretion, renal blood flow, and body temperature, as well as basic cellular processes<br />

such as DNA repair and the timing of the cell division cycle. The mammalian circadian system consists<br />

of many individual tissue-specific clocks (peripheral clocks) that are controlled by a master circadian<br />

pacemaker residing in the suprachiasmatic nuclei (SCN) of the brain. The periodic circadian rhythm is<br />

prominently manifested by the light-dark cycle, which is sensed by the visual system and processed by<br />

the SCN. The cellular circadian clockwork consists of interwoven positive and negative regulatory loops,<br />

or limbs. The positive limb includes the CLOCK and BMAL1 proteins, two transcription factors that bind<br />

E box enhancer elements and activate transcription of their target genes. The negative limb is formed<br />

by CRY and PER proteins, which inhibit CLOCK/BMAL1-mediated transcriptional activation. In tissues,<br />

roughly six to eight percent of all genes exhibit a circadian expression pattern. For example, expression<br />

of the nuclear receptor Rev-Erbα oscillates with circadian rhythm in liver cells. Rev-Erbα regulates<br />

expression of several key regulators of circadian rhythm, including BMAL1, ApoA-I, and ApoC-III.<br />

186 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

B<br />

Rev-Erbα (E1Y6D) Rabbit mAb #13418<br />

Normal Rabbit IgG #2729<br />

α-Synuclein (D37A6) XP ® Rabbit<br />

mAb #4179: IHC analysis of paraffinembedded<br />

mouse brain (A) using<br />

#4179. Confocal IF analysis of normal<br />

rat cerebellum using #4179 (green) (B).<br />

Blue pseudocolor = DRAQ5 ® #4084<br />

(fluorescent DNA dye).<br />

Rev-Erbα (E1Y6D) Rabbit mAb #13418: Chromatin IPs were performed with<br />

cross-linked chromatin from 4 x 10 6 Hep G2 cells and either 10 μl of #13418<br />

or 2 μl of Normal Rabbit IgG #2729 using SimpleChIP ® Enzymatic Chromatin IP<br />

Kit (Magnetic Beads) #9003. The enriched DNA was quantified by real-time PCR<br />

using human BMAL1 promoter primers, SimpleChIP ® Human NR1D1 Promoter<br />

Primers #13413, and SimpleChIP ® Human α Satellite Repeat Primers #4486.<br />

The amount of immunoprecipitated DNA in each sample is represented as a<br />

percent of the total input chromatin.<br />

Select Reviews<br />

Dzamko, N., Zhou, J., Huang, Y., and Halliday, G.M. (2014) Front. Mol. Neurosci. 7, 57. • Florio, M. and Huttner, W.B. (2014)<br />

Development 141, 2182−2194. • Franco, S.J. and Müller, U. (2013) Neuron 77, 19−34. • Kalsbeek, A., la Fleur, S., and<br />

Fliers, E. (2014) Mol. Metab. 3, 372−383. • Kojetin, D.J. and Burris, T.P. (2014) Nat. Rev. Drug Discov. 13, 197−216. •<br />

Spires-Jones, T.L. and Hyman, B.T. (2014) Neuron 82, 756−771. • Südhof, T.C. (2013) Neuron 80, 675−690. • Südhof, T.C.<br />

(2013) Nat. Med. 19, 1227−1231. • Tenreiro, S., Eckermann, K., and Outeiro, T.F. (2014) Front. Mol. Neurosci. 7, 42.<br />

Commonly Studied Neuroscience Targets<br />

Target M P<br />

A2B5<br />

•<br />

AMPA Receptor (GluR 1) •<br />

•<br />

Phospho-AMPA Receptor<br />

(GluR 1) (Ser845)<br />

AMPA Receptor (GluR 2/3/4)<br />

AMPA Receptor (GluR 2)<br />

Phospho-AMPA Receptor<br />

(GluR 2) (Tyr869/873/876)<br />

Phospho-AMPA Receptor<br />

(GluR 2) (Tyr876)<br />

AMPA Receptor (GluR 3)<br />

AMPA Receptor (GluR 4)<br />

(Arg860)<br />

APBA2<br />

ApoE<br />

ApoE4<br />

APP<br />

Phospho-APP (Thr668)<br />

APP/β-Amyloid<br />

β-Amyloid<br />

Arrestin 1/S-Arrestin<br />

Ataxin-1<br />

BACE<br />

β-Amyloid (pE3 Peptide)<br />

β-Amyloid (1-37 Specific)<br />

β-Amyloid (1-39 Specific)<br />

β-Amyloid (1-40 Specific)<br />

β-Amyloid (1-42 Specific)<br />

BMAL1<br />

Brn2/POU3F2<br />

BRSK1<br />

BRSK2<br />

Bassoon<br />

Calbindin<br />

CaMKI-δ<br />

CaMKII-α<br />

CaMKII (pan)<br />

Phospho-CaMKII (Tyr231)<br />

Phospho-CaMKII (Thr286)<br />

CaMKIV<br />

Phospho-CAMKK2 (Ser511)<br />

CASK<br />

Caspr2<br />

Cathepsin B<br />

CD13/APN<br />

CDK5<br />

CEND1<br />

CIRBP<br />

CK1δ<br />

CK1ε<br />

CLCN3<br />

• Phospho-CREB (Ser133)<br />

• CRMP-2<br />

• Phospho-CRMP-2 (Thr514)<br />

Dab1<br />

•<br />

• •<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

• •<br />

•<br />

• •<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

• •<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

Target M P<br />

CNPase<br />

• •<br />

Complexin-1<br />

Complexin-1/2<br />

CREB<br />

Phospho-Dab1 (Tyr220)<br />

Phospho-Dab1 (Tyr232)<br />

DAG Lipase α<br />

DARPP-32<br />

Phospho-DARPP-32 (Thr34)<br />

Phospho-DARPP-32 (Thr75)<br />

Phospho-DARPP-32 (Ser97)<br />

DCBLD2<br />

DDC<br />

Delta FosB<br />

DJ-1<br />

Dopamine β-Hydroxylase<br />

(DBH)<br />

Doublecortin<br />

Phospho-Doublecortin<br />

(Ser297)<br />

Phospho-Doublecortin<br />

(Ser334)<br />

Drebrin<br />

Drebrin A<br />

DYRK1A<br />

Dyrk1B<br />

Dysbindin<br />

EAAT1<br />

EAAT2<br />

EAAT3<br />

EGR1<br />

EGR3<br />

FABP7<br />

FE65<br />

GABA(B)R1<br />

GABA(B)R2<br />

GAD1<br />

GAD2<br />

GAP43<br />

GFAP<br />

GGA3<br />

Cleaved GGA3 (Asp313)<br />

GKAP<br />

Glutamate Dehydrogenase<br />

1/2<br />

GNB3<br />

GPR50<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• Munc18-1<br />

• Musashi<br />

Myelin Basic Protein<br />

•<br />

•<br />

•<br />

• •<br />

• •<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

• •<br />

• •<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

Target M P<br />

GRAF1<br />

GRK2<br />

GSTP1<br />

Homer1<br />

5-HTR1A<br />

5-HTR4<br />

Huntingtin<br />

KISS1R<br />

LIS1<br />

LRRK2<br />

MAG<br />

MAP2<br />

Phospho-MAP2 (Ser136)<br />

Phospho-MAP2<br />

(Thr1620/1623)<br />

MELK<br />

Mena<br />

Merlin<br />

Phospho-Merlin (Ser518)<br />

mGluR1<br />

mGluR2<br />

STOP<br />

Na Channel β1 Subunit<br />

NCS1<br />

Nestin<br />

NeuN<br />

NeuroD<br />

Neurofilament-H<br />

Neurofilament-L<br />

Neurofilament-M<br />

Neurogenin 2<br />

Neuropeptide Y<br />

Neuropilin-1<br />

Neuropilin-2<br />

NG2<br />

NGF<br />

NHERF1<br />

NHERF2<br />

Nicastrin<br />

NKCC1<br />

NMDAR1<br />

Phospho-NMDAR1 (Ser890)<br />

Phospho-NMDAR1 (Ser896)<br />

Phospho-NMDAR1 (Ser897)<br />

NMDAR2A<br />

Phospho-NMDAR2A<br />

(Tyr1246)<br />

NMDAR2B<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

• •<br />

• •<br />

• •<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

• •<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

chapter 08: Neuroscience<br />

These protein targets represent key<br />

nodes within neuroscience pathways<br />

and are commonly studied in<br />

neuroscience research. Primary<br />

antibodies, antibody conjugates, and<br />

antibody sampler kits containing these<br />

targets are available from <strong>CST</strong>.<br />

Listing as of September 2014. See our<br />

website for current product information.<br />

M Monoclonal Antibody<br />

P Polyclonal Antibody<br />

www.cellsignal.com/cstneuroscience 187


SYT SYT<br />

VAMP VAMP<br />

SYT SYT<br />

VAMP VAMP<br />

VAMP<br />

Syntaxin 1<br />

Syntaxin 1<br />

Dynamin<br />

Syntaxin 1<br />

Section I: Research Areas<br />

chapter 08: Neuroscience<br />

75<br />

2012–2014 citations<br />

<strong>CST</strong> antibodies for Phospho-CREB<br />

(Ser133) have been cited over 75 times<br />

in high-impact, peer-reviewed publications<br />

from the global research community.<br />

Target M P<br />

Phospho-NMDAR2B •<br />

(Tyr1070)<br />

Phospho-NMDAR2B •<br />

(Ser1284)<br />

Phospho-NMDAR2B •<br />

(Tyr1472)<br />

Nna1<br />

•<br />

nNOS<br />

• •<br />

Nogo-A<br />

•<br />

NT5E/CD73 •<br />

Oligophrenin-1 •<br />

Phospho-µ-Opioid Receptor •<br />

(Ser375)<br />

p35/25 •<br />

p39<br />

•<br />

p75NTR<br />

• •<br />

PARK9<br />

•<br />

Parkin<br />

• •<br />

PC1/3<br />

•<br />

PC2<br />

•<br />

PEN2<br />

• •<br />

PINK1<br />

•<br />

Plexin A1<br />

•<br />

Plexin A2 • •<br />

Plexin A3 •<br />

Plexin A4 •<br />

Presenilin-1 • •<br />

Presenilin-2 • •<br />

PSD93<br />

•<br />

Phospho-PSD93 (Tyr340) •<br />

PSD95<br />

• •<br />

Phospho-PSD95 •<br />

(Tyr236/Tyr240)<br />

Target M P<br />

Ras-GRF1<br />

Phospho-Ras-GRF1 (Ser916)<br />

RGS4<br />

Rhodopsin<br />

SAP102<br />

Secretagogin<br />

Semaphorin 3B<br />

Phospho-Semaphorin 4B<br />

(Ser825)<br />

Select Citations:<br />

Madiraju, A.K. et al. (2014) Metformin suppresses gluconeogenesis<br />

by inhibiting mitochondrial glycerophosphate<br />

dehydrogenase. Nature 510, 542–546.<br />

Villeda, S.A. et al. (2014) Young blood reverses age-related<br />

impairments in cognitive function and synaptic plasticity in<br />

mice. Nat. Med. 20, 659–663.<br />

White, A.C. et al. (2014) Stem cell quiescence acts as a<br />

tumour suppressor in squamous tumours. Nat. Cell Biol.<br />

16, 99–107.<br />

Kawai, M. et al. (2014) Sympathetic activation induces<br />

skeletal Fgf23 expression in a circadian rhythm-dependent<br />

manner. J. Biol. Chem. 289, 1457–1466.<br />

Parisiadou, L. et al. (2014) LRRK2 regulates synaptogenesis<br />

and dopamine receptor activation through modulation of PKA<br />

activity. Nat. Neurosci. 17, 367–376.<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

•<br />

SHANK2<br />

•<br />

Shootin1<br />

•<br />

SLC1A4<br />

•<br />

SNAP25<br />

• •<br />

SOD1<br />

• •<br />

Spinophilin • •<br />

SSTR1<br />

•<br />

Stargazin • •<br />

STEP<br />

• •<br />

Non-phospho-STEP (Ser221) •<br />

Synapsin<br />

• •<br />

Phospho-Synapsin (Ser9) •<br />

Synaptophysin • •<br />

SynGAP<br />

• •<br />

Syntaxin 1A<br />

•<br />

α-Synuclein • •<br />

α/β-Synuclein •<br />

Synaptotagmin • •<br />

Tau<br />

•<br />

Phospho-Tau (Thr181) •<br />

Phospho-Tau (Ser202) •<br />

Phospho-Tau (Ser396) •<br />

Target M P<br />

•<br />

Phospho-Tau (Ser400/<br />

Thr403/Ser404)<br />

TDP43<br />

Tenascin C<br />

TFAM<br />

Thy1<br />

TMP21<br />

Torsin A<br />

TPH-1<br />

Trk (pan)<br />

TrkA<br />

Phospho-TrkA (Tyr490)<br />

Phospho-TrkA (Tyr490)<br />

/TrkB (Tyr516)<br />

Phospho-TrkA (Tyr674/675)<br />

/TrkB (Tyr706/707)<br />

Phospho-TrkA (Tyr785)<br />

/TrkB (Tyr816)<br />

TrkB<br />

TrkC<br />

β3-Tubulin<br />

Tyrosine Hydroxylase<br />

Phospho-Tyrosine<br />

Hydroxylase (Ser31)<br />

Phospho-Tyrosine<br />

Hydroxylase (Ser40)<br />

UNC5B<br />

VAMP2<br />

VAMP3<br />

VGLUT1<br />

VGLUT2<br />

Vti1a<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

•<br />

•<br />

•<br />

•<br />

• •<br />

•<br />

Azeloglu, E.U. et al. (2014) Interconnected network motifs<br />

control podocyte morphology and kidney function. Sci Signal.<br />

7, ra12.<br />

Ma, L. et al. (2014) Cluster of differentiation 166 (CD166)<br />

regulated by phosphatidylinositide 3-Kinase (PI3K)/AKT signaling<br />

to exert its anti-apoptotic role via yes-associated protein<br />

(YAP) in liver cancer. J. Biol. Chem. 289, 6921–6933.<br />

Socodato, R. et al. (2014) The nitric oxide-cGKII system<br />

relays death and survival signals during embryonic retinal<br />

development via AKT-induced CREB1 activation. Cell Death<br />

Differ. 21, 915–928.<br />

Liu, J. et al. (2014) Insulin-like growth factor-1 and bone<br />

morphogenetic protein-2 jointly mediate prostaglandin E2-<br />

induced adipogenic differentiation of rat tendon stem cells.<br />

PLoS One 9, e85469.<br />

Antibody Validation Principles<br />

Please visit our website to learn more about what Antibody Validation means at Cell Signaling Technology.<br />

www.cellsignal.com/cstvalidation<br />

Vesicle Trafficking in Presynaptic Neurons: Synchronous Release<br />

Ca 2+<br />

Channel<br />

Ca 2+<br />

Channel<br />

SYT SYT<br />

VAMP VAMP<br />

SYT SYT<br />

VAMP VAMP<br />

SYT1<br />

Munc13<br />

RIM<br />

RIM-BP<br />

SYT SYT<br />

VAMP VAMP<br />

VAMP<br />

SYT SYT<br />

VAMP VAMP<br />

Munc13<br />

RIM<br />

RIM-BP<br />

Rab<br />

SYT1<br />

SYT1<br />

VAMP<br />

A<br />

Clathrin<br />

Docking<br />

Rab<br />

(NT)<br />

Munc18-1<br />

AP180<br />

AP-2<br />

SYT1<br />

SYT1<br />

VAMP<br />

C<br />

Super<br />

Priming<br />

(NT)<br />

SNAP25<br />

Complexin<br />

SYT1<br />

Amphiphysin<br />

Munc18-1<br />

SNAP25<br />

Vesicle<br />

Recycling<br />

Ca 2+<br />

Ca 2+<br />

Channel<br />

Ca 2+ Ca 2+<br />

Munc13<br />

RIM<br />

RIM-BP<br />

Rab<br />

Postsynaptic Neuron<br />

Ca 2+<br />

Channel<br />

Munc13<br />

RIM<br />

RIM-BP<br />

D<br />

Fusion<br />

and<br />

Release<br />

(NT)<br />

B<br />

Priming<br />

Rab<br />

(NT)<br />

VAMP<br />

VAMP<br />

SNAP25<br />

Ca 2+<br />

SYT1<br />

SNAP25<br />

Ca 2+<br />

SYT1<br />

Munc18-1<br />

Syntaxin 1<br />

Munc18-1<br />

Complexin<br />

SERT<br />

EAAT<br />

DAT<br />

NT<br />

Re-uptake<br />

Neuronal communication is a very connective process. Transfer of information between neurons occurs at the synapse, where the neuronal information is converted from<br />

electrical action potentials into neurochemical signals. The synapse comprises a presynaptic active zone (a clustering of vesicle fusion sites and calcium channels on the<br />

presynaptic cell membrane), the synaptic cleft, and the postsynaptic density, an electron-dense domain of the postsynaptic neuron specializing in the reception and integration<br />

of synaptic signals. Intracellular vesicles containing neurotransmitter (NT) rapidly fuse to the presynaptic membrane and release their contents into the synaptic cleft upon<br />

arrival of an action potential—a type of neurotransmission termed synchronous release. The docking, priming, and fusion of these vesicles is carried out by SNARE family and<br />

other chaperone proteins located on both the vesicle and presynaptic cell membrane. Synaptic vesicles dock to predetermined sites in the active zone through the interaction<br />

of vesicle-associated Rab3 (or Rab27) with RIM, which can bind to calcium channels directly and via RIM-BP (A). SNARE proteins might also play a role in docking based on<br />

studies of non-neuronal cells, but there is no conclusive evidence for such a role in mammalian neurons. The vesicle SNARE protein, VAMP (also called synaptobrevin), binds<br />

to SNARE proteins on the cell membrane, syntaxin 1 and SNAP25, priming the vesicle for fusion (B). Munc18-1 binds to monomeric syntaxin 1 as well the SNARE complex<br />

and assists with complex assembly. The co-chaperone protein complexin and the calcium-binding protein synaptotagmin 1 (SYT1) associate with SNARE proteins to form tight<br />

complexes, bringing the lipid membranes together (C). When an action potential in the presynaptic neuron opens voltage-gated calcium channels, calcium binds to SYT1<br />

and allows SYT1 to interact with the SNARE complex as well as the plasma membrane resulting in membrane fusion and release of NT into the synaptic cleft (D). The fast<br />

response to an action potential is due in part to the proteins RIM, RIM-BP, and Munc13, which form physical interactions between the vesicle, the cell membrane, and calcium<br />

channels, bringing the three necessary elements into close proximity. Released NT can be recycled through specific transporters such as EAATs (reuptake of glutamate) or a<br />

monoamine transporter, such as SERT (reuptake of serotonin) or DAT (reuptake of dopamine) back into the cytoplasm of the neuron.<br />

Select Reviews:<br />

Blakely, R.D. and Edwards, R.H. (2012) Cold Spring Harb. Perspect. Biol. 4, a005595. • Jahn, R. and Fasshauer, D. (2012) Nature 490, 201–207. • Saheki, Y. and De<br />

Camilli, P. (2012) Cold Spring Harb. Perspect. Biol. 4, a005645. • Südhof, T.C. (2013) Neuron 80, 675−690. • Südhof, T.C. (2013) Nat. Med. 19, 1227–1231. • Südhof,<br />

T.C. (2012) Neuron 75, 11–25.<br />

© 2015 Cell Signaling Technology, Inc. • We would like to thank Taulant Bacaj, Stanford School of Medicine, Palo Alto, CA for reviewing this diagram.<br />

188 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstpathways<br />

189


II<br />

Antibody<br />

Applications<br />

Studies of cellular regulation may employ a variety of immunodetection<br />

methods, with sample types spanning cultured cells, primary cells,<br />

and tissue samples and their lysates. Using multiple applications for<br />

investigation provides valuable complementary information and confirms<br />

data validity. The insights gained can, in turn, help you communicate a<br />

more comprehensive understanding of cellular signaling mechanisms to<br />

the research community. In this context it is important to understand the<br />

strengths and limitations of each method. At Cell Signaling Technology,<br />

it is our philosophy to share all our methods and tips that help ensure<br />

successful experiments. In this section of the <strong>CST</strong> <strong>Guide</strong>, we discuss<br />

and illustrate best practice approaches to overcoming obstacles commonly<br />

faced when performing antibody-based applications.<br />

Section Includes:<br />

Tips for Success<br />

Application Protocols<br />

Troubleshooting <strong>Guide</strong>s<br />

Protein Synthesis<br />

Key components of protein synthesis, including transcription and translational machinery,<br />

are illustrated at the junction between the nuclear pore and endoplasmic reticulum.<br />

www.cellsignal.com/cstlandscapes<br />

193


09<br />

Section II: ANTIBODY APPLICATIONS<br />

Chromatin Immunoprecipitation<br />

(ChIP)<br />

Optimal antibody concentration is critical for efficient ChIP.<br />

Using either too much or too little antibody can result in significantly lower enrichment, potentially leading<br />

to difficulty in detecting a low frequency, low stability binding event. Shown below are antibody titration<br />

data generated as part of <strong>CST</strong>’s rigorous ChIP validation process for p53 (1C12) Mouse mAb #2524.<br />

chapter 09: Chromatin Immunoprecipitation (ChIP)<br />

The ChIP assay is a powerful and versatile technique used for probing protein-DNA interactions within<br />

the natural chromatin context of the cell. The ChIP procedure isolates protein-DNA complexes that have<br />

been chemically crosslinked and harvested, using an antibody to enrich for specific protein associations<br />

by immunoprecipitation. After reversing the crosslinks, the DNA associated with a particular protein<br />

can be analyzed by PCR or ChIP-Seq.<br />

The success or failure of a ChIP experiment is highly dependent on the integrity of the chromatin, the<br />

quality of the epitope, and the specificity of the antibody. This is especially true for low abundance,<br />

low stability interactions—such as the binding of transcription factors (e.g., TCF4) and cofactors (e.g.,<br />

Ezh2) that may fall under the detection threshold if the integrity of the protein or DNA is compromised,<br />

or if the immunoenrichment relies on an antibody that is not highly specific to the target of interest.<br />

ChIP Tips for Success<br />

Enzymatic digestion better preserves chromatin integrity.<br />

Unlike sonication, enzymatic digestion using micrococcal nuclease does not expose the sample to<br />

harsh denaturing conditions (high heat and detergent), so it gently fragments the chromatin while<br />

protecting the integrity of the protein-DNA complex. As shown below, enzyme-digested chromatin<br />

displays a more robust enrichment of target DNA loci than does sonicated chromatin, particularly<br />

with low frequency interactions.<br />

Titration identifies the optimal amount of antibody to use per IP.<br />

% of total input chromatin<br />

0.8<br />

0.7<br />

0.6<br />

0.5<br />

0.4<br />

0.3<br />

0.2<br />

0.1<br />

0<br />

1 µl<br />

Optimal Antibody Concentration<br />

2.5 µl<br />

5 µl<br />

10 µl<br />

p53 (1C12) Mouse mAb #2524<br />

20 µl<br />

40 µl<br />

Normal Rabbit<br />

IgG #2729<br />

SimpleChIP ® Enzymatic<br />

Chromatin IP Kit (Magnetic<br />

Beads) #9003: Chromatin IPs<br />

were performed with cross-linked<br />

chromatin from 4 x 10 6 HCT 116 cells<br />

treated with UV (100 J/m 2 followed<br />

by a 3 hr recovery) and the indicated<br />

amounts of p53 (1C12) Mouse mAb<br />

#2524 using #9003. The enriched<br />

DNA was quantified by real-time<br />

PCR using SimpleChIP ® Human<br />

CDKN1A Promoter Primers #6449,<br />

human MDM2 intron 2 primers,<br />

and SimpleChIP ® Human α Satellite<br />

Repeat Primers #4486. The amount<br />

of immunoprecipitated DNA in each<br />

sample is presented as a percent of<br />

the total input chromatin.<br />

CDKN1A<br />

MDM2<br />

Sat1α<br />

The right positive control is essential for assay reliability.<br />

Target DNA loci are<br />

immunoprecipitated<br />

better from enzymedigested<br />

chromatin<br />

than sonicated<br />

chromatin.<br />

SimpleChIP ® Plus Enzymatic<br />

Chromatin IP Kit (Magnetic Beads)<br />

#9005: Chromatin IPs were performed<br />

with enzyme-digested or sonicated<br />

chromatin and the indicated ChIPvalidated<br />

antibodies using #9005.<br />

The enriched DNA was quantified by<br />

real-time PCR using primers to the<br />

designated loci. The amount of immunoprecipitated<br />

DNA in each sample<br />

is presented as a percent of the total<br />

input chromatin.<br />

GAPDH RPL30 HoxA1 HoxA2<br />

SimpleChIP<br />

Sonicated<br />

% of total input chromatin<br />

% of total input chromatin<br />

35<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

6<br />

5<br />

4<br />

3<br />

2<br />

High Abundance,<br />

High Stability Interactions<br />

Histone H3 (D2B12) XP ® Rabbit<br />

mAb (ChIP Formulated) #4620<br />

Low Abundance,<br />

Low Stability Interactions<br />

Rpb1 CTD (4H8)<br />

Mouse mAb #2629<br />

Tri-Methyl-Histone H3 (Lys4)<br />

(C42D8) Rabbit mAb #9751<br />

Tri-Methyl-Histone H3 (Lys27)<br />

(C36B11) Rabbit mAb #9733<br />

A Histone H3 antibody will immunoprecipitate all genomic loci, whether transcriptionally active or<br />

inactive, so it functions as a universal control for tracking assay efficiency and reagent performance.<br />

Although Rpb1 antibody is often used as a positive control in other ChIP kits, it only binds active loci<br />

and therefore may not be suitable when examining transcriptionally inactive regions of the genome.<br />

H3 is a more reliable control than Rpb1.<br />

% of total input chromatin<br />

20<br />

18<br />

16<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

Histone H3 (D2B12) XP ® Rabbit<br />

mAb (ChIP Formulated) #4620<br />

Rpb1 CTD (4H8) Mouse mAb #2629 Normal Rabbit IgG #2729<br />

Histone H3 (D2B12) XP ® Rabbit<br />

mAb (ChIP Formulated) #4620:<br />

ChIP was performed with 10 μg<br />

of cross-linked chromatin and the<br />

indicated antibodies. The enriched<br />

DNA was quantified by qPCR. The<br />

amount of immunoprecipitated DNA<br />

in each sample is presented as a<br />

percent of the total input chromatin.<br />

γ-actin<br />

GAPDH<br />

RPL30<br />

HoxA1<br />

HoxA2<br />

MYT1<br />

1<br />

0<br />

Ezh2 (D2C9) XP ®<br />

Rabbit mAb #5246<br />

SUZ12 (D39F6) XP ®<br />

Rabbit mAb #3737<br />

Normal Rabbit IgG #2729<br />

Benefits of Enzyme-based ChIP<br />

To learn more about the major advantages of enzymatic digestion using micrococcal nuclease compared to sonication,<br />

please go to www.cellsignal.com/chipbenefits<br />

194 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstchip<br />

195


Section II: ANTIBODY APPLICATIONS<br />

chapter 09: Chromatin Immunoprecipitation (ChIP)<br />

ChIP General Protocol<br />

Solutions and Reagents Included:<br />

Reagents Included in SimpleChIP ® and SimpleChIP ® Plus Enzymatic Chromatin IP Kits #9002, #9003, #9004, or #9005:<br />

Glycine Solution (10X), Buffer A (4X), Buffer B (4X), ChIP Buffer (10X), ChIP Elution Buffer (2X), 5 M NaCl, 0.5 M EDTA, 1 M DTT,<br />

DNA Binding Reagent A, DNA Wash Reagent B, DNA Elution Reagent C, DNA Spin Columns, Protease Inhibitor Cocktail (200X),<br />

RNAse A (10 mg/ml), Micrococcal Nuclease (2000 gel units/µl), Proteinase K (20 mg/ml), SimpleChIP ® Human RPL30 Exon 3<br />

Primers #7014, SimpleChIP ® Mouse RPL30 Intron 2 Primers #7015, Histone H3 (D2B12) XP ® Rabbit mAb (ChIP Formulated)<br />

#4620, Normal Rabbit IgG #2729, ChIP-Grade Protein G Magnetic Beads #9006 or ChIP-Grade Protein G Agarose Beads #9007<br />

Reagents Not Included:<br />

Formaldehyde (37%), Ethanol (96–100%), Isopropanol, 1X PBS (20X PBS #9808), Nuclease-free water, Taq DNA polymerase,<br />

dNTP Mix, For Kits #9002 and #9003 only: PMSF (#8553) (0.1 M stock), For Kits #9003 and #9005 only: 6-Tube Magnetic<br />

Separation Rack #7017<br />

I. Tissue Cross-linking and Sample Preparation<br />

IMPORTANT: Section I exclusive to use of SimpleChIP ® Plus Kits (#9004 & #9005)<br />

When harvesting tissue, remove unwanted material such as fat and necrotic material from the sample. Tissue can then be<br />

processed and cross-linked immediately, or frozen on dry ice for processing later. For optimal chromatin yield and ChIP results,<br />

use 25 mg of tissue for each IP to be performed. The chromatin yield does vary between tissue types and some tissues may<br />

require more than 25 mg for each IP. Please see Troubleshooting <strong>Guide</strong>, Section A for more information regarding the expected<br />

chromatin yield for different types of tissue. One additional chromatin sample should be processed for Analysis of Chromatin<br />

Digestion and Concentration (Section IV).<br />

Before starting:<br />

• Remove and warm 200X Protease Inhibitor Cocktail (PIC) and 10X glycine solution. Make sure PIC is completely thawed.<br />

• Prepare 3 ml of 1X Phosphate Buffered Saline (PBS) + 15 μl 200X PIC per 25 mg of tissue to be processed and place on ice.<br />

• Prepare 45 μl of 37% formaldehyde per 25 mg of tissue to be processed and keep at room temperature. Use fresh formaldehyde<br />

that is not past the manufacturer’s expiration date.<br />

A. Cross-linking<br />

1. Weigh the fresh or frozen tissue sample. Use 25 mg of tissue for each IP to be performed.<br />

2. Place tissue sample in a 60 mm or 100 mm dish and finely mince using a clean scalpel or razor blade. Keep dish on ice. It is<br />

important to keep the tissue cold to avoid protein degradation.<br />

3. Transfer minced tissue to a 15 ml conical tube.<br />

4. Add 1 ml of 1X PBS + PIC per 25 mg tissue to the conical tube.<br />

5. To crosslink proteins to DNA, add 45 μl of 37% formaldehyde per 1 ml of 1X PBS + PIC and rock at room temp for 20 min.<br />

Final formaldehyde concentration is 1.5%.<br />

6. Stop cross-linking by adding 100 μl of 10x glycine per 1 ml of 1X PBS + PIC and mix for 5 min at room temperature.<br />

7. Centrifuge tissue at 1,500 rpm in a bench top centrifuge for 5 min at 4°C.<br />

8. Remove supernatant and wash with 1 ml 1X PBS + PIC per 25 mg tissue.<br />

9. Repeat centrifugation (Section A, Step 7).<br />

10. Remove supernatant and resuspend tissue in 1 ml 1X PBS + PIC per 25 mg tissue and store on ice. Disaggregate tissue into<br />

single-cell suspension using a Medimachine (Section B) or Dounce homogenizer (Section C).<br />

B. Tissue Disaggregation Using Medimachine from BD Biosciences (part #340587)<br />

1. Cut off the end of a 1 ml pipette tip to enlarge the opening for transfer of tissue chunks.<br />

2. Transfer 1 ml of tissue resuspended in 1X PBS + PIC into the top chamber of a medicon filter (50 µm mesh; BD Part #340592).<br />

3. Grind tissue for 2 min according to manufacturer’s instructions.<br />

4. Collect cell suspension from the bottom chamber of the medicon using a 1 ml syringe and 18-gauge blunt needle.<br />

Transfer cell suspension to a 15 ml conical tube and place on ice.<br />

5. Repeat steps 2–4 until all the tissue is processed into a homogenous suspension.<br />

6. If more grinding is necessary, add more 1X PBS + PIC to tissue. Repeat steps 2–5 until all tissue is ground into a<br />

homogeneous suspension.<br />

7. Check for single-cell suspension by microscope (optional).<br />

8. Centrifuge cells at 1,500 rpm in a bench top centrifuge for 5 min at 4°C.<br />

9. Remove supernatant from cells and immediately continue with Nuclei Preparation and Chromatin Digestion (Section III).<br />

C. Tissue Disaggregation Using a Dounce Homogenizer<br />

1. Transfer tissue resuspended in 1X PBS + PIC to a Dounce homogenizer.<br />

2. Disaggregate tissue pieces with 20–25 strokes. Check for single-cell suspension by microscope (optional).<br />

3. Transfer cell suspension to a 15 ml conical tube and centrifuge at 1,500 rpm in a bench top centrifuge for 5 min at 4°C.<br />

4. Remove supernatant from cells and immediately continue with Nuclei Preparation and Chromatin Digestion (Section III).<br />

II. Cell Culture Cross-linking and Sample Preparation<br />

For optimal ChIP results, use approximately 4 x 10 6 cells for each IP to be performed. For HeLa cells, this is equivalent to half<br />

of a 15 cm culture dish containing cells that are 90% confluent in 20 ml of growth medium. One additional sample should be<br />

processed for Analysis of Chromatin Digestion and Concentration (Section IV). Include one extra dish of cells in experiment to<br />

be used for determination of cell number using a hemocytometer.<br />

196 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

Before starting:<br />

• Remove and warm 200X Protease Inhibitor Cocktail (PIC) and 10X Glycine Solution. Make sure PIC is completely thawed.<br />

• Prepare 2 ml of Phosphate Buffered Saline (PBS) + 10 μl 200X PIC per 15 cm dish to be processed and place on ice.<br />

• Prepare 40 ml of 1X PBS per 15 cm dish to be processed and place on ice.<br />

• Prepare 540 μl of 37% formaldehyde per 15 cm dish of cells to be processed and keep at room temperature. Use fresh<br />

formaldehyde that is not past the manufacturer’s expiration date.<br />

1. To crosslink proteins to DNA, add 540 μl of 37% formaldehyde to each 15 cm culture dish containing 20 ml medium. Swirl<br />

briefly to mix and incubate 10 min at room temperature. Final formaldehyde concentration is 1%. Addition of formaldehyde<br />

may result in a color change of the medium.<br />

2. Add 2 ml of 10X glycine solution to each 15 cm dish containing 20 ml medium, swirl briefly to mix, and incubate 5 min at<br />

room temperature. Addition of glycine may result in a color change of the medium.<br />

3. For suspension cells, transfer cells to a 50 ml conical tube, centrifuge at 1,500 rpm in a bench top centrifuge for 5 min<br />

at 4°C and wash pellet two times with 20 ml ice-cold 1X PBS. Remove supernatant and immediately continue with Nuclei<br />

Preparation and Chromatin Digestion (Section III).<br />

4. For adherent cells, remove media and wash cells two times with 20 ml ice-cold 1X PBS, completely removing wash from<br />

culture dish each time.<br />

5. Add 2 ml ice-cold 1X PBS + PIC to each 15 cm dish. Scrape cells into cold buffer. Combine cells from all culture dishes into<br />

one 15 ml conical tube.<br />

6. Centrifuge cells at 1,500 rpm in a bench top centrifuge for 5 min at 4°C. Remove supernatant and immediately continue<br />

with Nuclei Preparation and Chromatin Digestion (Section III).<br />

III. Nuclei Preparation and Chromatin Digestion<br />

One IP preparation is defined as 25 mg of disaggregated tissue or 4 x 10 6 tissue culture cells.<br />

Before starting:<br />

• Remove and warm 200X Protease Inhibitor Cocktail (PIC) and 1 M DTT. Make sure both are completely thawed and DTT<br />

crystals are completely in solution.<br />

• Remove and warm 10X ChIP buffer and ensure SDS is completely in solution.<br />

• Prepare 1 ml 1X buffer A (250 μl 4X buffer A + 750 μl water) + 0.5 μl 1 M DTT + 5 μl 200X PIC per IP prep and place on ice.<br />

• Prepare 1.1 ml 1X Buffer B (275 μl 4X buffer B + 825 μl water) + 0.55 μl 1 M DTT per IP prep and place on ice.<br />

• Prepare 100 μl 1X ChIP buffer (10 μl 10X ChIP Buffer + 90 μl water) + 0.5 μl 200X PIC per IP prep and place on ice.<br />

1. Resuspend cells in 1 ml ice-cold buffer A + DTT + PIC per IP prep. Incubate on ice for 10 min. Mix by inverting tube every<br />

3 min.<br />

2. Pellet nuclei by centrifugation at 3,000 rpm in a bench top centrifuge for 5 min at 4°C. Remove supernatant and resuspend<br />

pellet in 1 ml ice-cold buffer B + DTT per IP prep. Repeat centrifugation, remove supernatant, and resuspend pellet in 100 μl<br />

Buffer B + DTT per IP prep. Transfer sample to a 1.5 ml microcentrifuge tube, up to 1 ml total per tube.<br />

3. Add 0.5 μl of micrococcal nuclease per IP prep, mix by inverting tube several times and incubate for 20 min at 37°C with<br />

frequent mixing to digest DNA to length of approximately 150-900 bp. Mix by inversion every 3–5 min. The amount of micrococcal<br />

nuclease required to digest DNA to the optimal length may need to be determined empirically for individual tissues and<br />

cell lines (see Troubleshooting <strong>Guide</strong>, Section B). HeLa nuclei digested with 0.5 μl micrococcal nuclease per 4 x 10 6 cells and<br />

mouse liver tissue digested with 0.5 μl micrococcal nuclease per 25 mg of tissue gave the appropriate length DNA fragments.<br />

4. Stop digest by adding 10 μl of 0.5 M EDTA per IP prep and placing tube on ice.<br />

5. Pellet nuclei by centrifugation at 13,000 rpm in a microcentrifuge for 1 min at 4°C and remove supernatant.<br />

6. Resuspend nuclear pellet in 100 μl of 1X ChIP buffer + PIC per IP prep and incubate on ice for 10 min.<br />

7. Sonicate up to 500 μl of lysate per 1.5 ml microcentrifuge tube with several pulses to break nuclear membrane. Incubate<br />

samples for 30 sec on wet ice between pulses. Optimal conditions required for complete lysis of nuclei can be determined by<br />

observing nuclei on a light microscope before and after sonication.<br />

HeLa nuclei were completely lysed after 3 sets of 20-sec pulses using a VirTis Virsonic 100 Ultrasonic Homogenizer/Sonicator<br />

at setting 6 with a 1/8-inch probe. Alternatively, nuclei can be lysed by homogenizing the lysate 20 times in a Dounce<br />

homogenizer; however, lysis may not be as complete.<br />

8. Clarify lysates by centrifugation at 10,000 rpm in a microcentrifuge for 10 min at 4°C.<br />

9. Transfer supernatant to a new tube. This is the cross-linked chromatin preparation, which should be stored at -80°C until<br />

further use. Remove 50 μl of the chromatin preparation for Analysis of Chromatin Digestion and Concentration (Section IV).<br />

IV. Analysis of Chromatin Digestion and Concentration (Recommended Step)<br />

1. To the 50 μl chromatin sample (Step 9 in Section III), add 100 μl nuclease-free water, 6 μl 5 M NaCl, and 2 μl RNAse A. Vortex<br />

to mix and incubate samples at 37°C for 30 min.<br />

2. To each RNAse A-digested sample, add 2 μl Proteinase K. Vortex to mix and incubate samples at 65°C for 2 hr.<br />

3. Purify DNA from samples using DNA purification spin columns as described in Section VII.<br />

4. After purification of DNA, remove a 10 μl sample and determine DNA fragment size by electrophoresis on a 1% agarose gel<br />

with a 100 bp DNA marker. DNA should be digested to a length of approximately 150–900 bp (1–6 nucleosomes).<br />

5. To determine DNA concentration, transfer 2 μl of purified DNA to 98 μl nuclease-free water to give a 50-fold dilution and<br />

read the OD260. The concentration of DNA in μg/ml is OD260 x 2,500. DNA concentration should ideally be between 50 and<br />

200 μg/ml.<br />

NOTE: For optimal ChIP results, it is highly critical that the chromatin is of appropriate size and concentration. Over-digestion<br />

of chromatin may diminish signal in the PCR quantification. Under-digestion of chromatin may lead to increased background<br />

signal and lower resolution. Adding too little chromatin to the IP may result in diminished signal in the PCR quantification. A<br />

protocol for optimization of chromatin digestion can be found in the Troubleshooting <strong>Guide</strong>.<br />

www.cellsignal.com/cstprotocols 197


Section II: ANTIBODY APPLICATIONS<br />

chapter 09: Chromatin Immunoprecipitation (ChIP)<br />

ChIP Companion<br />

Products<br />

We offer a comprehensive list of<br />

companion products that are validated<br />

in-house with our protocols so that you<br />

can get the reliable reagents you need<br />

to complete your experiment.<br />

www.cellsignal.com/chipcompanions<br />

V. Chromatin IP<br />

For optimal ChIP results, use approximately 5–10 μg of digested, cross-linked chromatin (as determined in Section IV) per IP.<br />

This should be roughly equivalent to a single 100 μl IP prep from 25 mg of disaggregated tissue or 4 x 10 6 tissue culture cells.<br />

Typically, 100 μl of digested chromatin is diluted into 400 μl 1X ChIP Buffer prior to the addition of antibodies. However, if more<br />

than 100 μl of chromatin is required per IP, the cross-linked chromatin preparation does not need to be diluted as described<br />

below. Antibodies can be added directly to the undiluted chromatin preparation for IP of chromatin complexes.<br />

Before starting:<br />

• Remove and warm 200X Protease Inhibitor Cocktail (PIC). Make sure PIC is completely thawed.<br />

• Remove and warm 10X ChIP Buffer and ensure SDS is completely in solution.<br />

• Thaw digested chromatin preparation (Section III, Step 9) and place on ice.<br />

• Prepare low salt wash: 3 ml 1X ChIP buffer (300 μl 10X ChIP buffer + 2.7 ml H 2 0) per IP prep. Store at room temperature<br />

until use.<br />

• Prepare high salt wash: 1 ml 1X ChIP buffer (100 μl 10X ChIP buffer + 900 μl H 2 0) + 70 μl 5M NaCl per IP. Store at room<br />

temperature until use.<br />

1. In one tube, prepare enough 1X ChIP buffer for the dilution of digested chromatin into the desired number of IPs: 400 μl<br />

of 1X ChIP buffer (40 μl of 10X ChIP buffer + 360 μl H 2 0) + 2 μl 200X PIC per IP. When determining the number of IPs, remember<br />

to include the positive control Histone H3 (D2B12) XP ® Rabbit mAb and negative control Normal Rabbit IgG antibody<br />

samples. Place mix on ice.<br />

2. To the prepared 1X ChIP buffer, add the equivalent of 100 μl (5–10 μg of chromatin) of the digested, cross-linked chromatin<br />

preparation (Section III, Step 9) per IP. For example, for 10 IPs, prepare a tube containing 4 ml 1X ChIP Buffer (400 μl 10X<br />

ChIP buffer + 3.6 ml water) + 20 μl 200X PIC + 1 ml digested chromatin preparation.<br />

3. Remove a 10 μl sample of the diluted chromatin and transfer to a microfuge tube. This is your 2% input sample, which can<br />

be stored at -20°C until further use (Step 1 in Section VI).<br />

4. For each IP, transfer 500 μl of the diluted chromatin to a 1.5 ml microcentrifuge tube and add the immunoprecipitating antibody.<br />

The amount of antibody required per IP varies and should be determined by the user. For the positive control Histone<br />

H3 (D2B12) XP ® Rabbit mAb add 10 μl to the IP sample. For the negative control, Normal Rabbit IgG, add 1 μl (1 μg) to 2 μl<br />

(2 μg) to the IP sample. Incubate IP samples 4 hr to overnight at 4°C with rotation.<br />

5. a. Resuspend ChIP-Grade Protein G Agarose Beads by gently vortexing. Immediately add 30 μl of Protein G Agarose Beads to<br />

each IP reaction and incubate for 2 hr at 4°C with rotation.<br />

b. Resuspend ChIP-Grade Protein G Magnetic Beads by gently vortexing. Immediately add 30 μl of Protein G Magnetic Beads<br />

to each IP reaction and incubate for 2 hr at 4°C with rotation.<br />

6. a. Pellet Protein G Agarose Beads in each IP by brief 1 min centrifugation at 6,000 rpm in a microcentrifuge and remove<br />

supernatant.<br />

b. Pellet Protein G Magnetic Beads in each IP by placing the tubes in a Magnetic Separation Rack. Wait 1–2 min for solution<br />

to clear and then carefully remove supernatant.<br />

7. Wash Protein G Beads by adding 1 ml of low salt wash to the beads and incubate at 4°C for 5 min with rotation. Repeat<br />

Steps 6 and 7 two additional times for a total of 3 low salt washes.<br />

8. Add 1 ml of high salt wash to the beads and incubate at 4°C for 5 min with rotation.<br />

9. a. Pellet Protein G Agarose Beads in each IP by brief 1 min centrifugation at 6,000 rpm in a microcentrifuge. Remove<br />

supernatant and immediately proceed to Section VI.<br />

b. Pellet Protein G Magnetic Beads in each IP by placing the tubes in a Magnetic Separation Rack. Wait 1–2 min for solution to<br />

clear and then carefully remove supernatant and proceed to Section VI.<br />

VI. Elution of Chromatin from Antibody/Protein G Agarose Beads and Reversal of Cross-links<br />

Before starting:<br />

• Remove and warm 2X ChIP elution buffer in a 37°C water bath and ensure SDS is in solution.<br />

• Set a water bath or thermomixer to 65°C.<br />

• Prepare 150 μl 1X ChIP elution buffer (75 μl 2X ChIP elution buffer + 75 μl water) for each IP and the 2% input sample.<br />

1. Add 150 μl of the 1X ChIP elution buffer to the 2% input sample tube and set aside at room temperature until Step 6.<br />

2. Add 150 μl 1X ChIP elution buffer to each IP sample.<br />

3. Elute chromatin from the antibody/Protein G Beads for 30 min at 65°C with gentle vortexing (1,200 rpm). A thermomixer works<br />

best for this step. Alternatively, elutions can be performed at room temperature with rotation, but may not be as complete.<br />

4. a. Pellet Protein G Agarose Beads by brief 1 min centrifugation at 6,000 rpm in a microcentrifuge.<br />

b. Pellet Protein G Magnetic Beads by placing the tubes in a Magnetic Separation Rack and wait 1–2 min for solution to clear.<br />

5. Carefully transfer eluted chromatin supernatant to a new tube.<br />

6. To all tubes, including the 2% input sample from Step 1, reverse cross-links by adding 6 μl 5 M NaCl and 2 μl Proteinase K,<br />

and incubate 2 hr at 65°C. This incubation can be extended overnight.<br />

7. Immediately proceed to Section VII. Alternatively, samples can be stored at -20°C. However, to avoid formation of a precipitate,<br />

be sure to warm samples to room temperature before adding DNA binding reagent A (Section VII, Step 1).<br />

VII. DNA Purification Using Spin Columns<br />

Before starting:<br />

• Add 12 ml of isopropanol to DNA binding reagent A and 24 ml of ethanol (96–100%) to DNA wash reagent B before use.<br />

These steps only have to be performed once prior to the first set of DNA purifications.<br />

• Remove one DNA purification spin column and collection tube for each DNA sample (Section VI).<br />

1. Add 600 μl DNA binding reagent A to each DNA sample and vortex briefly.<br />

4 volumes of DNA binding reagent A should be used for every 1 volume of sample.<br />

2. Transfer 375 μl of each sample from Step 1 to a DNA purification spin column in collection tube.<br />

3. Centrifuge at 14,000 rpm in a microcentrifuge for 30 sec.<br />

4. Remove spin column from the collection tube and discard the liquid. Replace spin column in the collection tube.<br />

5. Transfer remaining 375 μl of each sample from Step 1 to the spin column in collection tube. Repeat Steps 3 and 4.<br />

6. Add 700 μl of DNA wash reagent B to the spin column in collection tube.<br />

7. Centrifuge at 14,000 rpm in a microcentrifuge for 30 sec.<br />

8. Remove spin column from the collection tube and discard the liquid. Replace spin column in the collection tube.<br />

9. Centrifuge at 14,000 rpm in a microcentrifuge for 30 sec.<br />

10. Discard collection tube and liquid. Retain spin column.<br />

11. Add 50 μl of DNA elution reagent C to each spin column and place into a clean 1.5 ml microcentrifuge tube.<br />

12. Centrifuge at 14,000 rpm in a microcentrifuge for 30 sec to elute DNA.<br />

13. Remove and discard DNA purification spin column. Eluate is now purified DNA. Samples can be stored at -20°C.<br />

VIII. Quantification of DNA by PCR<br />

Recommendations:<br />

• Use Filter-tip pipette tips to minimize risk of contamination.<br />

• The control primers included in the kit are specific for the human or mouse RPL30 gene and can be used for either standard<br />

PCR or quantitative real-time PCR. If the user is performing ChIPs from another species, it is recommended that the user<br />

design the appropriate specific primers to DNA and determine the optimal PCR conditions.<br />

• A Hot-Start Taq polymerase is recommended to minimize the risk of non-specific PCR products.<br />

• PCR primer selection is critical. Primers should be designed with close adherence to the following criteria:<br />

Primer length: 24 nucleotides<br />

Optimum Tm: 60°C<br />

Optimum GC: 50%<br />

Amplicon size: 150–200 bp (for standard PCR) 80–160 bp (for real-time quantitative PCR)<br />

Standard PCR Method:<br />

1. Label the appropriate number of 0.2 ml PCR tubes for the number of samples to be analyzed. These should include the 2%<br />

input sample, the positive control histone H3 sample, the negative control normal rabbit IgG sample, and a tube with no DNA<br />

to control for DNA contamination.<br />

2. Add 2 μl of the appropriate DNA sample to each tube.<br />

3. Prepare a master reaction mix as described below, making sure to add enough reagent for two extra tubes to account for<br />

loss of volume. Add 18 μl of master mix to each reaction tube.<br />

Reagent: Volume for 1 PCR Reaction (18 µl), Nuclease-free H 2 O: 12.5 µl, 10X PCR Buffer: 2.0 µl, 4 mM dNTP Mix: 1.0 µl,<br />

5 µM RPL30 Primers: 2.0 µl, Taq DNA Polymerase: 0.5 µl<br />

4. Start the following PCR reaction program:<br />

a. Initial denaturation 95°C 5 min<br />

b. Denature 95°C 30 sec<br />

c. Anneal 62°C 30 sec<br />

d. Extension 72°C 30 sec<br />

e. Repeat Steps b–d for a total of 34 cycles<br />

f. Final Extension 72°C 5 min<br />

5. Remove 10 μl of each PCR product for analysis by 2% agarose gel or 10% poly-acrylamide gel electrophoresis with a 100 bp<br />

DNA marker. The expected size of the PCR product is 161 bp for human RPL30 and 159 bp for mouse RPL30.<br />

Real-Time Quantitative PCR Method:<br />

1. Label the appropriate number of PCR tubes or PCR plates compatible with the model of PCR machine to be used. PCR<br />

reactions should include the positive control histone H3 sample, the negative control normal rabbit IgG sample, a tube with no<br />

DNA to control for contamination, and a serial dilution of the 2% input chromatin DNA (undiluted, 1:5, 1:25, 1:125) to create<br />

a standard curve and determine the efficiency of amplification.<br />

2. Add 2 μl of the appropriate DNA sample to each tube or well of the PCR plate.<br />

3. Prepare a master reaction mix as described below. Add enough reagents for two extra reactions to account for loss of volume.<br />

Add 18 μl of reaction mix to each PCR reaction tube or well.<br />

Reagent: Volume for 1 PCR Reaction (18 µl), Nuclease-free H 2 O: 6 µl 5 µM RPL30 Primers: 2 µl, 2X SYBR ® -Green Reaction<br />

Mix: 10 µl<br />

4. Start the following PCR reaction program:<br />

a. Initial denaturation 95°C, 3 min<br />

b. Denature 95°C, 15 sec<br />

c. Anneal and extension: 60°C, 60 sec<br />

d. Repeat steps b and c for a total of 40 cycles.<br />

5. Analyze quantitative PCR results using the software provided with the real-time PCR machine. Alternatively, one can calculate<br />

the IP efficiency manually using the Percent Input Method and the equation shown below. With this method, signals obtained<br />

from each IP are expressed as a percent of the total input chromatin.<br />

Percent Input = 2% x 2 (C[T] 2% Input Sample – C[T] IP Sample) C[T] = C T = Threshold cycle of PCR reaction<br />

198 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstprotocols<br />

199


Section II: ANTIBODY APPLICATIONS<br />

chapter 09: Chromatin Immunoprecipitation (ChIP)<br />

ChIP Troubleshooting <strong>Guide</strong><br />

A. Expected Chromatin Yield<br />

When harvesting cross-linked chromatin from tissue samples, the yield of chromatin can vary significantly between tissue types.<br />

The table below provides a range for the expected yield of chromatin from 25 mg of tissue compared to 4 x 10 6 HeLa cells, and<br />

the expected DNA concentration, as determined in Section IV of the protocol. For each tissue type, disaggregation using a BD <br />

Medimachine system (BD Biosciences) or a Dounce homogenizer yielded similar amounts of chromatin. However, chromatin<br />

processed from tissues disaggregated using the Medimachine typically gave higher IP efficiencies than chromatin processed<br />

from tissues disaggregated using a Dounce homogenizer. A Dounce homogenizer is strongly recommended for disaggregation of<br />

brain tissue, as the Medimachine does not adequately disaggregate brain tissue into a single-cell suspension. For optimal ChIP<br />

results, we recommend using 5–10 µg of digested, cross-linked chromatin per IP; therefore, some tissues may require harvesting<br />

more than 25 mg per each IP.<br />

Total Chromatin Yield<br />

Expected DNA Concentration<br />

Spleen<br />

20–30 µg per 25 mg tissue 200–300 µg/ml<br />

Liver<br />

10–15 µg per 25 mg tissue 100–150 µg/ml<br />

Kidney<br />

8–10 µg per 25 mg tissue 80–100 µg/ml<br />

Total Chromatin Yield<br />

Expected DNA Concentration<br />

Brain<br />

2–5 µg per 25 mg tissue 20–50 µg/ml<br />

Heart<br />

2–5 µg per 25 mg tissue 20–50 µg/ml<br />

HeLa<br />

10–15 µg per 4 x 10 6 cells 100–150 µg/ml<br />

B. Optimization of Chromatin Digestion<br />

Optimal conditions for the digestion of cross-linked chromatin DNA to 150–900 bp in length is highly dependent on the ratio of<br />

micrococcal nuclease to the amount of tissue or number of cells used in the digest. Below is a protocol for determination of the<br />

optimal digestion conditions for a specific tissue or cell type.<br />

1. Prepare cross-linked nuclei from 125 mg of tissue or 2 X 10 7 cells (equivalent of 5 IP preps), as described in Sections I, II,<br />

and III. Stop after Step 2 of Section III and proceed as described below.<br />

2. Transfer 100 μl of the nuclei preparation into 5 individual 1.5 ml microcentrifuge tubes and place on ice.<br />

3. Add 3 μl micrococcal nuclease stock to 27 μl of 1X Buffer B + DTT (1:10 dilution of enzyme).<br />

4. To each of the 5 tubes in Step 2, add 0 μl, 2.5 μl, 5 μl, 7.5 μl, or 10 μl of the diluted micrococcal nuclease, mix by inverting<br />

tube several times and incubate for 20 min at 37°C with frequent mixing.<br />

5. Stop each digest by adding 10 μl of 0.5 M EDTA and placing tubes on ice.<br />

6. Pellet nuclei by centrifugation at 13,000 rpm in a microcentrifuge for 1 min at 4°C and remove supernatant.<br />

7. Resuspend nuclear pellet in 200 μl of 1X ChIP buffer + PIC. Incubate on ice for 10 min.<br />

8. Sonicate lysate with several pulses to break nuclear membrane. Incubate samples for 30 sec on wet ice between pulses.<br />

Optimal conditions required for complete lysis of nuclei can be determined by observing nuclei on a light microscope before<br />

and after sonication. HeLa nuclei were completely lysed after 3 sets of 20 sec pulses using a VirTis Virsonic 100 Ultrasonic<br />

Homogenizer/Sonicator set at setting 6 with a 1/8-inch probe. Alternatively, nuclei can be lysed by homogenizing the lysate<br />

20 times in a Dounce homogenizer; however, lysis may not be as complete.<br />

9. Clarify lysates by centrifugation at 10,000 rpm in a microcentrifuge for 10 min at 4°C.<br />

10. Transfer 50 μl of each of the sonicated lysates to new microfuge tubes.<br />

11. To each 50 μl sample, add 100 μl nuclease-free water, 6 μl 5 M NaCl and 2 μl RNAse A. Vortex to mix and incubate<br />

samples at 37°C for 30 min.<br />

12. To each RNAse A-digested sample, add 2 μl Proteinase K. Vortex to mix and incubate sample at 65°C for 2 hr.<br />

13. Remove 20 μl of each sample and determine DNA fragment size by electrophoresis on a 1% agarose gel with a 100 bp<br />

DNA marker.<br />

14. Observe which of the digestion conditions produces DNA in the desired range of 150–900 base pairs (1–6 nucleosomes).<br />

The volume of diluted micrococcal nuclease that produces the desired size of DNA fragments using this optimization protocol<br />

is equivalent to 10 times the volume of micrococcal nuclease stock that should be added to one IP preparation (25 mg of<br />

disaggregated tissue cells or 4 X 10 6 tissue culture cells) to produce the desired size of DNA fragments. For example, if 5 μl<br />

of diluted micrococcal nuclease produces DNA fragments of 150–900 bp in this protocol, then 0.5 μl of stock micrococcal<br />

nuclease should be added to one IP preparation during the digestion of chromatin (Section III).<br />

15. If results indicate that DNA is not in the desired size range, then repeat optimization protocol, adjusting the amount of<br />

micrococcal nuclease in each digest accordingly. Alternatively, the digestion time can be changed to increase or decrease<br />

the extent of DNA fragmentation.<br />

Problem Possible Causes Recommendation<br />

Concentration of the<br />

digested chromatin is too<br />

low (low chromatin yield).<br />

Chromatin is under-digested<br />

and fragments are too large<br />

(greater than 900 bp). Large<br />

chromatin fragments can lead<br />

to increased background and<br />

lower resolution.<br />

Chromatin is over-digested<br />

and fragments are too small<br />

(exclusively 150 bp mononucleosome<br />

length). Complete<br />

digestion of chromatin to<br />

mono-nucleosome length DNA<br />

may diminish signal during PCR<br />

quantification, especially for<br />

amplicons greater than 150 bp<br />

in length.<br />

No product or very little product<br />

in the input PCR reactions.<br />

No product in the positive<br />

control histone H3-IP RPL30<br />

PCR reaction.<br />

Quantity of product in the<br />

negative control Rabbit IgG-IP<br />

and positive control histone<br />

H3-IP PCR reactions is equivalent<br />

(high background signal).<br />

No product in the Experimental<br />

Antibody-IP PCR reaction.<br />

Not enough tissue or cells were<br />

added to the chromatin digestion or<br />

cell nuclei were not completely lysed<br />

after digestion.<br />

Too many cells or not enough<br />

micrococcal nuclease was added to<br />

the chromatin digestion.<br />

Tissue or cells may have been over<br />

cross-linked. Cross-linking for longer<br />

than 10 min may inhibit digestion of<br />

chromatin.<br />

Not enough cells or too much<br />

micrococcal nuclease added to the<br />

chromatin digestion.<br />

Not enough DNA added to the PCR<br />

reaction or conditions are not optimal.<br />

PCR amplified region may span<br />

nucleosome-free region.<br />

Not enough chromatin added to the<br />

IP or chromatin is over-digested.<br />

Not enough chromatin or antibody<br />

added to the IP reaction or IP incubation<br />

time is too short.<br />

Incomplete elution of chromatin from<br />

Protein G beads.<br />

Too much or not enough chromatin<br />

added to the IP reaction. Alternatively,<br />

too much antibody added to<br />

the IP reaction.<br />

Too much DNA added to the PCR<br />

reaction or too many cycles of<br />

amplification.<br />

Not enough DNA added to the PCR<br />

reaction.<br />

Not enough antibody added to the<br />

IP reaction.<br />

Antibody does not work for ChIP.<br />

Add additional chromatin to each IP to give at least<br />

5 μg/IP and continue with protocol.<br />

Weigh tissue or count a separate plate of cells prior to<br />

cross-linking to determine accurate cell number. Some<br />

tissues may require processing of more than 25 mg per<br />

IP. The amount of tissue can be increased to 50 mg per<br />

IP, while still maintaining efficient chromatin fragmentation<br />

and extraction.<br />

Increase the number of sonications following chromatin<br />

digestion. Visualize cell nuclei under microscope before<br />

and after sonication to confirm complete lysis of nuclei.<br />

Weigh tissue or count a separate plate of cells prior to<br />

cross-linking to determine accurate cell number. Add<br />

less tissue or cells, or more micrococcal nuclease to<br />

the chromatin digest. See Section B for optimization of<br />

chromatin digestion.<br />

Perform a time course at a fixed formaldehyde concentration.<br />

Shorten the time of cross-linking<br />

to 10 min or less.<br />

Weigh tissue or count a separate plate of cells prior to<br />

cross-linking to determine accurate cell number. Add<br />

more tissue or cells, or less micrococcal nuclease to<br />

the chromatin digest. See Section B of troubleshooting<br />

guide for optimization of chromatin digestion.<br />

Add more DNA to the PCR reaction or increase the<br />

number of amplification cycles.<br />

Optimize the PCR conditions for experimental primer<br />

set using purified DNA from cross-linked and digested<br />

chromatin. Design a different primer set and decrease<br />

length of amplicon to less than 150 bp (see primer<br />

design recommendations in Protocol Section VIII).<br />

For optimal ChIP results, add 5–10 μg chromatin per IP.<br />

Be sure to add 5–10 μg of chromatin and 10 μl<br />

of antibody to each IP reaction and incubate with<br />

antibody overnight and an additional 2 hr after adding<br />

Protein G beads.<br />

Elution of chromatin from Protein G beads is optimal at<br />

65°C with frequent mixing to keep beads suspended<br />

in solution.<br />

For optimal ChIP results, add 5–10 µg of chromatin<br />

and 10 μl of histone H3 antibody to each IP reaction.<br />

Reduce the amount of normal rabbit IgG to 1 μl per IP.<br />

Add less DNA to the PCR reaction or decrease the<br />

number of PCR cycles. It is very important that<br />

the PCR products are analyzed within the linear<br />

amplification phase of PCR. Otherwise, the differences<br />

in quantities of starting DNA cannot be accurately<br />

measured. Alternatively, quantify immunoprecipitations<br />

using real-time quantitative PCR.<br />

Add more DNA to the PCR reaction or increase the<br />

number of amplification cycles.<br />

Typically a range of 1–5 μg of antibody are added to<br />

the IP reaction; however, the exact amount depends<br />

greatly on the individual antibody. Increase the amount<br />

of antibody added to the IP.<br />

Find an alternate antibody source.<br />

200 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

<strong>Guide</strong> to Successful ChIP<br />

Helpful tips and explanations to support your ChIP experiments are available in “A <strong>Guide</strong> to Successful Chromatin IP”.<br />

Request a copy at www.cellsignal.com/chipsuccess<br />

www.cellsignal.com/cstprotocols<br />

201


Events<br />

10<br />

Section II: ANTIBODY APPLICATIONS<br />

chapter<br />

Phosphorylation<br />

of intracellular<br />

Stat5 (Tyr694)<br />

is stimulated by<br />

GM-CSF treatment.<br />

Phospho-Stat5 (Tyr694)<br />

Phospho-Stat5 (Tyr694) (D47E7) XP ®<br />

Rabbit mAb #4322: Flow cytometric<br />

analysis of TF-1 cells, untreated (blue)<br />

or treated with hGM-CSF #8922 (green),<br />

using #4322.<br />

Btk (D3H5) Rabbit mAb #8547:<br />

Human whole blood was fixed and<br />

permeabilized as outlined in the <strong>CST</strong><br />

Flow Alternate Protocol and stained<br />

using #8547. Forward scatter (FSC)<br />

and side scatter (SSC) were used to<br />

gate on lymphocyte cells (A). Samples<br />

were co-stained using CD3-PE and<br />

CD19-APC to distinguish T and B<br />

cell subpopulations (B). B (red) and<br />

T (blue) cell population gates were<br />

applied to a histogram depicting the<br />

mean fluorescence intensity of Btk (C).<br />

Flow Cytometry (F)<br />

Flow cytometry is a well-established technology allowing phenotypic, molecular, and functional characterization<br />

of single cells in suspension. The unique multi-parameter capability of this platform enables<br />

the simultaneous detection and quantification of multiple intracellular and extracellular targets in the<br />

same sample. Flow cytometry is highly sensitive and can be used to detect cellular proteins within<br />

distinct sub populations from heterogeneous samples. Traditionally used for the phenotypic analysis of<br />

normal and malignant blood cells, flow cytometry is now also routinely used to study complex intracellular<br />

signaling.<br />

Intracellular flow cytometry can be used to analyze signaling changes in response to stimuli within<br />

specific cell types and subpopulations. Cells are fixed and permeabilized to allow antibody penetration<br />

and subsequent binding to targets without disrupting cellular morphology. Several fixation and permeabilization<br />

protocols have been developed for target-specific antibodies to enable intracellular staining<br />

alone or in combination with surface markers, and to maximize signal-to-noise.<br />

There are numerous methodologies available to perform intracellular flow cytometry. Below we discuss<br />

several key processes that will help achieve experimental success, particularly when studying posttranslational<br />

modifications (PTMs).<br />

Flow Cytometry Tips for Success<br />

Appropriate fixation and permeabilization methods are<br />

crucial for successful detection of intracellular targets.<br />

Flow cytometric analysis of PTMs, such as phosphorylation, requires fixation of cells within a critical<br />

window of pathway activation. Fixation using 4% formaldehyde (methanol-free) optimally crosslinks<br />

intracellular proteins and any associated modifications. Furthermore, enzymatic activity responsible for<br />

removing modifications such as phosphatases can be inactivated. A lower concentration of formaldehyde<br />

may be necessary for some targets if the extent of crosslinking masks the target epitope.<br />

Permeabilization with ice-cold 90% methanol added dropwise maintains structural cell integrity while<br />

allowing efficient antibody penetration without denaturing PTM epitopes that have previously been<br />

aldehyde fixed (1).<br />

Protocol alterations enable simultaneous detection of<br />

intracellular and extracellular targets by flow cytometry.<br />

Ex vivo stimulation of whole blood allows for perturbation of peripheral blood cells within the blood<br />

matrix, resulting in a more physiologically relevant model of pathway signaling and other cellular<br />

processes. Fixation in 4% formaldehyde stabilizes protein interactions and subsequent treatment with<br />

0.1% Trition X-100 lyses red blood cells (RBCs) (2). This allows permeabilization with 50% methanol<br />

to preserve surface marker integrity, enabling the simultaneous analysis of both surface and intracellular<br />

targets.<br />

Btk, an intracellular tyrosine kinase, is selectively expressed in B cells.<br />

SSC-H<br />

1000<br />

800<br />

600<br />

400<br />

200<br />

A 10 B 4<br />

100<br />

C<br />

0 200 400 600 800 1000<br />

FSC-H<br />

202 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

CD3-PE<br />

10 3<br />

10 2<br />

10 1<br />

T cells<br />

B cells<br />

Events<br />

10 0<br />

10 0 10 1 10 2 10 3 10 4 10 0 10 1 10 2 10 3 10 4<br />

CD19-APC<br />

Btk<br />

80<br />

60<br />

40<br />

20<br />

B cells<br />

T cells<br />

Consider the impact of basal modification levels<br />

when assessing signal transduction events.<br />

It is always important to account for the complexity of cellular systems when designing an experiment.<br />

In some instances, basal modification levels may be high and therefore, stimulation of the pathway of<br />

interest does not provide any additional detectable signal, which can be interpreted as a false negative<br />

result. This event is common with phosphorylated proteins. To address this issue, the fixed and permeabilized<br />

sample can be treated with phosphatase to remove phosphate groups from proteins and then<br />

examined for decreased signal that indicates high basal phosphorylation levels. Endogenous phosphorylation<br />

levels in live cells can be brought down by serum starvation or by kinase inhibitor treatment. Conversely,<br />

transient phosphorylation events may be difficult to capture due to endogenous phosphatase<br />

activity, in which case treatment with a phosphatase inhibitor can help preserve phosphorylation signal.<br />

Phosphatase treatment reveals basal<br />

phosphorylation levels of S6 ribosomal protein.<br />

Side Scatter<br />

Intracellular flow cytometry can be used<br />

to detect and quantify cytoplasmic cytokines.<br />

Adding Brefeldin A to a cell sample during stimulation prevents secretion of cytokines by blocking intracellular<br />

vesicle trafficking. Thus, cytokines accumulate in the cells, allowing for their detection. In the<br />

example shown, <strong>CST</strong>’s standard flow cytometry protocol was used to immunostain cell surface markers<br />

in conjunction with an intracellular cytokine Interferon-γ. With this protocol, further multiplexing using<br />

antibodies against phosphorylated proteins would allow the simultaneous analysis of phenotype,<br />

protein activation and functional endpoint, on a single cell basis, within a single sample.<br />

Cytoplasmic Interferon-γ can be monitored with intracellular flow cytometry.<br />

IFN-γ (D3H2) XP ® Rabbit mAb #8455:<br />

Flow cytometric analysis of human<br />

peripheral blood cells, untreated (left) or<br />

treated (right) with TPA #4174 (40 nM,<br />

4 hr), Ionomycin #9995 (2 μM, 4 hr),<br />

and Brefeldin A #9972 (1 μg/ml, last 3<br />

hr of stimulation), using a CD3 antibody<br />

and #8455. Analysis was performed on<br />

cells in the lymphocyte gate. The CD3<br />

antibody (Y axis) was used to identify T<br />

cells. Note the induction of IFN-γ (X axis)<br />

in treated, CD3+ cells (right).<br />

A B C<br />

CD3<br />

Phospho-S6 Ribosomal Protein (Ser235/236)<br />

10 4<br />

10 3<br />

10 2<br />

10 1<br />

10 0<br />

10 0 10 1 10 2 10 3 10 4 10 0 10 1 10 2 10 3 10 4<br />

IFN-γ<br />

References:<br />

1. Krutzik, P.O. and Nolan, G.P. (2003) Cytometry A. 55, 61–70.<br />

2. Chow, S., Hedley, D., Grom, P., Magari, R., Jacobberger, J.W., and Shankey, T.V. (2005) Cytometry A. 67, 4–17.<br />

10: Flow Cytometry (F)<br />

Phospho-S6 Ribosomal Protein<br />

(Ser235/236) (D57.2.2E) XP ® Rabbit<br />

mAb (Alexa Fluor ® 488 Conjugate)<br />

#4803: Human whole blood was fixed<br />

and permeabilized as outlined in the <strong>CST</strong><br />

Flow Alternate Protocol, and untreated<br />

(A), treated with λ phosphatase (B), or<br />

treated with TPA #4174 (C), before being<br />

stained using #4803.<br />

Alexa Fluor®<br />

Conjugated<br />

Antibodies<br />

For the most up-to-date list of Alexa<br />

Fluor ® conjugates that are optimized<br />

for flow cytometry, please go to<br />

www.cellsignal.com/alexafluor<br />

www.cellsignal.com/cstflow<br />

203


Section II: ANTIBODY APPLICATIONS<br />

chapter 10: Flow Cytometry (F)<br />

PE conjugated<br />

antibodies deliver<br />

robust Stoke’s<br />

shifts and are ideal<br />

for multiplexing.<br />

Events<br />

Phospho-Akt (Ser473)<br />

Anti-rabbit IgG (H+L), F(ab’) 2<br />

Fragment (PE Conjugate) #8885:<br />

Flow cytometric analysis of Jurkat<br />

cells, untreated (green) or treated<br />

with LY294002 #9901, Wortmannin<br />

#9951, and U0126 #9903 (blue), using<br />

Phospho-Akt (Ser473) (D9E) XP ® Rabbit<br />

mAb #4060 detected with #8885.<br />

Flow Cytometry General Protocol<br />

A. Solutions and Reagents<br />

NOTE: Prepare solutions with reverse osmosis deionized or equivalent grade water (dH 2 0).<br />

1. 20X Phosphate Buffered Saline (PBS): (#9808) To prepare 1 L 1X PBS: add 50 ml 20X PBS to 950 ml dH 2 O, mix.<br />

2. 16% Formaldehyde (methanol free)<br />

3. 100% Methanol<br />

4. Incubation Buffer: Dissolve 0.5 g Bovine Serum Albumin (BSA) (#9998) in 100 ml 1X PBS. Store at 4°C.<br />

5. Secondary Antibodies: Anti-mouse (#4408, #8890, #4410, #8887), Anti-rabbit (#4412, #8889, #4414, #8885), Anti-rat<br />

(#4416, #4418)<br />

B. Fixation<br />

1. Collect cells by centrifugation and aspirate supernatant.<br />

2. Resuspend cells in 0.5–1 ml 1X PBS. Add appropriate volume of formaldehyde to obtain a final concentration of<br />

4% (e.g. 750 µl 1X PBS + 250 µl 16% formaldehyde).<br />

3. Fix for 10 min in a 37°C water bath.<br />

4. Chill tubes on ice for 1 min.<br />

5. For extracellular staining with antibodies that do not require permeabilization, proceed to immunostaining (Section D)<br />

or store cells in 1X PBS with 0.1% sodium azide at 4°C; for intracellular staining, proceed to permeabilization (Section C).<br />

C. Permeabilization<br />

NOTE: This step is critical for many <strong>CST</strong> antibodies.<br />

1. Add ice-cold 100% methanol drop-wise to pre-chilled cells, while gently vortexing, to a final concentration of 90% methanol.<br />

Alternatively, remove fix prior to permeabilization by centrifugation and resuspend in 90% methanol as described above.<br />

2. Permeabilize for a minimum of 10 min on ice.<br />

3. Proceed with Immunostaining (Section D) or store cells at -20°C in 90% methanol.<br />

B. Preparation of Whole Blood (fixation, lysis, and permeabilization) for Immunostaining<br />

1. Aliquot 100 μl fresh whole blood per assay tube.<br />

2. OPTIONAL: Place tubes in rack in 37°C water bath for short-term treatments with ligands, inhibitors, drugs, etc.<br />

3. Add 65 μl of 10% formaldehyde to each tube.<br />

4. Vortex briefly and let stand for 15 min at room temperature.<br />

5. Add 1 ml of 0.1% Triton X-100 to each tube.<br />

6. Vortex and let stand for 30 min at room temperature.<br />

7. Add 1 ml incubation buffer.<br />

8. Pellet cells by centrifugation and aspirate supernatant.<br />

9. Repeat Steps 7 and 8.<br />

10. Resuspend cells in ice-cold 50% methanol in PBS (store methanol solution at -20°C until use).<br />

11. Incubate at least 10 min on ice.<br />

12. Proceed with staining or store cells at -20°C in 50% methanol.<br />

C. Staining Using Unlabeled Primary and Conjugated Secondary Antibodies<br />

NOTE: Account for isotype-matched controls for monoclonal antibodies or species matched IgG for polyclonal antibodies.<br />

1. Add 2–3 ml incubation buffer to each tube and wash by centrifugation. Repeat.<br />

2. Add primary antibodies diluted as recommended on datasheet or product webpage in incubation buffer.<br />

3. Incubate for 30–60 min at room temperature.<br />

4. Wash by centrifugation in 2–3 ml incubation buffer.<br />

5. Resuspend cells in fluorochrome-conjugated secondary antibody diluted in incubation buffer according to the manufacturer’s<br />

recommendations.<br />

6. Incubate for 30 min at room temperature.<br />

7. Wash by centrifugation in 2–3 ml incubation buffer.<br />

8. Resuspend cells in 0.5 ml PBS and analyze on flow cytometer.<br />

Cyclin B1<br />

Protocol Reference: Chow, S.,<br />

Hedley, D., Grom, P., Magari, R.,<br />

Jacobberger, J.W., and Shankey,<br />

T.V. (2005) Whole blood fixation<br />

and permeabilization protocol<br />

with red blood cell lysis for flow<br />

cytometry of intracellular phosphorylated<br />

epitopes in leukocyte<br />

subpopulations. Cytometry A.<br />

67, 4–17.<br />

PI/RNase Staining<br />

Solution labels DNA<br />

content without RNA<br />

crossreactivity.<br />

10 4<br />

10 3<br />

10 2<br />

10 1<br />

R-Phycoerythrin<br />

(PE) Conjugates<br />

For the most up-to-date list of ready<br />

to use flow validated antibodies conjugated<br />

to PE, eliminating the time and<br />

costs associated with PE conjugation<br />

and subsequent validation, go to<br />

www.cellsignal.com/peconjugates<br />

D. Immunostaining<br />

1. Aliquot 0.5–1x10 6 cells into each assay tube (by volume).<br />

2. Add 2 ml incubation buffer to each tube and wash by centrifugation. Repeat.<br />

3. Resuspend cells in 100 µl of diluted primary antibody prepared in incubation buffer at the recommended dilution.<br />

See individual antibody datasheet or product webpage for the appropriate dilution.<br />

4. Incubate for 1 hr at room temperature.<br />

5. Wash by centrifugation in 2 ml incubation buffer.<br />

6. If using a fluorochrome-conjugated primary antibody, resuspend cells in 0.5 ml 1X PBS and analyze on flow cytometer;<br />

for unconjugated or biotinylated primary antibodies, proceed to immunostaining (Step 9).<br />

7. Resuspend cells in fluorochrome-conjugated secondary antibody or fluorochrome-conjugated avidin, diluted in incubation<br />

buffer at the recommended dilution.<br />

8. Incubate for 30 min at room temperature.<br />

9. Wash by centrifugation in 2 ml incubation buffer.<br />

10. Resuspend cells in 0.5 ml 1X PBS and analyze on flow cytometer; alternatively, for DNA staining, proceed to optional DNA<br />

dye (Section E).<br />

E. Optional DNA Dye<br />

1. Resuspend cells in 0.5 ml of DNA dye (e.g. Propidium Iodide (PI)/RNase Staining Solution #4087).<br />

2. Incubate for at least 30 min at room temperature.<br />

3. Analyze cells in DNA staining solution on flow cytometer.<br />

Flow Cytometry Alternate Protocol<br />

(for combined staining of intracellular proteins and cell surface markers in blood)<br />

A. Solutions and Reagents<br />

NOTE: Prepare solutions with reverse osmosis deionized or equivalent grade water.<br />

1. 20X Phosphate Buffered Saline (PBS): (#9808) To prepare 1 L 1X PBS: add 50 ml 20X PBS to 950 ml dH 2<br />

O, mix.<br />

2. 16% Formaldehyde (methanol free)<br />

3. Triton X-100: To prepare 50 ml of 0.1% Triton X-100 add 50 µl Triton X-100 to 50 ml 1X PBS and mix well.<br />

4. 50% methanol<br />

5. Incubation Buffer: Dissolve 0.5 g Bovine Serum Albumin (BSA) (#9998) in 100 ml 1X PBS. Store at 4°C.<br />

6. Secondary Antibodies: Anti-mouse (#4408, #8890, #4410, #8887), Anti-rabbit (#4412, #8889, #4414, #8885), Anti-rat<br />

(#4416, #4418)<br />

Fluorochrome<br />

Reference Table<br />

Max. excitation<br />

wavelength (nm)<br />

Max. emission<br />

wavelength (nm)<br />

Excitation<br />

laser line (nm)<br />

Color<br />

Brilliant Violet 421 407 421 405 violet<br />

DAPI 345 455 360/405 blue<br />

Pacific Blue 404 456 360/407 blue<br />

Hoechst 355 465 360 blue<br />

Alexa Fluor ® 488 495 519 488 green<br />

FITC 495 519 488 green<br />

Pacific Orange 403 551 360/407 orange<br />

Cy 3 548 561 488/532/561 orange<br />

Alexa Fluor ® 555 555 565 488/532/561 orange<br />

Phycoerythrin (PE) 496/566 576 488/532/561 orange<br />

Texas Red ® 595 613 568/595 red<br />

PE-Texas Red ® 496/566 616 488/532 red<br />

Alexa Fluor ® 594 590 617 561/595 red<br />

Propidium iodide (PI) 536 617 488/532 red<br />

7-AAD 546 647 488/532 red<br />

Allophycocyanin (APC) 650 660 633/635 red<br />

Alexa Fluor ® 647 650 665 633/635 red<br />

Cy 5 647 665 633/635 red<br />

PE-Cy 5 496/546 666 488/532/561 red<br />

PerCP 482 678 488/532 red<br />

DRAQ7 599/644 678/694 635/647 red<br />

DRAQ5 ® 647 681/697 488/633/647 red<br />

PE-Cy 5.5 565 693 488 red<br />

APC-Cy 5.5 650 694 633/635/647 red<br />

PerCP-Cy ® 5.5 482 695 488 red<br />

DyLight 680 692 712 680/685 red<br />

Alexa Fluor ® 700 696 719 633/635 near-IR<br />

APC-Cy 7 650 785 633/635 near-IR<br />

PE-Cy 7 496/566 785 488/532/561 near-IR<br />

DyLight 800 777 794 785 near-IR<br />

Alexa Fluor ® 790 784 814 785 near-IR<br />

10 0 0 200 400 600 800 1000<br />

DNA (PI)<br />

Propidium Iodide (PI)/RNase Staining<br />

Solution #4087: Flow cytometric analysis<br />

of Jurkat cells using Cyclin B1 (D5C10)<br />

XP ® #12231 Rabbit mAb and #4087 (DNA<br />

content). Anti-rabbit IgG (H+L), F(ab’) 2<br />

Fragment (Alexa Fluor ® 488 Conjugate)<br />

#4412 was used as a secondary antibody.<br />

204 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstprotocols<br />

205


11<br />

Section II: ANTIBODY APPLICATIONS<br />

Immunofluorescence (IF)<br />

Fluorescent analysis of cells and tissues<br />

Immunofluorescence (IF) is a detection method that employs primary antibodies to reveal the localization<br />

of target proteins and analyze their relative expression levels in cultured cells or tissue sections. Primary<br />

antibodies can be directly labeled with a fluorescent tag (direct IF) or detected using a fluorescently<br />

labeled secondary antibody (indirect IF). The ability to detect multiple fluorescent tags as distinct colors<br />

within the same sample makes IF a powerful tool for investigating changes in subcellular localization,<br />

because protein location can be analyzed in relation to other labeled organelle markers or other cellular<br />

structures.<br />

Evaluate fixation and permeabilization<br />

protocols to achieve optimal staining.<br />

Cell and tissue samples may be preserved using either crosslinking or denaturing fixatives. (Note: not all<br />

samples are fixed for IF staining, such as in live cell staining.) The optimal fixation method is dependent<br />

upon the target protein, its subcellular localization, and in some cases the antibody that is used. Intracellular<br />

targets also require an additional permeabilization step. Detergents, such as Triton X-100 or Tween ®<br />

20 work well for most antibodies but in some cases, methanol permeabilization may be necessary.<br />

Optimizing fixation reagents can substantially improve staining results.<br />

Formaldehyde<br />

Methanol<br />

chapter 11: Immunofluorescence (IF)<br />

Methanol<br />

permeabilization<br />

improves intracellular<br />

immunostaining for<br />

some proteins.<br />

Triton X-100<br />

A number of factors can impact the ability<br />

to achieve bright and specific IF staining.<br />

• Antibody specificity and sensitivity • Target expression level • Species considerations<br />

• Permeabilization agent used • Type and degree of fixation • Dye selection<br />

• Antibody concentration and diluent • Cellular location of target<br />

• Detection method (direct, indirect, amplified)<br />

Keratin 8/18 (C51) Mouse mAb<br />

#4546: IF analysis of HeLa cells,<br />

fixed with formaldehyde (left) or<br />

methanol (right), using #4546<br />

(green). Red = Propidium Iodide<br />

(PI)/RNase Staining Solution #4087.<br />

Best with methanol fixation<br />

Methanol<br />

Nanog (D2A3) XP ® Rabbit mAb<br />

(Mouse Specific) #8822: Confocal<br />

IF analysis of F9 cells (Nanog positive)<br />

(A) and NIH/3T3 cells (Nanog negative)<br />

(B) was performed using #8822<br />

and competitor antibodies at the<br />

indicated concentrations.<br />

IF Tips for Success<br />

Use a well-characterized and specific<br />

antibody for accurate IF results.<br />

The primary antibody is an important reagent in successful IF experiments, because the specificity and<br />

sensitivity of the antibody dictates the quality of experimental data.<br />

Specificity of Nanog antibody is demonstrated by positive staining restricted<br />

to the nucleus and by absence of staining in a Nanog-negative cell line.<br />

A<br />

B<br />

<strong>CST</strong> #8822<br />

(0.25 µg/ml)<br />

Demonstrates specific nuclear<br />

staining in Nanog-expressing cells<br />

Crosslinking Fixatives<br />

Examples: formaldehyde, formalin, glutaraldehyde, glyoxal<br />

Note: Degree of crosslinking is dependent upon fixative<br />

concentration, temperature, and time of fixation.<br />

Advantages<br />

• Excellent tissue morphology<br />

AIF (D39D2) XP ® Rabbit mAb<br />

#5318: IF analysis of HeLa cells,<br />

fixed with formaldehyde (left) or<br />

methanol (right), using #5318<br />

(green). Red = Propidium Iodide<br />

(PI)/RNase Staining Solution #4087.<br />

Best with formaldehyde fixation<br />

• Inactivate enzymes such as proteases and phosphatases<br />

Limitations<br />

• Toxicity risk to user<br />

• Old formaldehyde solutions may induce autofluorescence<br />

PDI Antibody #2446: Confocal IF analysis<br />

of NIH/3T3 cells, permeabilized with<br />

0.3% Triton X-100 (top) or methanol<br />

(bottom), using #2446 (green) and<br />

β-Actin (8H10D10) Mouse mAb #3700<br />

(red). Blue pseudocolor = DRAQ5 ®<br />

#4084 (fluorescent DNA dye).<br />

Competitor antibodies used in accordance with manufacturer recommendations.<br />

Competitor 1<br />

(5.0 µg/ml)<br />

Shows cytoplasmic and nuclear staining<br />

in both positive and negative cell lines<br />

Competitor 2<br />

(2.0 µg/ml)<br />

Demonstrates some staining in the<br />

negative control and requires an<br />

8-fold higher IgG concentration<br />

Denaturing/Precipitating Fixatives<br />

Examples: ethanol, methanol, acetone, acetic acid<br />

Note: Denaturing fixatives may be combined with acetic<br />

acid to enhance tissue penetration.<br />

Advantages<br />

• Loss of soluble proteins can allow presentation of antigens normally inaccessible to antibodies<br />

• Work well with DNA stains<br />

• Do not require further permeabilization<br />

Limitations<br />

• May induce cell swelling or lysis, if not used cold<br />

• Loss of tertiary structure<br />

• Not advisable for use with CD markers<br />

• Protein loss may occur because of lack of crosslinking<br />

Permeabilization Agents<br />

For use after crosslinking fixatives, allows antibodies to penetrate cells and bind to intracellular antigens<br />

Detergent Examples: Triton X-100, Tween ® , Saponin,<br />

DOTMAC, SDS<br />

Alcohol Examples: ethanol, methanol<br />

Note: Alcohol permeabilization after fixation is not the<br />

same as alcohol fixation.<br />

• Typically not used with activation state-specific antibodies such as phospho-specific antibodies<br />

Detergent<br />

• Detergent permeabilization may require a separate step or may be included in blocking solution<br />

and antibody diluent (depending on the detergent used).<br />

Alcohol<br />

• Alcohol permeabilization may be required for some proteins.<br />

206 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstif<br />

207


Section II: ANTIBODY APPLICATIONS<br />

chapter 11: Immunofluorescence (IF)<br />

Perform a titration to determine<br />

optimal antibody concentration.<br />

It is important to use an antibody at its optimal concentration. Using a lower concentration (more<br />

dilute) will diminish positive signal, but just as importantly, using higher concentration (less dilute) will<br />

increase background and decrease the signal-to-noise ratio. <strong>CST</strong> scientists routinely perform titrations<br />

using positive and negative cell lines to identify the concentration that gives optimal signal with minimal<br />

background staining. This recommended antibody dilution information is determined and provided with<br />

every lot of an IF-validated antibody.<br />

Antibody titration curves determine optimal concentration<br />

for maximal signal-to-noise in IF results.<br />

A<br />

Mean Fluorescence Intensity<br />

1200<br />

1000<br />

800<br />

600<br />

400<br />

200<br />

Optimal concentration for highest<br />

signal-to-noise with minimal background.<br />

50.0<br />

45.0<br />

40.0<br />

35.0<br />

30.0<br />

25.0<br />

20.0<br />

15.0<br />

10.0<br />

5.0<br />

Signal-to-Noise<br />

Choose a detection method that suits your<br />

target protein abundance and sample type.<br />

Conjugated primary antibodies (direct detection) offer convenience and a shorter protocol. Secondary<br />

antibodies conjugated to fluorochromes (indirect detection) may provide stronger signal intensity<br />

because multiple secondary antibodies can bind each primary antibody, but require using primary<br />

antibodies from different species when examining multiple targets within the same sample. Depending<br />

on the target expression level and sample type, direct and indirect IF may still be too dim for some<br />

assays, necessitating further amplification, such as avidin/biotin or tyramide. Avidin/biotin can improve<br />

signal intensity, but it may not be possible to completely block all nonspecific signal from endogenous<br />

biotin. More dramatic signal amplification can be achieved using tyramide amplification, which utilizes<br />

HRP-conjugated secondary antibodies to catalyze the deposition of fluorochrome-conjugated tyramide<br />

around the target. Tyramide amplification is very useful when performing IF in formalin-fixed paraffinembedded<br />

(FFPE) tissues. Signal amplification helps distinguish true signal from autofluorescence and<br />

negates issues related to antigen quality or scarcity, from either unmasking or protein loss. Tyramide<br />

also enables multiplex staining with antibodies from the same species (1).<br />

Retina stained with two directly conjugated<br />

antibodies from the same species<br />

Synapsin-1 (D12G5) XP ® Rabbit mAb (Alexa Fluor ® 594 Conjugate) #13556:<br />

Confocal IF analysis of rat retina using #13556 (red) and Neurofilament-L (C28E10)<br />

Rabbit mAb (Alexa Fluor ® 488 Conjugate) #8024 (green). Blue pseudocolor =<br />

DRAQ5 ® #4084 (fluorescent DNA dye).<br />

0.25 0.5 0.75 1 1.25 1.5 1.75 2 2.25<br />

Concentration (µg/ml)<br />

MFI (+) MFI (–) Signal-to-Noise<br />

B<br />

Dye-conjugated<br />

secondary antibodies<br />

can improve signal<br />

intensity for low<br />

abundance protein<br />

targets.<br />

Neurofilament-L (C28E10) Rabbit<br />

mAb #2837: Confocal IF analysis of<br />

neuroepithelial clusters differentiated<br />

from human iPS cells, using #2837 detected<br />

with Anti-rabbit IgG (H+L), F(ab’) 2<br />

Fragment (Alexa Fluor ® 555 Conjugate)<br />

#4413 (blue) and β3-Tubulin (TU-20)<br />

Mouse mAb #4466 detected with<br />

Anti-mouse IgG (H+L), F(ab’) 2 Fragment<br />

(Alexa Fluor ® 488 Conjugate) #4408<br />

(red). Blue pseudocolor = DRAQ5 ®<br />

#4084 (fluorescent DNA dye).<br />

C<br />

Amplification enables detection of low abundance<br />

protein targets in paraffin-embedded tissue.<br />

0.031 µg/ml 0.063 µg/ml 0.125 µg/ml 0.25 µg/ml 1 µg/ml<br />

MUC1 (D9O8K) XP ® Rabbit mAb #14161: Graph depicting Mean Fluorescence Intensity (MFI) of ZR-75 cells (MUC1 expressing) and<br />

HCT 116 cells (MUC1 negative) using #14161, and calculated signal-to-noise (A). IF analysis of ZR-75 cells (B) and HCT 116 cells (C) at<br />

varying concentrations, as indicated, using #14161. Red = Propidium Iodide (PI)/RNase Staining Solution #4087.<br />

A<br />

B<br />

C<br />

E-Cadherin (24E10) Rabbit mAb<br />

#3195: IF analysis of FFPE human<br />

metastatic lymph node using #3195<br />

detected with a conjugated secondary<br />

antibody (A,B) or detection with Antirabbit<br />

IgG, HRP-linked Antibody #7074<br />

and a FITC-tyramide conjugate (C).<br />

Tissue Autofluorescence<br />

Low Exposure<br />

High Exposure<br />

Low Exposure<br />

Organelle Marker Samplers<br />

Organelle Marker Samplers provide a convenient collection of primary antibodies targeting well-established<br />

organelle associated proteins. For the most up-to-date listing of Organelle Marker Samplers, please go to<br />

www.cellsignal.com/organelles<br />

References:<br />

1. Toth, Z.E. and Mezey, E. (2007) J. Histochem. Cytochem. 55, 545–554.<br />

208 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstif<br />

209


Section II: ANTIBODY APPLICATIONS<br />

Cultured Cells (Immunocytochemistry, IF-IC) Protocol<br />

IMPORTANT: Please refer to the Applications section on the front page of the product’s datasheet or product webpage to<br />

determine if this product is validated and approved for use on cultured cell lines (IF-IC), frozen tissue sections (IF-F), or paraffinembedded<br />

samples (IF-P). Some of <strong>CST</strong>’s antibodies perform optimally using an alternate protocol. Please consult the datasheet<br />

or webpage for product-specific recommendations.<br />

A. Solutions and Reagents<br />

NOTE: Prepare solutions with reverse osmosis deionized (RODI) or equivalent grade water.<br />

1. 20X Phosphate Buffered Saline (PBS): (#9808) To prepare 1 L 1X PBS: add 50 ml 20X PBS to 950 ml dH 2 O, mix.<br />

NOTE: Adjust pH to 8.0.<br />

2. Formaldehyde, 16%, methanol free: (Polysciences, Inc., cat #18814), use fresh, store opened vials at 4°C in dark.<br />

Dilute 1 in 4 in 1X PBS to make a 4% formaldehyde solution.<br />

3. Blocking Buffer: (1X PBS/5% normal serum/0.3% Triton X-100): To prepare 10 ml, add 0.5 ml normal serum from the<br />

same species as the secondary antibody (e.g., Normal Goat Serum (#5425) and 0.5 ml 20X PBS to 9 ml dH 2 O) and mix<br />

well. While stirring, add 30 µl Triton X-100.<br />

4. Antibody Dilution Buffer: (1X PBS/1% BSA/0.3% Triton X-100): To prepare 10 ml, add 30 µl Triton X-100 to 10 ml<br />

1X PBS. Mix well then add 0.1g BSA (#9998), mix until well dissolved.<br />

5. Fluorochrome-conjugated Secondary Antibodies: (Anti-mouse #4408, #4409, #8890, #4410) (Anti-rabbit #4412, #4413,<br />

#8889, #4414) (Anti-rat #4416, #4417, #4418)<br />

6. Mounting Medium: ProLong ® Gold Antifade Reagent (#9071) or ProLong ® Gold Antifade Reagent with DAPI (#8961)<br />

B. Specimen Preparation<br />

I. Cultured Cell Lines (IF-IC)<br />

NOTE: Cells should be grown, treated, fixed and stained directly in multi-well plates, chamber slides or on coverslips.<br />

1. Aspirate liquid, then cover cells to a depth of 2–3 mm with 4% formaldehyde diluted in 1X PBS.<br />

NOTE: Formaldehyde is toxic, use only in a fume hood.<br />

2. Allow cells to fix for 15 min at room temperature.<br />

3. Aspirate fixative, rinse three times in 1X PBS for 5 min each.<br />

4. Proceed with Immunostaining (Section C).<br />

C. Immunostaining<br />

NOTE: All subsequent incubations should be carried out at room temperature unless otherwise noted in a humid light-tight box or<br />

covered dish/plate to prevent drying and fluorochrome fading.<br />

1. Block specimen in blocking buffer for 60 min.<br />

2. While blocking, prepare primary antibody by diluting as indicated on the product’s datasheet or webpage in antibody dilution<br />

buffer.<br />

3. Aspirate blocking solution, apply diluted primary antibody.<br />

4. Incubate overnight at 4°C.<br />

5. Rinse 3 times in 1X PBS for 5 min each. NOTE: If using a fluorochrome-conjugated primary antibody, proceed directly to<br />

Step 8.<br />

6. Incubate specimen in fluorochrome-conjugated secondary antibody diluted in antibody dilution buffer for 1–2 hr at room<br />

temperature in the dark.<br />

7. Rinse 3 times in 1X PBS for 5 min each.<br />

8. Coverslip slides with ProLong ® Gold Antifade Reagent (#9071) or ProLong ® Gold Antifade Reagent with DAPI (#8961).<br />

9. For best results, allow mountant to cure overnight at room temperature. For long-term storage, store slides flat at 4°C<br />

protected from light.<br />

Frozen/Cryostat Tissue Sections (IF-F) Protocol<br />

IMPORTANT: Please refer to the Applications section on the front page of the product’s datasheet to determine whether an<br />

antibody is validated and approved for use on cultured cell lines (IF-IC), frozen tissue sections (IF-F), or paraffin-embedded<br />

samples (IF-P). Some of <strong>CST</strong>’s antibodies perform optimally using an alternate protocol. Please consult the datasheet or webpage<br />

for product-specific recommendations.<br />

A. Solutions and Reagents<br />

NOTE: Prepare solutions with reverse osmosis deionized (RODI) or equivalent grade water.<br />

1. 20X Phosphate Buffered Saline (PBS): (#9808) To prepare 1 L 1X PBS, add 50 ml 20X PBS to 950 ml dH 2 O, mix.<br />

NOTE: Adjust pH to 8.0.<br />

2. Formaldehyde: 16%, methanol free: (Polysciences, Inc., cat #18814) Use fresh and store opened vials at 4°C in dark.<br />

Dilute in 1X PBS to make a 4% formaldehyde solution.<br />

3. Blocking Buffer (1X PBS/5% normal serum/0.3% Triton X-100): To prepare 10 ml, add 0.5 ml normal serum from<br />

the same species as the secondary antibody (e.g., Normal Goat Serum (#5425) and 0.5 ml 20X PBS to 9 ml dH 2 O and mix<br />

well. While stirring, add 30 µL Triton X-100.<br />

4. Antibody dilution buffer (1X PBS/1% BSA/0.3% Triton X-100): To prepare 10 ml, add 30 µL Triton X-100 to 10 ml<br />

1X PBS. Add 0.1 g BSA (#9998) and mix well until dissolved.<br />

5. Fluorochrome-conjugated Secondary Antibodies: Anti-mouse (#4408, #4409, #8890, #4410), Anti-rabbit (#4412,<br />

#4413, #8889, #4414), Anti-rat (#4416, #4417, #4418)<br />

6. Mounting Medium: ProLong ® Gold Antifade Reagent (#9071) or ProLong ® Gold Antifade Reagent with DAPI (#8961)<br />

B. Specimen Preparation<br />

1. For fixed frozen tissue, allow sections to air dry on the bench for 10 min, then proceed with Immunostaining (Section C).<br />

2. For fresh, unfixed frozen tissue, fix immediately as follows:<br />

a. Cover sections to a depth of 2–3 mm with 4% formaldehyde diluted in 1X PBS. NOTE: Formaldehyde is toxic, use only<br />

in a fume hood.<br />

b. Allow sections to fix for 15 min at room temperature.<br />

c. Aspirate liquid, rinse slides 3 times in 1X PBS for 5 min each.<br />

d. Proceed with Immunostaining (Section C).<br />

C. Immunostaining<br />

NOTE: All subsequent incubations should be carried out at room temperature unless otherwise noted in a humid light-tight box<br />

or covered dish/plate to prevent drying and fluorochrome fading.<br />

1. Block specimen in Blocking Buffer for 60 min.<br />

2. While blocking, prepare primary antibody by diluting as indicated on the product’s datasheet or webpage in antibody dilution<br />

buffer.<br />

3. Aspirate Blocking Buffer, then apply diluted primary antibody.<br />

4. Incubate overnight at 4°C.<br />

5. Rinse 3 times in 1X PBS for 5 min each. NOTE: If using a fluorochrome-conjugated primary antibody, proceed directly to<br />

Step 8.<br />

6. Incubate specimen in fluorochrome-conjugated secondary antibody diluted in antibody dilution buffer for 1 hr in the dark.<br />

7. Rinse 3 times in 1X PBS for 5 min each.<br />

8. Mount slides with ProLong ® Gold Antifade Reagent (#9071) or ProLong ® Gold Antifade Reagent with DAPI (#8961).<br />

9. For best results, allow mountant to cure overnight at room temperature. For long-term storage, store slides flat at 4°C<br />

protected from light.<br />

chapter 11: Immunofluorescence (IF)<br />

Phalloidin labels actin filaments<br />

and DRAQ5 ® labels DNA.<br />

Alexa Fluor ® 555 Phalloidin #8953: Confocal IF analysis of HeLa cells using COX IV<br />

(3E11) Rabbit mAb (Alexa Fluor ® 488 Conjugate) #4853 (green). Actin filaments were<br />

labeled with #8953 (red). Blue pseudocolor = DRAQ5 ® #4084 (fluorescent DNA dye).<br />

ProLong ® Gold Antifade Reagent<br />

extends fluorescent signal, allowing<br />

more time to capture images.<br />

ProLong ® Gold Antifade Reagent #9071: Confocal IF analysis of SNB19 cells using<br />

β–Actin (8H10D10) Mouse mAb #3700 (red), Vimentin (D21H3) XP ® Rabbit mAb (Alexa<br />

Fluor ® 488 Conjugate) #9854 (green), and COX IV (3E11) Rabbit mAb (Alexa Fluor ® 647<br />

Conjugate) #7561 (blue).<br />

210 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstprotocols<br />

211


Section II: ANTIBODY APPLICATIONS<br />

Paraffin Tissue Sections (IF-P) Protocol<br />

IMPORTANT: Please refer to the Applications section on the front page of the product’s datasheet to determine whether an antibody<br />

is validated and approved for use on cultured cell lines (IF-IC), frozen tissue sections (IF-F), or paraffin-embedded samples<br />

(IF-P). Some of <strong>CST</strong>’s antibodies perform optimally using an alternate protocol. Please consult the datasheet or webpage for<br />

product-specific recommendations.<br />

A. Solutions and Reagents<br />

NOTE: Prepare solutions with reverse osmosis deionized or equivalent grade water (dH 2 O).<br />

1. Xylene<br />

2. Ethanol, anhydrous denatured, histological grade (100% and 95%)<br />

3. Antigen Unmasking:<br />

a. 10 mM Sodium Citrate Buffer: To prepare 1 L add 2.94 g sodium citrate trisodium salt dihydrate (C 6 H 5 Na 3 O 7 •2H 2 O)<br />

to 1 L dH 2 O. Adjust pH to 6.0.<br />

b. 1 mM EDTA: To prepare 1 L add 0.372 g EDTA (C 10 H 14 N 2 O 8 Na 2 • 2 H 2 O) to 1 L dH 2 O. Adjust pH to 8.0.<br />

4. 10X Tris Buffered Saline with Tween ® 20 (TBST): (#9997) To prepare 1 L 1X TBST, add 100 ml 10X TBST to 900 ml<br />

dH 2 O, mix.<br />

5. Incubation Buffer (1X TBST/5% normal serum): To prepare 10 ml, add 0.5 ml normal serum from the same species as<br />

the secondary antibody (e.g., Normal Goat Serum (#5425) to 9.5 ml of 1X TBST, mix well.<br />

6. Fluorochrome-conjugated Secondary Antibodies: Anti-mouse (#4408, #4409, #8890, #4410), Anti-rabbit (#4412,<br />

#4413, #8889, #4414), Anti-rat (#4416, #4417, #4418)<br />

7. Mounting Medium: ProLong ® Gold Antifade Reagent (#9071) or ProLong ® Gold Antifade Reagent with DAPI (#8961)<br />

B. Deparaffinization/Rehydration<br />

NOTE: Do not allow slides to dry at any time during this procedure.<br />

1. Incubate sections in 3 washes of xylene for 5 min each.<br />

2. Incubate sections in 2 washes of 100% ethanol for 10 min each.<br />

3. Incubate sections in 2 washes of 95% ethanol for 10 min each.<br />

4. Wash sections 2 times in dH 2 O for 5 min each.<br />

C. Antigen Unmasking<br />

NOTE: Consult product datasheet for specific recommendation for the unmasking solution/protocol.<br />

1. For Citrate: Bring slides to a boil in 10 mM Sodium Citrate Buffer, pH 6.0; maintain at a sub-boiling temperature (95–98°C)<br />

for 10 min. Cool slides on bench top for 30 min.<br />

2. For EDTA: Bring slides to a boil in 1 mM EDTA, pH 8.0. Follow with 15 min at a sub-boiling temperature (95–98°C). No<br />

cooling is necessary.<br />

D. Immunostaining<br />

NOTE: All subsequent incubations should be carried out at room temperature unless otherwise noted in a humid light-tight box<br />

or covered dish/plate to prevent drying and fluorochrome fading.<br />

1. Wash sections 3 times in 1X TBST for 5 min each.<br />

2. Block specimen in Incubation Buffer for 60 min.<br />

3. While blocking, prepare primary antibody by diluting as indicated on the product’s datasheet or webpage in Incubation Buffer.<br />

4. Aspirate buffer from sections; apply diluted primary antibody.<br />

5. Incubate overnight at 4°C.<br />

6. Wash 3 times in 1X TBST for 5 min each. NOTE: If using a fluorochrome-conjugated primary antibody, proceed directly to<br />

Step 9.<br />

7. Incubate specimen in fluorochrome-conjugated secondary antibody diluted in Incubation Buffer for 1 hr at room temperature<br />

in the dark.<br />

8. Rinse 3 times in 1X TBST for 5 min each.<br />

9. Mount slides with ProLong ® Gold Antifade Reagent (#9071) or ProLong ® Gold Antifade Reagent with DAPI (#8961).<br />

10. For best results, allow mountant to cure overnight at room temperature. For long-term storage, store slides flat at 4°C<br />

protected from light.<br />

In-Cell Western Protocol<br />

A. Solutions and Reagents<br />

NOTE: Prepare solutions with reverse osmosis or equivalently purified water (dH 2 O).<br />

1. 10X Phosphate Buffered Saline (PBS): To prepare 1 L 1X PBS add 80 g sodium chloride (NaCl), 2 g potassium chloride<br />

(KCl), 14.4 g sodium phosphate, dibasic (Na 2 HPO 4 ) and 2.4 g potassium phosphate, monobasic (KH 2 PO 4 ) to 1 L dH 2 O. Adjust<br />

pH to 7.4.<br />

2. Formaldehyde, 16%, methanol-free: (Polysciences, Inc. #18814) Use fresh; store opened vials at 4°C in dark, dilute in<br />

1X PBS for use.<br />

3. Blocking Buffer: To prepare 25 ml, add 2.5 ml 10X PBS, 1.25 ml normal serum from the same species as the secondary<br />

antibody (e.g., normal goat serum (#5425), normal donkey serum) and 21.25 ml dH 2 O and mix well. While stirring, add 75 µl<br />

Triton X-100 (100%).<br />

4. Antibody Dilution Buffer: To prepare 40 ml, add 4 ml 10X PBS to 36 ml dH 2 O, mix. Add 0.4 g BSA and mix well. While<br />

stirring, add 120 µl Triton X-100 (100%).<br />

5. Fluorochrome-conjugated secondary antibody.<br />

NOTE: When using any primary or fluorochrome-conjugated secondary antibody for the first time, titrate the antibody to<br />

determine which dilution allows for the strongest specific signal with the least background for your sample.<br />

B. Fixation and Immunolabeling<br />

NOTE: Please refer to the IF protocol for each antibody to identify the recommended antibody-specific fixation and permeabilization<br />

conditions. Cells should be grown, treated, fixed, and stained directly in multi-well plates. To avoid edge effects from uneven<br />

distribution of cells in individual wells, let newly-seeded plates stand at room temperature for one hour before placing in 37°C<br />

incubator (for more information, see Lundholt, B.K., Scudder, K.M., and Pagliaro, L. (2003) A simple technique for reducing<br />

edge effect in cell-based assays. J. Biomol. Screen. 8, 566–570). To minimize false signal from scattered light and background<br />

fluorescence, use black-walled multi-well plates (e.g., BD Falcon, #353948). Avoid touching the inside of the wells with pipettes<br />

or aspirators as this may leave an artifact that will affect subsequent quantification. Instead, empty wells by shaking inverted<br />

plate sharply over waste receptacle and then blotting with clean paper towel. Volumes indicated below are for use with standard<br />

96-well plates. Adjust volume accordingly for plates with larger or smaller wells.<br />

1. Grow and treat cells as desired in multi-well plates.<br />

2. Fix cells with 50 µl 4% formaldehyde or 100% methanol, as recommended in the antibody-specific IF protocol.<br />

3. Allow cells to fix for 15 min at room temperature.<br />

4. Shake inverted plate over aldehyde waste receptacle and blot with clean paper towel.<br />

5. Rinse plate 3 times for 5 min each with room temperature PBS (100 µl/well).<br />

6. Block cells in Blocking Buffer for one hour at room temperature (50 µl/well).<br />

7. While blocking, prepare primary antibody by diluting in Antibody Dilution Buffer as indicated for Immunofluorescence (IF-IC)<br />

on datasheet.<br />

8. Shake out blocking solution and add diluted primary antibody (total volume 50 µl/well).<br />

NOTE: For double-labeling, prepare a cocktail of mouse and rabbit primary antibodies at their appropriate dilutions in<br />

Antibody Dilution Buffer.<br />

9. Cover plate and incubate overnight at 4°C.<br />

10. Rinse plate 3 times in PBS for 5 min each.<br />

11. Add fluorochrome-conjugated secondary antibody* diluted in Antibody Dilution Buffer (total volume 50 µl/well) and incubate<br />

for 1 hr at room temperature in the dark.<br />

NOTE: For double-labeling, prepare a cocktail of fluorochrome-conjugated anti-mouse and anti-rabbit secondary antibodies<br />

in Antibody Dilution Buffer.<br />

12. Rinse plate 3 times in PBS for 5 min each.<br />

13. To normalize for cell number, label nuclei using DRAQ5 ® diluted 1:1000 in PBS (total volume 50 µl/well). Incubate at least<br />

30 min at room temperature in the dark prior to scanning.<br />

NOTE: DRAQ5 ® can be emptied and the wells rinsed at least 1X with PBS for better optical resolution.<br />

14. Scan plate according to manufacturer’s directions. (Be sure bottom of plate has been wiped clean prior to scanning).<br />

Recommended Secondary Antibodies: Anti-Rabbit: Anti-Rabbit IgG (H+L) (DyLight ® 680 Conjugate) #5366, Anti-Rabbit IgG<br />

(H+L) (DyLight ® 800 Conjugate) #5151, Anti-Mouse: Anti-Mouse IgG (H+L) (DyLight ® 680 Conjugate) #5470, Anti-Mouse IgG<br />

(H+L) (DyLight ® 800 Conjugate) #5257<br />

chapter 11: Immunofluorescence (IF)<br />

IF Companion Products<br />

See our comprehensive list of IF companion products that are validated in-house with our protocols so that you can<br />

get the reliable reagents you need to complete your experiment. www.cellsignal.com/ifcompanion<br />

212 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstprotocols<br />

213


12<br />

Section II: ANTIBODY APPLICATIONS<br />

chapter<br />

Antibodies targeting<br />

the same protein<br />

may have different<br />

preferred methods<br />

of antigen retrieval.<br />

EGF Receptor (D38B1) XP ® Rabbit<br />

mAb #4267: IHC analysis of paraffinembedded<br />

human lung carcinoma<br />

using #4267 and an EGFR mouse<br />

mAb after antigen retrieval by boiling<br />

in citrate buffer (left), boiling in EDTA<br />

buffer (center), or digestion with<br />

pepsin (right).<br />

For #4267, superior signal is<br />

obtained with EDTA retrieval.<br />

However, for the competitor’s EGFR<br />

mouse mAb, signal is only achieved<br />

with pepsin digestion.<br />

Immunohistochemistry (IHC)<br />

Tissue Analysis<br />

Immunohistochemistry (IHC) is a common technique for morphological characterization of tumors or other<br />

tissue malignancies. IHC uses antibodies to detect and analyze protein expression while maintaining<br />

the composition, cellular characteristics, and structure of native tissue. Starting with high quality tissue<br />

samples is important for successful IHC results, so it is best to use freshly cut sections. It is also essential<br />

to include appropriate controls to assess antibody performance. Prior to testing on tissue, antibody<br />

specificity can be evaluated at the cellular level using a variety of cell lines and treatment conditions.<br />

• Total protein specificity can be assessed through the use of positively and negatively expressing<br />

cell lines.<br />

• Cells can be treated with biological or chemical modulators known to induce signaling changes<br />

to verify modification specificity, such as phosphorylation, acetylation, cleavage, etc.<br />

• Phospho-specific antibodies can be evaluated with phosphatase treatment.<br />

• Isotype control antibodies help rule out nonspecific staining of primary antibodies due to Fc<br />

receptor binding or other protein-protein interactions and should have the same immunoglobulin<br />

type as the test antibody.<br />

The IHC protocol includes variations in antigen retrieval methods, antibody diluents, and detection<br />

systems; the optimal method for these parameters must be determined experimentally for each<br />

primary antibody in order to produce the maximum signal.<br />

The most common problems encountered when performing IHC are weak specific staining or high<br />

background. Some challenges, such as incomplete or poor antigen retrieval, are broadly applicable to<br />

most antibodies, while other challenges are specific to certain species or protein modifications.<br />

IHC Tips for Success<br />

Antigen retrieval optimization can greatly improve staining.<br />

The crosslinks created during the fixation step can prevent antibody binding by inhibiting access to the<br />

antigen; therefore, it is important to reverse crosslinks using a procedure called antigen retrieval (also<br />

known as antigen unmasking or epitope retrieval). Antigen retrieval can be achieved through either<br />

a heat-induced method (heat-induced epitope retrieval; HIER) or through enzymatic digestion, and<br />

the optimal form of retrieval is specific for the unique antibody/antigen and not for the protein itself.<br />

Therefore, if you use more than one antibody against a particular protein target, the optimal retrieval<br />

conditions for each antibody must be determined individually.<br />

<strong>CST</strong> #4267<br />

Competitor’s mAb<br />

Citrate EDTA Pepsin<br />

Avoid casein-based solutions when<br />

using a phospho-specific antibody.<br />

Blocking is an important step in the IHC protocol because it prevents nonspecific background staining.<br />

Commercially available blocking solutions that contain casein should be avoided when working with<br />

phospho-specific primary antibodies as they tend to diminish signal intensity.<br />

Phospho-Histone H2A.X (Ser139)<br />

(20E3) Rabbit mAb #9718: IHC<br />

analysis of paraffin-embedded human<br />

breast carcinoma using #9718 after<br />

blocking with normal goat serum<br />

(NGS) (left) or a casein-based blocking<br />

solution (right).<br />

TBST/5% NGS<br />

Avoid same-species background.<br />

Casein<br />

When the primary antibody is from the same species as the sample being tested, the secondary antibody<br />

may bind endogenous IgG in some tissues, causing high background (mouse-on-mouse staining,<br />

for example). Including a control slide stained without the primary antibody can establish whether or<br />

not the secondary antibody is the source of the background.<br />

Ki-67 (D3B5) Rabbit mAb (Mouse<br />

Specific; IHC Formulated) #12202:<br />

IHC analysis of paraffin-embedded<br />

mouse lung using #12202 (left) or<br />

a competitor’s Ki-67 mouse mAb<br />

(right). Analysis with #12202 displays<br />

only nuclear staining, while analysis<br />

with the mouse mAb shows high<br />

background staining.<br />

<strong>CST</strong> #12202<br />

Competitor’s mouse mAb<br />

Use a polymer-based detection system to increase sensitivity<br />

and avoid endogenous biotin background staining.<br />

Traditional IHC detection methods use a biotinylated secondary antibody followed by exposure to an<br />

avidin-HRP complex prior to chromogenic detection. Biotin-based systems are prone to background<br />

staining, particularly in tissues that possess high levels of endogenous biotin, such as liver and kidney.<br />

Polymer-based detection reagents consist of enzymes and secondary antibodies directly conjugated to<br />

a polymer backbone. These systems eliminate false-positive staining due to endogenous biotin. The<br />

polymer-based system also improves sensitivity, enabling detection of low abundance targets. Signal-<br />

Stain ® Boost IHC Detection Reagents save time by eliminating one step in the detection procedure and<br />

are compatible with all peroxidase-based substrates.<br />

SignalStain ® Boost IHC Detection<br />

Reagent #8114: IHC analysis of<br />

paraffin-embedded human lung carcinoma<br />

using Sox2 (D6D9) XP ® Rabbit<br />

mAb #3579 and either biotin-based<br />

detection (left) or polymer-based<br />

detection #8114 (right).<br />

Competitor antibodies used in accordance with manufacturer recommendations.<br />

Biotin-based Detection<br />

Polymer-based Detection<br />

12: Immunohistochemistry (IHC)<br />

Casein block<br />

produces a lower<br />

overall signal when<br />

compared with<br />

TBST/5% NGS.<br />

When possible,<br />

avoid using primary<br />

antibodies raised in<br />

the same species as<br />

the tissue examined.<br />

Polymer-based<br />

detection is more<br />

sensitive than biotinbased<br />

systems.<br />

Competitor antibodies used in accordance with manufacturer recommendations.<br />

214 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstihc<br />

215


Section II: ANTIBODY APPLICATIONS<br />

chapter 12: Immunohistochemistry (IHC)<br />

IHC Tips &<br />

Techniques<br />

Videos<br />

For more in-depth help with IHC,<br />

please see our IHC Protocols and<br />

Troubleshooting videos at<br />

www.cellsignal.com/ihcvideo<br />

IHC Paraffin Protocol (using SignalStain ® Boost Detection Reagent)<br />

*IMPORTANT: Please refer to the Applications section on the front page of product datasheet or product webpage to determine<br />

whether a product is validated and approved for use on paraffin-embedded (IHC-P) tissue sections. Please see product datasheet<br />

or product webpage for appropriate antibody dilution and unmasking solution.<br />

A. Solutions and Reagents<br />

NOTE: Prepare solutions with reverse osmosis deionized or equivalent grade water (dH 2 O).<br />

1. Xylene<br />

2. Ethanol, anhydrous denatured, histological grade (100% and 95%)<br />

3. Deionized water (dH 2 O)<br />

4. Hematoxylin (optional)<br />

5. Wash Buffer: 1X Tris Buffered Saline with Tween ® 20 (TBST): To prepare 1 L 1X TBST: add 100 ml 10X TBST (#9997) to<br />

900 ml dH 2 O, mix.<br />

6. Antibody Diluent Options:*<br />

a. SignalStain ® Antibody Diluent: (#8112)<br />

b. TBST/5% normal goat serum: To 5 ml 1X TBST, add 250 µl Normal Goat Serum (#5425).<br />

c. PBST/5% normal goat serum: To 5 ml 1X PBST, add 250 µl Normal Goat Serum (#5425).<br />

1X Phosphate Buffered Saline (PBS): To prepare 1 L 1X PBS: add 50 ml 20X PBS (#9808) to 950 ml dH 2 O, mix.<br />

1X PBS/0.1% Tween ® 20 (PBST): To prepare 1 L 1X PBST: add 1 ml Tween ® 20 to 1 L 1X PBS and mix.<br />

7. Antigen Unmasking Options:*<br />

a. Citrate: 10 mM Sodium Citrate Buffer: To prepare 1 L, add 2.94 g sodium citrate trisodium salt dihydrate<br />

(C 6 H 5 Na 3 O 7 •2H 2 O) to 1 L dH 2 O. Adjust pH to 6.0.<br />

b. EDTA: 1 mM EDTA: To prepare 1 L add 0.372 g EDTA (C 10 H 14 N 2 O 8 Na 2 •2H 2 O) to 1 L dH 2 O. Adjust pH to 8.0.<br />

c. TE: 10 mM Tris/1 mM EDTA, pH 9.0: To prepare 1 L, add 1.21 g Tris base (C 4 H 11 NO 3 ) and 0.372 g EDTA<br />

(C 10 H 14 N 2 O 8 Na 2 •2H 2 O) to 950 ml dH 2 O. Adjust pH to 9.0, then adjust final volume to 1 L with dH 2 O.<br />

d. Pepsin: 1 mg/ml in Tris-HCl, pH 2.0.<br />

8. 3% Hydrogen Peroxide: To prepare 100 ml, add 10 ml 30% H 2 O 2 to 90 ml dH 2 O.<br />

9. Blocking Solution: TBST/5% Normal Goat Serum: to 5 ml 1X TBST, add 250 µl Normal Goat Serum (#5425).<br />

10. Detection System: SignalStain ® Boost IHC Detection Reagents (HRP, Mouse #8125; HRP, Rabbit #8114 )<br />

11. Substrate: SignalStain ® DAB Substrate Kit (#8059).<br />

B. Deparaffinization/Rehydration<br />

NOTE: Do not allow slides to dry at any time during this procedure.<br />

1. Deparaffinize/hydrate sections:<br />

a. Incubate sections in 3 washes of xylene for 5 min each.<br />

b. Incubate sections in 2 washes of 100% ethanol for 10 min each.<br />

c. Incubate sections in 2 washes of 95% ethanol for 10 min each.<br />

2. Wash sections 2 times in dH 2 O for 5 min each.<br />

C. Antigen Unmasking*<br />

NOTE: Consult product datasheet for specific recommendation for the unmasking solution/protocol.<br />

1. For Citrate: Bring slides to a boil in 10 mM sodium citrate buffer, pH 6.0; maintain at a sub-boiling temperature for 10 min.<br />

Cool slides on bench top for 30 min.<br />

2. For EDTA: Bring slides to a boil in 1 mM EDTA, pH 8.0: follow with 15 min at a sub-boiling temperature. No cooling<br />

is necessary.<br />

3. For TE: Bring slides to a boil in 10 mM Tris/1 mM EDTA, pH 9.0: then maintain at a sub-boiling temperature for 18 min.<br />

Cool at room temperature for 30 min.<br />

4. For Pepsin: Digest for 10 min at 37°C.<br />

D. Staining<br />

NOTE: Consult product datasheet for recommended antibody diluent.<br />

1. Wash sections in dH 2 O 3 times for 5 min each.<br />

2. Incubate sections in 3% hydrogen peroxide for 10 min.<br />

3. Wash sections in dH 2 O 2 times for 5 min each.<br />

4. Wash sections in wash buffer for 5 min.<br />

5. Block each section with 100–400 µl blocking solution for 1 hr at room temperature.<br />

6. Remove blocking solution and add 100–400 µl primary antibody diluted in recommended antibody diluent to each section*.<br />

Incubate overnight at 4°C.<br />

7. Equilibrate SignalStain ® Boost Detection Reagent to room temperature.<br />

8. Remove antibody solution and wash sections with wash buffer three times for 5 min each.<br />

9. Cover section with 1–3 drops SignalStain ® Boost Detection Reagent as needed. Incubate in a humidified chamber for<br />

30 min at room temperature.<br />

216 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

10. Wash sections three times with wash buffer for 5 min each.<br />

11. Add 1 drop (30 µl) SignalStain ® DAB Chromogen Concentrate to 1 ml SignalStain ® DAB Diluent and mix well before use.<br />

12. Apply 100–400 µl SignalStain ® DAB to each section and monitor closely. 1–10 min generally provides an acceptable<br />

staining intensity.<br />

13. Immerse slides in dH 2 O.<br />

14. If desired, counterstain sections with hematoxylin per manufacturer’s instructions.<br />

15. Wash sections in dH 2 O two times for 5 min each.<br />

16. Dehydrate sections:<br />

a. Incubate sections in 95% ethanol two times for 10 sec each.<br />

b. Repeat in 100% ethanol, incubating sections two times for 10 sec each.<br />

c. Repeat in xylene, incubating sections two times for 10 sec each.<br />

17. Mount sections with coverslips.<br />

For optimal results, always use the recommended primary<br />

antibody diluent, as indicated on the product datasheet.<br />

SignalStain ® Antibody Diluent<br />

#8112: IHC analysis of paraffinembedded<br />

human breast carcinoma<br />

(top) and HCC827 xenograft (bottom)<br />

using Phospho-Akt (Ser473) (D9E)<br />

XP ® Rabbit mAb #4060 or Phospho-<br />

EGF Receptor (Tyr1173) (53A5) Rabbit<br />

mAb #4407 after dilution in either<br />

#8112 (left) or TBST/5% NGS (right).<br />

A superior signal is achieved<br />

when #4060 is diluted in #8112 as<br />

compared with TBST/5% NGS.<br />

No one diluent enables the optimal<br />

performance of all antibodies.<br />

#4060<br />

#4407<br />

SignalStain ® #8112<br />

TBST-5% NGS<br />

IHC Frozen Protocol (using SignalStain ® Boost Detection Reagent)<br />

IMPORTANT: Please refer to the Applications section on the front page of product datasheet or product webpage to determine<br />

whether a product is validated and approved for use on frozen (IHC-F) tissue sections. Please see product datasheet or product<br />

webpage for appropriate antibody dilution.<br />

NOTE: Please see product datasheet and website for product-specific protocol recommendations.<br />

A. Solutions and Reagents<br />

NOTE: Prepare solutions with reverse osmosis deionized or equivalent grade water (dH 2 O).<br />

1. Xylene<br />

2. Ethanol (anhydrous denatured, histological grade 100% and 95%)<br />

3. Hematoxylin (optional)<br />

4. 1X Phosphate Buffered Saline (PBS): To prepare 1 L 1X PBS: add 50 ml 20X PBS (#9808) to 950 ml dH 2 O, mix.<br />

5. Fixative Options: For optimal fixative, please refer to the product datasheet.<br />

a. 10% neutral buffered formalin<br />

b. Acetone<br />

c. Methanol<br />

d. 3% Formaldehyde: To prepare 100 ml, add 18.75 ml 16% formaldehyde to 81.25 ml 1X PBS.<br />

6. Wash Buffer: 1X Tris Buffered Saline with Tween ® 20 (TBS): To prepare 1 L 1X TBS: add 100 ml 10X TBS (#12498) to 900<br />

ml dH 2 O, mix.<br />

7. Methanol/Peroxidase: To prepare, add 10 ml 30% H 2 O 2 to 90 ml methanol. Store at -20°C.<br />

8. Blocking Solution: 1X TBS/0.3% Triton X-100/5% Normal Goat Serum (#5425). To prepare, add 500 µl goat serum and<br />

30 µl Triton X-100 to 9.5 ml 1X TBS.<br />

9. Detection System: SignalStain ® Boost IHC Detection Reagents (HRP, Mouse #8125; HRP, Rabbit #8114 )<br />

10. Substrate: SignalStain ® DAB Substrate Kit (#8059).<br />

www.cellsignal.com/cstprotocols<br />

217


Section II: ANTIBODY APPLICATIONS<br />

B. Sectioning<br />

1. For tissue stored at -80°C: Remove from freezer and equilibrate at -20°C for approximately 15 min before attempting to<br />

section. This may prevent cracking of the block when sectioning.<br />

2. Section tissue at a range of 6–8 µm and place on positively charged slides.<br />

3. Allow sections to air dry on bench for a few min before fixing (this helps sections adhere to slides).<br />

C. Fixation Options<br />

NOTE: Consult product datasheet to determine the optimal fixative.<br />

1. After sections have dried on the slide, fix in optimal fixative as directed below.<br />

a. 10% Neutral Buffered Formalin: 10 min at room temperature. Proceed with staining procedure immediately (Section D).<br />

b. Cold Acetone: 10 min at -20°C. Air dry. Proceed with staining immediately (Section D).<br />

c. Methanol: 10 min at -20°C. Proceed with staining immediately (Section D).<br />

d. 3% Formaldehyde: 15 min at room temperature. Proceed with staining immediately (Section D).<br />

e. 3% Formaldehyde/methanol: 15 min at room temperature in 3% formaldehyde, followed by 5 min in methanol at<br />

-20°C (do not rinse in between). Proceed with staining immediately (Section D).<br />

D. Staining<br />

1. Wash sections in wash buffer 2 times for 5 min.<br />

2. Incubate for 10 min at room temperature in methanol/peroxidase.<br />

3. Wash sections in wash buffer 2 times for 5 min.<br />

4. Block each section with 100–400 µl blocking solution for 1 hr at room temperature.<br />

5. Remove blocking solution and add 100–400 µl primary antibody diluted in blocking solution to each section.<br />

6. Incubate overnight at 4°C.<br />

7. Equilibrate SignalStain ® Boost Detection Reagent to room temperature.<br />

8. Remove antibody solution and wash sections in wash buffer three times for 5 min each.<br />

9. Cover section with 1–3 drops SignalStain ® Boost Detection Reagent as needed. Incubate in a humidified chamber for 30<br />

min at room temperature.<br />

10. Wash sections 3 times with wash buffer for 5 min each.<br />

11. Add 1 drop (30 µl) SignalStain ® DAB Chromogen Concentrate to 1 ml SignalStain ® DAB Diluent and mix well before use.<br />

12. Apply 100–400 µl SignalStain ® DAB to each section and monitor closely. 1–10 min generally provides an acceptable<br />

staining intensity.<br />

13. Immerse slides in dH 2 O.<br />

14. If desired, counterstain sections with hematoxylin per manufacturer’s instructions.<br />

15. Wash sections in dH 2 O 2 times for 5 min each.<br />

16. Dehydrate sections:<br />

a. Incubate sections in 95% ethanol 2 times for 10 sec each.<br />

b. Repeat in 100% ethanol, incubating sections 2 times for 10 sec each.<br />

c. Repeat in xylene, incubating sections 2 times for 10 sec each.<br />

17. Mount sections with coverslips.<br />

Not all DAB substrates perform equally.<br />

<strong>CST</strong> #8059<br />

<strong>Guide</strong> to Successful IHC<br />

Competitor’s<br />

Competitor antibodies used in accordance with manufacturer recommendations.<br />

SignalStain ® DAB Substrate Kit #8059:<br />

IHC analysis of paraffin-embedded human<br />

breast carcinoma using Phospho-Stat3<br />

(Tyr705) (D3A7) XP ® Rabbit mAb #9145.<br />

Chromogenic detection was performed<br />

using #8059 (left) or DAB supplied by a<br />

competitor (right).<br />

Helpful tips and explanations to support your IHC experiments are available in “A <strong>Guide</strong> to Successful<br />

Immunohistochemistry”. Request a copy at www.cellsignal.com/ihcguide<br />

IHC Troubleshooting <strong>Guide</strong><br />

Suboptimal IHC staining is frequently resolved by adjusting relatively few variables. Adjustments to key steps within the protocol,<br />

such as antigen retrieval, can often resolve these common issues.<br />

Little or no staining:<br />

Cause<br />

Sample storage<br />

Tissue sections dried out<br />

Slide preparation<br />

Antigen unmasking/retrieval<br />

Unmasking/retrieval buffer<br />

Antibody dilution/diluent<br />

Incubation time<br />

Detection system<br />

Negative staining<br />

High background:<br />

Cause<br />

Slide preparation<br />

Peroxidase quenching<br />

Biotin block<br />

Blocking<br />

Antibody dilution/diluent<br />

Secondary cross reactivity<br />

Washes<br />

Solution<br />

Slides may lose signal over time in storage. This process is variable and dependent upon<br />

the protein target. The effect of slide storage on staining has not been established for every<br />

protein; therefore, it is best practice that slides are freshly cut before use. If slides must be<br />

stored, do so at 4°C. Do not bake slides before storage.<br />

It is vital that the tissue sections remain covered in liquid throughout the staining procedure.<br />

Inadequate deparaffinization may cause spotty, uneven background staining. Repeat the<br />

experiment with new sections using fresh xylene.<br />

Fixed tissue sections have chemical crosslinks between proteins that, dependent on the<br />

tissue and antigen target, may prevent antibody access or mask antigen targets. Antigen<br />

unmasking protocols may utilize a hot water bath, microwave, or pressure cooker. Antigen<br />

unmasking protocols utilizing a water bath are not recommended. Antigen unmasking<br />

performed with a microwave is preferred, though staining of particular tissues or antigen<br />

targets may require the use of a pressure cooker.<br />

Staining of particular tissues or antigen targets may require an optimized unmasking buffer.<br />

Refer to product datasheet for antigen unmasking buffer recommendations. Always prepare<br />

fresh 1X solutions daily.<br />

Consult <strong>CST</strong> product datasheet for the recommended dilution and diluent. Titration of the<br />

antibody may be required if a reagent other than the one recommended is used.<br />

Primary antibody incubation according to a rigorously tested protocol provides consistent,<br />

reliable results. <strong>CST</strong> antibodies have been developed and validated for optimal results<br />

when incubated overnight at 4°C.<br />

Polymer-based detection reagents, such as SignalStain ® Boost IHC Detection Reagents<br />

(#8114 and #8125), in conjunction with SignalStain ® DAB Substrate Kit (#8059), are more<br />

sensitive than avidin/biotin-based detection systems. Standard secondary antibodies directly<br />

conjugated with HRP may not provide sufficient signal amplification. Always verify the<br />

expiration date of the detection reagent prior to use.<br />

A complete lack of staining may indicate an issue with the antibody or protocol. Employ a<br />

high expressing positive control, such as paraffin-embedded cell pellets, to ensure that the<br />

antibody and procedure are working as expected.<br />

Phospho-specific antibodies in particular, or any antibody directed against a rarely<br />

expressed protein, may not stain 100% of the cases of a given indication. It is possible that<br />

the sample is truly negative.<br />

Solution<br />

Inadequate deparaffinization may cause spotty, uneven background staining. Repeat the<br />

experiment with new sections using fresh xylene.<br />

Endogenous peroxidase activity in samples may produce excess background signal if an<br />

HRP-based detection system is being used. Quench slides in a 3% H 2 O 2 solution, diluted in<br />

RODI water, for 10 min prior to incubation with the primary antibody.<br />

Using biotin-based detection systems with samples that have high levels of endogenous<br />

biotin, such as kidney and liver tissues, may be problematic. In this case, use a polymerbased<br />

detection system such as SignalStain ® Boost IHC Detection Reagents (#8114 and<br />

#8125). A biotin block may also be performed after the normal blocking procedure prior to<br />

incubation in primary antibody.<br />

Block slides with 1X TBST (#9997) with 5% Normal Goat Serum (#5425) for 30 min prior to<br />

incubation with the primary antibody.<br />

Consult <strong>CST</strong> product datasheet for the recommended dilution and diluent. Titration of the<br />

antibody may be required if a reagent other than the one recommended is used.<br />

The secondary antibody may bind endogenous IgG, causing high background, in some<br />

samples where the secondary antibody is raised in the same species as the sample being<br />

tested (mouse-on-mouse staining). Include a control slide stained without the primary<br />

antibody to confirm whether the secondary antibody is the source of the background.<br />

Adequate washing is critical for contrasting low background and high signal. Wash slides<br />

three times for 5 min with TBST (#9997) after primary and secondary antibody incubations.<br />

chapter 12: Immunohistochemistry (IHC)<br />

218 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstprotocols<br />

219


13<br />

Section II: ANTIBODY APPLICATIONS<br />

chapter<br />

Sandwich ELISA<br />

The enzyme-linked immunosorbent assay (ELISA) is a method for detecting a specific analyte in a<br />

heterogeneous biological fluid or cell extract. ELISA is a quantitative immunoassay typically perfomed<br />

in multi-well microplates, which allows for simultaneous analysis of multiple samples. In a sandwich<br />

ELISA, the target molecule is bound between a capture antibody immobilized on a microplate and a<br />

detection antibody that is detected by a secondary antibody conjugated to an enzymatic or other reporter<br />

system. The use of two antibodies recognizing different epitopes of the same protein results in a highly<br />

specific assay. Sandwich ELISA assays are useful for the screening of small molecule inhibitors without<br />

using expensive instrumentation or complex data analysis methods. Sandwich ELISA output can be<br />

measured by a number of optical methods, including colorimetric (absorbance) and chemiluminescent<br />

(RLU or relative light units). Chemiluminescent readouts show a higher dynamic range, especially at low<br />

target protein concentrations. The specifcity and sensitivity of the capture and detection antibodies are<br />

critical to ensuring high signal-to-background ratios. The capture and detection antibody pairs are also<br />

amenable to customization on plate-based and bead-based platforms.<br />

Rapid cell harvest and elevated phosphatase<br />

inhibitor concentrations may improve ELISA<br />

results for low abundance proteins.<br />

While it is important to achieve complete cell lysis for the investigation of both total and post-translationally<br />

modified protein targets, rigorous extraction methods must be balanced with preservation of<br />

the antibody-antigen interactions required for ELISA. Preserving the phosphorylation state of target<br />

epitopes can be more challenging when working with specific cell lines or tissues, as some cells<br />

produce higher levels of phosphatases than others. When investigating the phosphorylation state of<br />

low abundance proteins or targets that are difficult to detect, especially in high phosphatase-producing<br />

cell lines, it is sometimes necessary to replace the standard protocol for preparation of cell lysates<br />

with a rapid harvest method. This method omits the cell scraping and sonication steps and uses a lysis<br />

buffer that contains increased concentrations of Ser/Thr phosphatase inhibitors. For example, when<br />

initial ELISA results from phospho-targets produce a low signal, scientists at <strong>CST</strong> use a no scrape, no<br />

sonicate rapid harvest method combined with a lysis buffer (#7018) that contains increased amount of<br />

phosphatase inhibitors*, as a means to increase target-specific signal.<br />

13: Sandwich eLISA<br />

*Complete protocol and buffer components<br />

can be found on the PathScan ®<br />

Sandwich ELISA Lysis Buffer #7018<br />

product datasheet<br />

Improved ELISA detection<br />

of phospho-AMPKα (Thr172)<br />

was achieved using an<br />

alternative lysis buffer and<br />

a rapid extraction protocol.<br />

3<br />

2.5<br />

2<br />

1.5<br />

1<br />

0.5<br />

PathScan ® Phospho-AMPKα<br />

(Thr172) Sandwich ELISA Kit #7959:<br />

C2C12 cells, untreated or treated with<br />

H 2 0 2 (10 mM for 10 min), were lysed and<br />

harvested using Cell Lysis Buffer (10X)<br />

#9803 or PathScan ® Sandwich ELISA<br />

Lysis Buffer #7018 (which contains<br />

inhibitors of Ser/Thr phosphatases) and<br />

detected using #7959. Equal protein<br />

concentrations of cell extract were used<br />

in each assay. ELISA signal was greatest<br />

when cell lysis was performed with<br />

#7018 and a rapid harvest procedure<br />

with no cell scraping or sonication.<br />

0<br />

Scrape/Sonicate No Scrape/Sonicate Scrape/Sonicate No Scrape/Sonicate<br />

#9803<br />

#7018<br />

Cell Lysate total protein concentration (0.25 mg/ml)<br />

Untreated<br />

H 2 O 2 Treated<br />

Phosphatase<br />

inhibitors preserve<br />

the constitutive phosphorylation<br />

of Mer.<br />

ELISA Tips for Success<br />

For detection of intracellular targets, the success of an ELISA experiment depends critically on the<br />

quality and composition of the cell lysis buffer and the method used to generate the cell extract.<br />

Include protease and/or phosphatase inhibitors in lysis<br />

buffer to preserve protein levels and phosphorylation state.<br />

Using fresh cell lysis buffer containing appropriate protease and phosphatase inhibitors ensures that<br />

changes in the protein or phosphorylation levels are not due to proteolytic degradation of target proteins<br />

or loss of phosphorylation due to endogenous phosphatases.<br />

Absorbance 450nm<br />

3<br />

2<br />

1<br />

0<br />

Total Mer<br />

Phosho-Mer (panTyr)<br />

– + – +<br />

phosphatase<br />

inhibitor<br />

PathScan ® Phospho-Mer (panTyr) Sandwich ELISA Kit #13340: Constitutive<br />

phosphorylation of Mer in HCC827 cells lysed in the presence of protease inhibitors<br />

and phosphatase inhibitors (phospho lysate) is detected by #13340 (upper, right).<br />

In contrast, a low level of phospho-Mer protein is detected in HCC827 cells lysed<br />

in the presence of protease inhibitors and absence of phosphatase inhibitors<br />

(nonphospho lysate). Similar levels of total Mer protein from both nonphospho and<br />

phospho lysates are detected by PathScan ® Total Mer Sandwich ELISA Kit #13488<br />

(upper, left). Absorbance at 450 nm is shown in the top figure while corresponding<br />

western blots using Mer (D21F11) XP ® Rabbit mAb #4319 (left) and a phospho-Mer<br />

(Tyr749/753) antibody (right) are shown in the bottom figure. Phosphatase inhibitors<br />

include sodium pyrophosphate, β-glycerophosphate, and sodium orthovanadate.<br />

Nonphospho<br />

Phospho<br />

PathScan ® Sandwich ELISA Colorimetric Protocol<br />

NOTE: Refer to product-specific datasheet or product webpage for assay incubation temperature.<br />

A. Solutions and Reagents<br />

For lyophilized formulation<br />

NOTE: Prepare solutions with reverse osmosis deionized (RODI) or equivalent grade water.<br />

1. Microwell strips: Bring all to room temperature before use.<br />

2. Detection Antibody: Supplied lyophilized as a green colored cake or powder. Add 1.0 ml of Detection Antibody Diluent (green<br />

solution) to yield a concentrated stock solution. Incubate at room temperature for 5 min with occasional gentle mixing to fully<br />

reconstitute. To make the final working solution, add the full 1.0 ml volume of reconstituted Detection Antibody to 10.0 ml of<br />

Detection Antibody Diluent in a clean tube and gently mix. Unused working solution may be stored for 4 weeks at 4°C.<br />

3. HRP-Linked Antibody: Supplied lyophilized as a red colored cake or powder. Add 1.0 ml of HRP Diluent (red solution) to<br />

yield a concentrated stock solution. Incubate at room temperature for 5 min with occasional gentle mixing to fully reconstitute.<br />

To make the final working solution, add the full 1.0 ml volume of reconstituted HRP-Linked Antibody to 10.0 ml of HRP<br />

Diluent in a clean tube and gently mix. Unused working solution may be stored for 4 weeks at 4°C. NOTE: Some PathScan ®<br />

ELISA Kits may include HRP-Linked Streptavidin in place of HRP-Linked Antibody.<br />

4. Detection Antibody Diluent: Green colored diluent for reconstitution and dilution of the detection antibody (11 ml provided).<br />

5. HRP Diluent: Red colored diluent for reconstitution and dilution of the HRP‐Linked Antibody (11 ml provided).<br />

6. Sample Diluent: Blue colored diluent provided for dilution of cell lysates.<br />

7. 1X Wash Buffer: Prepare by diluting 20X Wash Buffer (included in each PathScan ® Sandwich ELISA Kit) in purified water.<br />

8. 1X Cell Lysis Buffer: Prepare 1X lysis buffer using either PathScan ® Sandwich ELISA buffer (ready to use 1X stock, #7018)<br />

or 10X Cell Lysis Buffer (#9803). To prepare 10 ml of 1X Cell Lysis Buffer, add 1 ml 10X Cell Lysis Buffer to 9 ml of dH 2 O,<br />

mix. Both buffers can be stored at 4°C for short-term use (1–2 weeks). Recommended: Add 1 mM phenylmethylsulfonyl<br />

fluoride (PMSF) (#8553) immediately before use. NOTE: Refer to product-specific datasheet or webpage for lysis buffer<br />

recommendation.<br />

9. TMB Substrate: (#7004)<br />

10. STOP Solution: (#7002)<br />

220 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstelisa<br />

221


Section II: ANTIBODY APPLICATIONS<br />

chapter 13: Sandwich eLISA<br />

For liquid formulation<br />

NOTE: Prepare solutions with reverse osmosis deionized (RODI) or equivalent grade water.<br />

1. 20X Phosphate Buffered Saline (PBS): (#9808) To prepare 1 L PBS: add 50 ml 10X PBS to 950 ml dH 2 O, mix.<br />

2. Bring all microwell strips to room temperature before use.<br />

3. Prepare 1X Wash Buffer by diluting 20X Wash Buffer (included in each PathScan ® Sandwich ELISA Kit) in dH 2 O.<br />

4. 1X Cell Lysis Buffer: 10X Cell Lysis Buffer (#9803): To prepare 10 ml of 1X Cell Lysis Buffer, add 1 ml of 10X Cell Lysis<br />

Buffer to 9 ml of dH 2 O, mix. PathScan ® Sandwich ELISA Lysis Buffer (#7018) 1X: This buffer is ready to use as is. Both buffers<br />

can be stored at 4°C for short-term use (1–2 weeks). Recommended: Add 1 mM phenylmethylsulfonyl fluoride (PMSF)<br />

(#8553) immediately before use. NOTE: Refer to product-specific datasheet or webpage for lysis buffer recommendation.<br />

5. TMB Substrate: (#7004)<br />

6. STOP Solution: (#7002)<br />

B. Preparing Cell Lysates<br />

For adherent cells<br />

1. Aspirate media when the culture reaches 80–90% confluence. Treat cells by adding fresh media containing regulator for<br />

desired time.<br />

2. Remove media and rinse cells once with ice-cold 1X PBS.<br />

3. Remove PBS and add 0.5 ml ice-cold 1X cell lysis buffer plus 1 mM PMSF to each plate (10 cm diameter) and incubate the<br />

plate on ice for 5 min.<br />

4. Scrape cells off the plate and transfer to an appropriate tube. Keep on ice.<br />

5. Sonicate lysates on ice.<br />

6. Microcentrifuge for 10 min (x14,000 rpm) at 4°C and transfer the supernatant to a new tube. The supernatant is the cell<br />

lysate. Store at -80°C in single-use aliquots.<br />

For suspension cells<br />

1. Remove media by low speed centrifugation (~1,200 rpm) when the culture reaches 0.5–1.0 x 10 6 viable cells/ml. Treat cells<br />

by adding fresh media containing regulator for desired time.<br />

2. Collect cells by low speed centrifugation (~1,200 rpm) and wash once with 5–10 ml ice-cold 1X PBS.<br />

3. Cells harvested from 50 ml of growth media can be lysed in 2.0 ml of 1X cell lysis buffer plus 1 mM PMSF.<br />

4. Sonicate lysates on ice.<br />

5. Microcentrifuge for 10 min (x14,000 rpm) at 4°C and transfer the supernatant to a new tube. The supernatant is the cell<br />

lysate. Store at -80°C in single-use aliquots.<br />

C. Test Procedure<br />

1. After the microwell strips have reached room temperature, break off the required number of microwells. Place the microwells<br />

in the strip holder. Unused microwells must be resealed in the storage bag and stored at 4°C immediately.<br />

2. Cell lysates can be undiluted or diluted with sample diluent (supplied in each PathScan ® Sandwich ELISA Kit, blue color).<br />

Individual datasheets or product webpage for each kit provide information regarding an appropriate dilution factor for lysates<br />

and kit assay results.<br />

3. Add 100 µl of each undiluted or diluted cell lysate to the appropriate well. Seal with tape and press firmly onto top of<br />

microwells. Incubate the plate for 2 hr at 37°C. Alternatively, the plate can be incubated overnight at 4°C.<br />

4. Gently remove the tape and wash wells:<br />

a. Discard plate contents into a receptacle.<br />

b. Wash 4 times with 1X wash buffer, 200 µl each time per well.<br />

c. For each wash, strike plates on fresh paper towels hard enough to remove the residual solution in each well, but do not<br />

allow wells to completely dry at any time.<br />

d. Clean the underside of all wells with a lint-free tissue.<br />

5. Add 100 µl of detection antibody (green color) to each well. Seal with tape and incubate the plate at 37°C for 1 hr.<br />

6. Repeat wash procedure (Section C, Step 4).<br />

7. Add 100 µl of HRP-linked secondary antibody (red color) to each well. Seal with tape and incubate the plate for 30 min at 37°C.<br />

8. Repeat wash procedure (Section C, Step 4).<br />

9. Add 100 µl of TMB substrate to each well. Seal with tape and incubate the plate for 10 min at 37°C or 30 min at 25°C.<br />

10. Add 100 µl of STOP solution to each well. Shake gently for a few seconds.<br />

NOTE: Initial color of positive reaction is blue, which changes to yellow upon addition of STOP solution.<br />

11. Read results<br />

a. Visual Determination: Read within 30 min after adding STOP solution.<br />

b. Spectrophotometric Determination: Wipe underside of wells with a lint-free tissue. Read absorbance at 450 nm<br />

within 30 min after adding STOP solution.<br />

222 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

Phospho cell lysates<br />

serve as convenient<br />

and well-validated<br />

positive controls for<br />

ELISA studies.<br />

PathScan ® Sandwich ELISA<br />

Control Phospho Cell Extracts I<br />

#7988: ELISA analysis of #7988 using<br />

multiple PathScan ® Sandwich ELISA<br />

kits. Samples were prepared using the<br />

standard ELISA protocol, to a final concentration<br />

of 0.25 mg/ml and assayed<br />

using the indicated ELISA kits.<br />

Absorbance 450nm<br />

5<br />

4.5<br />

4<br />

3.5<br />

3<br />

2.5<br />

2<br />

1.5<br />

1<br />

0.5<br />

0<br />

Phospho-Akt<br />

(Thr308)<br />

Total Bad<br />

Total NF-κB<br />

p65<br />

PathScan ® Sandwich ELISA Chemiluminescent Protocol<br />

NOTE: Refer to product-specific datasheets or product webpage for assay incubation temperature. This chemiluminescent ELISA<br />

is offered in low volume microplate. Samples and reagents only require 50 µl per microwell.<br />

A. Solutions and Reagents<br />

For lyophilized formulation<br />

NOTE: Prepare solutions with reverse osmosis deionized (RODI) or equivalent grade water.<br />

1. Microwell strips: Bring all to room temperature before use.<br />

2. Detection Antibody: Supplied lyophilized as a green colored cake or powder. Add 1.0 ml of Detection Antibody Diluent (green<br />

solution) to yield a concentrated stock solution. Incubate at room temperature for 5 min with occasional gentle mixing to fully<br />

reconstitute. To make the final working solution, add the full 1.0 ml volume of reconstituted Detection Antibody to 10.0 ml of<br />

Detection Antibody Diluent in a clean tube and gently mix. Unused working solution may be stored for 4 weeks at 4°C.<br />

3. HRP-Linked Antibody: Supplied lyophilized as a red colored cake or powder Add 1.0 ml of HRP Diluent (red solution) to<br />

yield a concentrated stock solution. Incubate at room temperature for 5 min with occasional gentle mixing to fully reconstitute.<br />

To make the final working solution, add the full 1.0 ml volume of reconstituted HRP-Linked Antibody to 10.0 ml of HRP<br />

Diluent in a clean tube and gently mix. Unused working solution may be stored for 4 weeks at 4°C. NOTE: Some PathScan ®<br />

ELISA Kits may include HRP-Linked Streptavidin in place of HRP-Linked Antibody.<br />

4. Detection Antibody Diluent: Green colored diluent for reconstitution and dilution of the detection antibody (5.5 ml provided).<br />

5. HRP Diluent: Red colored diluent for reconstitution and dilution of the HRP‐Linked Antibody (5.5 ml provided).<br />

6. Sample Diluent: Blue colored diluent provided for dilution of cell lysates.<br />

7. 1X Wash Buffer: Prepare by diluting 20X Wash Buffer (included in each PathScan ® Sandwich ELISA Kit) in purified water.<br />

8. 1X Cell Lysis Buffer: 10X Cell Lysis Buffer (#9803): To prepare 10 ml of 1X Cell Lysis Buffer, add 1 ml of 10X Cell Lysis<br />

Buffer to 9 ml of dH 2 O, mix. PathScan ® Sandwich ELISA Lysis Buffer (#7018) 1X: This buffer is ready to use as is. Both buffers<br />

can be stored at 4°C for short-term use (1–2 weeks). Recommended: Add 1 mM phenylmethylsulfonyl fluoride (PMSF)<br />

(#8553) immediately before use. NOTE: Refer to product-specific datasheet or webpage for lysis buffer recommendation.<br />

9. Luminol/Enhancer Solution and Stable Peroxide Buffer<br />

For liquid formulation<br />

NOTE: Prepare solutions with reverse osmosis deionized (RODI) or equivalent grade water.<br />

1. 20X Phosphate Buffered Saline (PBS): (#9808) To prepare 1 L 1X PBS: add 50 ml 10X PBS to 950 ml dH 2 O, mix.<br />

2. Bring all microwell strips to room temperature before use.<br />

3. Prepare 1X wash buffer by diluting 20X Wash Buffer (included in each PathScan ® Sandwich ELISA Kit) in dH 2 O.<br />

4. 1X Cell Lysis Buffer: 10X Cell Lysis Buffer (#9803): To prepare 10 ml of 1X Cell Lysis Buffer, add 1 ml of 10X Cell Lysis<br />

Buffer to 9 ml of dH 2 O, mix. PathScan ® Sandwich ELISA Lysis Buffer (#7018) 1X: This buffer is ready to use as is. Both buffers<br />

can be stored at 4°C for short-term use (1–2 weeks). Recommended: Add 1 mM phenylmethylsulfonyl fluoride (PMSF)<br />

(#8553) immediately before use. NOTE: Refer to product-specific datasheet or webpage for lysis buffer recommendation.<br />

5. 20X LumiGLO ® Reagent and 20X Peroxide: (#7003)<br />

B. Preparing Cell Lysates<br />

For adherent cells<br />

1. Aspirate media when the culture reaches 80–90% confluence. Treat cells by adding fresh media containing regulator for<br />

desired time.<br />

2. Remove media and rinse cells once with ice-cold 1X PBS.<br />

3. Remove PBS and add 0.5 ml ice-cold 1X Cell Lysis Buffer plus 1 mM PMSF to each plate (10 cm diameter) and incubate the<br />

plate on ice for 5 min.<br />

4. Scrape cells off the plate and transfer to an appropriate tube. Keep on ice.<br />

5. Sonicate lysates on ice.<br />

6. Microcentrifuge for 10 min (x14,000 rpm) at 4°C and transfer the supernatant to a new tube. The supernatant is the cell<br />

lysate. Store at -80°C in single-use aliquots.<br />

Total<br />

S6 RP<br />

p-S6<br />

(Ser235/236)<br />

p-CREB<br />

(Ser133)<br />

Total<br />

CREB<br />

p-PRAS40<br />

(Thr246)<br />

p-RSK1<br />

(Ser380)<br />

www.cellsignal.com/cstprotocols<br />

223


Section II: ANTIBODY APPLICATIONS<br />

chapter 13: Sandwich eLISA<br />

Validation of <strong>CST</strong><br />

Cell Lysis Buffer<br />

for detection of an<br />

intracellular target<br />

For suspension cells<br />

1. Remove media by low speed centrifugation (~1,200 rpm) when the culture reaches 0.5–1.0 x 10 6 viable cells/ml. Treat cells<br />

by adding fresh media containing regulator for desired time.<br />

2. Collect cells by low speed centrifugation (~1,200 rpm) and wash once with 5–10 ml ice-cold 1X PBS.<br />

3. Cells harvested from 50 ml of growth medium can be lysed in 2.0 ml of 1X cell lysis buffer plus 1 mM PMSF.<br />

4. Sonicate lysates on ice.<br />

5. Microcentrifuge for 10 min (x14,000 rpm) at 4°C and transfer the supernatant to a new tube. The supernatant is the cell<br />

lysate. Store at -80°C in single-use aliquots.<br />

C. Test Procedure<br />

1. After the microwell strips have reached room temperature, break off the required number of microwells. Place the microwells<br />

in the strip holder. Unused microwells must be resealed in the storage bag and stored at 4°C immediately.<br />

2. Cell lysates can be used undiluted or diluted with sample diluent (supplied in each PathScan ® Sandwich ELISA Kit, blue<br />

color). Individual datasheets or product webpage for each kit provide information regarding an appropriate dilution factor for<br />

lysates and kit assay results.<br />

3. Add 50 µl of each undiluted or diluted cell lysate to the appropriate well. Seal with tape and press firmly onto top of<br />

microwells. Incubate the plate for 2 hr at room temperature. Alternatively, the plate can be incubated overnight at 4°C.<br />

4. Gently remove the tape and wash wells:<br />

a. Discard plate contents into a receptacle.<br />

b. Wash 4 times with 1X Wash Buffer, 150 µl each time per well.<br />

c. For each wash, strike plates on fresh paper towels hard enough to remove the residual solution in each well, but do not<br />

allow wells to dry completely at any time.<br />

d. Clean the underside of all wells with a lint-free tissue.<br />

5. Add 50 µl of detection antibody (green color) to each well. Seal with tape and incubate the plate at room temperature for 1 hr.<br />

6. Repeat wash procedure (Section C, Step 4).<br />

7. Add 50 µl of HRP-linked secondary antibody (red color) to each well. Seal with tape and incubate the plate at room<br />

temperature for 30 min.<br />

8. Repeat wash procedure (Section C, Step 4).<br />

9. Prepare detection reagent working solution by mixing equal parts 2X LumiGLO ® Reagent and 2X Peroxide.<br />

10. Add 50 µl of the detection reagent working solution to each well.<br />

11. Use a plate-based luminometer set at 425 nm to measure Relative Light Units (RLU) within 1–10 min following addition of<br />

the substrate.<br />

a. Optimal signal intensity is achieved when read within 10 min.<br />

RLU (x10 5 )<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

PathScan ® Sandwich ELISA Antibody Pair Protocol<br />

NOTE: This protocol includes plate preparation<br />

6<br />

4<br />

2<br />

0<br />

0.01 0.02 0.03<br />

0.1 0.2 0.3 0.4<br />

Protein conc. of lysate (mg/ml)<br />

0.5<br />

Cell Lysis Buffer (10X) #9803: The relationship between protein<br />

concentration of lysates from A-431 cells, untreated or treated with<br />

Human Epidermal Growth Factor (hEGF) #8916, and immediate light<br />

generation with chemiluminescent substrate using PathScan ® Total<br />

EGF Receptor Chemiluminescent Sandwich ELISA Kit #7297. After<br />

starvation, A-431 cells (85% confluence) were treated with hEGF<br />

(100 ng/ml, 5 min at 37°C) and then lysed using #9803. Graph<br />

inset corresponding to the shaded area shows high sensitivity and a<br />

linear response at the low protein concentration range.<br />

EGF-treated<br />

Control<br />

A. Solutions and Reagents<br />

NOTE: Prepare solutions with reverse osmosis deionized (RODI) or equivalent grade water.<br />

1. 20X Phosphate Buffered Saline (PBS): (#9808) To prepare 1 L 1X PBS: add 50 ml 20X PBS to 950 ml dH 2 O, mix.<br />

2. Wash Buffer: 1X PBS/0.05% Tween ® 20, (20X PBST #9809)<br />

3. Blocking Buffer: 1X PBS/0.05% Tween ® 20, 1% BSA<br />

4. 1X Cell Lysis Buffer: 10X Cell Lysis Buffer (#9803): To prepare 10 ml of 1X Cell Lysis Buffer, add 1 ml of 10X Cell Lysis<br />

Buffer to 9 ml of dH 2 O, mix. PathScan ® Sandwich ELISA Lysis Buffer (#7018) 1X: This buffer is ready to use as is. Both buffers<br />

can be stored at 4°C for short-term use (1–2 weeks). Recommended: Add 1 mM phenylmethylsulfonyl fluoride (PMSF)<br />

(#8553) immediately before use. NOTE: Refer to product-specific datasheet or webpage for lysis buffer recommendation.<br />

5. Bovine Serum Albumin (BSA): (#9998)<br />

6. TMB Substrate: (#7004)<br />

7. STOP Solution: (#7002)<br />

NOTE: Reagents should be made fresh daily.<br />

B. Preparing Cell Lysates<br />

For adherent cells<br />

1. Aspirate media when the culture reaches 80–90% confluence. Treat cells by adding fresh media containing regulator for<br />

desired time.<br />

2. Remove media and rinse cells once with ice-cold 1X PBS.<br />

3. Remove PBS and add 0.5 ml ice-cold 1X Cell Lysis Buffer plus 1 mM PMSF to each plate (10 cm diameter) and incubate the<br />

plate on ice for 5 min.<br />

4. Scrape cells off the plate and transfer to an appropriate tube. Keep on ice.<br />

5. Sonicate lysates on ice.<br />

6. Microcentrifuge for 10 min (x14,000 rpm) at 4°C and transfer the supernatant to a new tube. The supernatant is the cell<br />

lysate. Store at -80°C in single-use aliquots.<br />

For suspension cells<br />

1. Remove media by low speed centrifugation (~1,200 rpm) when the culture reaches 0.5–1.0 x 10 6 viable cells/ml. Treat cells<br />

by adding fresh media containing regulator for desired time.<br />

2. Collect cells by low speed centrifugation (~1,200 rpm) and wash once with 5–10 ml ice-cold 1X PBS.<br />

3. Cells harvested from 50 ml of growth media can be lysed in 2.0 ml of 1X cell lysis buffer plus 1 mM PMSF.<br />

4. Sonicate lysates on ice.<br />

5. Microcentrifuge for 10 min (x14,000 rpm) at 4°C and transfer the supernatant to a new tube. The supernatant is the cell<br />

lysate. Store at -80°C in single-use aliquots.<br />

C. Coating Procedure<br />

1. Rinse microplate with 200 µl of dH 2 O, discard liquid. Blot on paper towel to make sure wells are dry.<br />

2. Dilute capture antibody 1:100 in 1X PBS. For a single 96 well plate, add 100 µl of capture antibody stock to 9.9 ml 1X PBS.<br />

Mix well and add 100 µl/well. Cover plate and incubate overnight at 4°C (17–20 hr).<br />

3. After overnight coating, gently uncover plate and wash wells:<br />

a. Discard plate contents into a receptacle.<br />

b. Wash 4 times with wash buffer, 200 µl each time per well. For each wash, strike plates on fresh paper towels hard<br />

enough to remove the residual solution in each well, but do not allow wells to completely dry at any time.<br />

c. Clean the underside of all wells with a lint-free tissue.<br />

4. Block plates. Add 150 µl of blocking buffer/well, cover plate, and incubate at 37°C for 2 hr.<br />

5. After blocking, wash plate (Section C, Step 3). Plate is ready to use.<br />

D. Test Procedure<br />

1. Lysates can be used undiluted or diluted in blocking buffer. 100 µl of lysate is added per well. Cover plate and incubate at<br />

37°C for 2 hr.<br />

2. Wash plate (Section C, Step 3).<br />

3. Dilute detection antibody 1:100 in blocking buffer. For a single 96 well plate, add 100 µl of detection antibody Stock to<br />

9.9 ml of blocking buffer. Mix well and add 100 µl/well. Cover plate and incubate at 37°C for 1 hr.<br />

4. Wash plate (Section C, Step 3).<br />

5. Secondary antibody, either streptavidin anti-mouse or anti-rabbit-HRP, is diluted 1:1000 in blocking buffer. For a single<br />

96 well plate, add 10 µl of secondary antibody stock to 9.99 ml of blocking buffer. Mix well and add 100 µl/well. Cover and<br />

incubate at 37°C for 30 min.<br />

6. Wash plate (Section C, Step 3).<br />

7. Add 100 µl of TMB substrate per well. Cover and incubate at 37°C for 10 min.<br />

8. Add 100 µl of STOP solution per well. Shake gently for a few seconds.<br />

9. Read plate on a microplate reader at absorbance 450 nm.<br />

a. Visual Determination: Read within 30 min after adding STOP solution.<br />

b. Spectrophotometric Determination: Wipe underside of wells with a lint-free tissue. Read absorbance at 450 nm<br />

within 30 min after adding STOP solution.<br />

<strong>CST</strong> Technical Support<br />

When you contact <strong>CST</strong> for technical support, you can be confident you will be working with a colleague you can trust,<br />

striving to provide the highest quality products and services to you, our customer. Please see our Technical Support<br />

resource page online for contact information. www.cellsignal.com/cstsupport<br />

224 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstprotocols<br />

225


14<br />

Section II: ANTIBODY APPLICATIONS<br />

Western Blotting (WB)<br />

Protein Detection<br />

Western blotting (WB) or immunoblotting is the most common technique used for monitoring protein<br />

expression in cells or tissue. WB can be used to measure total or modification-specific protein levels<br />

qualitatively or quantitatively by chemiluminescent or fluorescent detection. The WB protocol comprises<br />

multiple steps beginning with preparation of cell lysate and separation of cellular proteins by molecular<br />

weight on a polyacrylamide gel matrix (SDS-PAGE). The separated proteins are then transferred to a<br />

nitrocellulose or polyvinylidene fluoride (PVDF) membrane and detected using target-specific antibodies.<br />

Prior to detection, the membrane is incubated in a blocking solution to prevent nonspecific antibody<br />

binding. A wide variety of primary antibodies can be used to probe western blots including modificationspecific,<br />

motif-specific, or total protein antibodies. Proteins can be detected indirectly using an<br />

unconjugated primary antibody specific to the target of interest coupled with a secondary antibody<br />

conjugated to either an enzyme (horseradish peroxidase or HRP) for chemiluminescent detection or an<br />

infrared dye for fluorescent detection. Alternatively, primary antibodies directly conjugated to HRP can<br />

be used to detect proteins in a one-step protocol.<br />

The most common challenges presented by WB analysis are weak signal and high background.<br />

The tips below will help you address the most common factors responsible for these problems.<br />

WB Tips for Success<br />

Incubate with primary antibody overnight<br />

to improve antibody-target binding.<br />

Overnight incubation of membrane and primary antibody can greatly enhance signal simply by allowing<br />

more time for antibody-antigen binding. We have found that overnight incubation, with gentle agitation,<br />

at 4°C results in stronger antibody specific signal.<br />

Primary antibody incubation overnight at<br />

4°C yields significantly increased antibody<br />

binding compared to a 2 hr incubation.<br />

Phospho-Akt (Ser473) Antibody #9271: WB analysis of extracts from HeLa<br />

cells, untreated or treated with LY294002 #9901 or Human Insulin-like Growth<br />

Factor I (hIGF-I) #8917, using #9271 (top) or PKC Antibody #2058 (bottom).<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

16 hr, 4°C<br />

2 hr, RT<br />

Phospho-<br />

Akt (Ser473)<br />

PKCδ<br />

- - +<br />

- - + LY294002<br />

- + + - + + hIGF-I<br />

Dilute secondary antibody in stronger<br />

blocking agents to reduce nonspecific signal.<br />

We suggest diluting secondary antibody in 5% nonfat milk in TBST rather than in 5% BSA in TBST. Milk<br />

offers stronger blocking of nonspecific binding, hence the BSA-based dilution yields significantly higher<br />

background than the milk-based dilution.<br />

Diluting secondary antibody in milk yields<br />

lower background levels because milk is a<br />

stronger blocking agent.<br />

Phospho-Stat3 (Tyr705) Antibody #9131: WB analysis of extracts from Jurkat<br />

cells, untreated or treated with Human Interferon-α1 (hIFN-α1) #8927, using #9131.<br />

Blots were incubated in Anti-rabbit IgG, HRP-linked Antibody #7074 diluted in either<br />

5% nonfat milk in TBST or 5% BSA in TBST, as indicated.<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

Milk<br />

BSA<br />

– + – +<br />

Weak signal of mid-to-high molecular weight proteins.<br />

chapter 14: Western Blotting (WB)<br />

Phospho-<br />

Stat3<br />

Due to their large size, high molecular weight proteins do not typically transfer as efficiently as smaller<br />

proteins. Wet transfer often provides better transfer of mid-to-high molecular weight proteins than<br />

semi-dry or dry transfer methods. Additionally, because methanol acts as a fixative, decreasing the<br />

methanol concentration in transfer buffer from 20% to 5% and increasing transfer time to 3 hours can<br />

also improve transfer efficiency of proteins >200 kDa.<br />

Wet transfer provides better capture of mid-to-high<br />

molecular weight proteins and a stronger WB signal.<br />

Phospho-Stat3 (Tyr705) (D3A7)<br />

XP ® Rabbit mAb #9145: WB<br />

analysis of extracts from HeLa<br />

cells, untreated or treated with<br />

Human Interferon-α1 (hIFN-α1)<br />

#8927, using #9145 or a<br />

phospho-Stat3 (Tyr705) antibody<br />

from a competitor. Blots were<br />

transferred using either traditional<br />

wet transfer methods (left) or<br />

iBlot ® dry transfer system (right).<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

HeLa<br />

Wet Transfer<br />

Dry Transfer<br />

HeLa HeLa HeLa<br />

hIFN-α1<br />

Phospho-<br />

Stat3 (Tyr705)<br />

Sonicate lysates to increase nuclear protein recovery.<br />

20<br />

kDa<br />

100<br />

80<br />

Sonication thoroughly disrupts both cell and nuclear membranes, improving cell lysis and resulting in<br />

higher protein levels in clarified lysate. This enhanced membrane disruption is especially important<br />

for nuclear proteins, which may not be completely released through detergent use alone. If you do not<br />

have access to a probe sonicator, passing samples through a fine gauge needle will also serve to break<br />

membranes and shear DNA.<br />

– +<br />

<strong>CST</strong> #9145<br />

– + – + – +<br />

Alternate<br />

Provider<br />

<strong>CST</strong> #9145<br />

Alternate<br />

Provider<br />

hIFN-α1<br />

60<br />

50<br />

40<br />

30<br />

Lysate sonication is critical to the observation<br />

of nuclear and chromatin-bound proteins.<br />

20<br />

10<br />

-<br />

Sonicated<br />

+ - + sorbitol<br />

Unsonicated<br />

Phospho-<br />

Histone H3<br />

(Ser10)<br />

Phospho-Histone H3 (Ser10) Antibody #9701: WB analysis of extracts from CKR/PAEC cells,<br />

untreated or treated with sorbitol and either sonicated or without sonication, using #9701.<br />

WB Protocols and Troubleshooting Videos<br />

For more in-depth help with WB, please see our online WB Protocols and Troubleshooting videos.<br />

www.cellsignal.com/wbvideo<br />

226 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstwb<br />

227


Section II: ANTIBODY APPLICATIONS<br />

chapter 14: Western Blotting (WB)<br />

WB General Protocol<br />

For western blots, incubate membrane with diluted primary antibody in either 5% w/v BSA or nonfat dry milk, 1X TBS, 0.1%<br />

Tween ® 20 at 4°C with gentle shaking, overnight. NOTE: Please refer to primary antibody datasheet or product webpage for<br />

recommended primary antibody dilution buffer and recommended antibody dilution.<br />

A. Solutions and Reagents<br />

NOTE: Prepare solutions with reverse osmosis deionized (RODI) or equivalent grade water.<br />

1. 20X Phosphate Buffered Saline (PBS): (#9808) To prepare 1 L 1X PBS: add 50 ml 20X PBS to 950 ml dH 2 O, mix.<br />

2. 10X Tris Buffered Saline (TBS): (#12498) To prepare 1 L 1X TBS: add 100 ml 10X to 900 ml dH 2 O, mix.<br />

3. 1X SDS Sample Buffer: Blue Loading Pack (#7722) or Red Loading Pack (#7723) Prepare fresh 3X reducing loading buffer<br />

by adding 1/10 volume 30X DTT to 1 volume of 3X SDS loading buffer. Dilute to 1X with dH 2 O.<br />

4. 10X Tris-Glycine SDS Running Buffer: (#4050) To prepare 1 L 1X Running Buffer: add 100 ml 10X running buffer to<br />

900 ml dH 2 O, mix.<br />

5. 10X Tris-Glycine Transfer Buffer: (#12539) To prepare 1 L 1X Transfer Buffer: add 100 ml 10X Transfer Buffer to 200 ml<br />

methanol + 700 ml dH 2 O, mix.<br />

6. 10X Tris Buffered Saline with Tween ® 20 (TBST): (#9997) To prepare 1 L 1X TBST: add 100 ml 10X TBST to 900 ml<br />

dH 2 O, mix.<br />

7. Nonfat Dry Milk: (#9999)<br />

8. Blocking Buffer: 1X TBST with 5% w/v nonfat dry milk; for 150 ml, add 7.5 g nonfat dry milk to 150 ml 1X TBST and<br />

mix well.<br />

9. Wash Buffer: (#9997) 1X TBST<br />

10. Bovine Serum Albumin (BSA): (#9998)<br />

11. Primary Antibody Dilution Buffer: 1X TBST with 5% BSA or 5% nonfat dry milk as indicated on primary antibody<br />

datasheet; for 20 ml, add 1.0 g BSA or nonfat dry milk to 20 ml 1X TBST and mix well.<br />

12. Biotinylated Protein Ladder Detection Pack: (#7727)<br />

13. Prestained Protein Marker, Broad Range (Premixed Format): (#7720)<br />

14. Blotting Membrane and Paper: (#12369) This protocol has been optimized for nitrocellulose membranes. Pore size 0.2 µm<br />

is generally recommended.<br />

15. Secondary Antibody Conjugated to HRP: anti-rabbit (#7074); anti-mouse (#7076)<br />

16. Detection Reagent: LumiGLO ® chemiluminescent reagent and peroxide (#7003) or SignalFire ECL Reagent (#6883)<br />

B. Protein Blotting<br />

A general protocol for sample preparation.<br />

1. Treat cells by adding fresh media containing regulator for desired time.<br />

2. Aspirate media from cultures; wash cells with 1X PBS; aspirate.<br />

3. Lyse cells by adding 1X SDS sample buffer (100 µl per well of 6-well plate or 500 µl for a 10 cm diameter plate).<br />

Immediately scrape the cells off the plate and transfer the extract to a microcentrifuge tube. Keep on ice.<br />

4. Sonicate for 10–15 sec to complete cell lysis and shear DNA (to reduce sample viscosity).<br />

5. Heat a 20 µl sample to 95–100°C for 5 min; cool on ice.<br />

6. Microcentrifuge for 5 min.<br />

7. Load 20 µl onto SDS-PAGE gel (10 cm x 10 cm). NOTE: Loading of prestained molecular weight markers (#7720, 10 µl/lane)<br />

to verify electrotransfer and biotinylated protein ladder (#7727, 10 µl/lane) to determine molecular weights are recommended.<br />

8. Electrotransfer to nitrocellulose membrane (#12369).<br />

C. Membrane Blocking and Antibody Incubations<br />

NOTE: Volumes are for 10 cm x 10 cm (100 cm 2 ) of membrane; for different sized membranes, adjust volumes accordingly.<br />

I. Membrane Blocking<br />

1. (Optional) After transfer, wash nitrocellulose membrane with 25 ml TBS for 5 min at room temperature.<br />

2. Incubate membrane in 25 ml of blocking buffer for 1 hr at room temperature.<br />

3. Wash three times for 5 min each with 15 ml of TBST.<br />

For HRP Conjugated Primary Antibodies<br />

1. Incubate membrane and primary antibody (at the appropriate dilution as recommended in the product datasheet) in 10 ml<br />

primary antibody dilution buffer with gentle agitation overnight at 4°C.<br />

2. Wash 3 times for 5 min each with 15 ml of TBST.<br />

3. Incubate with Anti-biotin, HRP-linked Antibody (#7075 at 1:1000–1:3000), to detect biotinylated protein markers, in 10 ml<br />

of blocking buffer with gentle agitation for 1 hr at room temperature.<br />

4. Wash 3 times for 5 min each with 15 ml of TBST.<br />

5. Proceed with Detection (Section D).<br />

For Biotinylated Primary Antibodies<br />

1. Incubate membrane and primary antibody (at the appropriate dilution as recommended in the product datasheet) in 10 ml<br />

primary antibody dilution buffer with gentle agitation overnight at 4°C.<br />

2. Wash 3 times for 5 min each with 15 ml of TBST.<br />

3. Incubate membrane with Streptavidin-HRP (#3999 at the appropriate dilution) in 10 ml of blocking buffer with gentle<br />

agitation for 1 hr at room temperature.<br />

4. Wash 3 times for 5 min each with 15 ml of TBST.<br />

5. Proceed with Detection (Section D).<br />

Do not add Anti-biotin, HRP-linked Antibody for detection of biotinylated protein markers. There is no need. The Streptavidin-HRP<br />

will also visualize the biotinylated markers.<br />

D. Detection of Proteins<br />

1. Incubate membrane with 10 ml LumiGLO ® (0.5 ml 20X LumiGLO ® #7003, 0.5 ml 20X Peroxide, and 9.0 ml dH 2 O) or<br />

10 ml SignalFire #6883 (5 ml Reagent A, 5 ml Reagent B) with gentle agitation for 1 min at room temperature.<br />

2. Drain membrane of excess developing solution (do not let dry), wrap in plastic wrap and expose to x-ray film. An initial<br />

10 sec exposure should indicate the proper exposure time. NOTE: Due to the kinetics of the detection reaction, signal is<br />

most intense immediately following incubation and declines over the following 2 hr.<br />

Phosphatase inhibitors help preserve phosphorylation.<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

60<br />

50<br />

A<br />

0 3 6 20 0 3 6 20 hrs<br />

B<br />

Phospho-Akt<br />

(Ser 473)<br />

Selection <strong>Guide</strong> for Chemiluminescent Detection<br />

Akt<br />

Protease/Phosphatase Inhibitor Cocktail (100X) #5872:<br />

WB analysis of extracts from NIH/3T3 cells, serum-starved<br />

overnight and treated with hPDGF-BB #8912 (100 ng/ml, 5<br />

min), prepared in lysis buffer in the absence of phosphatase<br />

inhibitors (A) or with #5872 added (B) and was incubated at<br />

37˚C for the indicated time following cell lysis, using Phospho-<br />

Akt (Ser473) (D9E) XP ® Rabbit mAb #4060 (upper) or Akt (pan)<br />

(C67E7) Rabbit mAb #4691 (lower).<br />

Reagent Sensitivity Recommended Application<br />

LumiGLO ®<br />

Sub-picogram sensitivity for recombinant protein General use detection reagent,<br />

Reagent #7003<br />

most versatile option<br />

SignalFire ECL<br />

Reagent #6883<br />

SignalFire Plus ECL<br />

Reagent #12630<br />

SignalFire Elite ECL<br />

Reagent #12757<br />

Mid-picogram sensitivity for recombinant protein<br />

Low-picogram sensitivity for recombinant protein,<br />

with strong signal lasting for many hours<br />

Femtogram sensitivity for recombinant protein, with<br />

low nanogram sensitivity for target protein in cell<br />

extracts<br />

General use detection reagent,<br />

most economical option<br />

Strong and long-lasting signal, useful for<br />

reading assays performed in large batches<br />

Extremely intense signal enables detection<br />

of low abundance endogenous proteins<br />

II. Primary Antibody Incubation<br />

Proceed to one of the following specific set of steps depending on the primary antibody used.<br />

For Unconjugated Primary Antibodies<br />

1. Incubate membrane and primary antibody (at the appropriate dilution and diluent as recommended in the product datasheet)<br />

in 10 ml primary antibody dilution buffer with gentle agitation overnight at 4°C.<br />

2. Wash 3 times for 5 min each with 15 ml of TBST.<br />

3. Incubate membrane with the species appropriate HRP-conjugated secondary antibody (#7074 or #7076 at 1:2000) and<br />

anti-biotin, HRP-linked Antibody (#7075 at 1:1000–1:3000) to detect biotinylated protein markers in 10 ml of blocking<br />

buffer with gentle agitation for 1 hr at room temperature.<br />

4. Wash 3 times for 5 min each with 15 ml of TBST.<br />

5. Proceed with Detection (Section D).<br />

228 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

<strong>CST</strong> Technical Support<br />

When you contact <strong>CST</strong> for technical support, you can be confident you will be working with a colleague you can trust,<br />

striving to provide the highest quality products and services to you, our customer. Please see our Technical Support<br />

resource page online for contact information. www.cellsignal.com/cstsupport<br />

www.cellsignal.com/cstprotocols<br />

229


Section II: ANTIBODY APPLICATIONS<br />

chapter 14: Western Blotting (WB)<br />

WB Fluorescent Protocol<br />

For western blots, incubate membrane with diluted primary antibody in either 5% w/v BSA or nonfat dry milk, 1X TBS, 0.1%<br />

Tween ® 20 at 4°C with gentle shaking, overnight.<br />

NOTE: Two-color western blots require primary antibodies from different species and appropriate secondary antibodies labeled<br />

with different dyes. Overlap of epitopes may cause interference and should be considered in two color western blots. If the primary<br />

antibodies require different primary antibody incubation buffers, test each primary individually in both buffers to determine<br />

the optimal one for the dual-labeling experiment.<br />

A. Solutions and Reagents<br />

NOTE: Prepare solutions with reverse osmosis deionized (RODI) or equivalent grade water.<br />

1. 20X Phosphate Buffered Saline (PBS): (#9808) To prepare 1 L 1X PBS: add 50 ml 20X PBS to 950 ml dH 2<br />

O, mix.<br />

2. 10X Tris Buffered Saline (TBS): (#12498) To prepare 1 L 1X TBS: add 100 ml 10X TBS to 900 ml dH 2 O, mix.<br />

3. 1X SDS Sample Buffer: Blue Loading Pack (#7722) or Red Loading Pack (#7723) Prepare fresh 3X reducing loading buffer<br />

by adding 1/10 volume 30X DTT to 1 volume of 3X SDS loading buffer. Dilute to 1X with dH 2 O.<br />

4. 10X Tris-Glycine SDS Running Buffer: (#4050) To prepare 1 L 1X Running Buffer: add 100 ml 10X running buffer 900 ml<br />

dH 2 O, mix.<br />

5. 10X Tris-Glycine Transfer Buffer: (#12539) To prepare 1 L 1X Transfer Buffer: add 100 ml 10X Transfer Buffer 200 ml<br />

methanol + 700 ml dH 2 O, mix.<br />

6. 10X Tris Buffered Saline with Tween ® 20 (TBST-10X): (#9997) To prepare 1 L 1X TBST: add 100 ml 10X TBST to<br />

900 ml dH 2<br />

O, mix.<br />

7. Nonfat Dry Milk: (#9999)<br />

8. Blocking Buffer: 1X TBS with 5% w/v nonfat dry milk; for 150 ml, add 7.5 g nonfat dry milk to 150 ml 1X TBS and mix<br />

well. Tween ® 20 should not be present in the Blocking Buffer because it is auto-fluorescent and increases non-specific<br />

background. After the blocking step, Tween ® 20 can be reintroduced to subsequent diluent buffers.<br />

9. Wash Buffer: 1X TBST<br />

10. Bovine Serum Albumin (BSA): (#9998)<br />

11. Primary Antibody Dilution Buffer: 1X TBST with 5% BSA or 5% nonfat dry milk as indicated on primary antibody<br />

datasheet; for 20 ml, add 1.0 g BSA or nonfat dry milk to 20 ml 1X TBST and mix well.<br />

12. Secondary Antibody Dilution Buffer: 1X TBST with 5% nonfat dry milk; for 20 ml, add 1.0 g nonfat dry milk to 20 ml<br />

1X TBST and mix well. (Secondary antibodies; anti-rabbit #5151 and #5366; anti-mouse #5257 and #5470)<br />

13. Prestained Protein Marker, Broad Range (Premixed Format): (#7720)<br />

14. Blotting Membrane and Paper: (#12369) This protocol has been optimized for nitrocellulose membranes (recommended).<br />

Pore size 0.2 µm is generally recommended.<br />

B. Protein Blotting<br />

A general protocol for sample preparation.<br />

1. Treat cells by adding fresh media containing regulator for desired time.<br />

2. Aspirate media from cultures; wash cells with cold 1X PBS; aspirate.<br />

3. Lyse cells by adding 1X SDS sample buffer (100 µl per well of 6-well plate or 500 µl per plate of 10 cm diameter plate).<br />

Immediately scrape the cells off the plate and transfer the extract to a microcentrifuge tube. Keep on ice.<br />

4. Sonicate for 10–15 sec to complete cell lysis and shear DNA (to reduce sample viscosity).<br />

5. Heat a 20 µl sample to 95–100°C for 5 min; cool on ice.<br />

6. Microcentrifuge for 5 min.<br />

7. Load 20 µl onto SDS-PAGE gel (10 cm x 10 cm).<br />

NOTE: Loading of prestained molecular weight markers (#7720, 10 µl/lane) is recommended to verify electrotransfer and to<br />

determine molecular weights. Prestained markers are autofluorescent at near-infrared wavelengths.<br />

8. Electrotransfer to nitrocellulose membrane (#12369).<br />

C. Membrane Blocking and Antibody Incubations<br />

NOTE: Volumes are for 10 cm x 10 cm (100 cm 2 ) of membrane; for different sized membranes, adjust volumes accordingly.<br />

1. (Optional) After transfer, wash nitrocellulose membrane with 25 ml TBS for 5 min at room temperature.<br />

2. Incubate membrane in 25 ml of blocking buffer for 1 hr at room temperature. CRITICAL STEP: Do not include Tween ® 20 in<br />

blocking buffer (Section A, Step 8).<br />

3. Wash 3 times for 5 min each with 15 ml of TBST.<br />

4. Incubate membrane and primary antibody (at the appropriate dilution as recommended in the product datasheet) in 10 ml<br />

primary antibody dilution buffer with gentle agitation overnight at 4°C.<br />

5. Wash 3 times for 5 min each with 15 ml of TBST.<br />

6. Incubate membrane with fluorophore-conjugated secondary antibody (#5470, #5257, #5366, #5151) (1:5000–1:25,000<br />

dilution of 1 mg/ml stock) in 10 ml of secondary antibody dilution buffer with gentle agitation for 1 hr at room temperature.<br />

7. Wash 3 times for 5 min each with 15 ml of TBST.<br />

In the absence of protease inhibitors, β-Catenin signal<br />

fades within 3 hours after harvest. In the presence of<br />

protease inhibitors, β-Catenin is significantly slowed.<br />

Protease/Phosphatase Inhibitor Cocktail (100X) #5872:<br />

WB analysis of extracts from NIH/3T3 cells, prepared in lysis<br />

buffer in the absence of protease inhibitors (A) or with #5872<br />

added (B), and incubated at 37ºC for the indicated time points,<br />

using β-Catenin (D10A8) XP ® Rabbit mAb #8480. In the absence<br />

of protease inhibitors, β-Catenin signal fades within 3 hr after<br />

harvest, indicating protein degradation. In the presence of the<br />

protease inhibitor cocktail, the β-Catenin degradation is slowed<br />

significantly and signal is still present at 20 hr following harvest.<br />

Loading Control Antibodies<br />

It is important to be certain that WB band intensities reflect the experimental treatment and are not<br />

attributable to variability in preparing or loading cell extracts. Loading control antibodies should detect a<br />

relatively abundant protein that does not vary significantly under experimental conditions. For example,<br />

the “housekeeping” enzyme GAPDH is commonly employed as a loading control for short-term signaling<br />

studies, but it may not be the optimal loading control in samples where metabolic pathways are affected.<br />

This table will help you select loading controls on the basis of sample type and subunit molecular<br />

weight. By choosing a control with a subunit molecular weight different than your target protein, you<br />

have the option of detecting the loading control in the same lane as your target protein.<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

0 3 6 20 hrs<br />

β-Catenin<br />

degradation<br />

product<br />

kDa<br />

Whole Cell /<br />

Cytoplasmic Nuclear Mitochondrial Cytoskeletal Plasma Membrane<br />

Serum &<br />

Extracellular Fluids<br />

230 Myosin IIb<br />

135 Cadherin<br />

124 Vinculin Vinculin<br />

116 PARP<br />

100 α-Actinin NaK-ATPase<br />

90 HSP 90<br />

89 PARP<br />

85 CD71<br />

77 Transferrin<br />

74 Lamin A/C<br />

70 HSP 70<br />

68 Lamin B1<br />

63 Lamin A/C<br />

62 HDAC 1<br />

60 HSP 60 HSP 60<br />

52 α-Tubulin<br />

45 MEK1/2 Lamin B1 β-Actin<br />

38 TBP<br />

37 GAPDH<br />

36 PCNA<br />

35 β-Tubulin<br />

34 VDAC/Porin<br />

24 Caveolin-1<br />

21 Caveolin-1<br />

19 Cofilin<br />

17 Histone H3 COX IV<br />

9 Profilin 1<br />

5 Profilin 1<br />

A<br />

0 3 6 20<br />

B<br />

D. Detection of Proteins<br />

1. Drain membrane of excess TBST and allow to dry. CRITICAL STEP: Membrane must be dry for fluorescent detection.<br />

2. Scan membrane using an appropriate fluorescent scanner following the manufacturer’s recommendations.<br />

230 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys. www.cellsignal.com/cstprotocols<br />

231


Section II: ANTIBODY APPLICATIONS<br />

chapter 14: Western Blotting (WB)<br />

WB Troubleshooting <strong>Guide</strong><br />

High Background:<br />

General background is high or nonspecific bands that appear after a short exposure of blot to film. Be conscious of<br />

the fact that motif antibodies, post-translational modifications, and splice variants may result in multiple banding.<br />

Cause<br />

Solution<br />

Lysate<br />

preparation<br />

Membrane<br />

Primary antibody dilution<br />

and/or incubation<br />

Inadequate<br />

washing<br />

Secondary antibody<br />

dilution and/or incubation<br />

Detection reagent<br />

Exposure time<br />

Freshly prepared samples result in fewer nonspecific and degradation bands, and therefore<br />

yield a cleaner blot. In general, tissue extracts tend to contain more background bands and<br />

degradation products than cell line extracts due to connective tissue. Using fresh, sonicated<br />

and clarified tissue extracts may lessen background. Samples should always be lysed in<br />

appropriate buffers that include protease inhibitors and phosphatase inhibitors for phosphotargets<br />

(Phosphatase Inhibitor Cocktail (100X) #5870, Protease/Phosphatase Inhibitor Cocktail<br />

(100X) #5872, Protease Inhibitor Cocktail #5871). We recommend the use of Cell Lysis Buffer<br />

(#9803) or RIPA Buffer (#9806), which contains stronger detergent, for sample preparation<br />

and protein concentration quantification. SDS loading buffer (#7722 or #7723) may be<br />

used for whole cell lysis without protein quantification. Chaps Cell Extract Buffer (#9852) is<br />

used to prepare cytoplasmic cell fractions and is useful for studying caspase signaling. Cell<br />

Fractionation Kit (#9038) is used to prepare cytoplasmic, membrane/organelle, and nuclear/<br />

cytoskeletal cell fractions.<br />

Use high quality nitrocellulose membrane (Nitrocellulose Sandwiches #12369). Pore size<br />

0.2 µm is generally recommended; membranes with a pore size of 0.45 µm are not recommended<br />

for proteins smaller than 30 kDa. PVDF membranes may yield higher background<br />

than nitrocellulose. Nylon membranes are not recommended for western blotting. Block for<br />

1 hr at room temperature in 5% nonfat dry milk in TBST.<br />

Incubate primary antibody overnight at 4°C in TBST at the recommended dilution with the<br />

recommended blocking agent (either 5% BSA or 5% nonfat dry milk). For individual antibodies<br />

please consult the product datasheet for recommended dilution buffer.<br />

Washing for less time than the recommended 3 times 5 min in TBST is common, and can<br />

result in high background. We recommend that washes be timed to ensure accuracy. Additionally,<br />

low Tween ® 20 can contribute to high background; we recommend 0.1% Tween ® 20.<br />

This applies to washing steps after both primary and secondary antibody incubations.<br />

Some secondary antibodies bind nonspecifically to proteins in cell extracts. To assess the<br />

quality of a secondary antibody, perform a blot (through to film exposure) without primary antibody.<br />

Serial dilutions of the secondary antibody can be performed on blots with the same cell<br />

extracts and primary antibody to optimize secondary antibody concentration. Always incubate<br />

the secondary antibody in 5% nonfat dry milk in TBST for 1 hr at room temperature, diluting<br />

the secondary antibody in BSA yields higher levels of background bands. <strong>CST</strong> secondary<br />

antibodies have already been optimized and do not require titration.<br />

Chemiluminescent detection requires that high purity water be used to dilute concentrated<br />

reagents. Use only purified water with organic and inorganic impurities removed. We recommend<br />

using RODI water. Additionally, ensure that the membrane is never allowed to dry out<br />

during the antibody incubation process prior to detection reagent exposure.<br />

Film exposure times of more than 30 sec lead to increased background signal. To avoid long exposure<br />

times, it is important to use cell lines or tissues with adequate protein expression levels<br />

and use recommended primary antibody incubation conditions (see primary antibody dilution<br />

and/or incubation section). If necessary, use treatment to induce expression or modification.<br />

Low Signal:<br />

The protein of interest cannot be detected after a short exposure of blot to film.<br />

Cause<br />

Solution<br />

Lysate preparation 20–30 µg total protein from whole cell extracts per lane is usually sufficient for detection. If<br />

basal levels of target protein, or protein modification are low, it may be necessary to induce<br />

expression or modification via chemical stimulant. You may want to investigate alternative cell<br />

lines or tissues in which the protein of interest is more abundant. Samples should always be<br />

lysed in appropriate buffers that include protease inhibitors and phosphatase inhibitors, for<br />

phospho-targets (Phosphatase Inhibitor Cocktail (100X) #5870, Protease/Phosphatase Inhibitor<br />

Cocktail (100X) #5872, Protease Cocktail #5871). Lysates should always be sonicated to<br />

ensure efficient protein extraction of chromatin and membrane-bound targets. Please visit the<br />

Controls Table on our website for suggested positive controls and treatments.<br />

Primary antibody dilution<br />

and/or incubation<br />

SDS-PAGE Gel selection<br />

Transfer and Membrane<br />

Blocking<br />

Washing<br />

Secondary antibody<br />

dilution and/or incubation<br />

Incubate primary antibody overnight at 4°C in TBST at the recommended dilution with the<br />

recommended blocking agent (5% BSA or 5% nonfat dry milk). The use of alternate blocking<br />

agents, such as gelatin, serum, protein-free blocking agents, casein, or mixed blocking agents<br />

may reduce target signal intensily. For optimal results with individual antibodies, please consult<br />

the product datasheet for recommended dilution buffer.<br />

In general, Tris-Glycine gels are recommended. However, for high molecular weight proteins<br />

we recommend Tris-Acetate gels and associated buffers. Proteins transfer more efficiently<br />

from 1 mm gels than from 1.5 mm gels. The use of thicker gels can result in incomplete<br />

transfer of high molecular weight proteins, so we recommend monitoring transfer efficiency<br />

and implementing modifications of transfer conditions to optimize for the size of your protein<br />

target of interest.<br />

Incomplete transfer can be corrected by longer transfer or by the use of higher voltage. In general,<br />

we recommend including 20% methanol in transfer buffer, and performing a wet transfer<br />

for 1.5 hr at 70 V. For high molecular weight proteins, we recommend reducing the methanol<br />

in transfer buffer to 5% to improve transfer efficiency and avoid fixing large proteins in the<br />

gel matrix, as well as increasing transfer time to 3 hr at 70 V. Over transfer (protein transfer<br />

through membrane) can be problematic when examining smaller proteins. For small proteins,<br />

it is important to use a 0.2 µm pore size membrane and wet transfer for 1.5 hr at 70 V, not a<br />

0.45 µm pore size membrane or transferring overnight, which may result in over transfer and<br />

a lack of small protein signal.<br />

Blocking the membrane for too long can obscure antigenic epitopes and prevent the antibody<br />

from binding. Block for only 1 hr at room temperature.<br />

Washing for longer than the recommended 3 times 5 min is a common issue and can result<br />

in reduced signal. We recommend that washes be timed to ensure accuracy. TBST should<br />

contain 0.1% Tween ® 20. This applies to washing steps after both primary and secondary<br />

antibody incubations. Additionally, we recommend washing in TBST, as washing in PBST has<br />

been shown to result in reduced signal.<br />

Serial dilutions of the secondary antibody can be performed on blots with the same cell<br />

extracts and primary antibody to optimize secondary antibody concentration. Always incubate<br />

the secondary antibody in 5% nonfat dry milk in TBST for 1 hr at room temperature. Avoid<br />

including sodium azide in HRP-conjugated secondary antibody buffers as azide inhibits HRP<br />

activity. <strong>CST</strong> secondary antibodies have already been optimized and do not require titration.<br />

Detection reagent<br />

Use biotinylated molecular weight standards that can be detected with anti-biotin-HRP as<br />

positive controls for chemiluminescent detection.<br />

WB detection of<br />

subcellular protein<br />

markers verifies<br />

effectiveness of<br />

detergent-based<br />

cell fractionation.<br />

kDa<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

A B C D<br />

1 2 3 4 1 2 3 4 1 2 3 4 1 2 3 4<br />

Cell Fractions<br />

1. Whole cell lysates<br />

2. Cytoplasmic<br />

3. Membrane<br />

4. Nuclear/Cytoskeletal<br />

Cell Fractionation Kit #9038: WB analysis of HeLa cell fractions were prepared using #9038. Subcellular localization was determined<br />

using antibodies from the Cell Fractionation Antibody Sampler Kit #11843 [MEK1/2 (D1A5) Rabbit mAb #8727 (A), AIF (D39D2) XP ® Rabbit<br />

mAb #5318 (B), Histone H3 (D1H2) XP ® Rabbit mAb #4499 (C), and Vimentin (D21H3) XP ® Rabbit mAb #5741 (D)].<br />

<strong>CST</strong> Technical Support<br />

When you contact <strong>CST</strong> for technical support, you can be confident you will be working with a colleague you can trust,<br />

striving to provide the highest quality products and services to you, our customer. Please see our Technical Support<br />

resource page online for contact information. www.cellsignal.com/cstsupport<br />

232 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstprotocols<br />

233


15<br />

Section II: ANTIBODY APPLICATIONS<br />

For targets that run<br />

near 50 kDa, use<br />

light chain-specific or<br />

conformation-specific<br />

secondary antibody to<br />

avoid obscuring the<br />

intended target band.<br />

Immunoprecipitation (IP)<br />

Protein Enrichment<br />

Immunoprecipitation (IP) uses antibodies bound to protein A or G beads to isolate target proteins from<br />

a cell lysate. IP is often used as an enrichment strategy to study low abundance proteins that cannot<br />

be detected directly by western blot. IP is also used to identify members of protein complexes, as<br />

proteins associated with the target protein will co-precipitate with the antibody-bound beads. Proteins<br />

are either eluted off the beads prior to western blot, or the bead-bound protein can be directly analyzed<br />

by western blot.<br />

The stringency of IP buffers is key for experimental success. High stringency buffers that contain<br />

anionic detergents such as SDS may work for IP but are not recommended for co-IP experiments, as<br />

they dissociate protein complexes. Medium and low stringency buffers that contain nonionic detergents<br />

such as NP-40 or Triton X-100 are better suited for co-IP experiments because they allow protein<br />

associations to remain intact. Successful IP and co-IP experiments require specific and sensitive antibodies<br />

to minimize enrichment of non-specific proteins. While unconjugated antibodies are commonly<br />

used for IP experiments, alternative detection options include biotinylated primary antibodies combined<br />

with streptavidin beads or primary antibodies directly conjugated to IP beads. IP beads are available in<br />

agarose, Sepharose ® , or magnetic-bead formats. Agarose and Sepharose beads have a higher protein<br />

binding capacity, but require multiple centrifugation steps, which increases experimental time and<br />

can lead to loss of beads due to pipetting. Magnetic beads are a convenient option as the beads are<br />

precipitated using a magnetic rack, thereby eliminating several centrifugation steps.<br />

A common challenge presented by IP analysis is the presence of an IgG chain band obscuring the<br />

western blot band of interest. The tips below offer options to clarify your results by avoiding crossreacting<br />

IgG bands.<br />

IP Tips for Success<br />

Use conformation-specific secondary antibodies for WB analysis<br />

of an IP if the target size is similar to an IgG subunit size.<br />

The antibody used for IP remains in the sample and often appears on the subsequent western blot as<br />

bands at 50 kDa (IgG heavy chain) and 25 kDa (IgG light chain). The conformation-specific secondary<br />

antibody does not recognize denatured heavy or light IgG chains, so the 25 and 50 kDa bands do not<br />

appear on the western blot. The light chain-specific secondary antibody only recognizes the IgG light<br />

chain, so the 50 kDa heavy chain does not appear on the blot.<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

A B C<br />

1 2 1 2 1 2<br />

Heavy<br />

Chain<br />

PRAS40<br />

Where possible, use primary antibodies<br />

from different species for the IP and the WB.<br />

IgG chains from the IP antibody are recognized by the secondary antibody when the western blot<br />

primary antibody is raised in the same host species (For example, the IgG chains will be detected by<br />

the anti-rabbit secondary antibody when rabbit antibodies are used for IP and western blot). Using IP<br />

and western blot antibodies raised in different species avoids secondary antibody recognition of the IgG<br />

heavy chain and light chain from the IP.<br />

234 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

Light<br />

Chain<br />

Mouse Anti-rabbit IgG (Light-Chain Specific) (L57A3) mAb<br />

#3677: IP of PRAS40 from untreated HeLa cells using PRAS40 (D23C7)<br />

Rabbit mAb #2691. Secondary antibodies include Mouse Anti-rabbit<br />

IgG (Conformation Specific) (L27A9) mAb #3678 (A), #3677 (B), and<br />

Anti-rabbit IgG, HRP-linked Antibody #7074 (C). The bound Mouse<br />

Anti-Rabbit IgG mAb was detected by Anti-mouse IgG, HRP-linked<br />

Antibody #7076 (A,B). The positions of the reduced and denatured<br />

rabbit IgG heavy and light chains are indicated.<br />

Lanes<br />

1. 10% input of untreated HeLa cells<br />

2. IP of PRAS40 from untreated HeLa cells using PRAS40 (D23C7)<br />

Rabbit mAb #2691<br />

Akt (pan) (C67E7) Rabbit mAb #4691 and Akt<br />

(pan) (40D4) Mouse mAb #2920: IP of Akt from 293<br />

cells treated with Human Insulin-like Growth Factor I<br />

(hIGF-I) #8917. WB analysis performed with #4691 and<br />

Anti-rabbit IgG, HRP-linked Antibody #7074 (A) or with<br />

#2920 and Anti-mouse IgG, HRP-linked Antibody #7076<br />

(B). Note in lane 5 that the rabbit IgG heavy chain is<br />

recognized by #7074 (A), but not by #7076 (B).<br />

Lanes<br />

1. Beads and 293 + hIGF-I lysate control<br />

2. Rabbit IgG control<br />

3. Mouse IgG control<br />

4. 10% input of 293 + hIGF-I lysate (no IP)<br />

5. IP: Akt (pan) (C67E7) Rabbit mAb #4691<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

IP Native Protein Protocol<br />

This protocol is intended for IP of native proteins for analysis by western immunoblotting or kinase activity.<br />

20<br />

A<br />

B<br />

1 2 3 4 5 1 2 3 4 5<br />

A. Solutions and Reagents<br />

NOTE: Prepare solutions with reverse osmosis deionized (RODI) or equivalent grade water.<br />

1. 20X Phosphate Buffered Saline (PBS): (#9808) To prepare 1 L of 1X PBS, add 50 ml 20X PBS to 950 ml dH 2<br />

O, mix.<br />

2. 10X Cell Lysis Buffer: (#9803) To prepare 10 ml of 1X cell lysis buffer, add 1 ml cell lysis buffer to 9 ml dH 2<br />

O, mix.<br />

NOTE: Add 1 mM PMSF (#8553) immediately prior to use.<br />

3. 3X SDS Sample Buffer: Blue Loading Pack (#7722) or Red Loading Pack (#7723) Prepare fresh 3X reducing loading buffer<br />

by adding 1/10 volume 30X DTT to 1 volume of 3X SDS loading buffer.<br />

4. Protein A or G Agarose Beads (For unconjugated primary antibodies): Use Protein A (#9863, #8687) for rabbit IgG IP<br />

and Protein G (#8740) for mouse IgG IP. NOTE: Magnetic beads (#8687, #8740) require 6-Tube Magnetic Separation Rack<br />

(#7017).<br />

5. Immobilized Streptavidin (Bead Conjugate) (For biotinylated antibodies): (#3419) Gently vortex vial and use 10 µl per IP.<br />

6. 10X Kinase Buffer (for kinase assays): (#9802) To prepare 1 ml of 1X kinase buffer, add 100 µl 10X kinase buffer to<br />

900 µl dH 2 O, mix.<br />

7. ATP (10 mM) (for kinase assays): (#9804) To prepare 0.5 ml of ATP (200 µM), add 10 µl ATP (10 mM) to 490 µl 1X<br />

kinase buffer.<br />

B. Preparing Cell Lysates<br />

1. Aspirate media. Treat cells by adding fresh media containing regulator for desired time.<br />

2. To harvest cells under nondenaturing conditions, remove media and rinse cells once with ice-cold 1X PBS.<br />

3. Remove PBS and add 0.5 ml ice-cold 1X cell lysis buffer to each plate (10 cm) and incubate on ice for 5 min.<br />

4. Scrape cells off the plate and transfer to microcentrifuge tubes. Keep on ice.<br />

5. Sonicate on ice 3 times for 5 sec each.<br />

6. Microcentrifuge for 10 min at 4°C, 14,000 x g and transfer the supernatant to a new tube. The supernatant is the cell lysate.<br />

If necessary, lysate can be stored at -80°C.<br />

C. Immunoprecipitation<br />

Cell Lysate Pre-Clearing (Optional step for unconjugated and biotinylated antibodies.)<br />

1. Add 10–30 µl of 50% bead slurry, either Protein A or G agarose or magnetic beads (for unconjugated primary antibodies) or<br />

10 µl streptavidin beads (#3419; for biotinylated antibodies), to 200 µl cell lysate at 1 mg/ml.<br />

2. Incubate with rotation at 4°C for 30–60 min.<br />

3. Microcentrifuge for 10 min at 4°C. Transfer the supernatant to a fresh tube.<br />

4. Proceed to one of the following specific set of steps depending on the primary antibody used.<br />

Using Unconjugated Primary Antibodies<br />

1. Add primary antibody (at the appropriate dilution as recommended in the product datasheet) to 200 µl cell lysate at 1 mg/ml.<br />

Incubate with gentle rocking overnight at 4°C.<br />

2. Add either protein A or G agarose or magnetic beads (10–30 µl of 50% bead slurry). Incubate with gentle rocking for 1–3 hr<br />

at 4°C for agarose beads, or 10–30 min for magnetic beads.<br />

3. Microcentrifuge for 30 sec at 4°C. Wash pellet five times with 500 µl of 1X cell lysis buffer. Keep on ice between washes.<br />

4. Proceed to Analyze by Western Immunoblotting or Analyze by Kinase Assay (Section D).<br />

Using Biotinylated Primary Antibodies<br />

1. Add biotinylated antibody (at the appropriate dilution as recommended in the product datasheet) to 200 µl cell lysate<br />

at 1 mg/ml. Incubate with gentle rocking overnight at 4°C.<br />

2. Gently mix Immobilized Streptavidin (Sepharose ® Bead Conjugate #3419) and add 10 µl of slurry. Incubate with gentle<br />

rocking for 2 hr at 4°C.<br />

3. Microcentrifuge for 30 sec at 4°C. Wash pellet 5 times with 500 µl of 1X cell lysis buffer. Keep on ice during washes.<br />

4. Proceed to Sample Analysis (Section D).<br />

chapter 15: Immunoprecipitation (IP)<br />

Akt<br />

IgG<br />

heavy<br />

chain<br />

IgG<br />

light<br />

chain<br />

Use a mouse derived<br />

western blot antibody<br />

to avoid recognition<br />

of IP antibody rabbit<br />

IgG chains.<br />

Use magnetic beads<br />

to reduce protocol<br />

time by skipping<br />

centrifugation steps.<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

1 2<br />

HC<br />

Syntaxin 6<br />

Protein A Magnetic Beads #8687:<br />

IP of Syntaxin 6 from COS-7 cells using<br />

Syntaxin 6 (C34B2) Rabbit mAb #2869<br />

and #8687. WB analysis was performed<br />

using Syntaxin 6 (C34B2) Rabbit mAb<br />

#2869. Magnetic beads shorten the IP<br />

protocol by eliminating centrifugation<br />

steps after washing and also reduce loss<br />

of beads due to pipetting.<br />

Lanes<br />

1. IP pellet<br />

2. IP supernatant<br />

www.cellsignal.com/cstip<br />

235


Section II: ANTIBODY APPLICATIONS<br />

Using Immobilized Antibodies (Sepharose ® Bead Conjugate)<br />

1. Vortex gently and add immobilized bead conjugate (10 µl) to 200 µl cell lysate at 1 mg/ml. Incubate with gentle rocking<br />

overnight at 4°C.<br />

2. Microcentrifuge for 30 sec at 4°C. Wash pellet five times with 500 µl of 1X cell lysis buffer. Keep on ice during washes.<br />

3. Proceed to Sample Analysis (Section D).<br />

Using Immobilized Antibodies (Magnetic Bead Conjugate)<br />

1. Vortex gently and add immobilized bead conjugate (10 µl) to 200 µl cell lysate at 1 mg/ml. Incubate with gentle rocking<br />

overnight at 4°C.<br />

2. Pellet magnetic beads by placing the tubes in a magnetic separation rack (#7017) and wait 1–2 min for solution to clear.<br />

Wash pellet five times with 500 µl of 1X cell lysis buffer. Keep on ice during washes.<br />

3. Proceed to Sample Analysis (Section D).<br />

D. Sample Analysis<br />

Proceed to one of the following specific set of steps. NOTE: For magnetic beads, do not centrifuge. Instead use a magnetic<br />

separation rack (#7017).<br />

For Analysis by Western Immunoblotting<br />

1. Resuspend the pellet with 20 µl 3X SDS sample buffer. Vortex, then microcentrifuge for 30 sec.<br />

2. Heat the sample to 95–100°C for 2–5 min and microcentrifuge for 1 min at 14,000 x g.<br />

3. Load the sample (15–30 µl) on a 4–20% gel for SDS-PAGE.<br />

4. Analyze sample by WB General Protocol (Section B, Step 7).<br />

NOTE: For proteins with molecular weights near 50 kDa, we recommend using Mouse Anti-rabbit IgG (Light-Chain Specific)<br />

(L57A3) mAb #3677 or Mouse Anti-rabbit IgG (Conformation Specific) (L27A9) mAb #3678 as a secondary antibody to<br />

minimize masking produced by denatured heavy chains. For proteins with molecular weights near 25 kDa, Mouse Anti-rabbit<br />

IgG (Conformation Specific) (L27A9) mAb #3678 or Mouse Anti-rabbit IgG (Conformation Specific) (L27A9) mAb (HRP Conjugate)<br />

#5127 is recommended.<br />

For Analysis by Kinase Assay<br />

1. Wash pellet twice with 500 µl 1X kinase buffer. Keep on ice.<br />

2. Suspend pellet in 40 µl 1X kinase buffer supplemented with 200 µM ATP and appropriate substrate.<br />

3. Incubate for 30 min at 30°C.<br />

4. Terminate reaction with 20 µl 3X SDS sample buffer. Vortex, then microcentrifuge for 30 sec.<br />

5. Transfer supernatant containing phosphorylated substrate to another tube.<br />

6. Heat the sample to 95–100°C for 2–5 min and microcentrifuge for 1 min at 14,000 x g.<br />

7. Load the sample (15–30 µl) on SDS-PAGE (4–20%).<br />

IP Denatured Protein Protocol<br />

This protocol is intended for immunoprecipitation of denatured proteins where the epitope of interest may not be accessible in<br />

the native conformation.<br />

A. Solutions and Reagents<br />

NOTE: Prepare solutions with reverse osmosis or equivalently purified water (dH 2 O).<br />

1. Denaturing Cell Lysis Buffer: 50 mM Tris (pH 7.5), 70 mM β-Mercaptoethanol (β-ME) Add β-ME just prior to use. Pre-boil<br />

for 10 min.<br />

2. 10X Cell Lysis Buffer: (#9803) 20 mM Tris (pH 7.5), 150 mM NaCl, 1 mM EDTA, 1 mM EGTA, 1% Triton X-100, 2.5 mM<br />

sodium pyrophosphate, 1 mM β-glycerophosphate, 1 mM Na 3 VO 4 , 1 µg/ml Leupeptin<br />

To prepare 10 ml of 1X Cell Lysis Buffer, add 1 ml of 10XCell Lysis Buffer to 9 ml dH 2 O, mix. NOTE: Add 1 mM PMSF (#8553)<br />

immediately prior to use.<br />

3. Protein A or G Agarose Beads (For unconjugated primary antibodies): Use Protein A (#9863, #8687) for rabbit IgG IP<br />

and Protein G (#8740) for mouse IgG IP.<br />

4. 3X SDS Loading Buffer: (#7722) 187.5 mM Tris-HCl (pH 6.8 at 25°C), 6% w/v SDS, 30% glycerol, 150 mM DTT, 0.03%<br />

w/v bromophenol blue<br />

B. Preparing Cell Lysates<br />

1. Aspirate media. Treat cells by adding fresh media containing regulator for desired time.<br />

2. To harvest cells under denaturing conditions, remove media and rinse cells once with 1X PBS.<br />

3. Remove 1X PBS and add 0.4 ml Denaturing Cell Lysis Buffer (preboiled for 10 min immediately prior to use) to each plate<br />

(150 x 25 mm).<br />

4. Immediately scrape cells off the plate and transfer to 2.5 ml tubes.<br />

5. Boil for 10 min.<br />

6. Add 4 volumes (1.6 ml) 1X ice-cold Cell Lysis Buffer. This is the cell lysate. If necessary, lysate can be stored at –80°C.<br />

C. Immunoprecipitation<br />

1. Add primary antibody to 200 µl cell lysate; incubate with gentle rocking overnight at 4°C.<br />

2. Add Protein A Agarose Beads (20 µl of 50% bead slurry). Incubate with gentle rocking for 1–3 hr at 4°C.<br />

3. Wash pellet 5 times with 500 µl of 1X Cell Lysis Buffer. microcentrifuge for 30 sec at 4°C. Keep on ice during washes.<br />

4. Resuspend the pellet with 20 µl 3X SDS Loading Buffer. Vortex, then microcentrifuge for 30 sec.<br />

5. Heat the sample to 95–100°C for 2–5 min and microcentrifuge for 1 min at 14,000 x g.<br />

6. Load the sample (15–30 µl) on a 4–20% gel for SDS-PAGE.<br />

7. Analyze sample by WB General Protocol (Section B, Step 7).<br />

chapter 15: Immunoprecipitation (IP)<br />

Use IgG isotype controls to estimate the<br />

nonspecific binding of primary antibodies.<br />

Rabbit (DA1E) mAb IgG XP ® Isotype Control #3900: IP of XAF1 from Jurkat cells<br />

treated with Human Interferon-α1 (hIFN-α1) #8927 (10 ng/ml, overnight) using #3900<br />

(lane 2) or #13805 (lane 3). WB analysis was performed using XAF1 (E1E4O) Rabbit mAb<br />

#13805. In this example, there is minimal nonspecific binding of primary antibodies.<br />

Lanes<br />

1. 10% Lysate input<br />

2. Rabbit (DA1E) mAb IgG XP ® Isotype Control #3900<br />

3. XAF1 (E1E4O) Rabbit mAb #13805<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

1 2 3<br />

IgG heavy<br />

chain<br />

XAF1<br />

20<br />

– + –<br />

– – +<br />

Rabbit (DA1E) mAb IgG<br />

XP ® Isotype Control<br />

XAF1 (E1E4O) Rabbit mAb<br />

<strong>CST</strong> Technical Support<br />

When you contact <strong>CST</strong> for technical support, you can be confident you will be working with a colleague you can trust,<br />

striving to provide the highest quality products and services to you, our customer. Please see our Technical Support<br />

resource page online for contact information. www.cellsignal.com/cstsupport<br />

236 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstprotocols<br />

237


16<br />

Section II: ANTIBODY APPLICATIONS<br />

chapter<br />

Control Treatments and Cell Lines<br />

This table displays cell lines, treatments, and <strong>CST</strong> Control Cell Extracts that can be used as positive<br />

controls for activation-state specific antibodies. The information found in this table should be seen as a<br />

starting point for troubleshooting.<br />

Autophagy<br />

LC3A<br />

LC3B<br />

Cell Line Treatment Control<br />

HeLa, KNRK,<br />

NIH/3T3<br />

HeLa, KNRK,<br />

NIH/3T3<br />

16: Control Treatments and Cell Lines<br />

Chloroquine (50 μM for 20 hr) #11972<br />

Chloroquine (50 μM for 20 hr) #11972<br />

Phospho-ULK1 (Ser555) MCF7 Oligomycin (0.5 µM for 30 min)<br />

To demonstrate phospho-specificity,<br />

cell extracts or nitrocellulose<br />

membranes can be subjected to<br />

phosphatase treatment.<br />

All UV treatments were carried out<br />

using a Stratagene Stratalinker ® UV<br />

Crosslinker.<br />

Where “transfected” is indicated<br />

in the cell line column, transfected<br />

levels of the respective target<br />

protein were detected.<br />

Control<br />

Treatments<br />

by Target<br />

Please visit our website for the<br />

most up-to-date listing of control<br />

treatments and cell lines.<br />

www.cellsignal.com/cstcontrols<br />

Cell Line Treatment Control<br />

Adhesion and Extracellular Matrix<br />

Phospho-Afadin (Ser1718) A-431 Serum-starve overnight,<br />

hEGF (100 ng/ml for 5 min)<br />

Phospho-Catenin δ-1 (Tyr228/Tyr904) A-431 Serum-starve overnight,<br />

hEGF (100 ng/ml for 15 min)<br />

Phospho-Connexin 43 (Ser368) COS-7 Serum-starve overnight,<br />

TPA (200 nM for 30 min)<br />

Phospho-FAK (Tyr397/Tyr576/Tyr577/Tyr925) A549 Untreated<br />

Phospho-p130 Cas (Tyr165/Tyr249/Tyr410) NIH/3T3 Untreated<br />

Phospho-Paxillin (Tyr118)<br />

C2C12, A-431,<br />

C6 NIH/3T3<br />

Untreated<br />

Apoptosis<br />

Cleaved Caspase-1 (Asp297) THP-1 TPA (80 nM overnight) plus<br />

LPS (1 µg/ml for 8 hr)<br />

Cleaved Caspase-3 (Asp175) HeLa Serum-starve overnight,<br />

#9663<br />

Staurosporine (1 μM for 3 hr)<br />

Cleaved Caspase-6 (Asp162) HeLa Serum-starve overnight,<br />

#9663<br />

Staurosporine (1 μM for 3 hr)<br />

Cleaved Caspase-7 (Asp198) HeLa Serum-starve overnight,<br />

#9663<br />

Staurosporine (1 μM for 3 hr)<br />

Cleaved Caspase-8 (Asp391/Asp384/Asp387) Jurkat Etoposide (25 μM for 5 hr) #2043<br />

Cleaved Caspase-9 (Asp315/Asp330) Jurkat Etoposide (25 μM for 5 hr) #2043<br />

Cleaved DFF45 (Asp224) Jurkat Etoposide (25 µM for 5 hr) #2043<br />

Cleaved Lamin A (Asp230) HeLa Serum-starve overnight,<br />

#9663<br />

Staurosporine (1 μM for 4 hr)<br />

Cleaved Lamin A (Small Subunit) HeLa Serum-starve overnight,<br />

#9663<br />

Staurosporine (1 μM for 4 hr)<br />

Cleaved PARP (Asp214) HeLa Serum-starve overnight,<br />

#2043<br />

Staurosporine (1 µM for 3 hr)<br />

Cleaved α-Fodrin (Asp1185) HeLa Serum-starve overnight,<br />

#9663<br />

Staurosporine (1 μM for 4 hr)<br />

Phospho-AP2M1 (Thr156) HeLa H 2 O 2 (4 mM for 30 min) #9663<br />

Phospho-Bad (Ser112/Ser136/Ser155) COS-7 Serum-starve overnight,<br />

TPA (200 nM for 30 min)<br />

Phospho-Bcl-2 (Ser70) Jurkat Paclitaxel (1 μM for 20 hr)<br />

Phospho-Bcl-2 (Thr56) RL-7 Nocadazole (1 μg/ml for 20 hr)<br />

Phospho-Bim (Ser55/Ser69) A20 Serum-starve overnight,<br />

TPA (200 nM for 30 min)<br />

Phospho-c-Myc (Thr58/Ser62) A-431 Serum-starve overnight,<br />

TPA (200 nM for 30 min)<br />

Phospho-Caspase-9 (Thr125) MCF7 hEGF (10 ng/ml for 30 min)<br />

Phospho-FADD (Ser194) Jurkat Inhibit with hydroxyurea<br />

(4.0 mM for 24 hr); activate with<br />

Nocodazole (1.0 μg/ml) for 24 hr<br />

Phospho-Lamin A/C (Ser22) THP-1 Nocadazole (1 μg/ml for 20 hr)<br />

Phospho-Mcl-1 (Ser159/Thr163) H929 MG132 (10 μM for 2 hr)<br />

Phospho-Mst1 (Thr183)/Mst2 (Thr180) WEHI Staurosporine (1 µM for 30 min–3 hr)<br />

Phospho-PAR-4 (Thr163) HeLa Calyculin A (100 nM for 5 min)<br />

Phospho-PEA-15 (Ser104) C6 TPA (200 nM for 1 hr)<br />

238 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

#9293<br />

Calcium, cAMP and Lipid Signaling<br />

Phospho-cPLA2 (Ser505) HeLa Serum-starve overnight,<br />

TPA (200 nM for 15 min)<br />

Phospho-IP3 Receptor (Ser1756)<br />

mouse & rat<br />

brains<br />

Phospho-MARCKS (Ser152/Ser156) HeLa, NIH/3T3 Serum-starve overnight,<br />

TPA (200 nM for 15 min)<br />

Phospho-PKA C (Thr197) HeLa Untreated<br />

Phospho-PKC (pan) HeLa, NIH/3T3 Serum-starve overnight,<br />

TPA (200 nM for 15 min)<br />

Phospho-PKD/PKCμ (Ser744/Ser748/Ser916) HeLa, NIH/3T3 Serum-starve overnight,<br />

TPA (200 nM for 15 min)<br />

Phospho-PKM2 (Tyr105) A549 Untreated<br />

Phospho-PLCβ3 (Ser537/Ser1105) HeLa Serum-starve overnight,<br />

TPA (200 nM for 15 min)<br />

Phospho-PLCγ1 (Tyr783/Ser1248) NIH/3T3 Serum-starve overnight,<br />

PDGF (50 ng/ml for 30 min)<br />

Phospho-PLCγ2 (Tyr759/Tyr1217) Ramos, Raji IgM (12 µg/ml for 2 min)<br />

Phospho-PLD1 (Thr147/Ser561) HeLa Serum-starve overnight,<br />

TPA (200 nM for 15 min)<br />

Phospho-PRK1 (Thr774)/PRK2 (Thr816) HeLa Untreated<br />

Cell Cycle/Checkpoint Control<br />

Acetyl-p53 (Lys379/Lys382) MCF7 Doxorubicin (0.5 µM for 24 hr);<br />

400 nM TSA can be used in conjunction<br />

to stabilize activated protein<br />

Phospho-53BP1 (Ser25/Ser29/Thr543/Ser1778) HT-29 UV (100 mJ/cm 2 ), 1 hr recovery<br />

Phospho-ATM (Ser1981) 293 IR (10 Gy), 1 hr recovery<br />

Phospho-ATR (Ser428) HeLa UV (100 mJ/cm 2 ), 1 hr recovery<br />

Phospho-ATRIP (Ser224) HeLa Untreated<br />

Phospho-Aurora A (Thr288)/Aurora B (Thr232)/Aurora HT-29 Nocodazole (100 ng/ml for 18 hr)<br />

C (Thr198)<br />

Phospho-BRCA1 (Ser1524) HeLa UV (100 mJ/cm 2 ), 1 hr recovery<br />

Phospho-cdc2 (Thr14/Tyr161) HeLa Nocodazole (100 ng/ml for 18 hr)<br />

Phospho-cdc2 (Thr15) HeLa Untreated<br />

Phospho-cdc25C (Thr48/Ser216) HT-29 Nocodazole (100 ng/ml for 18 hr)<br />

Phospho-CDK2 (Thr160) HeLa Hydroxyurea (10 mM for 16 hr)<br />

Phospho-CDK9 (Thr186) HeLa Untreated<br />

Phospho-Chk1 (Ser280/Ser296/Ser317/Ser345) HeLa UV (100 mJ/cm 2 ), 1 hr recovery<br />

Phospho-Chk2 (Ser19/Ser33/Ser35/Thr68/Thr387/ HeLa UV (100 mJ/cm 2 ), 1 hr recovery<br />

Thr432/Ser516)<br />

Phospho-Cyclin B1 (Ser133/Ser147) HeLa Nocodazole (100 ng/ml for 18 hr)<br />

Phospho-Cyclin D1 (Thr286) HT-1080 MG132 (10 µM for 4 hr)<br />

Phospho-Cyclin E (Thr62) MCF7 Hydroxyurea (10 mM for 8 hr)<br />

Phospho-FANCD2 (Ser222) HeLa UV (100 mJ/cm 2 ), 1 hr recovery<br />

Phospho-LATS1 (Ser909) HeLa TPA (200 nM for 30 min)<br />

Phospho-LATS1 (Thr1079) HeLa Okadaic Acid (1 μM for 1 hr)<br />

Phospho-MDM2 (Ser166) MCF7 hIGF-I (100 ng/ml for 15 min)<br />

Phospho-Mre11 (Ser676) HeLa UV (100 mJ/cm 2 ), 1 hr recovery<br />

Phospho-Myt1 (Ser83) HeLa Nocodazole (100 ng/ml for 18 hr)<br />

#9160<br />

#9160<br />

#9160<br />

#9160<br />

#9160<br />

#9160<br />

www.cellsignal.com/cstcontrols<br />

239


Section II: ANTIBODY APPLICATIONS<br />

chapter 16: Control Treatments and Cell Lines<br />

Control<br />

Treatments<br />

by Target<br />

Please visit our website for the<br />

most up-to-date listing of control<br />

treatments and cell lines.<br />

www.cellsignal.com/cstcontrols<br />

Cell Line Treatment Control<br />

Phospho-NPM (Ser4/Thr95/Thr199) HeLa Nocodazole (100 ng/ml for 18 hr)<br />

Phospho-NuMA (Ser395) M059K UV (100 mJ/cm 2 ), 1 hr recovery<br />

Phospho-p53 (Ser33/Thr81/Ser315) HeLa Nocodazole (100 ng/ml for 18 hr)<br />

Phospho-p53 (Ser392) 293 Hydroxyurea (20 mM for 30 hr)<br />

Phospho-p53 (Ser6/Ser9/Ser15/Thr18<br />

293 UV (100 mJ/cm 2 ), 1 hr recovery #9253<br />

/Ser20/Ser37/Ser46/Ser315)<br />

Phospho-p57 Kip2 (Thr310) HeLa Dexamethasone (50 nM for 16 hr)<br />

Phospho-p63 (Ser160/Ser162) ME-180 Nocodazole (100 ng/ml for 18 hr)<br />

Phospho-p73 (Tyr99) HT-1376 Pervanadate (1 mM for 20 min)<br />

Phospho-p95/NBS1 (Ser343) HeLa UV (100 mJ/cm 2 ), 1 hr recovery<br />

Phospho-PBK/TOPK (Thr9) HT-29 Nocodazole (100 ng/ml for 18 hr)<br />

Phospho-Pin1 (Ser16) OVCAR8 Forskolin (30 µM for 30 min)<br />

Phospho-PNK1 (Ser114/Thr118) HeLa UV (100 mJ/cm 2 ), 1 hr recovery<br />

Phospho-PP1α (Thr320)<br />

HeLa, COS-7, Untreated<br />

NIH/3T3, C6<br />

Phospho-Rad17 (Ser645) COS-7 UV (100 mJ/cm 2 ), 1 hr recovery<br />

Phospho-Rb (Ser608/Ser780/Ser795/Ser807/Ser811) HT-29 Nocodazole (100 ng/ml for 18 hr);<br />

treatment induces slight phosphoshift<br />

Phospho-RCC1 (Ser11) HT-29 Thymidine (2 mM for 16 hr) followed<br />

by Nocodazole (100 ng/ml for 18 hr)<br />

Phospho-Rpb1 CTD (Ser2/5) MCF7 Doxorubicin (0.5 µM for 30 hr)<br />

Phospho-SMC1 (Ser360/Ser957) 293 UV (100 mJ/cm 2 ), 1 hr recovery<br />

Phospho-TACC3 (Ser558) HT-29 Thymidine (2 mM for 16 hr),<br />

add fresh media<br />

Phospho-TIF1β (Ser824) HeLa IR (10 Gy), 1 hr recovery<br />

Phospho-TLK1 (Ser695) SK-N-MC Serum-starve overnight<br />

Phospho-Wee1 (Ser642) A-431 Serum-starve overnight,<br />

hEGF (100 nM for 5 min)<br />

Chromatin/Epigenetic Regulation<br />

Acetyl- and Phospho-Histone H3 (Lys9/Ser10) NIH/3T3 Serum-starve plus TSA (400 nM<br />

overnight), add 20% serum for<br />

15 min, then add Calyculin A<br />

(100 nM for 15 min)<br />

Acetyl-CBP (Lys1535)/p300 (Lys1499) NIH/3T3 TSA (400 nM overnight)<br />

Acetyl-Histone H2A (Lys5) NIH/3T3 TSA (400 nM overnight)<br />

Acetyl-Histone H2B (Lys5/Lys12/Lys20) NIH/3T3 TSA (400 nM overnight)<br />

Acetyl-Histone H3 (Lys9/Lys14/Lys18/Lys23) NIH/3T3 TSA (400 nM overnight)<br />

Acetyl-Histone H4 (Lys5/Lys8/Lys12) NIH/3T3 TSA (400 nM overnight)<br />

Methyl-Histone H3 (Arg2) MCF7 Untreated<br />

Mono/Di/Tri-Methyl-Histone H3<br />

NIH/3T3 Untreated<br />

(Lys4/Lys27/Lys9/Lys36/Lys79)<br />

Mono/Di/Tri-Methyl-Histone H4 (Lys20) NIH/3T3 Untreated<br />

Phospho-CTDSPL2 (Ser104) HeLa Untreated<br />

Phospho-DBC1 (Thr454) 293 UV (100 mJ/cm 2 ), 4 hr recovery<br />

Phospho-HDAC3 (Ser424) NIH/3T3 Untreated<br />

Phospho-HDAC4 (Ser246/Ser632)/HDAC5 (Ser259/ HeLa Serum-starve overnight, Pervanadate<br />

Ser498)/HDAC7 (Ser155/Ser486)<br />

Phospho-Histone H2A.X (Ser139/Tyr142) 293 UV (40 mJ/cm 2 ), 2 hr recovery<br />

Phospho-Histone H3 (Thr3/Ser10/Thr11/Ser28) NIH/3T3 Serum-starve overnight, add 20%<br />

serum for 15 min, then Calyculin A<br />

(100 nM ) 15 min<br />

Phospho-HP1γ (Ser83) HeLa IBMX (30 µM) plus Forskolin<br />

(500 µM) for 30 min<br />

Phospho-SATB1 (Ser47) Jurkat Untreated<br />

Phospho-SirT1 (Ser27/Ser47) 293 Untreated<br />

Cell Line Treatment Control<br />

Cytoskeletal Regulation<br />

Acetyl-α-Tubulin (Lys40) HeLa TSA (400 nM for 16 hr)<br />

Phospho-Caveolin-1 (Tyr14) HeLa Serum-starve overnight, H 2 O 2<br />

(500 μM for 15 min)<br />

Phospho-Cofilin (Ser3)<br />

C2C12, Untreated<br />

NIH/3T3, HeLa<br />

Phospho-Cortactin (Tyr421) HeLa Serum-starve overnight, H 2 O 2<br />

(2 nM for 2 min)<br />

Phospho-CrkII (Tyr221) K-562 Untreated<br />

Phospho-DRP1 (Ser616) HeLa Nocadazole (100 ng/ml for 16 hr)<br />

Phospho-DRP1 (Ser637) PC-12 Serum-starve overnight,<br />

Forskolin (20 µM for 1 hr)<br />

Phospho-Ezrin (Tyr353) A-431 Serum-starve overnight,<br />

hEGF (100 ng/ml for 10 min)<br />

Phospho-Ezrin (Tyr567)/Radixin (Thr564)<br />

/Moesin (Thr558)<br />

C2C12,<br />

NIH/3T3, HeLa<br />

Untreated<br />

Phospho-Filamin A (Ser2152) 293 Untreated<br />

Phospho-MARK Family (Activation Loop) Raji, BaF3 Untreated<br />

Phospho-Myosin IIa (Ser1943)<br />

HeLa, A-431, Untreated<br />

293T<br />

Phospho-Myosin Light Chain 2 (Thr18/Ser19) C2C12 Untreated<br />

Phospho-MYPT1 (Ser507/Ser668/Thr853) HeLa Serum-starve overnight,<br />

Calyculin A (100 nM for 30 min)<br />

Phospho-Na, K-ATPase a1 (Ser16/Ser23) PC-12 Serum-starve overnight,<br />

TPA (200 nM for 30 min)<br />

Phospho-Na, K-ATPase a1 (Tyr10) A-431 Serum-starve overnight,<br />

hEGF (100 ng/ml for 5 min)<br />

Phospho-PAK1 (Ser144/Ser199/Ser204/Thr423)/PAK2<br />

(Ser20/Ser141/Ser192/Ser197/Thr402)<br />

Phospho-PAK4 (Ser474)/PAK5 (Ser602)/PAK6 (Ser560)<br />

guinea pig<br />

neutrophils<br />

guinea pig<br />

neutrophils<br />

fMLP (1 µM for 30 sec)<br />

fMLP (1 µM for 30 sec)<br />

Phospho-Rac1/cdc42 (Ser71) A-431 Serum-starve overnight,<br />

hEGF (100 ng/ml for 10 min)<br />

Phospho-RanBP3 (Ser58) HeLa Serum-starve overnight,<br />

hEGF (100 ng/ml for 10 min)<br />

Phospho-Stathmin (Ser16/Ser38) HeLa Nocadazole (100 ng/ml for 16 hr)<br />

Phospho-TCTP (Ser46) HT-29 2 mM Thymidine for 16 hr, then<br />

wash plate, add fresh media and<br />

Nocodazole (100 ng/ml overnight)<br />

Phospho-Troponin I (Cardiac) (Ser23/24)<br />

Primary cardiac Isoproterenol (1 µM for 5 min)<br />

myocytes<br />

Phospho-VASP (Ser157/Ser239) A-431 Serum-starve overnight,<br />

Forskolin (10 µM for 30 min)<br />

Phospho-Vimentin (Ser56) HeLa Paclitaxel (100 ng/ml for 20 hr)<br />

Phospho-Vimentin (Ser82) C2C12, C6 Untreated<br />

Developmental Biology<br />

Cleaved Notch1 (Val1744)<br />

MOLT-4, Jurkat Untreated<br />

Phospho-LRP6 (Ser1490) HeLa Wnt3a-conditioned media for 5 hr<br />

(Conditioned media from L cells<br />

transfected with Wnt3a ligand)<br />

Phospho-NDRG1 (Ser330) Jurkat Calyculin A (100 nM for 20 min)<br />

Phospho-NDRG1 (Thr346) C2C12 Insulin (30 min)<br />

Phospho-Numb (Ser276) Ramos TPA (200 nM for 30 min)<br />

Phospho-Smad1(Ser206/Ser463/Ser465) HeLa Serum-starve overnight,<br />

BMP (50 ng/ml for 30 min)<br />

Phospho-Smad2 (Ser245/Ser250/Ser255/Ser465/Ser467) HeLa Serum-starve overnight,<br />

TPA (200 nM for 30 min)<br />

Phospho-Smad3 (Ser423/Ser425) HT-1080 Serum-starve overnight,<br />

TGF-β (10 ng/ml for 30 min)<br />

Phospho-Smad5 (Ser463/Ser465) HeLa Serum-starve overnight,<br />

BMP (50 ng/ml for 30 min)<br />

#12052<br />

#12052<br />

240 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstcontrols<br />

241


Section II: ANTIBODY APPLICATIONS<br />

chapter 16: Control Treatments and Cell Lines<br />

Control<br />

Treatments<br />

by Target<br />

Please visit our website for the<br />

most up-to-date listing of control<br />

treatments and cell lines.<br />

www.cellsignal.com/cstcontrols<br />

Cell Line Treatment Control<br />

Phospho-Smad8 (Ser426/Ser428) HeLa Serum-starve overnight,<br />

BMP (50 ng/ml for 30 min)<br />

Phospho-YAP (Ser127) HeLa Untreated<br />

Phospho-β-Catenin (Ser33/Ser37/Thr41/Ser45) SW480 Untreated<br />

Phospho-β-Catenin (Ser552/Ser675) SK-N-MC Forskolin (50 µM for 30 min)<br />

Immunology and Inflammation<br />

Cleaved IL-1β (Asp116) THP-1 Differentiate with TPA (200 nM<br />

for 24 hr), rest for 24 hr, then<br />

treat with LPS (1 µg/ml for 24 hr).<br />

Protein is secreted into media.<br />

Phospho-5-Lipoxygenase (Ser663)<br />

mouse & Untreated<br />

rat brain<br />

Phospho-AML1 (Ser249) Jurkat, HEL Untreated<br />

Phospho-BLNK (Tyr96) Ramos Anti-IgM (12 µg/ml for 2 min)<br />

Phospho-Btk (Ser180) THP-1 Serum-starve overnight,<br />

H 2 O 2 (2 mM for 2 min)<br />

Phospho-Btk (Tyr223) Ramos Anti-IgM (12 μg/ml for 10 min)<br />

Phospho-CD19 (Tyr531) Ramos Anti-IgM (12 μg/ml for 2 min)<br />

Phospho-CrkL (Tyr207) K-562 Untreated<br />

Phospho-HS1 (Tyr397) Ramos Serum-starve overnight,<br />

IgM (12 µg/ml for 10 min)<br />

Phospho-IKKα/β (Ser176/Ser180) HeLa hTNF-α (20 ng/ml for 10 min) #9243<br />

Phospho-IKKγ (Ser376) HeLa hTNF-α (20 ng/ml for 10 min) #9243<br />

Phospho-IRAK1 (Thr209/Ser376/Thr387) HeLa, 293, Untreated<br />

MCF7<br />

Phospho-IRF-3 (Ser396) Raw 264.7 LPS (1 μg/ml for 2 hr)<br />

Phospho-IκBα (Ser32/36) HeLa hTNF-α (20 ng/ml for 5 min) #9243<br />

Phospho-IκBβ (Ser19/Ser23) HeLa hTNF-α (20 ng/ml) plus<br />

Calyculin A (50 nM for 5 min)<br />

Phospho-IκBε (Ser18/Ser22) HeLa hTNF-α (20 ng/ml) plus<br />

Calyculin A (50 nM for 5 min)<br />

Phospho-Jak1 (Tyr1022/1023) HT-29 Serum-starve overnight,<br />

IL-4 (100 ng/ml for 5–10 min)<br />

Phospho-Jak2 (Tyr1007/Tyr1008) TF-1 Serum-starve overnight, GM-CSF<br />

(25 ng/ml for 15 min); GM-CSF<br />

(2 ng/ml) should be included during<br />

growth and serum-starvation<br />

Phospho-Jak2 (Tyr221/Tyr1007/Try1008) CTLL-2 Untreated<br />

Phospho-LAT (Tyr171/Tyr191) Jurkat Anti-CD3 (10 μg/ml for 2 min)<br />

Phospho-Lck (Tyr505) Jurkat Anti-CD3 (10 μg/ml for 2 min)<br />

#9243<br />

#9243<br />

Phospho-Lyn (Tyr507) Ramos Serum-starve overnight,<br />

anti-IgM (12 μg/ml for 2 min)<br />

Phospho-NF-κB p105 (Ser933) HeLa hTNF-α (20 ng/ml for 5 min) #9243<br />

Phospho-NF-κB p65 (Ser276) Jurkat Calyculin A (100 nM for 30 min) #9273<br />

Phospho-NF-κB p65 (Ser468/Ser536) HeLa hTNF-α (20 ng/ml for 5 min)<br />

Phospho-p40phox (Thr154) THP-1 Serum-starve overnight,<br />

TPA (200 nM for 15 min)<br />

Phospho-RelB (Ser552) Jurkat TPA (200 nM for 30 min)<br />

Phospho-RIP2 (Ser176) THP-1 Treat overnight with TPA (50 ng/ml),<br />

then culture in fresh medium 48 hr<br />

to form differentiated macrophages.<br />

Induce with LPS (1 µg/ml for 10 min).<br />

Phospho-SLP76 (Ser376) Jurkat Anti-CD3 (10 μg/ml for 2 min)<br />

Phospho-SHIP1 (Tyr1020) Ramos Anti-human IgM (12 µg/ml for 2 min)<br />

Phospho-SHIP2 (Tyr986/Tyr987/Tyr1135) K-562 Untreated<br />

Phospho-Stat1 (Tyr701/Ser727) HeLa Serum-starve overnight,<br />

IFN-α (100 ng/ml for 5 min)<br />

#9173<br />

Cell Line Treatment Control<br />

Phospho-Stat2 (Tyr690) HeLa Serum-starve overnight,<br />

IFN-α (100 ng/ml for 15 min)<br />

Phospho-Stat3 (Tyr705/Ser727) HeLa Serum-starve overnight,<br />

#9133<br />

IFN-α (100 ng/ml for 5 min)<br />

Phospho-Stat4 (Tyr693) NK-92 Cytokine-starve overnight, IL-2<br />

(10 ng/ml for 15 min); IL-2 (5 ng/ml)<br />

should be included during growth<br />

Phospho-Stat5 (Tyr694) HeLa Serum-starve overnight,<br />

#9353<br />

IFN-α (100 ng/ml for 5 min)<br />

Phospho-Stat6 (Tyr641) HeLa Serum-starve overnight,<br />

IL-4 (100 ng/ml for 15 min)<br />

Phospho-Tyk2 (Tyr1054/Tyr1055) HeLa Serum-starve overnight,<br />

IFN-α (100 ng/ml for 15 min)<br />

Phospho-Zap-70 (Tyr319/Tyr493)/Syk (Tyr352) Jurkat Anti-CD3 (10 ng/ml for 2 min)<br />

MAP Kinase Signaling<br />

Phospho-A-Raf (Ser299) HeLa Serum-starve overnight,<br />

TPA (200 nM for 15 min)<br />

#9160<br />

Phospho-ATF-2 (Thr69/Thr71) NIH/3T3 Anisomycin (25 μg/ml for 30 min) #9223<br />

Phospho-B-Raf (Ser445) HeLa Serum-starve overnight,<br />

#9160<br />

TPA (200 nM for 15 min)<br />

Phospho-Bcr (Tyr177) K-562 Untreated (K-562 cell line<br />

contains Bcr-Abl fusion)<br />

Phospho-c-Abl (Tyr89/Tyr204/Tyr245/Tyr412/Thr735) K-562 Untreated (K-562 cell line<br />

contains Bcr-Abl fusion)<br />

Phospho-c-Fos (Ser32) HeLa Serum-starve overnight,<br />

TPA (200 nM for 4 hr)<br />

Phospho-c-Jun (Ser63/Ser73/Thr91/Ser243) NIH/3T3 UV (40 mJ/cm 2 ), 30 min recovery #9263<br />

Phospho-c-Jun (Thr93) NIH/3T3 Untreated<br />

Phospho-c-Raf (Ser259/Ser289/Ser296/Ser301/Ser338) HeLa Serum-starve overnight,<br />

TPA (200 nM for 15 min)<br />

Phospho-DUSP1/MKP1 (Ser359) HeLa ALLN (50 µM for 5–6 hr)<br />

Phospho-Elk-1 (Ser383)<br />

recombinant<br />

protein<br />

GST-Elk1 fusion protein<br />

phosphorylated in vitro by Erk1/2<br />

Phospho-Erk5 (Thr218/Tyr220) NIH/3T3 Serum-starve overnight, PDGF<br />

(100 ng/ml for 5 min); IP with<br />

#12950 prior to western<br />

Phospho-FRA1 (Ser265) HeLa Serum-starve overnight,<br />

TPA (200 nM for 4 hr)<br />

Phospho-FRS2-α (Tyr196/Tyr436) NIH/3T3 Serum-starve overnight, hFGF<br />

basic/FGF2 (100 ng/ml for 10 min)<br />

Phospho-GRB10 (Tyr67) H-4-II-E Serum-starve overnight,<br />

HGF (40 ng/ml for 10 min)<br />

Phospho-KSR1 (Ser392) HeLa Untreated<br />

#9160<br />

#9183<br />

Phospho-MAPKAPK-2 (Thr222/Thr334) HeLa Serum-starve overnight, UV<br />

(40 mJ/cm 2 ), 20 min recovery<br />

Phospho-MEK1 (Thr286/Ser298) HeLa Serum-starve overnight,<br />

#9160<br />

Nocodazole (0.2 μg/ml for 16 hr) for<br />

Thr286; TPA (200 nM for 15 min)<br />

for Ser298<br />

Phospho-MEK1/2 (Ser217/Ser221) HeLa Serum-starve overnight, TPA #9160<br />

(200 nM for 15 min)<br />

Phospho-MKK3/MKK6 (Ser189/Ser207) NIH/3T3 UV (40 mJ/cm 2 ), 30 min recovery #9233<br />

Phospho-MKK7 (Ser271/Thr275) U-937 Sorbitol (400 mM for 15 min)<br />

Phospho-MSK1 (Ser376/Thr581) 293 UV (40 mJ/cm 2 ), 30 min recovery<br />

Phospho-p38 MAPK (Thr180/Tyr182) C6 Anisomycin (25 μg/ml for 20 min) #9213<br />

Phospho-p44/42 MAPK (Erk1/2) (Thr202/Tyr204) Jurkat TPA (200 nM for 10 min) #9194<br />

Phospho-p90RSK (Thr359/Ser363/Ser380/Thr573) HeLa TPA (200 nM for 30 min) #9160<br />

Phospho-PTPα (Tyr789)<br />

293, COS-7, Untreated<br />

C6, C2C12<br />

Phospho-Pyk2 (Tyr402) Jurkat Serum-starve overnight, anti-CD3<br />

plus anti-CD28 (both 1 µg/ml for<br />

10 min)<br />

242 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstcontrols<br />

243


Section II: ANTIBODY APPLICATIONS<br />

chapter 16: Control Treatments and Cell Lines<br />

Control<br />

Treatments<br />

by Target<br />

Please visit our website for the<br />

most up-to-date listing of control<br />

treatments and cell lines.<br />

www.cellsignal.com/cstcontrols<br />

Cell Line Treatment Control<br />

Phospho-RSK2 (Ser227) HeLa TPA (200 nM for 30 min) #9160<br />

Phospho-RSK2 (Tyr529) SK-N-MC Serum-starve overnight,<br />

FGF (100 ng/ml for 10 min)<br />

Phospho-RSK3 (Thr356/Ser360) HeLa Serum-starve overnight,<br />

TPA (200 nM for 15 min)<br />

Phospho-SAPK/JNK (Thr183/Tyr185) 293 UV (40 mJ/cm 2 ),<br />

30 min recovery time<br />

Phospho-SEK1/MKK4 (Ser257/Thr261) HeLa UV (50 mJ/cm 2 ),<br />

30 min recovery time<br />

Phospho-SEK1/MKK4 (Ser80) NIH/3T3 PDGF (100 ng/ml for 5 min)<br />

Phospho-Shc (Tyr239/Tyr240/Tyr317) Hep G2 Serum-starve overnight,<br />

hEGF (100 ng/ml for 5 min)<br />

Phospho-SHP-2 (Tyr542/Tyr580) NIH/3T3 Serum-starve overnight,<br />

PDGF (100 ng/ml for 5 min)<br />

Phospho-Src (Tyr527) COLO201 Untreated<br />

Phospho-Src Family (Tyr416) COLO201 Serum-starve overnight,<br />

treat 5 min 20% FBS<br />

Phospho-SRC-3 (Thr24) MCF7 Untreated<br />

Phospho-SRF (Ser103) HeLa Serum-starve overnight,<br />

TPA (200 nM for 15 min)<br />

Phospho-TAK1 (Thr184/Thr187/Ser412) HeLa hTNF-α or IL-1β (20 ng/ml), both<br />

with Calyculin A (100 nM) for 10 min<br />

Phospho-TNK1 (Tyr277) KARPAS-299 Untreated (KARPAS-299 cell lines<br />

contains NPM-ALK fusion)<br />

Phospho-β-Arrestin 1 (Ser412) 293 Untreated<br />

Cellular Metabolism<br />

Acetyl-CoA Carboxylase C2C12 Untreated<br />

Phospho-Acetyl-CoA Carboxylase (Ser79) C2C12 Serum-starve overnight,<br />

AICAR (0.5 mM for 30 min)<br />

Phospho-AMPKα (Thr172) C2C12 Serum-starve overnight,<br />

AICAR (0.5 mM for 30 min)<br />

Phospho-AMPKα1 (Ser485) C2C12 Serum-starve overnight,<br />

AICAR (0.5 mM for 30 min)<br />

Phospho-AMPKβ1 (Ser108/Ser182) C2C12 Serum-starve overnight,<br />

AICAR (0.5 mM for 30 min)<br />

Phospho-AS160 (Thr642) HeLa Serum-starve overnight,<br />

hIGF-I (100 ng/ml for 15 min)<br />

Phospho-ATP-Citrate Lyase (Ser454) NIH/3T3 Serum-starve overnight,<br />

PDGF (50 ng/ml for 10 min)<br />

Phospho-C/EBPα (Ser21)<br />

differentiated<br />

3T3-L1<br />

Phospho-C/EBPα (Thr222/Thr226) U-937 Untreated<br />

Phospho-C/EBPβ (Ser105) PC-12 Untreated<br />

Phospho-C/EBPβ (Thr235)<br />

differentiated<br />

3T3-L1<br />

Serum-starve overnight,<br />

insulin (100 nM for 45 min)<br />

Serum-starve overnight,<br />

insulin (100 nM for 10 min)<br />

Phospho-Glycogen Synthase (Ser641) 293 Serum-starve overnight,<br />

insulin (100 nM for 5 min)<br />

Phospho-HSL (Ser563/Ser565/Ser660)<br />

differentiated<br />

3T3-L1<br />

Isoproterenol (10 µM for 20 min)<br />

Phospho-IGF-I Receptor β (Tyr1316/Tyr980/Tyr1131/<br />

Tyr1135)<br />

Phospho-IRS-1 (Ser302/Ser307/Ser318/Ser332<br />

/Ser336/Ser612/Ser636/Ser639/Ser1101)<br />

293 Serum-starve overnight,<br />

hIGF-I (50 ng/ml for 5 min)<br />

MCF7 Serum-starve overnight,<br />

insulin (100 nM for 5 min)<br />

Motif Antibodies<br />

Acetylated-Lysine COS-7 TSA (0.4 μM for 18 hr)<br />

Acetylated-Lysine (Ac-K2-100) COS-7 TSA (0.4 μM for 18 hr)<br />

Cleaved Caspase Substrate HeLa Staurosporine (1 µM for 3 hr)<br />

Mono-Methyl Arginine HCT116 Untreated<br />

#9160<br />

#9253<br />

#9160<br />

#9158<br />

#9158<br />

#9158<br />

#9158<br />

Phospho-(Ser) 14-3-3 Binding Motif Jurkat Calyculin A (100 nM for 30 min) #9273<br />

Cell Line Treatment Control<br />

Phospho-(Ser) Arg-X-Tyr/Phe-X-pSer Motif Jurkat Calyculin A (100 nM for 30 min) #9273<br />

Phospho-(Ser) CDKs Substrate HeLa Nocadazole (1 μg/ml for 12 hr)<br />

Phospho-(Ser) PKC Substrate HeLa TPA (200 nM for 30 min) #9160<br />

Phospho-(Ser/Thr) AMPK Substrate 293 Serum starved for 1 hr #9158<br />

Phospho-(Ser/Thr) ATM/ATR Substrate 293 UV (50 mJ/cm 2 ), 30 min recovery #9253<br />

Phospho-(Ser/Thr) PDK1 Docking Motif Jurkat Calyculin A (100 nM for 30 min) #9273<br />

Phospho-(Ser/Thr) Phe Jurkat Calyculin A (100 nM for 30 min) #9273<br />

Phospho-(Ser/Thr) PKD Substrate HeLa TPA (200 nM for 30 min) #9160<br />

Phospho-(Thr) MAPK/CDK Substrate Jurkat Nocadazole (1 μg/ml for 12 hr)<br />

Phospho-Akt Substrate (RXRXXS*/T*) Jurkat Calyculin A (100 nM for 30 min) #9273<br />

Phospho-PKA Substrate (RRXS/T) Jurkat Calyculin A (100 nM for 30 min) #9273<br />

Phospho-Threonine NIH/3T3 PDGF-BB (100 ng/ml for 5 min)<br />

Phospho-Threonine-X-Arginine Jurkat Calyculin A (100 nM for 30 min) #9273<br />

Phospho-Tyrosine Jurkat Pervanadate (1 mM for 30 min)<br />

Neuroscience<br />

Phospho-AMPA Receptor (GluR 2) (Tyr869/873/876) rat brain ischemia and reperfusion<br />

Phospho-APP (Thr668) HeLa Nocodazole (1 µg/ml for 18 hr)<br />

Phospho-CaMKII (Thr286) HeLa TPA (200 nM for 15 min) #9160<br />

Phospho-CREB (Ser133) SK-N-MC Forskolin (30 µM) and IBMX<br />

(0.5 mM for 30 min)<br />

Phospho-CRMP-2 (Thr514) rat brain Untreated<br />

Phospho-DARPP-32 (Thr34/Thr75)<br />

rat & mouse PKA (12,500 U/100 μl for 1 hr)<br />

brain<br />

Phospho-Doublecortin (Ser297) fetal rat brain Untreated<br />

Phospho-Doublecortin (Ser334)<br />

rat & mouse Untreated<br />

brain<br />

Phospho-MAP2 (Ser136/Thr1620/Thr1623) PC-12 Positive control: Nocodazole<br />

(1 µg/ml for 16 hr)<br />

Phospho-MAPK/CDK Substrates (PXSP or SPXR/K) HeLa Nocadazole (1 μg/ml for 12 hr)<br />

Phospho-Merlin (Ser518)<br />

rat, mouse, & Untreated<br />

human brain<br />

Phospho-NMDAR2A (Tyr1246) rat brain Untreated<br />

Phospho-NMDAR2B (Tyr1070/Tyr1472) rat brain Untreated<br />

Phospho-PSD93 (Tyr340) rat brain Untreated<br />

Phospho-PSD95 (Tyr236/Tyr240) rat brain Untreated<br />

Phospho-Semaphorin 4B (Ser825) MKN-45 Untreated<br />

Phospho-Synapsin (Ser9) PC-12 NGF (100 ng/ml for 2 hr)<br />

Phospho-Tau (Ser396)<br />

mouse & rat Untreated<br />

brains<br />

Phospho-TrkA (Tyr490/Tyr674/Tyr785) PC-12 NGF (100 ng/ml for 2–5 min)<br />

Phospho-TrkB (Tyr516/Tyr706/Tyr707/Tyr816) PC-12 NGF (100 ng/ml for 2–5 min)<br />

Phospho-Tyrosine Hydroxylase (Ser31/Ser40)<br />

rat, mouse, &<br />

human brain,<br />

SH-SY5Y, PC-<br />

12, Neuro-2a<br />

Untreated<br />

Nuclear Receptor Signaling<br />

Phospho-Estrogen Receptor α (Ser104/Ser106/Ser118) MCF7 Serum-starve overnight, hEGF<br />

(100 ng/ml) plus estradiol (100 nM)<br />

for 30 min<br />

Phospho-Glucocorticoid Receptor (Ser211) A549 Serum-starve overnight,<br />

dexamethasone (100 nM for 1 hr)<br />

Phospho-Nur77 (Ser351) Jurkat TPA (40 nM), A23187 (2 µM),<br />

both for 4 hr<br />

Phospho-Progesterone Receptor (Ser190) T-47D Serum-starve overnight,<br />

promegestone (100 nM for 1 hr)<br />

#9193<br />

244 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstcontrols<br />

245


Section II: ANTIBODY APPLICATIONS<br />

chapter 16: Control Treatments and Cell Lines<br />

Cell Line Treatment Control<br />

PI3 Kinase/Akt Signaling<br />

Phospho-Akt (Ser473) Jurkat Untreated or Calyculin A (100 nM<br />

for 30 min); alternative: serumstarve,<br />

insulin (100 nM for 10 min)<br />

works for most cell lines<br />

#9273<br />

Phospho-Akt (Thr308/Thr450) Jurkat Calyculin A (100 nM for 30 min) #9273<br />

Phospho-Drosophila p70 S6 Kinase (Thr398) S2 hEGF (100 ng/ml for 30 min)<br />

Phospho-eNOS (Ser113/Thr495) BAEC Untreated<br />

Phospho-eNOS (Ser1177) BAEC VEGF (50 ng/ml for 5 min)<br />

Phospho-FoxO1 (Thr24/Ser256/Ser319) HeLa Serum-starve overnight,<br />

20% serum for 30 min<br />

Phospho-FoxO3a (Thr32/Ser318/Ser321/Ser253) HeLa Serum-starve overnight,<br />

20% serum for 30 min<br />

Phospho-FoxO4 (Ser193) HeLa Serum-starve overnight,<br />

20% serum for 30 min<br />

Phospho-Gab1 (Tyr307/Tyr627) Hep G2 Serum-starve overnight,<br />

HGF (40 ng/ml for 10 min)<br />

Phospho-Gab2 (Ser159/Tyr452) H-4-II-E Serum-starve overnight,<br />

HGF (40 ng/ml for 10 min)<br />

Phospho-GSK-3α (Ser21) NIH/3T3 Serum-starve overnight,<br />

PDGF (100 ng/ml for 30 min)<br />

Phospho-GSK-3β (Ser9) NIH/3T3 Serum-starve overnight,<br />

PDGF (100 ng/ml for 30 min)<br />

Phospho-GSK-3β (Thr390) HeLa Paclitaxel (100 nM/ml for 20 hr)<br />

Phospho-mTOR (Ser2448/Ser2481) MCF7 Serum-starve overnight,<br />

insulin (100 nM for 5–10 min)<br />

Phospho-p70 S6 Kinase<br />

(Thr389/Ser371/Thr421/Ser424)<br />

MCF7<br />

Phospho-PDK1 (Ser241) Jurkat, HeLa Untreated<br />

Phospho-PI3K p85 (Tyr458)/p55 (Tyr199) NIH/3T3/Src Untreated<br />

Serum-starve overnight,<br />

insulin (100 nM for 5–10 min)<br />

Phospho-PRAS40 (Thr246) NIH/3T3 Serum-starve overnight,<br />

insulin (100 nM for 5–10 min)<br />

Phospho-PTEN (Ser380/Thr382/Thr383) HeLa Untreated<br />

Phospho-Raptor (Ser792) 293 Oligomycin (0.5 µM for 30 min)<br />

Phospho-Rictor (Thr1135) HeLa Serum-starve overnight,<br />

hIGF-I (50 ng/ml for 30 min)<br />

Phospho-SGK1 (Ser78) SK-MEL-28 H 2 O 2 (4 mM for 15 min)<br />

Phospho-SGK3 (Thr320) MCF7 H 2 O 2 (4 mM for 15 min)<br />

Phospho-Tuberin/TSC2<br />

NIH/3T3 PDGF (50 ng/ml for 30 min)<br />

(Ser939/Ser1254/Thr1462/Tyr1571)<br />

Phospho-WNK1 (Thr60) HT-29 Serum-starve overnight,<br />

hIGF-I (100 ng/ml for 30 min)<br />

Phospho-YB1 (Ser102) MCF7 Serum-starve overnight,<br />

hIGF-I (100 ng/ml for 30 min)<br />

Vesicle Trafficking and Protein Folding<br />

Phospho-HSP27 (Ser15/Ser78/Ser82) HeLa Serum-starve overnight, UV<br />

(40 mJ/cm 2 ), 20 min recovery<br />

Phospho-HSP90α (Thr5/Thr7) HeLa Serum-starve overnight, UV<br />

(40 mJ/cm 2 ), 20 min recovery<br />

Phospho-PERK (Thr980) AR42J Thapsigargin (1 μM for 20 min)<br />

#9203<br />

#9203<br />

Cell Line Treatment Control<br />

Phospho-Etk (Tyr40) Jurkat Serum-starve overnight, anti-CD3<br />

(1 μg/ml) plus anti-CD28 (0.5 μg/ml)<br />

for 10 min<br />

Phospho-FLT3 (Tyr589/Tyr591/Tyr842/Tyr969) SEM Untreated<br />

Phospho-HER2/ErbB2 (Tyr877/Tyr1221/Tyr1222/<br />

Tyr1248)<br />

MCF7<br />

Serum-starve overnight,<br />

neuregulin (100 ng/ml for 5 min)<br />

Phospho-HER3/ErbB3 (Tyr1197/Tyr1222/Tyr1289) MCF7 Serum-starve overnight,<br />

neuregulin (100 ng/ml for 5 min)<br />

Phospho-HER4/ErbB4 (Tyr984/Tyr1284) MCF7 Serum-starve overnight,<br />

neuregulin (100 ng/ml for 5 min)<br />

Phospho-M-CSF Receptor (Tyr699/Tyr723/Tyr809/Tyr923) NKM-1 Untreated<br />

Phospho-Met (Tyr1003/Tyr1234/Tyr1235/Tyr1349) H-4-II-E Serum-starve overnight,<br />

HGF (40 ng/ml for 10 min)<br />

Phospho-PDGF Receptor α (Tyr754/Tyr849/Tyr1018) NIH/3T3 Serum-starve overnight,<br />

PDGF-β (100 ng/ml for 5 min)<br />

Phospho-PDGF Receptor β<br />

(Tyr740/Tyr751/Tyr771/Tyr857/Tyr1009/Tyr1021)<br />

NIH/3T3<br />

Phospho-Ret (Tyr905) TT Untreated<br />

Serum-starve overnight,<br />

PDGF-β (100 ng/ml for 5 min)<br />

Phospho-Ros (Tyr2274) HCC78 HCC78 cells express a<br />

SLC3482-Ros fusion protein<br />

Phospho-VEGF Receptor 2 (Tyr1175/Tyr1212) HUVEC Serum-starve overnight,<br />

VEGF (100 ng/ml for 5 min)<br />

Translational Control<br />

Methyl-PABP1 (Arg455/Arg460) HeLa, C6,<br />

COS-7<br />

Untreated<br />

Phospho-4E-BP1 (Thr37/Thr46/Ser65/Thr70) HeLa Untreated<br />

Phospho-eEF2 (Thr56) C6 Forskolin (10 µM for 1 hr)<br />

Phospho-eEF2k (Ser366)<br />

HeLa, COS-7, Untreated<br />

NIH/3T3, C6<br />

Phospho-eIF2α (Ser51) HeLa Thapsigargin (300 nM for 30 min)<br />

Phospho-eIF4B (Ser422) NIH/3T3 Serum-starve overnight,<br />

20% calf serum for 20 min<br />

Phospho-eIF4E (Ser209) MCF7 Serum-starve overnight,<br />

insulin (100 nM for 5–10 min)<br />

Phospho-eIF4G (Ser1108) HeLa Serum-starve overnight,<br />

insulin (100 nM for 5–10 min)<br />

Phospho-elF4B (Ser406) NIH/3T3 Untreated<br />

Phospho-Mnk1 (Thr197/Thr202) HeLa Serum-starve overnight, 20%<br />

serum for 20 min or Anisomycin<br />

(25 µg/ml for 30 min)<br />

Phospho-PPIG (Ser374) Jurkat Untreated<br />

Phospho-S6 Ribosomal Protein<br />

(Ser235/Ser236/Ser240/Ser244)<br />

MCF7<br />

Serum-starve overnight, insulin<br />

(100 nM for 5–10 min)<br />

Ubiquitin and Ubiquitin-like proteins<br />

Phospho-BAP1 (Ser592) HeLa Serum-starve overnight,<br />

UV (50 mJ/cm 2 ), 1 hr recovery<br />

Phospho-c-Cbl (Tyr731/Tyr774) K-562 Untreated<br />

#2904<br />

#9203<br />

#9213<br />

#9203<br />

Control<br />

Treatments<br />

by Target<br />

Please visit our website for the<br />

most up-to-date listing of control<br />

treatments and cell lines.<br />

www.cellsignal.com/cstcontrols<br />

Tyrosine Kinase<br />

Phospho-ALK (Tyr1078/Tyr1096/Tyr1278<br />

/Try1282/Tyr1283/Tyr1586/Tyr1604)<br />

KARPAS-299<br />

Untreated (KARPAS-299 cell<br />

lines contains NPM-ALK fusion)<br />

Phospho-Axl (Tyr702) NCI-H1299 Gas6 (400 ng/mL for 5 min)<br />

Phospho-c-Kit (Tyr703/Tyr719) NCI-H526 Serum-starve overnight,<br />

hSCF (100 ng/ml for 5 min)<br />

Phospho-EGF Receptor (Thr669/Tyr845/Tyr998<br />

/Tyr992/Tyr1045/Ser1046 /Ser1047/Tyr1068<br />

/Tyr1086/Tyr1148/Tyr1173)<br />

HT-29<br />

Serum-starve overnight,<br />

hEGF (100 ng/ml for 5 min)<br />

#5634<br />

246 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/cstcontrols<br />

247


Section I: Research Areas<br />

chapter 08: Neuroscience<br />

Amyloid Plaque and Neurofibrillary Tangle Formation in Alzheimer’s Disease<br />

Dopamine Signaling in Parkinson’s Disease<br />

PEN-2<br />

Aph<br />

Nicastrin<br />

Presenilin<br />

APP<br />

Intracellular<br />

Domain<br />

γ-<br />

Secretase<br />

Normal State<br />

Nuclear translocation<br />

Transcriptional regulation<br />

AICD<br />

Normal<br />

Axon<br />

Pruning<br />

α-<br />

Secretase<br />

APP<br />

Alzheimer’s Disease State<br />

Apoptosis<br />

Apoptosis<br />

Casp-6<br />

Neurofibrillary<br />

Tangles<br />

Tau<br />

Akt<br />

Aberrant<br />

Neuronal<br />

Death<br />

p35<br />

β-<br />

APP<br />

Secretase<br />

Calpain<br />

CDK5<br />

p25<br />

GSK-3α/β<br />

Destabilized<br />

Microtubules<br />

Me<br />

Hyperphosphorylation<br />

of Tau<br />

AICD<br />

γ-<br />

APP Secretase<br />

Exosome-mediated<br />

Secretion of Tau<br />

Activation of PKC,<br />

PKA, Erk2<br />

ROS<br />

Formation<br />

Normal State<br />

Tyrosine<br />

Tyrosine<br />

Hydroxylase<br />

Dopa<br />

Decarboxylase<br />

Dopamine<br />

L-DOPA<br />

Parkinson’s Disease State<br />

JNK<br />

Apoptosis<br />

MKK4/7<br />

mTORC2<br />

Akt<br />

Dopamine<br />

Dopamine<br />

PINK1<br />

Cell<br />

Metabolism<br />

Parkin<br />

Survival<br />

Apoptosis ROS<br />

LRRK2 Mutation<br />

DJ-1<br />

PINK1 Mutation<br />

α-synuclein<br />

Aggregation<br />

Lewy Bodies<br />

REDD1<br />

Misfolding<br />

α-synuclein Ub<br />

Proteosomal Degradation<br />

Cell Stress,<br />

Cytokines<br />

Environmental Toxins,<br />

Neurotoxins<br />

Genetic<br />

Mutations<br />

Microglia<br />

Activation<br />

Increased<br />

Neuronal Survival,<br />

Neutrite Outgrowth,<br />

Synaptic sAPPα<br />

Plasticity,<br />

Cell Adhesion<br />

sAPPα<br />

Glucose NMDAR<br />

Transporters<br />

Glc<br />

Na +<br />

DR6<br />

N-APP<br />

AMPAR Acetylcholine<br />

Receptors:<br />

nAChR<br />

mAChR<br />

Ca 2+<br />

Glucose NMDAR AMPAR<br />

Transporters<br />

Casp-3,-9<br />

Apoptosis<br />

sAPPβ<br />

p53, Bad,<br />

Bax production<br />

and activation<br />

Oxidative Stress<br />

Aβ40/42<br />

nAChR<br />

mAChR<br />

Amyloid Plaques<br />

Aβ Misfolding,<br />

Aggregation<br />

Lipid<br />

Peroxidation<br />

ROS<br />

Formation<br />

Membrane Damage<br />

Microglia<br />

Activation<br />

Inflammatory<br />

Cytokines<br />

β,γ<br />

D2-type<br />

Gα i<br />

CDK5<br />

cAMP<br />

PP1<br />

AC<br />

DARPP-32<br />

DARPP-32<br />

Gα s<br />

D1-type D2-type D1-type<br />

PKA<br />

β,γ<br />

Transcription<br />

AMPAR<br />

NMDAR<br />

Ca 2+<br />

Na +<br />

CREB CREM ATF<br />

AMPAR<br />

NMDAR<br />

Post-Synaptic Signaling Blocked<br />

Neurodegeneration<br />

Alzheimer’s disease is one of the most common neurodegenerative diseases worldwide. Clinically, it is characterized by the presence of extracellular amyloid plaques and<br />

intracellular neurofibrillary tangles, resulting in neuronal dysfunction and cell death. Central to this disease is the differential processing of the integral membrane protein APP<br />

(Amyloid β Precursor Protein) in the normal versus disease state. In the normal state, APP is initially cleaved by α-secretase to generate sAPPα and a C83 carboxy-terminal<br />

fragment. The presence of sAPPα is associated with normal synaptic signaling and results in synaptic plasticity, learning and memory, emotional behaviors, and neuronal<br />

survival. In the disease state, APP is cleaved sequentially by β-secretase and γ-secretase to release an extracellular fragment called Aβ40/42. This neurotoxic fragment<br />

frequently aggregates and results in Aβ40/42 oligomerization and plaque formation. Aβ40/42 aggregation results in blocked ion channels, disruption of calcium homeostasis,<br />

mitochondrial oxidative stress, impaired energy metabolism and abnormal glucose regulation, and ultimately neuronal cell death. Alzheimer’s disease is also characterized<br />

by the presence of neurofibrillary tangles. These tangles are the result of hyperphosphorylation of the microtubule-associated protein Tau. GSK-3β and CDK5 are the kinases<br />

primarily responsible for phosphorylation of Tau, although other kinases such as PKC, PKA, and Erk2 are also involved. Hyperphosphorylation of Tau results in the dissociation<br />

of Tau from the microtubule, leading to microtubule destabilization and oligomerization of the Tau protein within the cell. Neurofibrillary tangles form as a result of Tau<br />

oligomerization and lead to apoptosis of the neuron.<br />

Select Reviews:<br />

Bossy-Wetzel, E., Schwarzenbacher, R., and Lipton, S.A. (2004) Nat. Med. 10, 2–9. • Chen, J.X. and Yan, S.S. (2010) J. Alzheimers Dis. 2, 569–578. • Claeysen, S.,<br />

Cochet, M., Donneger, R., Dumuis, A., Bockaert, J., and Giannoni, P. (2012) Cell. Signal. 24, 1831–1840. • Marcus, J.N. and Schachter, J. (2011) J. Neurogenet. 25,<br />

127–133. • Müller, W.E., Eckert, A., Kurz, C., Eckert, G.P., and Leuner, K. (2010) Mol. Neurobiol. 41, 159–171. • Nizzari, M., Thellung, S., Corsaro, A., Villa, V., Pagano,<br />

A., Porcile, C., Russo, C., and Florio, T. (2012) J. Toxicol. 2012, 187297. • Thinakaran, G. and Koo, E.H. (2008) J. Biol. Chem. 283, 29615–29619.<br />

Parkinson’s disease is the second most prevalent neurodegenerative disorder. Clinically, this disease is characterized by bradykinesia, resting tremors, and rigidity due to<br />

loss of dopaminergic neurons within the substania nigra section of the ventral midbrain. In the normal state, release of the neurotransmitter dopamine in the presynaptic<br />

neuron results in signaling in the postsynaptic neuron through D1- and D2-type dopamine receptors. D1 receptors signal through G proteins to activate adenylate cyclase,<br />

causing cAMP formation and activation of PKA. D2-type receptors block this signaling by inhibiting adenylate cyclase. Parkinson’s disease can occur through both genetic<br />

mutation (familial) and exposure to environmental and neurotoxins (sporadic). Recessively inherited loss-of-function mutations in parkin, DJ-1, and PINK1 cause mitochondrial<br />

dysfunction and accumulation of reactive oxidative species (ROS), whereas dominantly inherited missense mutations in α-synuclein and LRRK2 may affect protein degradation<br />

pathways, leading to protein aggregation and accumulation of Lewy bodies. Mitochondrial dysfunction and protein aggregation in dopaminergic neurons may be responsible<br />

for their premature degeneration. Another common feature of the mutations in α-synuclein, Parkin, DJ-1, PINK1, and LRRK2 is the impairment in dopamine release and<br />

dopaminergic neurotransmission, which may be an early pathogenic precursor prior to death of dopaminergic neurons. Exposure to environmental and neurotoxins can also<br />

cause mitochondrial functional impairment and release of ROS, leading to a number of cellular responses including apoptosis and disruption of protein degradation pathways.<br />

There is also an inflammatory component to this disease, resulting from activation of microglia that cause the release of inflammatory cytokines and cell stress. This microglia<br />

activation causes apoptosis via the JNK pathway and by blocking the Akt signaling pathway via REDD1.<br />

Select Reviews:<br />

Dauer, W. and Przedborski, S. (2003) Neuron 39, 889–909. • Girault, J.A. and Greengard, P. (2004) Arch. Neurol. 61, 641–644. • Patten, D.A., Germain, M., Kelly, M.A.,<br />

and Slack, R.S. (2010) J. Alzheimers Dis. 20, 357–367. • Imai, Y. and Lu, B. (2011) Curr. Opin. Neurobiol. 21, 935–941. • Springer, W. and Kahle, P.J. (2011) Autophagy<br />

7, 266–278.<br />

© 2009–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Christopher Phiel, Univ. of Colorado, Denver, CO and Prof. Jeff Kuret, Ohio State Univ., Columbus, OH for reviewing this diagram.<br />

190 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

© 2009–2015 Cell Signaling Technology, Inc. • We would like to thank Prof. Jie Shen, Harvard Medical School, Boston, MA, for reviewing this diagram.<br />

www.cellsignal.com/cstpathways 191


III<br />

Workflow<br />

Tools<br />

The biomedical research workflow is often a multi-dimensional rather<br />

than linear process. Depending on the question you are trying to answer,<br />

the process may include surveying a new cell type or disease model to<br />

identify biologically relevant alterations or performing detailed functional<br />

analysis of a single protein or post-translationally modified site.<br />

One of the keys to success is finding the right tool to answer each question<br />

that emerges in your investigation. In the following pages, we invite<br />

you to explore several research tools that can deliver valuable information<br />

and insights as you navigate the various phases of your research projects.<br />

Section Includes:<br />

Explore research tools<br />

How these tools can benefit your research<br />

Example data<br />

Modulation<br />

pg 266<br />

Localization &<br />

Classification<br />

pg 269<br />

Screening &<br />

Quantification<br />

pg 272<br />

Profiling<br />

pg 264<br />

Monitoring<br />

pg 274<br />

Identification<br />

pg 262<br />

Research<br />

Verification<br />

pg 277<br />

Discovery<br />

pg 258<br />

Customization<br />

pg 278<br />

Investigation<br />

pg 254<br />

Exploration<br />

pg 250<br />

Vesicle Trafficking<br />

Multiple levels shown of key pathways and structures involved in ER and Golgi-mediated<br />

trafficking and protein processing, including post-translational modifications.<br />

www.cellsignal.com/cstlandscapes<br />

249


17<br />

Section III: Workflow Tools<br />

Exploration<br />

using PhosphoSitePlus® online database<br />

How can I explore what is known about the post-translational<br />

modifications (PTMs) occurring on my protein of interest<br />

The PhosphoSitePlus ® (PSP) resource is a comprehensive protein modification knowledgebase,<br />

including experimentally determined PTMs from peer-reviewed journals and analytical tools that enable<br />

researchers to explore protein sequences, structures, mutations, pathways, and regulation. Created<br />

with grant support from the NIH and Cell Signaling Technology (<strong>CST</strong>), and curated by <strong>CST</strong> scientists,<br />

this database is continuously updated and features a dynamic, open, and highly interactive interface.<br />

It is designed to facilitate the study of the roles of PTMs in normal and pathological cellular processes,<br />

and to help accelerate the discovery of critical disease biomarkers and drug targets.<br />

Home Page<br />

Starting point for interacting with PhosphoSitePlus®<br />

Users can choose from two types of Simple Searches, three Advanced Searches, and three<br />

Browsing Interfaces.<br />

A. Simple Searches for Protein or Kinase Substrates<br />

In addition to the protein search, which will lead to a Protein Page, the substrate search asks the user<br />

to enter the name of a kinase and returns a list of experimentally verified in vivo and in vitro sites<br />

phosphorylated by the selected kinase. The preferred substrate sequences can be summarized and<br />

viewed as a Sequence Logo.<br />

B. Advanced Search and Browsing Options<br />

Search interfaces give users the power to explore what is known about proteins and their PTMs.<br />

Browse interfaces enable users to retrieve data from thousands of high-throughput (HTP) mass<br />

spectrometry experiments.<br />

chapter 17: Exploration<br />

PhosphoSitePlus ® allows a researcher<br />

to search for or download:<br />

• Modified proteins, modification sites and sequences associated with specific protein types,<br />

molecular weights, protein domains, treatments, tissues, diseases, cell lines, and references<br />

• Sets of known protein kinase substrates from in vivo and in vitro experiments<br />

• Specific sequences containing modified residues<br />

• Scripts to generate molecular structures (PyMOL of DeLano Scientific LLC and Chimera) in which<br />

all modified residues are labeled and color-coded<br />

• Datasets of PTMs that overlap known mutation sites (nsSNPs), enabling the researcher to identify<br />

those that have the potential to rewire signaling pathways<br />

• Datasets of modification sites observed in MS-MS experiments associated with specific diseases,<br />

cell lines, or tissues<br />

• Complete datasets of all modification sites reported to regulate downstream processes<br />

• Pathway datasets in Cytoscape ® of The Cytoscape Consortium and BioPAX formats<br />

• Sequence logos and motif analyses of sets of modification sites generated within PSP or<br />

submitted by the user<br />

Search Proteins or Sequences by:<br />

• Name or accession ID<br />

• Protein type or domain<br />

• Cellular component<br />

• Molecular weight range<br />

• Sequence/motif<br />

• List of peptides<br />

Search References by:<br />

• Author, PubMed ID, or protein name<br />

Search for Sites by:<br />

• Response to treatments<br />

• Tissues, cell line, or cell type<br />

• Correlation with disease state<br />

• Protein type, cellular component<br />

• Protein function or biological process<br />

Comparative Site Search:<br />

• Identify sites observed in one<br />

set of conditions but not others<br />

• Sequence, motif, or domain<br />

• Protein molecular weight<br />

Downloads Datasets:<br />

• Sites and metadata of major<br />

modification type<br />

• Disease mutations and genetic<br />

variants that overlap PTMs<br />

• Sites reported to regulate<br />

downstream processes<br />

• Sites regulated by specified<br />

treatments<br />

• Pathway information in Cytoscape ®<br />

and BioPAX formats<br />

Browse and Download HTP<br />

Data Associated with:<br />

• Specific diseases<br />

• Cell lines<br />

• Tissue<br />

Sequence Analysis Tools:<br />

• Sequence Logo Generator<br />

• Motif Analyzer<br />

Navigation and Content<br />

Interactive content is delivered in four increasingly granular interfaces. Navigation between the interfaces<br />

is seamless. Each page has links back to the Homepage.<br />

A<br />

Home Page; is the launch site for<br />

searching, browsing, static downloads,<br />

and informational pages.<br />

Protein Page; opens when the<br />

user selects the name of a protein<br />

from a Search Results page<br />

Home Page<br />

Protein Page<br />

Modification Site Page<br />

B<br />

Modification Site Page; opens<br />

when a user clicks on a specific site<br />

either from the Protein Page or from<br />

a Search Results page.<br />

Curated Information Page<br />

Curated Information Page;<br />

opens when a user clicks on a<br />

specific reference on a Modification<br />

Site Page, and presents the most<br />

granular information in PSP.<br />

250 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/exploration<br />

251


Section III: Workflow Tools<br />

chapter 17: Exploration<br />

Protein Page<br />

Provides information about a protein and its PTMs.<br />

Modification Site Page<br />

Summarizes information on single site from all records.<br />

C. Overview:<br />

• Brief description of protein function<br />

• Protein type and cellular component<br />

• Chromosomal location of human<br />

ortholog<br />

• Molecular function and<br />

biological process<br />

• Accession IDs, synonyms,<br />

and gene symbols<br />

• Molecular weight, isoelectric<br />

point, and pI calculator<br />

• Associated molecular structures<br />

and viewer<br />

• <strong>CST</strong> pathways and antibodies<br />

• Structural viewer<br />

D. Linked Resources Include:<br />

• Pathways: STRING, Reactome,<br />

NetworKIN<br />

• Expression: Protein Atlas, BioGPS<br />

• Scansite: Predictor of Kinases and<br />

Interactors<br />

• KinBase: Kinase Database at The<br />

Salk Institute<br />

• Structures: RCSB PDB, Pfam<br />

• NextGen Protein Resource: neXtProt<br />

E. Sites Implicated In:<br />

• Biological and molecular regulation,<br />

with links to associated site pages<br />

F. Modification Sites<br />

and Domains:<br />

• Zoomable linear diagram<br />

with sequence<br />

• Domains and modification<br />

sites mapped<br />

• Domain names linked to Pfam<br />

C<br />

D<br />

E<br />

F<br />

H<br />

I<br />

J<br />

Curated Information Page<br />

Experimental details on all sites in a single record.<br />

H. Site Information:<br />

•For most sites identified at <strong>CST</strong>, links<br />

to MS/MS spectra are included<br />

• Scansite predictions provide potential<br />

kinases and binding partners<br />

• Blast links allow site comparison<br />

against NCBI, UniProt, or PDB<br />

I. Experimental Summary<br />

Sections:<br />

May include information about how<br />

the site was experimentally characterized;<br />

diseases, cell lines and tissues<br />

in which it was observed; upstream<br />

control by treatments, receptors and<br />

other cellular proteins, and enzymes<br />

that may directly modify the site<br />

(in vivo and in vitro)<br />

J. References:<br />

Information curated from the literature<br />

contains links to the PubMed entry.<br />

Information curated from MS/MS<br />

experiments at <strong>CST</strong> or other institutions<br />

indicates the biological sample studied<br />

and treatments. If performed at <strong>CST</strong><br />

with PTMScan ® Technology, the<br />

antibody used for peptide enrichment<br />

prior to MS/MS is indicated.<br />

Links to the Curated Information Page<br />

are provided for most modification sites.<br />

G. Table of Sites, Sequences,<br />

and References:<br />

• Modification sites and surrounding<br />

sequences (+/- 7 AA) from parent<br />

protein, orthologs, and isoforms<br />

• Red characters indicate modified<br />

sites with links to associated records<br />

• First column (SS): the number<br />

of records using site-specific,<br />

low-throughput techniques<br />

• Second column (MS): the number of<br />

records using mass spectrometrybased<br />

high-throughput techniques<br />

G<br />

Download PyMOL and/or Chimera script<br />

Highlight modified residues<br />

K<br />

L<br />

K. Record and Sites:<br />

Standard bibliographic information<br />

about the record links to its PubMed<br />

entry. A list of modification sites<br />

associated with this record links to<br />

the the details curated for each site.<br />

L. Details Curated for<br />

each Modification Site:<br />

• Methods used to characterize site<br />

• Upstream Regulation<br />

– Kinases, phosphatases, receptors<br />

and signaling intermediates<br />

that regulate modification<br />

– Treatments that regulate<br />

modification<br />

• Downstream Regulation<br />

– Effects on protein function and<br />

biological processes due to<br />

modification<br />

– Protein-protein interactions directly<br />

influenced by modification<br />

• Disease Relevance of Modification<br />

252 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/exploration<br />

253


18 Investigation<br />

Section III: Workflow Tools<br />

using motif and post-translational<br />

modification-specific antibodies<br />

Motif and post-translational modification (PTM) antibodies<br />

can help answer two major scientific questions.<br />

1. How can I measure global changes in PTMs or protein kinase activity in response<br />

to cellular treatments<br />

2. How can I investigate PTMs on a protein of interest without complex in vivo labeling<br />

experiments or where no target-specific antibody is available<br />

Post-translational modifications of proteins catalyzed by kinases, acetylases, methylases, ubiquitin<br />

ligases, and other enzymes are key regulators of cell signaling and can be valuable targets for therapeutic<br />

intervention in disease states. Antibodies recognizing specific protein modification sites (e.g.<br />

Phospho-Akt, Acetyl-Histone) are well-known and valuable tools for studying PTMs. To augment these<br />

tools, <strong>CST</strong> has developed motif and PTM-specific antibodies, a distinct class of PTM reagents that<br />

have a broader target specificity and can therefore be used for a different set of applications.<br />

Motif Antibodies<br />

PTMs catalyzed by a specific modification enzyme are often constrained to a peptide sequence pattern,<br />

or motif, that reflects the substrate specificity of that enzyme. For example, substrates of the<br />

protein kinase Akt generally contain an RXRXXS motif in which Akt phosphorylates the serine residue.<br />

Antibodies that detect the motif RXRXXpS can be used to detect a broad spectrum of Akt substrates.<br />

Motif antibodies, proprietary to <strong>CST</strong>, recognize a modified residue in the context of a specific peptide<br />

sequence motif. These highly specialized antibodies were originally developed for the identification<br />

and quantification of PTMs catalyzed by kinases from several branches of the human kinome. More<br />

recently, individual motif antibodies have been developed to recognize several other post-translational<br />

modifications such as methyl-arginine and caspase cleavage sites.<br />

PTM-specific Antibodies<br />

PTM-specific antibodies differ from motif antibodies in that they recognize a specific protein posttranslational<br />

modification, but are not restricted to a specific sequence motif surrounding the modification<br />

site. Examples of PTM-specific antibodies from <strong>CST</strong> include Phospho-Tyrosine (P-Tyr-1000) Rabbit<br />

mAb #8954 and Acetylated-Lysine (Ac-K 2 -100) Rabbit mAb #9814. Together, motif and PTM-specific<br />

antibodies provide a robust toolbox for the identification and classification of PTMs and their effects on<br />

cellular regulation.<br />

How can motif and PTM-specific antibodies be used<br />

• Quantitatively identify the PTM alterations of cellular proteins due to up- or down-regulation<br />

of a specific kinase, acetylase, methylase, nitrosylase, or ubiquitin ligase.<br />

• Motif and PTM antibodies can be used in early-stage investigations where a high quality<br />

antibody may not be available for a specific modified protein target. Assay target-specific<br />

PTM alterations via immunoprecipitation (IP) enrichment of the target protein followed by<br />

western blot (WB) analysis using the motif or PTM-specific antibody.<br />

• Screen candidate therapeutics using in vitro kinase and phosphatase assays based on a<br />

motif or PTM-specific antibody.<br />

One example of the application of a motif antibody involves the response of Jurkat cells to the PP1 and<br />

PP2A protein phosphatase inhibitor Calyculin A. The response was studied by 2D gel electrophoresis<br />

followed by WB analysis using a <strong>CST</strong> motif antibody recognizing the phospho-serine 14-3-3 binding<br />

motif. The data reveal a pronounced increase in overall phosphorylation levels. Based on this initial<br />

study, individual proteins of interest are selected for further study.<br />

Specific kinases in the human kinome phosphorylate<br />

target proteins at distinct substrate sequence motifs.<br />

CHED<br />

Haspin<br />

STLK3<br />

MAST4<br />

chapter 18: Investigation<br />

TK<br />

FGFR2 FGFR3<br />

TrkC<br />

EphB2<br />

TrkB<br />

FGFR1<br />

FGFR4<br />

EphB1<br />

TrkA<br />

FLT1/VEGFR1<br />

ATM<br />

EphA5<br />

KDR/VEGFR2<br />

MuSK ROR2<br />

Fms/CSFR<br />

Atypical<br />

ROR1<br />

EphB3<br />

Ret<br />

Kit<br />

EphA3<br />

DDR2<br />

Mer<br />

ATM<br />

EphA4<br />

DDR1<br />

Tyro3/<br />

Axl<br />

FLT4<br />

Sky<br />

ATR<br />

PDGFRα<br />

EphA6<br />

IGF1R IRR<br />

FLT3<br />

PDGFRβ<br />

mTOR/FRAP<br />

InsR<br />

Yes<br />

EphB4<br />

Met<br />

EGFR<br />

PIKK<br />

HER2/ErbB2<br />

Ron<br />

DNAPK<br />

Src<br />

MLK3<br />

Ros<br />

SMG1<br />

EphA7<br />

ALK<br />

MLK1<br />

Lyn<br />

LTK<br />

Tie2<br />

Tie1<br />

TRRAP<br />

HCK<br />

Fyn<br />

RYK<br />

HER4<br />

EphA8<br />

MLK4<br />

CCK4/PTK7<br />

TKL<br />

MLK2<br />

Lck<br />

Fgr<br />

Ack Tnk1 Tyk2<br />

Jak1<br />

HER3<br />

Jak2<br />

EphA2<br />

Jak3<br />

BLK<br />

ANKRD3 SgK288<br />

DLK<br />

Syk Zap70/SRK<br />

EphA1<br />

PYK2/FAK2<br />

LZK<br />

FAK<br />

Lmr1<br />

ALK4<br />

ITK<br />

Lmr2<br />

C-Raf/Raf1<br />

FRK<br />

TGFβR1<br />

ZAK<br />

BRaf<br />

TEC<br />

EphB6<br />

KSR<br />

Srm<br />

RIPK2<br />

KSR2<br />

TXK<br />

Brk<br />

BTK<br />

Lmr3<br />

ALK7<br />

IRAK3<br />

IRAK1<br />

LIMK1<br />

ARaf<br />

BMPR1B<br />

Etk/BMX<br />

EphA10<br />

LIMK2 TESK1 ILK<br />

BMPR1A<br />

CTK<br />

RIPK3<br />

TSK2<br />

TAK1<br />

HH498<br />

ALK1<br />

CSK CK2a1<br />

PAK1<br />

ALK2<br />

Abl2/Arg<br />

IRAK2<br />

ActR2<br />

Abl Fes<br />

ActR2B<br />

STE<br />

Fer<br />

RIPK1<br />

Jak3~b<br />

TGFβR2<br />

LRRK2<br />

MEKK2/MAP3K2<br />

Jak2~b<br />

LRRK1<br />

MISR2<br />

MEKK3/MAP3K3<br />

Tyk2~b<br />

SuRTK106<br />

IRAK4<br />

BMPR2<br />

ASK/MAP3K5<br />

ANPα/NPR1<br />

MAP3K8<br />

Jak1~b<br />

MAP3K7<br />

ANPβ/NPR2<br />

KHS1<br />

MOS<br />

KHS2<br />

HSER<br />

SgK496<br />

WNK1<br />

WNK3<br />

MEKK6/MAP3K6<br />

DYRK2<br />

Mst4<br />

PBK<br />

MAP3K4<br />

DYRK3<br />

GUCY2D<br />

WNK2<br />

DYRK4<br />

DYRK1A<br />

NRBP1<br />

GUCY2F<br />

NRBP2 MEKK1/MAP3K1 OSR1<br />

DYRK1B<br />

WNK4<br />

MLKL<br />

PERK/PEK<br />

SgK307<br />

SLK<br />

PKR<br />

LOK<br />

HIPK1 HIPK3<br />

GCN2 SgK424<br />

TAO1<br />

SCYL3 TAO2<br />

HIPK2<br />

SCYL1<br />

Tpl2/COT<br />

SCYL2<br />

NIK<br />

TAO3<br />

PAK1<br />

CLK4 HIPK4<br />

PRP4<br />

HRI<br />

PAK3<br />

CLIK1<br />

PAK2<br />

IRE1<br />

CLK2 CLK1<br />

PAK4<br />

MAP2K5<br />

PAK5/PAK7<br />

CLIK1L<br />

IRE2<br />

CLK3<br />

TBCK<br />

PAK6<br />

MAP2K7<br />

MEK1/MAP2K1<br />

RNAseL<br />

GCN2~b<br />

MEK2/MAP2K2<br />

TTK<br />

MSSK1<br />

SgK071<br />

KIS<br />

GRK2<br />

CMGC<br />

SRPK2<br />

MYT1<br />

SEK1/MAP2K4 MKK3/MKK6<br />

SRPK1<br />

CK2α1<br />

Wee1<br />

MAK<br />

CK2α2<br />

SgK196<br />

CDC7<br />

Wee1B<br />

CK1δ<br />

ICK<br />

PRPK<br />

TTBK1<br />

CK1ε<br />

GSK3β<br />

MOK<br />

TTBK2<br />

GSK3α<br />

CK1α1<br />

CDKL3<br />

CK1α2<br />

CDKL2<br />

PINK1<br />

SgK493<br />

SgK269<br />

VRK3<br />

CK1γ2<br />

CDKL1 CDKL5<br />

ERK7<br />

SgK396<br />

SgK223<br />

CDKL4 Erk4<br />

Slob<br />

CK1γ1<br />

Erk3<br />

SgK110<br />

PIK3R4<br />

CK1γ3<br />

NLK<br />

SgK069<br />

Bub1<br />

Erk5<br />

SBK<br />

BubR1<br />

Erk1/<br />

IKKα<br />

p44MAPK<br />

IKKβ<br />

VRK1<br />

CDK7<br />

IKKε<br />

VRK2<br />

Erk2/<br />

Erk2<br />

CK1<br />

PLK4<br />

PITSLRE<br />

TBK1/NAK<br />

p42MAPK p38γ<br />

MPSK1<br />

JNK1<br />

p38δ<br />

JNK2<br />

TLK2<br />

JNK3 CDK10<br />

GAK<br />

TLK1<br />

PLK3<br />

p38β<br />

AAK1<br />

CDK8 CDK11<br />

CAMKK1<br />

ULK3<br />

PLK1<br />

p38α<br />

PLK2<br />

CDK4<br />

CCRK<br />

BIKE<br />

CAMKK2<br />

BARK1/GRK2<br />

CDK6<br />

ULK1<br />

BARK2/GRK3 RHOK/GRK1<br />

Fused<br />

GRK5<br />

PFTAIRE2<br />

SgK494<br />

ULK2 ULK4<br />

GRK6 GRK4<br />

PFTAIRE1<br />

CDK9<br />

Nek6<br />

RSKL1<br />

PCTAIRE2<br />

Nek7 Nek10<br />

SgK495<br />

PASK<br />

PDK1<br />

RSKL2<br />

Nek8<br />

MSK1<br />

RSK1/p90RSK<br />

PCTAIRE1<br />

CDK5 CRK7<br />

PCTAIRE3<br />

Nek9<br />

LKB1<br />

MSK2<br />

RSK4 RSK2<br />

Chk1<br />

p70S6K<br />

RSK3<br />

Nek2<br />

Akt2/PKBβ<br />

AurA/Aur2<br />

p70S6Kβ<br />

Akt1/PKBα<br />

cdc2/CDK1<br />

Nek11<br />

CDK3<br />

CDK2<br />

Nek4<br />

AurB/Aur1<br />

Trb3 Pim1<br />

AurC/Aur3<br />

Pim2<br />

LATS1<br />

Nek3<br />

Pim1Trb2<br />

Pim3<br />

Nek5<br />

Trad Trio<br />

LATS2<br />

NDR1<br />

Trb1<br />

Obscn~b<br />

NDR2<br />

Nek1<br />

YANK1<br />

SPEG~b<br />

MAST3<br />

MASTL<br />

Obscn<br />

STK33<br />

YANK2 PKN1<br />

YANK3<br />

SPEG<br />

TTN<br />

MAST2<br />

SgK085<br />

caMLCK<br />

MAPKAPK2<br />

skMLCK<br />

smMLCK<br />

DRAK2<br />

DRAK1<br />

DAPK2<br />

DAPK3<br />

DAPK1<br />

SSTK<br />

TSSK3<br />

TSSK1<br />

TSSK2<br />

AMPKα2<br />

AMPKα1<br />

BRSK2<br />

CAMK<br />

BRSK1<br />

SNARK<br />

ARK5<br />

QSK<br />

SNRK<br />

NIM1<br />

SIK<br />

QIK<br />

TSSK4<br />

MELK<br />

MARK4<br />

MARK3<br />

MARK1<br />

MARK2<br />

HUNK<br />

PKD2/PKCµ<br />

PKD1<br />

PKD3/PKCν<br />

MNK1<br />

MNK2<br />

RSK4~b<br />

RSK1~b<br />

RSK2~b<br />

RSK3~b<br />

CASK<br />

MAPKAPK5<br />

MAPKAPK2<br />

MAPKAPK3<br />

MSK2~b<br />

MSK1~b<br />

Chk2/Rad53<br />

STRAD/STLK5<br />

STLK6<br />

CaMKIβ<br />

CaMKIγ<br />

MAST1<br />

DCAMKL3<br />

DCAMKL1<br />

DCAMKL2<br />

VACAMKL<br />

PhKγ1<br />

PhKγ2<br />

PSKH1<br />

CaMKIIγ<br />

PSKH2<br />

CaMKIIα<br />

CaMKIIβ<br />

CaMKIIδ<br />

CaMKIV<br />

CaMKIα<br />

CaMKIδ<br />

GRK7<br />

HPK1<br />

GCK<br />

Akt3/PKBγ<br />

SGK1<br />

SGK2<br />

SGK3<br />

PKG2<br />

PKN1/PRK1<br />

PKG1<br />

PKN2/PRK2<br />

PKN3<br />

PRKY<br />

PKCδ<br />

PKCθ<br />

PRKX<br />

PKCη<br />

PKCε<br />

PKCι<br />

PKCζ<br />

PKAγ<br />

PKAα<br />

PKCγ<br />

PKAβ<br />

AGC<br />

ROCK1<br />

PKCα<br />

ROCK2<br />

PKCβ<br />

DMPK<br />

CRIK<br />

DMPK2<br />

MRCKβ<br />

MRCKα<br />

Representative phosphorylation motifs from the PhosphoSitePlus ® online resource. Logos for individual motifs are superimposed on the kinase dendrogram developed in collaboration<br />

between Gerard Manning and Cell Signaling Technology (1). These and over 100 other kinase motifs can be generated for kinases that have 15 or more unique protein substrates reported<br />

in the literature, and these are curated into the PhosphoSitePlus ® knowledge base (2).<br />

References<br />

1. Manning, G., et al. (2002) Science 298, 1912–1934.<br />

2. Hornbeck, P.V., et al. (2012) Nucleic Acids Res. 40, 261–270.<br />

NRK/ZC4<br />

Mst1<br />

Mst2<br />

TNIK/ZC2<br />

MYO3A<br />

MYO3B<br />

YSK1<br />

Mst3<br />

HGK/ZC1<br />

MINK/ZC3<br />

Akt<br />

254 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/investigation<br />

255


Section III: Workflow Tools<br />

chapter 18: Investigation<br />

Treatment with<br />

Calyculin A induces<br />

a broad increase in<br />

phosphorylation at<br />

phospho-Ser 14-3-3<br />

substrate motif sites.<br />

How <strong>CST</strong> motif and PTM-specific<br />

antibodies can benefit your research:<br />

• Multi-pathway Analysis: motif antibodies allow the detection of a broad range of substrates for<br />

a single kinase, so multiple pathways can be analyzed simultaneously using a single antibody.<br />

SDS-PAGE<br />

→<br />

• Multiplex Applications: many <strong>CST</strong> motif antibodies are validated for IP, peptide ELISA (E-P),<br />

and/or immunohistochemistry (IHC) in addition to WB, enabling flexibility in experimental design<br />

and the possibility of confirming results via different applications.<br />

• Proteomic Analysis: <strong>CST</strong> PTMScan ® services and kits integrate motif and PTM-specific<br />

antibodies with mass spectrometry (MS) to enable the discovery and analysis of hundreds<br />

to thousands of PTM sites using standard LC-MS techniques.<br />

Untreated<br />

→<br />

IPGE<br />

Calyculin A<br />

→<br />

IPGE<br />

Phospho-(Ser) 14-3-3 Binding<br />

Motif Antibody #9601: WB analysis of<br />

extracts from Jurkat cells, untreated or<br />

Calyculin A-treated (0.1 µM for 30 min),<br />

using #9601. Proteins were separated<br />

by 2-D electrophoresis prior to blotting.<br />

#9646 Phospho-Akt Substrate (RXXS*/T*) (110B7E) Rabbit mAb<br />

IP †<br />

RXX(S*/T*)<br />

(Sepharose ® Bead Conjugate)<br />

#9611 Phospho-(Ser/Thr) Akt Substrate Antibody W, IP † , IHC-P, E-P (K/R)XX(S*/T*)<br />

#10001 Phospho-Akt Substrate (RXRXXS*/T*) (23C8D2) Rabbit mAb W, E-P RXRXX(S*/T*)<br />

#5759 Phospho-(Ser/Thr) AMPK Substrate (P-S/T 2 -102) Rabbit mAb W, IP † , E-P (L/M)XRXX(S*/T*),<br />

RXX(S*/T*)<br />

#2909 Phospho-(Ser/Thr) ATM/ATR Substrate (4F7) Rabbit mAb W L(S*/T*)Q<br />

#2851 Phospho-(Ser/Thr) ATM/ATR Substrate Antibody W, IP † , IHC-P, E-P L(S*/T*)Q<br />

#6966 Phospho-(Ser/Thr) ATM/ATR Substrate (S*/T*QG) (P-S/T2-100) Rabbit mAb W, IP † (S*/T*)QG, (S*/T*)Q<br />

#8738 Phospho-(Ser/Thr) CK2 Substrate (P-S/T 3 -100) Rabbit mAb W (S*/T*)DXE<br />

#9634 Phospho-(Ser/Thr) PDK1 Docking Motif (18A2) Mouse mAb W, IP † , E-P (F/K)XX(F/Y)(S*/T*)(F/Y)<br />

#9631 Phospho-(Ser/Thr) Phe Antibody W, IP † , E-P (F/Y/W)(S*/T*)(F)<br />

#9624 Phospho-PKA Substrate (RRXS*/T*) (100G7E) Rabbit mAb W, IP † , E-P (K/R)(K/R)X(S*/T*)<br />

#9621 Phospho-(Ser/Thr) PKA Substrate Antibody W, IP † , IHC-P, E-P (K/R)(K/R)X(S*/T*)<br />

#4381 Phospho-(Ser/Thr) PKD Substrate Antibody W, E-P LXRXX(S*/T*)<br />

Phospho-Tyrosine Application Motif Recognized<br />

#8954 Phospho-Tyrosine (P-Tyr-1000) Rabbit mAb W, IP † , IF-IC, F Y*<br />

#9411 Phospho-Tyrosine Mouse mAb (P-Tyr-100) W, IP † , IHC-P, IF-P, Y*<br />

IF-F, IF-IC, F, E-P<br />

#9414 Phospho-Tyrosine Mouse mAb (P-Tyr-100) (Alexa Fluor ® 488 Conjugate) F Y*<br />

#9415 Phospho-Tyrosine Mouse mAb (P-Tyr-100) (Alexa Fluor ® 647 Conjugate) IF-IC, F Y*<br />

#9417 Phospho-Tyrosine Mouse mAb (P-Tyr-100) (Biotinylated) W, IP † , IHC-P Y*<br />

Antibody recognition<br />

of the PKA substrate<br />

motif is phosphorylation-dependent.<br />

+ + + +<br />

– – – + – – – –<br />

Phospho-(Ser/Thr) PKA Substrate<br />

Antibody #9621: WB analysis of extracts<br />

from A-431 cells, phosphorylated in vitro<br />

by protein kinase A (A), Erk2 (B) or cdc2/<br />

cyclin A (C), plus or minus PKA inhibitor<br />

(PKI), using #9621.<br />

Lanes<br />

1. PKA<br />

2. Erk2<br />

3. cdc2/Cyclin A<br />

A B C<br />

Cell Extract<br />

PKI<br />

#5465 Phospho-Tyrosine Mouse mAb (P-Tyr-100) (HRP Conjugate) W, E-P Y*<br />

Motif and PTM-specific antibodies available from <strong>CST</strong><br />

As with our more traditional antibodies, <strong>CST</strong>’s motif and PTM antibody development team invests significant<br />

time and resources ensuring that these reagents are highly specific for the motif or PTM of interest.<br />

These antibodies are validated by a number of methods including WB, peptide arrays, and peptide IP<br />

followed by LC-MS analysis to confirm that the antibody performs to the highest possible standard.<br />

#8095 Phospho-Tyrosine Mouse mAb (P-Tyr-100) (Magnetic Bead Conjugate) IP † Y*<br />

#9419 Phospho-Tyrosine Mouse mAb (P-Tyr-100) (Sepharose ® Bead Conjugate) IP † Y*<br />

#9416 Phospho-Tyrosine Mouse mAb (P-Tyr-102) W, IP † , IHC-P, F, E-P Y*<br />

Nitro-Tyrosine Application Motif Recognized<br />

#9691 Nitro-Tyrosine Antibody W Y-NO2<br />

EGF stimulates<br />

phosphorylation of<br />

tyrosine residues in<br />

many cellular proteins.<br />

Phospho-Serine Application Motif Recognized<br />

#9615 Phospho-(Ser) Kinase Substrate Antibody Sampler Kit<br />

#9606 Phospho-(Ser) 14-3-3 Binding Motif (4E2) Mouse mAb W, IP † , E-P (K/R)XXS*P<br />

#9601 Phospho-(Ser) 14-3-3 Binding Motif Antibody W, IP † , IHC-P, E-P (K/R)XXS*P<br />

#9607 Phospho-ATM/ATR Substrate (S*Q) (D23H2/D69H5) Rabbit mAb W, IP † S*Q<br />

#6967 Phospho-(Ser) PKC Substrate (P-S 3 -101) Rabbit mAb W, IP † , E-P (K/R)XS*X(K/R)<br />

Acetylated Lysine Application Motif Recognized<br />

#9814 Acetylated-Lysine (Ac-K 2 -100) Rabbit mAb W, IP † , ChIP, E-P Ac-K<br />

#6952 Acetylated-Lysine (Ac-K 2 -100) Rabbit mAb (HRP Conjugate) W Ac-K<br />

#9681 Acetylated-Lysine Mouse mAb (Ac-K-103) W, E-P Ac-K<br />

#9441 Acetylated-Lysine Antibody W, IP † , IHC-P, IF-IC, Ac-K<br />

ChIP, E-P<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

#2325 Phospho-MAPK/CDK Substrates (PXS*P or S*PXR/K) (34B2) Rabbit mAb W, IP † , IHC-P, E-P PXS*P, S*PX(K/R)<br />

#5501 Phospho-MAPK/CDK Substrates (PXS*P or S*PXR/K) (34B2) Rabbit mAb IP †<br />

PXS*P, S*PX(R/K)<br />

(Sepharose ® Bead Conjugate)<br />

#9477 Phospho-(Ser) CDKs Substrate (P-S 2 -100) Rabbit mAb W, IP † , E-P (K/H)S*P<br />

#2261 Phospho-(Ser) PKC Substrate Antibody W, IP † , E-P (K/R)XS*(Hyd)(K/R)<br />

#2981 Phospho-(Ser) Arg-X-Tyr/Phe-X-pSer Motif Antibody W, IP † , E-P RX(F/Y)XS*<br />

Phospho-Threonine Application Motif Recognized<br />

#9391 Phospho-Threonine-Proline Mouse mAb (P-Thr-Pro-101) W, IHC-P, E-P T*P<br />

#5243 Phospho-PLK Binding Motif (ST*P) (D73F6) Rabbit mAb W, IP † , E-P ST*P<br />

#2351 Phospho-Threonine-X-Arginine Antibody W, IHC-P, E-P T*X(K/R)<br />

#3004 Phospho-Thr-Pro-Glu (C32G12) Rabbit mAb W, IP † , E-P T*PE, T*P<br />

#9386 Phospho-Threonine (42H4) Mouse mAb W, IP † , E-P T*<br />

#9381 Phospho-Threonine Antibody (P-Thr-Polyclonal) W, IP † , E-P T*<br />

#8781 Phospho-Threonine Antibody (P-Thr-Polyclonal)<br />

IP † T*<br />

(Sepharose Bead Conjugate)<br />

#6949 Phospho-Threonine Antibody (P-Thr-Polyclonal) (HRP Conjugate) W, E-P T*<br />

Mono-/Di-Methyl-Arginine Application Motif Recognized<br />

#8015 Mono-Methyl Arginine (Me-R 4 -100) Rabbit mAb W, IP † , E-P Me-R<br />

#8711 Mono-Methyl Arginine (R*GG) (D5A12) Rabbit mAb W, IP † , E-P Me-RGG<br />

#13522 Asymmetric Di-Methyl Arginine Motif [adme-R] Rabbit mAb W ADMe-R<br />

#13222 Symmetric Di-Methyl Arginine Motif [sdme-RG] Rabbit mAb W SDMe-RG<br />

Ubiquitin Linkage Application Motif Recognized<br />

#8081 K48-Linkage Specific Polyubiquitin (D9D5) Rabbit mAb W Polyubiquitin formed<br />

by K48 linkage<br />

#12805 K48-linkage Specific Polyubiquitin (D9D5) Rabbit mAb (HRP Conjugate) W Polyubiquitin formed<br />

by K48 linkage<br />

#4289 K48-linkage Specific Polyubiquitin Antibody W Polyubiquitin formed<br />

by K48 linkage<br />

#5621 K63-Linkage Specific Polyubiquitin (D7A11) Rabbit mAb W Polyubiquitin formed<br />

by K63 linkage<br />

#12930 K63-linkage Specific Polyubiquitin (D7A11) Rabbit mAb (HRP Conjugate) W Polyubiquitin formed<br />

by K63 linkage<br />

60<br />

50<br />

40<br />

30<br />

– + hEGF<br />

Phospho-Tyrosine (P-Tyr-1000) Rabbit<br />

mAb #8954: WB analysis of extracts from<br />

A-431 cells, untreated or treated with<br />

Human Epidermal Growth Factor (hEGF)<br />

#8916 (100 ng/ml, 5 min), using #8954.<br />

†<br />

IP denotes suitability for standard IP, not for peptide immunoaffinity enrichment prior to LC-MS/MS.<br />

Phospho-Serine/Threonine Application Motif Recognized<br />

#9920 Phospho-(Ser/Thr) Kinase Substrate Antibody Sampler Kit<br />

#9614 Phospho-Akt Substrate (RXXS*/T*) (110B7E) Rabbit mAb W, IP † , IHC-P, E-P RXX(S*/T*)<br />

#6950 Phospho-Akt Substrate (RXXS*/T*) (110B7E) Rabbit mAb (HRP Conjugate) W RXX(S*/T*)<br />

#8050 Phospho-Akt Substrate (RXXS*/T*) (110B7E) Rabbit mAb<br />

(Magnetic Bead Conjugate)<br />

IP †<br />

RXX(S*/T*)<br />

PTMScan® Proteomics Services<br />

Quantitative PTM profiling — see page 258 or visit our website. www.cellsignal.com/discovery<br />

256 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/investigation<br />

257


19<br />

Section III: Workflow Tools<br />

chapter<br />

Discovery<br />

using peptide immunoaffinity enrichment and LC-MS/MS<br />

to study post-translational modifications (PTMs)<br />

How can I use PTM-specific proteomics to enable discovery<br />

of novel disease drivers and identification of biomarkers<br />

How can I profile changes in specific PTM sites on key target<br />

proteins to explore changes in critical signaling events<br />

Post-translational modifications such as phosphorylation, acetylation, methylation, ubiquitination, and<br />

others are critical regulators of protein activity and function. Understanding the role of PTMs in disease<br />

states and developing novel biomarkers and therapeutics are areas of intense research. However,<br />

researchers who study PTMs are challenged by their inherent low levels, which make them difficult to<br />

identify and quantify from whole cell or native samples.<br />

This challenge can be overcome by using immunoaffinity enrichment to isolate specific peptides<br />

containing PTMs of interest from a protease-digested cell or tissue extract. LC-MS/MS analysis is then<br />

employed to identify and quantify the isolated peptides. PTMScan ® Technology, proprietary to <strong>CST</strong>, utilizes<br />

the specificity of PTM-specific and motif antibodies to enrich target peptides from the background<br />

of non-modified endogenous peptides, enabling the identification of modified proteins that otherwise<br />

may not be detected. Hundreds to thousands of post-translationally modified peptides can be identified<br />

and quantified in a single experiment. This approach can be used to study modifications such as<br />

phosphorylation, acetylation, methylation, ubiquitination, succinylation, or caspase-mediated cleavage.<br />

The highly conserved nature of PTMs and substrate motifs across many different species also makes<br />

this approach applicable to many different model systems.<br />

PTM Site Discovery and Profiling<br />

• Discover candidate biomarkers linked to activation or repression of a specific class of PTMs.<br />

• Perform a comprehensive proteomic survey of thousands of PTM sites associated with a wide<br />

variety of organisms or a disease model system, integrating the results into known signaling<br />

pathways or helping to define novel signaling networks.<br />

• Detect substrates of novel signaling proteins (kinases, phosphatases, ubiquitin ligases,<br />

deubiquitinases, acetyl transferases, methyl transferases, or succinyl transferases).<br />

• Profile and quantify global effects of a candidate therapeutic on a specific type of PTM,<br />

identifying nodes of interest for further study.<br />

• Understand how cross-talk among various PTMs (phosphorylation, ubiquitination, acetylation,<br />

methylation) is related to a particular biological response or involved in cell development or<br />

differentiation.<br />

• Analyze downstream effects of targeted gene silencing on signaling and potential activation<br />

of alternative pathways.<br />

• Identify protein-protein interaction binding partners along with their corresponding PTMs.<br />

Discover Low Abundance PTM Sites<br />

Immunoaffinity enrichment allows you to identify low abundance modifications by selectively capturing<br />

the PTM in the context of the motif of interest from a population of endogenous peptides. The specificity<br />

of immunoaffinity enrichment complements that of IMAC, a charge-based metal affinity method<br />

commonly used for phospho-peptides. IMAC is driven by general coordination chemistry dictated by<br />

the immobilized metal ion (1). IMAC can therefore lead to enrichment of peptides from more abundant<br />

proteins for subsequent LC-MS/MS analysis, but not so readily for less abundant phospho-peptides.<br />

Complementary methods: immunoaffinity<br />

and IMAC enrichment for phospho-peptides.<br />

A total of 27,372 unique phosphopeptides<br />

were isolated from MKN-45<br />

cells using both IMAC and motif<br />

antibody enrichment strategies. In<br />

this study, distinct sets of modified<br />

peptides were identified by the two<br />

affinity enrichment methods, with<br />

only 5.6% overlap between the MS/<br />

MS identified peptides.<br />

Identification of Biomarkers<br />

PTMScan Technology has been used to identify novel biomarkers in a number of different model systems<br />

(2–6). One example of the application of PTMScan Technology was conducted in a large-scale<br />

survey of tyrosine kinase activity in lung cancer, performed at <strong>CST</strong>. For this study, we used a phosphotyrosine<br />

motif antibody to analyze changes in phosphorylation across the proteome in non-small cell<br />

lung carcinoma (NSCLC) cell lines and tissues.<br />

We surveyed the phospho-tyrosine status of receptor tyrosine kinases (RTK) and non-receptor tyrosine<br />

kinases in 41 NSCLC cell lines and over 150 NSCLC tumors. Over 50 tyrosine kinases and more than<br />

2,500 downstream substrates that play roles in NSCLC growth and progression were identified. Two<br />

very exciting findings from this study were the identification of novel ALK and ROS1 C-terminal fusion<br />

proteins (EML4-ALK, CD74-ROS, SLC34A2-ROS) in some NSCLC cell lines and tumors (7). Further<br />

investigation of ALK and ROS1 in other cancers led to the identification of a novel FN1-ALK fusion<br />

protein in ovarian cancer and a FIG-ROS1 fusion in cholangiocarcinoma (8,9).<br />

As one consequence of this study, a companion diagnostic immunohistochemistry (IHC) assay for<br />

NSCLC was developed. In 2011, the diagnostic IHC assay was approved in the U.S. for patients with tumor<br />

samples that stain positive for ALK fusion protein expression for crizotinib treatment of NSCLC (10).<br />

ALK fusion protein<br />

detection in human<br />

lung carcinoma<br />

ALK (D5F3 ® ) XP ® Rabbit mAb #3633:<br />

IHC analysis of paraffin-embedded<br />

human lung carcinoma with high (left)<br />

and low levels (right) of ALK fusion<br />

protein expression using #3633.<br />

All Antibodies*<br />

(13,480)<br />

Carcinoma-associated fusion proteins<br />

discovered using PTMScan ® Technology<br />

5.6%<br />

IMAC<br />

(13,892)<br />

*Phospho-Tyrosine, Basophilic Mix, Proline Mix, and Serine/Threonine Mix<br />

19: Discovery<br />

Discovery workflow<br />

for phospho-tyrosine<br />

modifications<br />

altered in NSCLC<br />

Cell or Tissue Samples<br />

Kinase Family Targeting<br />

Immunoprecipitation<br />

Using Motif Antibody<br />

LC-MS/MS and<br />

Bioinformatic Analysis<br />

258 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/discovery<br />

259


Section III: Workflow Tools<br />

chapter 19: Discovery<br />

How do I select the most relevant PTM to study<br />

The first step to discovery of novel PTM alterations in response to disease states or drug treatments<br />

is selecting the appropriate class of modifications for enrichment. If you are not certain which specific<br />

classes of modifications are altered in your experimental system, you can first perform a series of<br />

western blot analyses employing a panel of motif antibodies that detect a range of different PTMs<br />

(KinomeView ® ).<br />

What type of data can I expect<br />

from PTMScan ® kits and services<br />

The data set generated by a PTMScan service experiment includes not only quantification of PTM<br />

changes, but also the identity of each protein and specific location of each modification site. For ease<br />

of interpretation, the experimental results can be viewed at a global level as a scatter plot, as a detailed<br />

table view (below), or as a domain map for a selected protein. Using the appropriate LC-MS/MS platforms<br />

and informatics processing, the PTMScan kits can be used to generate comparable results.<br />

Identify Key Targets from Thousands of Peptides<br />

The process of data refinement can be illustrated by a simple scenario. A typical PTMScan proteomic<br />

analysis focused on a single class of PTM might identify 1,000–3,000 unique modified peptides. Of<br />

these, perhaps 50–100 might exhibit a clear change in modification that correlates with tissue type or<br />

with the expected biology and pathway associated with the cell treatment employed in the study. With<br />

further WB and/or ELISA experimentation, you can shorten this list of candidate PTM sites to a limited<br />

subset of actionable targets (1,4,6).<br />

Identify 1,000-3,000 unique PTM-containing peptides<br />

Adjust signal:noise cutoff<br />

to optimize results<br />

Representative PTMScan ® data, revealing the protein identity,<br />

site location, and relative abundance for each PTM detected.<br />

Normalized Fold Change<br />

SU11274 vs.<br />

DMSO Control<br />

Staurosporine<br />

vs. DMSO Control<br />

Protein<br />

Name Site -7/+ 7 Peptide Upstream Kinase<br />

-5.0 -4.6 EphA2 897 RVSIRLPsTSGSEGV LPS*T*SGSEGVPFR Akt1<br />

-13.6 -2.1 FOXO1A 319 TFRPRTSsNASTISG TSS*NASTISGR Akt1<br />

-158.0 -7.2 FOXO4 32 QSRPRSCtWPLPRPE SCT*WPLPRPEIANQPSEPPEVEPDLGEK Akt1<br />

-3.4 1.8 QIK 358 DGRQRRPsTIAEQTV RPS*TIAEQTVAK Akt1, Akt2<br />

-13.3 -29.4 S6 235,<br />

236,<br />

240<br />

-7.0 -24.5 S6 236,<br />

240<br />

IAKRRRLsSLRASTS RLS*S*LRAS*TSK Akt1, Akt2,<br />

P70S6KB, PKACA,<br />

PKCA, PKCD<br />

AKRRRLSsLRASTSK RLSS*LRAS*TSK Akt1, Akt2,<br />

P70S6KB, PKACA,<br />

PKCA, PKCD<br />

2.6 1.1 BRAF 365 GQRDRSSsAPNVHIN SSS*APNVHINTIEPVNIDDLIR Akt1, Akt3<br />

-7.0 -9.4 GSK3B 9 SGRPRTTsFAESCKP TTS*FAESCKPVQQPSAFGSMK Akt1, AurA,<br />

CAMK2B, GSK3B,<br />

KHS1, PKACA,<br />

PKCA<br />

-5.3 N.D. GSK3B 9, 21 SGRPRTTsFAESCKP TTS*FAESCKPVQQPS*AFGSMK Akt1, AurA,<br />

CAMK2B, GSK3B,<br />

KHS1, PKACA,<br />

PKCA<br />

-21.3 -3.0 PEA-15 116 KDIIRQPsEEEIIKL DIIRQPS*EEEIIK Akt1, CAMK2A,<br />

CK2A1<br />

-2.1 -2.9 GSK3A 21 SGRARTSsFAEPGGG TSS*FAEPGGGGGGGGGGPGGSASGPGGTGGGK Akt1, CAMK2B,<br />

PKACA, PKCA,<br />

PKCB<br />

-10.3 -1.8 RANBP3 126 VKRERTSsLTQFPPS TSS*LTQFPPSQSEER Akt1, ERK1, RSK2,<br />

p90RSK<br />

2.7 2.5 eIF4B 422 RERSRTGsESSQTGT TGS*ESSQTGTSTTSSR Akt1, p70S6K,<br />

p90RSK<br />

4.8 2.5 eIF4B 422,<br />

425<br />

RERSRTGsESSQTGT TGS*ESS*QTGTSTTSSR Akt1, p70S6K,<br />

p90RSK<br />

Table view presentation of data from PTMScan ® analysis of MKN-45 cells treated with SU11274 or staurosporine. Shown<br />

are representative data for the basophilic Akt substrate motif RXRXX(s/t) or RXX(s/t). Relative abundance changes of 2.5-fold or greater<br />

(treated versus control) for phosphorylated peptides are indicated by green (increase) or light red (reduction) highlighting. Incrementally<br />

darker highlighting in this image indicates abundance changes in the ranges of 2.5- to 24.9-fold and of greater than 25-fold, respectively.<br />

Analyze in context of known biology<br />

of treatment or system<br />

Map results using protein<br />

interaction database service<br />

to reveal novel pathways<br />

WB and/or<br />

ELISA validation<br />

Potential<br />

key nodes<br />

(


20<br />

Section III: Workflow Tools<br />

chapter<br />

Identification<br />

using Chromatin IP (ChIP) to identify protein-DNA interactions<br />

In a second example, the Phospho-Stat3 (Tyr705) (D3A7) XP ® Rabbit mAb #9145 and SimpleChIP ®<br />

Enzymatic Chromatin IP Kit (Magnetic Beads) #9003 are used to identify promoters with Stat3 binding<br />

sites. As shown in the figure, the active form of the Stat3 transcription factor—Phospho-Stat3<br />

(Tyr705)—binds the c-Fos and IRF-1 promoters, but not the negative control α satellite repeat element.<br />

20: IDENTIFICATION<br />

Acetylation of<br />

histone H3 at Lys27 is<br />

associated with active<br />

regions of the genome.<br />

% of total input chromatin<br />

35<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

GAPDH<br />

RPL30 AFM α Satellite<br />

SimpleChIP ® Enzymatic Chromatin IP<br />

Kit (Magnetic Beads) #9003: ChIPs were<br />

performed with cross-linked chromatin<br />

from 4 x 10 6 HeLa cells and either 5 μl<br />

of Acetyl-Histone H3 (Lys27) (D5E4) XP ®<br />

Rabbit mAb #8173 or 2 μl of Normal Rabbit<br />

IgG #2729 using #9003. The enriched<br />

DNA was quantified by real-time PCR using<br />

SimpleChIP ® Human GAPDH Exon 1 Primers<br />

#5516, SimpleChIP ® Human RPL30 Exon 3<br />

Primers #7014, SimpleChIP ® Human AFM<br />

Intron 1 Primers #5098, and SimpleChIP ®<br />

Human α Satellite Repeat Primers #4486.<br />

The amount of immunoprecipitated DNA in<br />

each sample is presented as a percent of<br />

the total input chromatin.<br />

Acetyl-Histone H3 (Lys27)<br />

(D5E4) XP ® Rabbit mAb #8173<br />

Normal Rabbit IgG #2729<br />

How can I analyze the complex interactions<br />

between chromatin and the proteins that bind to it<br />

Chromatin immunoprecipitation (ChIP) is a powerful tool for analyzing protein-DNA interactions within<br />

the natural chromatin context of the cell. The study of these interactions is critical for understanding<br />

the transcriptional regulators and epigenetic modifications central to the regulation of gene expression.<br />

The ChIP method identifies DNA-protein interactions<br />

using a procedure that can be summarized as follows:<br />

1. Chemical cross-linking of proteins to DNA in cells or tissues<br />

2. Tissue disruption and DNA fragmentation to the nucleosome level<br />

3. Immuno-enrichment of target protein (e.g. histone, transcription factor)<br />

and associated DNA sequences<br />

4. DNA purification and analysis<br />

Quantitative PCR (qPCR) Analysis<br />

Traditional ChIP, which analyzes the captured DNA by qPCR, is used to test interactions between known<br />

chromatin-associated proteins and known genetic loci, using an antibody specific to a protein of interest<br />

and PCR primers to specific genes or promoter regions. ChIP can also be used to study epigenetic<br />

changes using histone acetylation- or methylation-specific antibodies.<br />

Sequencing Analysis<br />

The ChIP method can be combined with sequencing (ChIP-seq) to identify novel regions of the genome<br />

associated with a particular transcription factor, chromatin regulator, or histone modification. Unlike traditional<br />

ChIP that requires a predetermined idea of the loci under investigation because primers to that<br />

region are necessary for subsequent DNA analysis by PCR, in ChIP-seq, all captured DNA fragments can<br />

be identified and analyzed directly by next generation sequencing. This powerful technique is extremely<br />

useful for large-scale epigenomic mapping and allows the researcher to take a genome-wide approach<br />

in the identification of loci associated with a particular transcription factor or histone modification.<br />

ChIP assays can be used to:<br />

• Identify multiple proteins associated with a specific region of the genome,<br />

or many regions of the genome associated with a particular protein<br />

• Determine the specific order of recruitment of various protein factors to a gene promoter<br />

• Identify active and inactive regions of the genome through<br />

associations with specific histone modifications<br />

• Analyze binding of transcription factors, transcription co-factors,<br />

DNA replication factors, and DNA repair proteins<br />

• Study epigenetic aberrations associated with disease<br />

For example, the SimpleChIP ® Enzymatic Chromatin IP Kit (Magnetic Beads) #9003 from Cell Signaling<br />

Technology was used to investigate genetic loci associated with acetylated-histone H3 at Lys27. As<br />

shown in the graph, the ChIP-validated Acetyl-Histone H3 (Lys27) (D5E4) XP ® Rabbit mAb isolates DNA<br />

fragments containing the active GAPDH and RPL30 genes, but not the inactive AFM gene or α satellite<br />

repeat element, illustrating that this histone modification associates with active regions of the genome.<br />

SimpleChIP ® Enzymatic Chromatin IP Kit (Magnetic Beads) #9003:<br />

ChIPs were performed with cross-linked chromatin from 4 x 10 6 Hep G2 cells<br />

starved overnight and treated with IL-6 (100 ng/ml) for 30 min, and either 10 μl<br />

of Phospho-Stat3 (Tyr705) (D3A7) XP ® Rabbit mAb #9145 or 2 μl of Normal<br />

Rabbit IgG #2729 using #9003. The enriched DNA was quantified by real-time<br />

PCR using human IRF-1 promoter primers, SimpleChIP ® Human c-Fos Promoter<br />

Primers #4663, and SimpleChIP ® Human α Satellite Repeat Primers #4486. The<br />

amount of immunoprecipitated DNA in each sample is presented as a percent of<br />

the total input chromatin.<br />

How SimpleChIP ® and SimpleChIP ®<br />

Plus Kits can benefit your research:<br />

• Higher Chromatin Quality: enzymatic digestion of chromatin is milder than sonication,<br />

better preserving chromatin integrity and improving detection of low-abundance targets<br />

(transcription factors, co-factors)<br />

% of total input chromatin<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

c-FOS<br />

IRF-1<br />

α Satellite<br />

• Appropriate Controls: reliably monitor ChIP efficiency in both active and inactive chromatin<br />

with included positive control Histone H3 antibody<br />

• Sample Flexibility: analyze chromatin from cultured cells or tissue samples using<br />

SimpleChIP ® Plus Kits<br />

• Rigorously Validated Antibodies: antibody specificity, sensitivity, and ChIP-optimized<br />

concentrations are key contributors to a successful ChIP experiment<br />

ChIP-Seq analysis of transcription factor TCF4 binding and<br />

Tri-Methyl Histone H3 (Lys4) enrichment at several gene loci.<br />

Number of sequencing reads<br />

Number of sequencing reads<br />

50<br />

25<br />

0<br />

80<br />

40<br />

0<br />

50<br />

25<br />

0<br />

80<br />

40<br />

0<br />

A<br />

C<br />

CCND1<br />

AXIN2<br />

ACTG1<br />

SimpleChIP ® Plus Enzymatic Chromatin IP Kit (Magnetic Beads) #9005: ChIPs were performed with cross-linked chromatin from 4 x 10 6<br />

HCT 116 cells and either 10 μl of Tri-Methyl-Histone H3 (Lys4) (C42D8) Rabbit mAb #9751 (upper) or 10 μl of TCF4 (C48H11) Rabbit mAb #2569<br />

(lower), using #9005. DNA sequencing libraries were generated using the NEBNext ® ChIP-Seq Library Prep Master Mix Set for Illumina (E6240)<br />

and 10 ng of DNA from each IP. The DNA libraries were sequenced using the Illumina MiSeq Sequencer and the obtained sequences were mapped<br />

to the UCSC Human Genome Assembly (hg19). The data was visualized using Integrative Genomics Viewer (IGV) and this figure shows enrichment<br />

of H3K4me3 and TCF4 binding at the CCND1 (A), c-Myc (B), AXIN2 (C) and ACTG1 (D) gene loci.<br />

B<br />

MYC<br />

D<br />

The Phospho-Stat3<br />

transcription factor<br />

binds to c-Fos and<br />

IRF-1 promoters.<br />

Phospho-Stat3 (Tyr705)<br />

(D3A7) XP ® Rabbit mAb #9145<br />

Normal Rabbit IgG #2729<br />

262 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/identification<br />

263


21<br />

Section III: Workflow Tools<br />

Profiling<br />

using antibody arrays<br />

How can you profile the signaling pathways and specific<br />

protein modifications at work in your experimental system<br />

An antibody array allows you to simultaneously profile and quantify multiple phosphorylation sites or<br />

other pathway readouts in a high throughput and economical way. Arrays must contain high quality<br />

antibody pairs, carefully selected for coverage of the most relevant targets, in order to provide relevant<br />

and accurate data.<br />

Antibody arrays can be used to:<br />

• Characterize a new cell line or disease model<br />

• Analyze possible effects of a new cellular stimulus<br />

• Investigate compensatory pathways used by tumors to evade targeted therapies<br />

• Identify off-target effects of small molecule or biologic therapeutics<br />

For example, in the simple experiment (seen right), a single variable—in this case, human IGF-I treatment—is<br />

monitored for effects on signaling nodes within the Akt pathway using the PathScan ® Akt<br />

Signaling Antibody Array from <strong>CST</strong>. The data show that treatment with a single dose of human IGF-I<br />

changes the levels of phosphorylation in Akt (Thr308 and Ser473), S6 Ribosomal Protein (Ser235/236),<br />

GSK-3α (Ser21), and several other proteins.<br />

However, experimental designs are not always simple; answering more refined follow-up questions<br />

may require you to test multiple biological stimulators, dosages, time points, or cell samples. Antibody<br />

array kits from <strong>CST</strong> come with 2 slides containing up to 16 pads each, allowing for comprehensive<br />

experimental design.<br />

How PathScan ® Antibody Arrays can benefit your research:<br />

• Multiplex Format: quickly analyze dozens of highly relevant signaling nodes<br />

across multiple pathways simultaneously<br />

• Multiple Array Pads: test up to 32 experimental variables in parallel<br />

• Calibrated Dynamic Range: permits detection of low-abundance targets<br />

without interference from high-abundance readouts<br />

• Rigorously Validated Antibody Pairs: generate accurate data and minimize background<br />

• Low Sample Volume: use as little as 50 μl of sample per pad<br />

Effect of human IGF-I on multiple downstream Akt signaling pathway nodes<br />

RFU (x10 4 )<br />

75<br />

60<br />

45<br />

30<br />

15<br />

0<br />

1<br />

1. Akt (Thr308)<br />

2. Akt (Ser473)<br />

3. S6 (Ser235/236)<br />

MCF7<br />

2 3 4 5 6 7 8 9 10 11 12 13 14 15 16<br />

4. AMPKa (Thr172)<br />

5. PRAS40 (Thr246)<br />

6. mTOR (Ser2481)<br />

Akt Antibody Array target map<br />

Targets<br />

Phosphorylation Site<br />

+ Positive Control N/A<br />

– Negative Control N/A<br />

1. Akt Thr308<br />

2. Akt Ser473<br />

3. S6 Ribosomal Protein Ser235/236<br />

4. AMPKa Thr172<br />

5. PRAS40 Thr246<br />

6. mTOR Ser2481<br />

7. GSK-3a Ser21<br />

8. GSK-3b Ser9<br />

9. p70 S6 Kinase Thr389<br />

10. p70 S6 Kinase Thr421/Ser424<br />

11. Bad Ser112<br />

12. RSK1 Ser380<br />

13. PTEN Ser380<br />

14. PDK1 Ser241<br />

15. Erk1/2 Thr202/Tyr204<br />

16. 4E-BP1 Thr37/46<br />

1<br />

3<br />

9<br />

15<br />

7. GSK-3a (Ser21)<br />

8. GSK-3b (Ser9)<br />

9. p70 S6K (Thr389)<br />

MCF7 + hIGF-I<br />

10. p70 S6K<br />

(Thr421/Ser424)<br />

11. Bad (Ser112)<br />

2<br />

7<br />

10<br />

12. RSK1 (Ser380)<br />

13. PTEN (Ser380)<br />

14. PDK1 (Ser241)<br />

+ 1 1 2 2 +<br />

3 3 4 4 5 5<br />

6 6 7 7 8 8<br />

9 9 10 10 11 11<br />

12 12 13 13 14 14<br />

+ 15 15 16 16 –<br />

15. Erk1/2<br />

(Thr202/Tyr204)<br />

16. 4E-BP1<br />

(Thr37/46)<br />

chapter 21: Profiling<br />

PathScan ® Akt Signaling<br />

Antibody Array Kit (Fluorescent<br />

Readout) #9700: MCF7 cells were<br />

grown to 85% confluency and then<br />

serum starved overnight. Cells were<br />

either untreated or treated with<br />

Human Insulin-like Growth Factor I<br />

(hIGF-I) #8917 (100 ng/ml, 20 min).<br />

Cell extracts were prepared and<br />

analyzed using #9700.<br />

MCF7<br />

MCF7 + hIGF-I<br />

Images were acquired using the<br />

LI-COR ® Biosciences Odyssey ®<br />

imaging system. This graph displays<br />

pixel intensity quantified using the<br />

LI-COR ® Image Studio v2.0 array<br />

analysis software.<br />

Using PathScan ® Akt<br />

Signaling Antibody Array Kit<br />

(Fluorescent Readout) #9700.<br />

Antibody arrays are<br />

based upon the sandwich<br />

immunoassay principle.<br />

PathScan ® Antibody Array Kits Chemiluminescent Fluorescent<br />

Akt Signaling #9474 #9700<br />

EGFR Signaling #12622 #12785<br />

Immune Cell Signaling #13792 #13788<br />

Intracellular Signaling #7323 #7744<br />

RTK Signaling #7982 #7949<br />

Stress and Apoptosis Signaling #12856 #12923<br />

Th1/Th2/Th17 Cytokine #13047 #13124<br />

<strong>CST</strong> Technical Support<br />

When you contact <strong>CST</strong> for technical support, you can be confident you will be working with a colleague you can trust,<br />

striving to provide the highest quality products and services to you, our customer. Please see our Technical Support<br />

resource page online for contact information. www.cellsignal.com/cstsupport<br />

264 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/profiling<br />

265


22<br />

Section III: Workflow Tools<br />

Modulation<br />

using chemical activators and inhibitors,<br />

cytokines, and growth factors<br />

Inhibition of Signaling Using Chemical Modulators<br />

gefitinib<br />

Wortmannin<br />

LY294002<br />

GF Growth Factors, Insulin, etc.<br />

membrane<br />

chapter 22: Modulation<br />

How can I activate or inhibit specific signaling nodes<br />

within a cell to study its biology or role in disease<br />

Chemical modulators, cytokines, and growth factors are important tools for manipulating the activity of<br />

key signaling nodes or pathways and are often used for dissecting the signaling pathways that regulate<br />

basic cellular functions or underlie disease. Small molecule chemical inhibitors are useful for achieving<br />

pathway inhibition, and the choice of inhibitor depends on the objective of the study. Some chemical<br />

inhibitors act at the receptor level, blocking all signaling emanating from a single ligand receptor complex<br />

and thus inhibiting multiple pathways at once. For example, the tyrosine kinase inhibitor gefitinib<br />

inhibits EGFR and prevents signaling through the MAPK, Akt, and many other pathways activated by<br />

the receptor. Other chemical inhibitors target a single pathway by inhibiting a key node within the cascade<br />

such as inhibition of the MAPK pathway using the MEK1 inhibitor PD184352 or U0126. Chemical<br />

inhibitors are used in a broad range of applications; however, recent research has largely focused on<br />

understanding their roles in the complex signaling networks of cancer and other disease states such as<br />

diabetes. Many of these agents are now in clinical development as candidate therapeutics.<br />

The ability to activate a pathway can be equally important. This can be achieved using chemical activators<br />

such as AICAR, which activates the AMPK pathway, or by treating cells with various growth factors<br />

or cytokines that are signaling receptor ligands.<br />

mLST8<br />

SIN1<br />

mTOR<br />

FKBP12<br />

GβL<br />

Rictor<br />

PRAS40<br />

Thr246 P<br />

Rapamycin<br />

P<br />

Tyr373 Tyr376<br />

Ser241 P P P PIP3<br />

PDK1<br />

PIP 3<br />

P Akt/PKB<br />

Thr308<br />

P<br />

Ser473<br />

Ser2481<br />

P<br />

GβL<br />

Raptor<br />

mTOR<br />

PTEN<br />

Ser939 P<br />

Rheb<br />

TSC1<br />

p85<br />

GF-R<br />

PI3K Grb<br />

IRS<br />

P<br />

TSC2<br />

P<br />

P P<br />

Ser1254<br />

Thr1462<br />

Tyr1571<br />

Sos Ras<br />

Ser2448<br />

P<br />

Ser371<br />

Thr229<br />

P P<br />

S6K P Thr421<br />

Thr389 P P Ser424<br />

mLST8<br />

Ser259<br />

Ser236<br />

Ser235<br />

P P P Ser240<br />

P Ser244<br />

S6<br />

cytoplasm<br />

P Ser259<br />

Raf P Ser338<br />

Ser217<br />

P<br />

MEK1/2<br />

P<br />

Ser221<br />

Thr202 P P Tyr204<br />

MAPK/ERK1/2<br />

PD98059<br />

(MEK1)<br />

U0126<br />

Cytokines and<br />

Growth Factors<br />

Cytokines and Growth Factors from<br />

<strong>CST</strong> are produced and bio-assayed<br />

in-house, ensuring the highest<br />

standards of quality and consistency.<br />

www.cellsignal.com/ck&gf<br />

Chemical modulators, cytokines,<br />

and growth factors can be used to:<br />

• Study the role of specific signaling nodes in disease<br />

• Inhibit a signaling pathway to investigate signaling networks or study activation<br />

of alternative pathways (e.g. cancer resistance mechanisms)<br />

• Activate or inhibit an individual signaling node to identify kinase substrates or protein interactions<br />

• Study immune response or cellular differentiation associated with a particular cytokine<br />

For example, the chemical modulator crizotinib was used to inhibit activity of the receptor tyrosine<br />

kinase ALK in KARPAS-299 cells. As shown in the western blot below, phosphorylation of ALK at<br />

Tyr1604, which is a readout for ALK activity, was completely inhibited by treating cells with 1000 nM<br />

crizotinib. Lower doses were less effective, with 100 nM crizotinib resulting in partial inhibition of ALK<br />

activity, while phospho-ALK levels in the 10 nM and 1 nM crizotinib-treated samples were similar to<br />

the untreated control.<br />

The receptor tyrosine kinase<br />

ALK is inhibited by crizotinib.<br />

Crizotinib #4401: WB analysis of extracts from KARPAS-299 cells, untreated<br />

or treated with #4401 (1 hr) at the indicated concentrations, using Phospho-ALK<br />

(Tyr1604) Antibody #3341 (upper) or ALK (D5F3 ® ) XP ® Rabbit mAb #3633 (lower).<br />

kDa<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

0<br />

1000<br />

100<br />

10<br />

1<br />

Phospho-ALK<br />

(Tyr1604)<br />

ALK (D5F3)<br />

Crizotinib (nM)<br />

Commonly Used Chemical Modulators<br />

Gene Expression, Epigenetics, Nuclear Function<br />

#2112 Cycloheximide Blocks protein biosynthesis by inhibiting protein elongation<br />

#5927 Doxorubicin Inhibits DNA and RNA synthesis by intercalating the DNA helix; inhibits topoisomerase I<br />

#2200 Etoposide Inhibits topoisomerase II resulting in DNA breakage; induces apoptosis<br />

#9676 Leptomycin B Inhibits nuclear export activity of XPO1/exportin 1<br />

#9950 Trichostatin A (TSA) Inhibits class I and class II histone deactylases<br />

Cell Growth and Death<br />

Cell Growth and Translation<br />

RTKs<br />

#4401 Crizotinib Inhibitor of ALK and ROS1 (ATP-competitive inhibitor)<br />

#5083 Erlotinib Inhibits EGFR (ATP-competitive inhibitor)<br />

#4765 Gefitinib Inhibits EGFR (active site inhibitor)<br />

#9084 Imatinib Inhibits Bcr-Abl, PDGFR, c-Kit (active site inhibitor)<br />

#12121 Lapatinib Inhibits EGFR and HER2 (ATP-competitive inhibitor)<br />

#8705 Sorafenib Inhibits VEGFR and PDGFR; inhibits Raf kinases; induces autophagy<br />

#12328 Sunitinib Broad RTK inhibitor (PDGFR, VEGFR, c-KIT, RET, CSF-1R, FLT-3/CD135)<br />

#9842 Tyrphostin AG 1478 Inhibits EGFR<br />

#12998 Vatalanib Broad RTK inhibitor (VEGFR, PDGFR-B, c-KIT)<br />

MAP Kinase<br />

#2222 Anisomycin Inhibits protein synthesis; activates stress-activated kinases (SAPK/JNK, p38-MAPK)<br />

#11916 Chelerythrine Chloride Cell-permeable inhibitor of PKC; Activates SAPK/JNK and p38-MAPK; induces apoptosis in<br />

some cell lines<br />

#12147 PD184352 Highly selective, noncompetitive inhibitor of MEK1 and MEK2<br />

#9900 PD98059 Highly selective inhibitor of MEK1 and MEK2; binds inactive forms and prevent activation by<br />

upstream kinases<br />

#9903 U0126 Highly selective inhibitor of MEK1 and MEK2<br />

#8158 SB202190 Cell-permeable, highly specific inhibitor of p38-MAPK (ATP-competitive inhibitor)<br />

#5633 SB203580 Cell-permeable inhibitor of p38-MAPK (inhibits PDK1 at higher concentrations)<br />

#8705 Sorafenib Inhibits VEGFR and PDGFR; inhibits Raf kinases; induces autophagy<br />

#8177 SP600125 Cell-permeable, highly specific inhibitor of JNK-family kinases<br />

PD184352 inhibits<br />

MEK1/2 and blocks<br />

signaling through the<br />

MAPK pathway.<br />

kDa<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

– + +<br />

–<br />

–<br />

2<br />

+ +<br />

0.2<br />

0.02<br />

Phospho-p44/42<br />

MAPK (Erk1/2)<br />

(Thr202/Tyr204)<br />

p44/42 MAPK<br />

(Erk1/2)<br />

TPA<br />

PD184352 (µM)<br />

PD184352 #12147: WB analysis of<br />

extracts from HeLa cells, serum-starved<br />

overnight and untreated or treated with<br />

TPA #4174 (200 nM, 20 min) either with<br />

or without #12147 pretreatment (1 hr)<br />

at the indicated concentrations, using<br />

Phospho-p44/42 MAPK (Erk1/2) (Thr202/<br />

Tyr204) (D13.14.4E) XP ® Rabbit mAb<br />

#4370 (upper) or p44/42 MAPK (Erk1/2)<br />

(137F5) Rabbit mAb #4695 (lower).<br />

266 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/modulation<br />

267


Section III: Workflow Tools<br />

PI3 Kinase/Akt Pathway<br />

#12017 Everolimus Specific inhibitor of mTORC1 protein complex<br />

#9901 LY294002 Reversible but potent inhibitor of PI3K family members<br />

#9904 Rapamycin Specific inhibitor of mTORC1 protein complex<br />

#9951 Wortmannin Irreversible, potent inhibitor of PI3K faimly members<br />

Other Kinases and Phosphatases<br />

#9902 Calyculin A Cell permeable, selective inhibitor of PP1 and PP2A<br />

#9052 Dasatinib Tyrosine kinase inhibitor (Abl, Bcr/Abl, Src, c-KIT, Ephrin receptors)<br />

#9084 Imatinib Tyrosine kinase inhibitor (Abl, Bcr/Abl, c-KIT, PDGFR)<br />

#12209 Nilotinib Tyrosine kinase inhibitor (Abl, Bcr/Abl, c-KIT, LCK, Ephrin receptors, DDR1/2, PDGFR-B)<br />

#5934 Okadaic Acid Cell permeable, selective inhibitor of PP1, PP2A, and PP2B<br />

#9493 PKC412 Very broad spectrum protein kinase inhibitor<br />

(conventional PKCs, some RTKs, Syk, Cdk1/B, c-Src, etc)<br />

#9885 Roscovitine Potent and selective inhibitor of CDK1, 2, and 5 (ATP-competitive)<br />

Cellular Metabolism<br />

#9944 AICAR AMP analog that activates AMPK<br />

#9996 Oligomycin ATP synthase inhibitor; inhibits oxidative phosphorylation<br />

Calcium, cAMP, and Lipid Signaling<br />

#9841 Bisindolylmaleimide I, Potent inhibitor of PKC family members<br />

Hydrochloride<br />

#9973 Cyclosporin A Inhibits calcineurin<br />

#9974 FK-506 Inhibits calcineurin<br />

#3828 Forskolin Potent activator of adenylate cyclase, used to increase levels of cAMP<br />

#12060 Gö6976 Potent inhibitor of calcium-dependent PKC family members<br />

#9844 H-89, Dihydrochloride Broad spectrum Ser/Thr kinase inhibitor used to inhibit PKA; also inhibits the activity of<br />

p70S6K, MSK, and ROCKII and others<br />

#9995 Ionomycin, Calcium Salt Calcium ionophore; activates calcium-/calmodulin-dependent<br />

kinase and calcium-dependent PKCs<br />

#9953 Staurosporine Very broad, ATP-competitive protein kinase inhbitor (PKC, PKA, Src, CaM kinase, etc.)<br />

#4174 TPA (12-O-Tetradecanoylphorbol-13-Acetate)<br />

Cell permeable, potent activator of PKC; also used to activate MAPK<br />

Cell Biology<br />

#8132 17-AAG Inhibits HSP90 chaperone activity resulting in degradation of HSP90 client proteins<br />

#2204 Bortezomib Proteasome inhibitor<br />

#9972 Brefeldin A Inhibits ER to Golgi protein transport; triggers UPR and apoptosis<br />

#9886 Docetaxel Inhibits microtubule depolymerization; prevents cell division<br />

#9843 Geldanamycin Inhibits HSP90 chaperone activity resulting in degradation of HSP90 client proteins<br />

#2194 MG-132 Proteasome inhibitor<br />

#2190 Nocodazole Inhibits microtubule polymerization; induces cell cycle arrest in G2/M-phase<br />

#9807 Paclitaxel Inhibits microtubule depolymerization; prevents cell division<br />

#12758 Thapsigargin Inhibits ER calcium-ATPases; induces ER stress<br />

#12819 Tunicamycin Inhibits glycoprotein synthesis and causes G1 cell cycle arrest<br />

Growth factor activation of signaling<br />

with IGF-I followed by chemical inhibition<br />

of mTOR using everolimus<br />

hIGF-I #8917 and Everolimus #12017: WB analysis of extracts from HeLa<br />

cells, serum-starved overnight and treated with #8917 (100 ng/ml, 10 min) either<br />

with or without #12017 pretreatment (1 hr) at the indicated concentrations, using<br />

Phospho-S6 Ribosomal Protein (Ser235/236) (D57.2.2E) XP ® Rabbit mAb #4858<br />

(upper) or S6 Ribosomal Protein (5G10) Rabbit mAb #2217 (lower).<br />

268 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

kDa<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

– +<br />

– –<br />

+ +<br />

1000<br />

100<br />

Phospho-S6<br />

Ribosomal<br />

Protein<br />

(Ser235/236)<br />

S6 Ribosomal<br />

Protein<br />

+ + hIGF-I<br />

Everolimus (nM)<br />

10<br />

1<br />

23<br />

Localization & Classification<br />

using conjugated primary antibodies<br />

How can I visualize protein location or simultaneously<br />

analyze intracellular signaling events within cell populations<br />

Antibody conjugation is the process by which a fluorescent dye, small molecule, enzyme, or solid<br />

matrix label is attached covalently to an antibody. Fluorescent dye conjugates are routinely used<br />

for detection and quantification of cell surface and intracellular proteins within cell populations by<br />

flow cytometry (FC) and visualization of protein location by immunofluorescence (IF). Other antibody<br />

conjugates, such as those labeled with small molecules, enzymes, or beads are used for detection or<br />

analysis of proteins within cell lysates by western blot (WB), ELISA, or immunoprecipitation (IP). For all<br />

labels, the key benefit of conjugated primary antibodies is the ability to directly detect target proteins,<br />

thereby eliminating the need for a secondary antibody.<br />

Conjugated primary antibodies can be used to:<br />

• Save time by eliminating the secondary antibody incubation and wash steps<br />

• Eliminate potential background attributable to secondary antibody<br />

• Multiplex using primary antibodies from the same host species<br />

• Perform high throughput screening with one-step staining protocol<br />

Conjugation Types<br />

Labels Examples Applications<br />

Fluorescent dyes Alexa Fluor ® , Pacific Blue , PE, FITC, APC Flow, IF<br />

Small molecule Biotin WB, ELISA<br />

Enzyme HRP, alkaline phosphatase WB, ELISA, dot blot<br />

Solid matrix Sepharose ® or magnetic beads IP, cell sorting<br />

Conjugation Chemistry<br />

The conjugation process commonly occurs by one of two methods. First, a label can be covalently<br />

attached to an antibody at primary amine groups on exposed lysine residues within the immunoglobulin<br />

protein. Although this conjugation method is highly effective, the exact location and density of label can<br />

change from antibody to antibody and potentially for each labeling reaction due to the high number of<br />

lysine residues within the average antibody molecule. In addition, conjugation of lysine residues in the<br />

variable region can disrupt antibody-antigen binding. A second method of conjugation occurs at sulfhydryl<br />

groups from cysteine residues forming the disulfide bonds that stabilize antibody structure. An<br />

initial chemical reduction step disrupts the bonds and makes these groups available for conjugation.<br />

Researchers typically target the disulfide bonds within the hinge region for this type of conjugation,<br />

as the single location within the antibody molecule lends itself to consistent and uniform labeling. The<br />

optimal conjugation chemistry is unique for each antibody and must be determined experimentally.<br />

The Importance of Optimization<br />

Conjugated antibodies must be properly validated in order to generate maximum signal:noise ratio. In<br />

addition to determining optimal label chemistry, it is also important to consider optimal label density.<br />

Each conjugation reaction can produce variations in degree of labeling (DOL)—that is, the number of<br />

label molecules attached to a single antibody. Insufficient DOL will result in a weak signal, whereas<br />

a higher DOL may create an overall brighter signal with higher background, resulting in lower signal<br />

to noise. As shown in the graph, antibodies with the highest DOL do not necessarily perform better<br />

than those antibodies with a lower DOL. Optimized DOL is critical when working with low abundance<br />

proteins or low affinity antibodies in order to avoid false negative data.<br />

www.cellsignal.com/localization 269


Section III: Workflow Tools<br />

chapter 23: Localization & classification<br />

Optimal antibody-to-dye ratio is critical for maximum performance.<br />

Custom conjugations are optimized by degree of<br />

labeling testing to identify the optimal antibody:<br />

dye molecule ratio, resulting in conjugates with<br />

maximum performance.<br />

DOL 2.34<br />

DOL 4.28<br />

DOL 5.51<br />

DOL 7.27<br />

How <strong>CST</strong> conjugated antibodies can benefit your research:<br />

• Rigorously Validated: optimal conjugation chemistry and DOL are determined for each antibody<br />

in their intended application, producing conjugates with maximal performance and eliminating<br />

additional optimization steps<br />

• Highly Reproducible Results: extensive testing and rigorous validation protocols minimize<br />

lot-to-lot variation<br />

• Assay Flexibility: custom conjugation is available for Alexa Fluor ® 488, 555, 594, or 647 dyes;<br />

PE; Pacific Blue dye; Sepharose ® or magnetic beads; biotin; and HRP labels<br />

Fluorescent Dye Antibody Conjugates<br />

Fluorescent signal is produced when light energy from a laser or metal halide lamp at a specific<br />

wavelength is absorbed by a fluorochrome (excitation) and then released (emission), producing light at<br />

distinct wavelengths that can be measured by fluorescence detectors.<br />

Fluorescent dyes can be used in IF to produce images of cell or tissue structures, and are often used<br />

to study changes in protein localization and/or expression in response to a cell stimulus. Fluorescent<br />

dye conjugates can be used in FC to analyze single cell signaling events, and are particularly useful<br />

for multiplex assays because multiple primary antibodies conjugated to different fluorochromes can be<br />

used to identify protein targets within a single sample regardless of host species, greatly increasing<br />

assay flexibility.<br />

Fluorescent dye conjugated antibody used<br />

to visualize α-Synuclein expression<br />

α-Synuclein (D37A6) XP ® Rabbit mAb (Alexa Fluor ® 488 Conjugate) #5496:<br />

Confocal IF analysis of normal rat cerebellum using #5496 (green). Blue pseudocolor<br />

= DRAQ5 ® #4084 (fluorescent DNA dye).<br />

S/N Ratio<br />

45<br />

40<br />

35<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

0 1 2 3 4 5<br />

Concentration (µg/ml)<br />

Multiplex analysis: decreased expression of Sox2 and<br />

Oct-4 pluripotency markers by day 5 in RA-induced NTERA-2 cells<br />

Sox2<br />

A B C<br />

Oct-4A<br />

Fluorescent dye conjugated antibody used to detect<br />

etoposide-induced apoptotic cell populations<br />

Cleaved Caspase-3 (Asp175) (D3E9) Rabbit mAb (Pacific Blue Conjugate)<br />

#8788: Flow cytometric analysis of Jurkat cells, untreated (blue) or etoposide-treated<br />

(green), using #8788.<br />

Events<br />

Cleaved Caspase-3 (Asp175)<br />

(Pacific Blue Conjugate)<br />

Small Molecule, Enzyme, and Bead Conjugates<br />

Conjugation of an antibody to the horseradish peroxidase (HRP) enzyme is a common strategy for<br />

detecting proteins in cell lysates by WB or ELISA. HRP catalyzes the oxidation of luminescent or chromogenic<br />

substrates into detectable light or colored products. Small molecule conjugates such as biotin<br />

take advantage of the natural affinity between biotin and streptavidin and can be detected directly by<br />

streptavidin-HRP. Conjugating an antibody to a Sepharose ® or magnetic bead allows for the physical<br />

separation of the detected protein from a whole cell lysate by centrifugation or magnetic force. Bead<br />

conjugates can be used to enrich low abundance proteins or to study protein-protein interactions using<br />

an antibody to a target of interest and analyzing proteins that co-precipitate.<br />

HRP enzyme labeled antibody<br />

used for direct WB detection<br />

kDa<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

1 2 3 4<br />

Cytochrome c (D18C7) Rabbit<br />

mAb (HRP Conjugate) #12959:<br />

WB analysis of extracts from various<br />

cell lines using #12959.<br />

Lanes<br />

1. HeLa 3. C2C12<br />

2. NIH/3T3 4. C6<br />

Oct-4A (C30A3) Rabbit mAb<br />

(Alexa Fluor ® 488 Conjugate)<br />

#5177: Flow cytometric analysis of<br />

NTERA-2 cells, treated with 10 μM<br />

retinoic acid (RA) for 5 days to induce<br />

neuronal differentiation, using #5177<br />

and Sox2 (D6D9) XP ® Rabbit mAb<br />

(Alexa Fluor ® 647 Conjugate) #5067.<br />

At 0 days (A), 3 days (B), and 5 days<br />

(C) of treatment, cells were harvested,<br />

fixed, and permeabilized according to<br />

the <strong>CST</strong> standard flow protocol and<br />

analyzed on a 4-laser Gallios Flow<br />

Cytometer (Beckman Coulter).<br />

10<br />

Cytochrome c<br />

Bead conjugates are used for protein enrichment by IP.<br />

Fluorescent dye<br />

conjugated antibody<br />

used to detect<br />

nuclear induction<br />

of phospho-p53.<br />

Phospho-p53 (Ser15) (16G8) Mouse<br />

mAb (Alexa Fluor ® 647 Conjugate)<br />

#8695: Confocal IF analysis of HT-29<br />

cells, untreated (left) or UV-treated<br />

(right), using #8695 (blue pseudocolor).<br />

Actin filaments were labeled with<br />

DyLight 554 Phalloidin #13054 (red).<br />

Phospho-Stat3 (Tyr705) (D3A7) XP ® Rabbit mAb (Sepharose ® Bead Conjugate)<br />

#4074: IP of HeLa cell lysates, untreated or treated with Human Interferon-α1 (hIFN-α1)<br />

#8927, using XP ® Rabbit (DA1E) IgG (Sepharose ® Bead Conjugate) #3423 and #4074.<br />

The WB was probed using Phospho-Stat3 (Tyr705) (3E2) Mouse mAb #9138.<br />

Lanes<br />

1. Rabbit (DA1E) mAb IgG XP ® Isotype Control<br />

(Sepharose ® Bead Conjugate) #3423<br />

2. Phospho-Stat3 (Tyr705) (D3A7) XP ® Rabbit mAb<br />

(Sepharose ® Bead Conjugate) #4074<br />

kDa<br />

200<br />

140<br />

100<br />

80<br />

60<br />

50<br />

40<br />

30<br />

1<br />

2<br />

– + – +<br />

Phospho-<br />

Stat3<br />

(Tyr705)<br />

hIFN-α1<br />

270 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/localization<br />

271


24<br />

Section III: Workflow Tools<br />

Screening & Quantification<br />

using ELISA to screen and quantify<br />

cellular responses at the target level<br />

How can I quantify levels of total and post-translationally<br />

modified proteins in response to cell stimuli<br />

The sandwich enzyme-linked immunosorbent assay (ELISA) is a quantitative, microplate-based assay<br />

that detects changes in abundance of a target protein within a cell sample using a capture antibody<br />

coated to the plate and a detection antibody to generate signal. Unlike a western blot that achieves<br />

specificity by using a single antibody combined with separation of protein by size, an ELISA is highly<br />

specific because it uses two antibodies recognizing different epitopes of the same protein. ELISAs are<br />

broadly utilized in both research and clinical settings; here, we focus on their application in research for<br />

the study of intracellular signaling using phospho-specific and total protein antibodies. ELISAs can also<br />

be used to quantify epigenetic changes using histone acetylation- or methylation-specific antibodies.<br />

The quantitative nature, high sensitivity, plate-based format, and ease-of-use make ELISAs a commonly<br />

used tool for screening in drug discovery and development.<br />

Sandwich ELISAs can be used to:<br />

• Screen therapeutic compounds for effects on post-translational modification of target proteins<br />

• Measure relative changes in total or post-translationally modified protein levels in response<br />

to an external stimuli such as a therapeutic compound<br />

• Assess effects of a cell stimulus at multiple time points or dosages<br />

• Quantitatively measure activation of signaling proteins in a high throughput format<br />

• Confirm data generated by western blot or other antibody-based applications<br />

For example, the PathScan ® Phospho-Bcl-2 (Ser70) Sandwich ELISA Kit from Cell Signaling Technology<br />

(<strong>CST</strong>) was used to assess phosphorylation of Bcl-2 at Ser70 in Jurkat cells treated with a single dose<br />

of paclitaxel. As shown in the figure, paclitaxel treatment increases phosphorylation of Bcl-2 at Ser70<br />

more than 2-fold relative to untreated control without affecting the level of total Bcl-2 protein. The<br />

changes quantified by ELISA were independently observed by western blot.<br />

Assessing the effects of growth factor or cytokine stimulation using ELISA<br />

PathScan ® Phospho-Akt (Thr308) Chemiluminescent<br />

Sandwich ELISA Kit #7135: Relationship between protein<br />

concentration of lysates from untreated and PDGF-treated<br />

NIH/3T3 cells and immediate light generation with chemiluminescent<br />

substrate is shown. Cells (80% confluence)<br />

were treated with PDGF #9909 (50 ng/ml) and lysed after<br />

incubation at 37ºC for 5 min. Graph inset corresponding to<br />

the shaded area shows high sensitivity and a linear response<br />

at the low protein concentration range.<br />

PDGF-treated<br />

Untreated<br />

RLU (x10 5 )<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

0<br />

0.0<br />

PathScan ® Phospho-Stat3 (Tyr705) Sandwich ELISA Kit #7300: Treatment<br />

of HeLa cells with IFN-α stimulates phosphorylation of Stat3 at Tyr705, detected<br />

by #7300, but does not affect the level of total Stat3 protein, detected by a Stat3<br />

antibody via western analysis. OD 450 readings are shown in the top figure, while<br />

the corresponding western blot using Phospho-Stat3 (Tyr705) (3E2) Monoclonal<br />

Antibody #9138 or Stat3 antibody, is shown in the figure to the right.<br />

Untreated<br />

IFN-α-treated<br />

70<br />

0.1<br />

0.2 0.3 0.4 0.5 0.6<br />

Protein conc. of lysate (mg/ml)<br />

Absorbance 450nm<br />

4<br />

3<br />

2<br />

1<br />

0<br />

14<br />

10<br />

6<br />

2<br />

Phospho-Stat3<br />

(Tyr705)<br />

chapter 24: screening & Quantification<br />

0.02 0.06 0.1<br />

0.7 0.8<br />

Phospho-<br />

Stat3<br />

(Tyr705)<br />

Stat3<br />

– + IFN-α<br />

How PathScan ® Sandwich ELISA Kits can benefit your research:<br />

• Highly Sensitive: detect low abundance target proteins and generate measurable linear<br />

responses at low lysate concentrations<br />

• Rigorously Validated Antibody Pairs: generate accurate data and minimize background,<br />

ensuring reliable results<br />

• Multiple Detection Options: colorimetric or chemiluminescent detection options are available<br />

• Specialized Packaging: bulk orders and custom 384-plate size are available<br />

Paclitaxel treatment of Jurkat cells results<br />

in phosphorylation of Bcl-2 at Ser70.<br />

High sensitivity ELISAs produce a broad dose response<br />

and are linear in low concentration ranges.<br />

PathScan ® Phospho-Bcl-2 (Ser70) Sandwich ELISA Kit #11874:<br />

Treatment of Jurkat cells with Paclitaxel stimulates phosphorylation of Bcl-2<br />

at Ser70, as detected by #11874, but does not affect the levels of total Bcl-2<br />

detected by PathScan ® Total Bcl-2 Sandwich ELISA Kit #12030. Jurkat cells<br />

were untreated or treated with λ phosphatase or Paclitaxel #9807 (1 mM,<br />

20 hr, 37°C). The absorbance readings at 450 nm are shown in the top<br />

figure, while the corresponding western blots using Phospho-Bcl-2 (Ser70)<br />

(5H2) Rabbit mAb #2827 (left panel) or Bcl-2 (D55G8) Rabbit mAb (Human<br />

Specific) #4223 (right panel) are shown in the bottom figure.<br />

λ phosphatase-treated<br />

Untreated<br />

Paclitaxel-treated<br />

Absorbance 450nm<br />

4<br />

3<br />

2<br />

1<br />

0<br />

Phospho-Bcl-2<br />

(Ser70)<br />

Total Bcl-2<br />

PathScan ® Phospho-EGF Receptor (Tyr1068) Sandwich ELISA<br />

Kit #7240: The relationship between protein concentration of lysates<br />

from untreated and EGF-treated A-431 cells and kit assay optical<br />

density readings. After starvation, A-431 cells (85% confluence) were<br />

treated with EGF #8916 (100 ng/ml, 5 min, 37°C) and then lysed.<br />

EGF-treated<br />

Control<br />

2.5<br />

1.5<br />

1.0<br />

0.5<br />

Absorbance 450nm 2.0<br />

0<br />

0.05 0.10 0.15 0.20 0.25<br />

Protein conc. of lysate (mg/ml)<br />

0.30<br />

– – +<br />

+ – –<br />

– – +<br />

+ – –<br />

Paclitaxel<br />

λ Phosphatase<br />

In a second example, the PathScan ® Phospho-Akt (Thr308) Chemiluminescent Sandwich ELISA Kit<br />

#7135 was used to examine the effects growth factor stimulation on phosphorylation of Akt at Thr308,<br />

an indicator of Akt activation and signaling. As shown in the figure, PDGF-mediated activation of Akt<br />

was quantified along a range of lysate levels, demonstrating the kit’s high sensitivity and a linear<br />

response at low protein concentrations. A similar assessment of the effects of cytokine stimulation on<br />

phosphorylation of Stat3 at Tyr705 was performed using #7300.<br />

272 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

<strong>CST</strong> Technical Support<br />

When you contact <strong>CST</strong> for technical support, you can be confident you will be working with a colleague you can trust,<br />

striving to provide the highest quality products and services to you, our customer. Please see our Technical Support<br />

resource page online for contact information. www.cellsignal.com/cstsupport<br />

www.cellsignal.com/screening<br />

273


25<br />

Cell<br />

Section III: Workflow Tools<br />

Monitoring<br />

using cell assays to monitor the health of<br />

cells and their response to multiple challenges<br />

How can you monitor cell state or cellular response<br />

to a specific treatment within a population of cells<br />

Cell assays are a diverse set of research tools used to assess cell functions such as growth, apoptosis,<br />

or signaling at the cellular level. Cell-based assays are commonly used in a broad array of biological<br />

disciplines. In general, these assays are highly sensitive and easy to perform. Many cell-based assays are<br />

conducted in 96-well plate format, allowing the researcher to process a large number of samples quickly.<br />

Cell assays can be used to:<br />

• Perform large-scale screening of therapeutic compounds or other stimuli<br />

• Evaluate cell response to multiple drug dosages at various time points<br />

• Complement data obtained by antibody-based applications<br />

• Determine if treatment data obtained through other experiments correlates<br />

to cell proliferation, senescence, or death<br />

Proliferation and Viability Assays<br />

Cell proliferation and viability assays allow a researcher to ask basic questions about cell status or<br />

responses to physiological stimuli or challenges without having to focus on the underlying molecular<br />

mechanism or individual protein target. These assays have broad applications in both basic research and<br />

drug discovery. Measuring cell proliferation and viability is often the first step in examining the effects<br />

of an unknown compound on cell health. These assays can also be used to investigate the effects of<br />

known growth factors or cytotoxic agents in combination with a test compound, or on cells containing<br />

genetic mutations or knockdowns within key signaling nodes.<br />

Absorbance 450nm<br />

• BrdU Assays measure cell proliferation by treating cells with a pyrimidine analog, BrdU, which<br />

incorporates into the DNA of replicating cells and can be measured using a BrdU antibody.<br />

• XTT and Resazurin Assays measure cell viability by colorimetric or fluorescent detection of<br />

reduced tetrazolium salt, an indicator of living cells.<br />

Doxorubicin-treated cells are viable but do not proliferate.<br />

4.0<br />

3.0<br />

2.0<br />

1.0<br />

XTT<br />

BrdU<br />

% Inhibition<br />

10<br />

0<br />

-3 -2 -1 0 1 2 -10 -2 -1 0 1 2<br />

110<br />

90<br />

70<br />

50<br />

30<br />

Log (Doxorubicin, µM)<br />

XTT Cell Viability Kit #9095 and BrdU Cell Proliferation Assay Kit #6813: C2C12 cells were seeded at 2x10 4 cells/well in a 96-well<br />

plate and incubated overnight. Cells were then treated with various concentrations of doxorubicin overnight. The cytotoxicity was measured<br />

using #9095 (red) and #6813 (blue) as shown in the left panel. The percentage inhibition in each assay was calculated and plotted in the<br />

right panel. Doxorubicin treatment can lead to cell DNA damage followed by cell cycle arrest.<br />

Death Assays<br />

Identifying agents that induce or prevent cell death is critical for studies in cancer, cardiovascular<br />

research, and other pathologies.<br />

• The Annexin V-FITC Early Apoptosis Detection Assay differentiates between early and<br />

late apoptosis using flow cytometry; annexin V binding to phosphatidylserine indicates early<br />

apoptosis, whereas late apoptotic cells will have lost plasma membrane integrity, allowing<br />

propidium iodide to passively enter the cell.<br />

• The Caspase-3 Activity Assay monitors caspase-3 activity as a readout for apoptosis by<br />

measuring cleavage of a fluorescent caspase-3 substrate.<br />

Camptothecin-treated<br />

cells undergo apoptosis.<br />

Annexin V-FITC Early Apoptosis<br />

Detection Kit #6592: Flow cytometric<br />

analysis of Jurkat cells untreated (left) or<br />

treated with camptothecin (10µM, 4 hr;<br />

right) using #6592.<br />

Propidium Iodide<br />

Late Apoptotic/<br />

Necrotic Cells<br />

Live Cells<br />

Untreated<br />

Early Apoptotic Cells<br />

Annexin V-FITC<br />

Late Apoptotic/<br />

Necrotic Cells<br />

Live Cells<br />

Camptothecin-treated<br />

Early Apoptotic Cells<br />

Second Messenger Assays<br />

Second messenger assays allow a researcher to evaluate signaling mechanisms at work in a cell<br />

population by assessing levels of cyclic adenosine 3’, 5’ monophosphate (cAMP) and cyclic guanosine<br />

3’, 5’ monophosphate (cGMP). cAMP and cGMP are involved in numerous cell signaling pathways,<br />

and alterations in concentration of these molecules are associated with several disease states such as<br />

cardiovascular disease, neuronal disorders, and cancer. cAMP and cGMP are generated through the<br />

activity of adenylyl cyclase (AC) or guanylyl cyclase (GC), respectively, and are degraded by phosphodiesterases<br />

(PDEs).<br />

• cAMP and cGMP Assays can be used to infer enzymatic activity of AC/GC or PDEs by<br />

measuring levels of cAMP or cGMP through a competition enzyme-linked immunoassay.<br />

A cAMP assay is a competition ELISA used to<br />

measure cAMP levels in forskolin-treated CHO cells.<br />

Absorbance 450nm<br />

4.0<br />

3.0<br />

2.0<br />

0.5mM IBMX<br />

No IBMX<br />

% Activity<br />

30<br />

1.0<br />

EC 50:<br />

10<br />

0.58 µM (No IBMX)<br />

0.09 µM (0.5 mM IBMX)<br />

0<br />

-3 -2 -2 -1 0 1 2 3 -4 -3 -2 -1 0 1 2 3<br />

110<br />

90<br />

70<br />

50<br />

Log (Forskolin, µM)<br />

Cyclic AMP XP ® Assay Kit #4339: Treatment of CHO cells with Forskolin #3828 increases cAMP concentration as detected by #4339.<br />

CHO cells were seeded at 4x10 4 cells/well in a 96-well plate and incubated overnight. Cells were either left untreated or pretreated with<br />

0.5 mM IBMX for 30 min prior to forskolin treatment (15 min) and lysed with 1X Cell Lysis Buffer #9803. The absorbance values (left) and<br />

percentage of activity (right) are shown above.<br />

The percentage of activity is calculated as follows: % activity=100x[(A-A basal )/(A max -A basal )], where A is the sample absorbance, A max<br />

is the absorbance at maximum stimulation (i.e., high forskolin concentration), and A basal is the absorbance at basal level (no forskolin).<br />

Forskolin directly activates adenylyl cyclases and increases cellular cAMP concentration. IBMX is a non-specific inhibitor of cAMP and<br />

cGMP phosphodiesterases and promotes accumulation of cAMP and cGMP in cells.<br />

chapter 25: Monitoring<br />

BrdU, XTT and<br />

Resazurin Assay Kits<br />

#6813 BrdU Cell Proliferation<br />

Assay Kit<br />

#5492 BrdU Cell Proliferation<br />

Chemiluminescent Assay Kit<br />

#11884 Resazurin Cell Viability Kit<br />

#9095 XTT Cell Viability Kit<br />

#9860 Senescence β-Galactosidase<br />

Staining Kit<br />

Annexin V-FITC and<br />

Caspase-3 Assay Kits<br />

#6592 Annexin V-FITC Early<br />

Apoptosis Detection Kit<br />

#5723 Caspase-3 Activity Assay Kit<br />

cAMP and cGMP Assay Kits<br />

#4339 Cyclic AMP XP ® Assay Kit<br />

#8019 Cyclic AMP XP ®<br />

Chemiluminescent Assay Kit<br />

#4360 Cyclic GMP XP ® Assay Kit<br />

#8020 Cyclic GMP XP ®<br />

Chemiluminescent Assay Kit<br />

GTPase Detection Kits<br />

#8816 Active Arf1 Detection Kit<br />

#8819 Active Cdc42 Detection Kit<br />

#8815 Active Rac1 Detection Kit<br />

#8818 Active Rap1 Detection Kit<br />

#8821 Active Ras Detection Kit<br />

#8820 Active Rho Detection Kit<br />

274 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/monitoring 275


Section III: Workflow Tools<br />

Small GTPase Detection Assays<br />

Like second messenger assays, small GTPase detection assays allow a researcher to evaluate cell<br />

signaling by measuring the presence of activated Ras GTPase as a readout for signaling activity. The<br />

Ras GTPase superfamily consists of five protein families (Ras, Rho, Rab, Arf, and Ran) whose members<br />

cycle between the active, GTP-bound, and inactive, GDP-bound, state. GTPases play an important role<br />

in cell cycle progression, cell survival, and actin cytoskeletal organization and are actively studied in<br />

many fields, including tumor biology.<br />

• GTPase Detection Assays measure activation of a single GTPase in the cell using a GST-fusion<br />

protein to bind the activated form of GTP-bound Rac1 (or other GTPase), which can then be<br />

immunoprecipitated with glutathione resin and analyzed by western blot.<br />

GTPase detection assays assess levels<br />

of active GTPase in a cell lysate.<br />

Active Rac1 Detection Kit #8815: NIH/3T3 cell lysates (500 µl at 1 mg/ml) were treated in vitro<br />

with GTPγS or GDP to activate or inactivate Rac1 (refer to optional step C in protocol). The lysates<br />

were then incubated with glutathione resin and GST-PAK1-PBD (lanes 2 and 3). GTPγS-treated<br />

lysate was also incubated without GST-PAK1-PBD in the presence of glutathione resin as a<br />

negative control (lane 4). WB analysis of cell lysate (20 µg, lane 1) or 20 µl of the eluted samples<br />

(lanes 2–4) was performed using a Rac1 Mouse mAb. Anti-mouse IgG, HRP-linked Antibody<br />

#7076 was used as the secondary antibody.<br />

1 2 3 4<br />

– + – – GDP<br />

– – + + GTPγS<br />

– + + – GST-PAK1-PBD<br />

26<br />

Verification<br />

using siRNA-mediated knockdown to verify function<br />

How can I ask a highly targeted question about protein<br />

or antibody function in the context of the whole cell<br />

Small interfering RNAs (siRNA) are short segments (19–27 nucleotides) of double stranded RNA that<br />

silence expression of a single gene in a sequence-specific manner. This is achieved when a single<br />

strand of the RNA duplex associates with the RNA-induced silencing complex (RISC), a multiprotein<br />

complex containing endonucleases that bind and degrade mRNA at a sequence complementary to the<br />

siRNA. Thus, transfection of a specific siRNA molecule into living cells can result in specific inhibition of<br />

expression of the corresponding protein.<br />

siRNA can be used to:<br />

• Verify an antibody is detecting its intended target<br />

• Analyze the functional role of a node within a signaling pathway<br />

• Validate a potential therapeutic target<br />

• Explore gene function in the context of a disease or biological system<br />

The GTP-bound<br />

GTPase pull-down<br />

process can be<br />

divided into 3 steps.<br />

Step 1: Mix sample, binding protein,<br />

and glutathione resin in the spin cup<br />

and incubate at 4ºC to allow GTP-bound<br />

GTPase binding to the glutathione resin<br />

through GST-linked binding protein.<br />

Step 2: Remove unbound proteins by<br />

centrifugation.<br />

Step 3: Elute glutathione resin-bound<br />

GTPase with SDS buffer. The eluted<br />

sample can then be analyzed by WB.<br />

sample<br />

spin cup<br />

collection tube<br />

= Other Proteins<br />

= GTP-bound GTPase<br />

= GDP-bound GTPase<br />

= Binding Protein<br />

= Glutathione<br />

Step 1 Step 2<br />

Step 3<br />

- mix sample with resin<br />

and binding protein<br />

- incubate<br />

wash<br />

elute<br />

Western blot<br />

For example, in the experiment below, SignalSilence ® p27 Kip1 siRNA from Cell Signaling Technology is<br />

used to verify antibody specificity. The robust signal obtained from p27 Kip1 (D69C12) XP ® Rabbit mAb<br />

#3686 in the presence of control siRNA (-) is diminished when p27 Kip1 expression is silenced using<br />

two different p27 Kip1 siRNAs (+), indicating the antibody specifically detects the p27 Kip1 protein.<br />

siRNA-mediated knockdown<br />

verifies p27 Kip1<br />

antibody specificity.<br />

kDa<br />

80<br />

60<br />

50<br />

40<br />

30<br />

20<br />

50<br />

40<br />

I<br />

II<br />

– + +<br />

p27<br />

Kip1<br />

β-Actin<br />

p27 Kip1<br />

siRNA<br />

SignalSilence ® p27 Kip1 siRNA I #12324<br />

and siRNA II #12410: WB analysis of extracts<br />

from HeLa cells, transfected with 100 nM<br />

SignalSilence ® Control siRNA (Unconjugated)<br />

#6568 (-), #12324 (+), or #12410 (+), using p27<br />

Kip1 (D69C12) XP ® Rabbit mAb #3686 (upper)<br />

or β-Actin (D6A8) Rabbit mAb #8457 (lower). The<br />

p27 Kip1 (D69C12) XP ® Rabbit mAb confirms<br />

silencing of p27 Kip1 expression, while the β-Actin<br />

(D6A8) Rabbit mAb is used as a loading control.<br />

SignalSilence ® siRNA<br />

Experimental Controls<br />

SignalSilence ® Control siRNA<br />

(Fluorescein Conjugate) #6201<br />

Transfection efficiency<br />

SignalSilence ® Control siRNA<br />

(Unconjugated) #6568<br />

siRNA specificity<br />

How SignalSilence ® siRNA can benefit your research:<br />

• Two siRNAs Available for Most Targets: confirm inhibition at the functional protein<br />

level using two equally potent sequences<br />

• Rigorously Validated Duplexes: confidently inhibit expression of your intended protein<br />

• Appropriate Controls: monitor transfection efficiency with a fluorescein-labeled<br />

nontargeted siRNA and specificity with an unconjugated control<br />

• Target Proteins From Different Species: inhibit expression of human or mouse targets<br />

A positive control<br />

siRNA confirms c-Myc<br />

siRNA specificity in the<br />

target cells, suggesting<br />

that absence of c-Myc<br />

expression is due to<br />

specific knockdown.<br />

SignalSilence ® Control<br />

siRNA (Unconjugated) #6568:<br />

Confocal IF analysis of HeLa<br />

cells, transfected with #6568<br />

(left) or SignalSilence ® c-Myc<br />

siRNA I #6341 (right), using<br />

c-Myc (D84C12) XP ® Rabbit mAb<br />

#5605 (green). Actin filaments<br />

were labeled with DyLight 554<br />

Phalloidin #13054 (red).<br />

276 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

www.cellsignal.com/verification 277


27<br />

Section III: Workflow Tools<br />

Customization<br />

antibodies in a carrier-free<br />

formulation for specific assay platforms<br />

How can I obtain an antibody formulated for<br />

the special requirements of my assay platform<br />

Researchers studying protein activities and associations can choose from a wide array of high throughput<br />

assay platforms. Many of these require antibodies in specific concentrations or in formulations free<br />

of BSA or other carrier proteins that are typically found in off-the-shelf antibody storage buffers. Custom<br />

antibody formulations from Cell Signaling Technology (<strong>CST</strong>) are highly specific, in-house validated<br />

antibodies that are ready to be labeled or used directly in your assay platform, providing the flexibility<br />

necessary for your unique experiment.<br />

Single peak indicates the antibody is intact and shows no cleavage or degradation.<br />

AU<br />

0.070<br />

0.065<br />

0.060<br />

0.055<br />

0.050<br />

0.045<br />

0.040<br />

0.035<br />

0.030<br />

0.025<br />

0.020<br />

Single peak indicates that the<br />

antibody is intact and shows<br />

no cleavage or degradation.<br />

chapter 27: Customization<br />

PD-L1 (E1L3N ® ) XP ® Rabbit mAb<br />

#13684: Size exclusion chromatogram<br />

of #13684 formulated in PBS buffer.<br />

Custom formulated antibodies can be used to:<br />

• Label antibodies with fluorochromes, lanthanides, or molecules for use on specific platforms such<br />

as AlphaScreen ® , DELFIA ® or HTRF assays<br />

• Develop pair-based sandwich assays for use on specific platforms such as Bio-Plex ® or Luminex ®<br />

• Develop custom plate-based sandwich ELISA kits<br />

• Design custom multiplex assays where several protein readouts are assayed using multiple targetspecific<br />

antibodies<br />

0.015<br />

0.010<br />

0.005<br />

0.000<br />

2 4 6 8 10 12 14 16 18 20 22 24 26 28 30<br />

minutes<br />

<strong>CST</strong> can supply antibodies in phosphate buffered saline (PBS) or borate buffer at your specified concentration.<br />

Once the antibodies are formulated in the specified buffer, we validate them for molecular integrity<br />

by size exclusion chromatography and application-specific functionality using WB, IP, peptide ELISA,<br />

flow cytometry, IHC, or IF. The application testing is dependent on the proposed use of the antibody.<br />

Peptide ELISA confirms phospho-specificity of multiple antibody lots.<br />

How custom formulated antibodies<br />

from <strong>CST</strong> can benefit your research:<br />

• <strong>CST</strong> is ISO9001:2008 certified for the production and supply of monoclonal antibodies, ensuring<br />

reproducible, traceable product quality<br />

• Antibodies are supplied in formulations adaptable to multiple platforms<br />

• Each antibody is custom manufactured with complete documentation support to ensure<br />

product integrity<br />

• Dedicated custom antibody personnel actively manage the supply chain for full transparency from<br />

initial enquiries to production and post-sales collaborative support. Key features of the supply chain<br />

process include:<br />

– Specific lots of antibody material can be reserved for long-term multiple purchases<br />

– Custom packaging ensures that the material is available in an experiment-or<br />

platform-ready format<br />

– <strong>CST</strong> freezer-to-customer traceability ensures peace of mind and guarantees product integrity<br />

and performance<br />

– Priority Alert Plus shipping with SenseAware SM technology from FedEx minimizes product transit<br />

time and allows continuous monitoring of temperature during shipment<br />

– Scientists specializing in unique application support are available to support custom<br />

product clients<br />

• All catalog antibodies are available in bulk volumes, which ensures long term supply.<br />

Lot reservation is available to reserve a single lot of any of our reagents.<br />

Phospho-Stat1 (Tyr701) (58D6) Rabbit mAb #9167: Peptide ELISA analysis of multiple lots of #9167, using plates<br />

coated with phospho-peptides (Tyr-P) or non-phospho-peptides (NP). The data shows minimal lot-to-lot variation.<br />

278 For Research Use Only. Not For Use in Diagnostic Procedures. See pages 302 & 303 for Pathway Diagrams, Application, and Reactivity keys.<br />

<strong>CST</strong> Custom Formulated Antibodies<br />

<strong>CST</strong>’s primary antibodies are available in a carrier free formulation at custom concentrations that allow antibody<br />

labeling, conjugation or custom assays. Visit our website to learn more. www.cellsignal.com/custom<br />

www.cellsignal.com/customization<br />

279


IV<br />

Additional<br />

Information<br />

Section Includes:<br />

About <strong>CST</strong><br />

Nature Conservancy<br />

Publications by <strong>CST</strong> Scientists<br />

Index<br />

Diagram & Table Keys<br />

Contact Information<br />

280<br />

Adhesion<br />

Depiction of cytoskeletal and extracellular structures and proteins involved in adhesion, including<br />

an adherens junction and integrin-mediated cellular interaction with the extracellular matrix.<br />

www.cellsignal.com/cstlandscapes<br />

281


28<br />

Section IV: Additional Information<br />

About <strong>CST</strong><br />

Behind the Antibodies<br />

The preceding sections in this book offer proof that we are serious about both advancing science and<br />

the quality of our products. But what about the company and people behind the antibodies If you met<br />

someone from <strong>CST</strong>, you would learn that we are scientists ourselves, we value the respect of our peers,<br />

are very proud of the work we do, and are just as interested as the people who buy our products in the<br />

outcome of their research. Like everyone else, we have our own back-story about who we are, how we<br />

came to be where we are now, and our dreams for the future. We’d like to introduce you to that <strong>CST</strong>.<br />

Science is our passion, and it’s how we pay the bills—we create and sell antibodies and related<br />

products that can be used in research aspiring to improve human health. But we are also citizens of our<br />

local and global communities. We are parents who care about the quality of science taught at our local<br />

schools, accountants who care about the environment, business leaders concerned with the footprint<br />

we leave behind, and scientists curious about art.<br />

As a private company we have the freedom to do things differently. We can invest our profits in our<br />

own translational research programs. We can focus on long-term goals without having to answer to<br />

the stock market each quarter. But we don’t just throw money around and hope to make a difference.<br />

We hire people who are very good at what they do and disciplined enough to do it both in and out of<br />

the lab. We have several volunteer committees within <strong>CST</strong> that independently determine how to spend<br />

funds we have set aside for various initiatives. It is what sets us apart from many other businesses.<br />

Corporate Social Responsibility<br />

Corporate Donations Committee<br />

As a company, <strong>CST</strong> sets aside money to help local organizations share in our success and find additional<br />

funding for selected initiatives. How that money is spent is determined by our employees. Our Corporate<br />

Donations Committee (CDC) is a group of enthusiastic volunteers at our US locations who decide on their<br />

own where that money can do the most good. Twice a year the CDC reviews requests and distributes<br />

funds to local schools, non-profit organizations, and charities.<br />

It’s a special culture where the responsibility of granting money is handed to employees. Involving<br />

employees in some of <strong>CST</strong>’s philanthropy helps develop the kind of culture that makes a difference and<br />

empowers them to feel part of the <strong>CST</strong> mission.<br />

Education in Science<br />

Naturally, we think that science education is important to keep discovery alive in the future. Because<br />

most of us can think of a teacher or an experience that sparked our interest in science, <strong>CST</strong>’s Education<br />

in Science program supports local public high schools by giving teachers access to much needed<br />

grant money for resources that keep science fun and engaging.<br />

At the college level, our Summer Internship Program provides interns from local colleges and communities<br />

with a positive work experience alongside our associates within the lab and outside of the<br />

lab. It’s gratifying (and fun) to see some of these interns return as full-time employees after completing<br />

their degree.<br />

The Arts<br />

Exchanging ideas in a supportive environment fosters the kind of creativity that both scientists and<br />

artists thrive in. Creative people are appreciated at <strong>CST</strong>, and the three gallery spaces in our two US<br />

facilities exhibit the creativity of some of New England’s finest artists. Our artist receptions and the discussions<br />

about their work always bring a different perspective in viewing artwork. A short walk through<br />

our labs and office spaces reveals walls filled with the work of artists that have previously exhibited at<br />

<strong>CST</strong> and which continue to enrich anyone who cares to stop and have a closer look.<br />

You can see why we think <strong>CST</strong> is a special kind of company. We are about more than making money.<br />

We are trying to build a business that’s not only great for the customers, but also rewarding for our<br />

employees and appreciated as a member of our local and global communities.<br />

We hire smart people with a commitment and passion to succeed and make a difference. These are<br />

the people behind the antibodies and every product we make.<br />

Environmental Commitment<br />

As a global citizen, <strong>CST</strong> recognizes that our business has real impact on the planet. We are working to<br />

lower our carbon footprint and limit the impact we make on the environment. In addition to incorporating<br />

sustainability into our own operations, <strong>CST</strong> also works within our communities to help restore<br />

ecosystems by supporting environmental organizations working in the field to promote conservation.<br />

This has been part of our mission since <strong>CST</strong> was founded in 1999.<br />

Sustainability<br />

We’re not forced into being good stewards of the environment. We’re doing it because we care.<br />

For example:<br />

Transportation Management Plan<br />

Our Transportation Management Plan (TMP) uses financial incentives and up to two days off work to<br />

promote alternative transportation such as carpooling, walking, biking, or taking public transportation.<br />

This has helped engaged and concerned employees lower our carbon emissions from commuting by<br />

nearly 8% in less than two years.<br />

Responsible Marketing and Packaging<br />

Since 2009, our Massachusetts locations have been shipping our products using a new shipping<br />

cooler composed from cardboard, mineral wool, rock wool, slag wool, and biodegradable plastic. This<br />

new shipping cooler has the same thermal properties as a Styrofoam box but can degrade in a landfill.<br />

Employee Community Garden Program<br />

<strong>CST</strong>’s community garden program provides fresh summer produce for the employees and our in-house<br />

cafeterias at our two Massachusetts locations. In the Netherlands, our employees enjoy fresh locally<br />

sourced fruit.<br />

The examples above are only a few examples of how we are working to protect our planet. Integral to<br />

all of these sustainability efforts is <strong>CST</strong>’s employee run Green Committee. The vision of this committee<br />

is to understand the company’s environmental impacts, create awareness within the organization,<br />

and help develop strategies to minimize environmental impact. This committee continues to find<br />

sustainable business solutions and develops successful, award-winning programs that have made a<br />

difference in our industry and our communities.<br />

Environmental Awareness and Support<br />

Moving science forward by helping solve important questions about disease and cancer is our mission<br />

but ultimately, human health depends on the health of our planet. It would be misguided to cure<br />

ourselves of disease, only to find we had nowhere to live, not enough food to eat or no clean air to<br />

breathe. Understanding the interactions that shape the living planet on a global level is just as important<br />

to us as the complex molecular interactions involved in cellular signaling.<br />

Each year for the <strong>CST</strong> Nature Calendar, we select a specific environmental theme that highlights not<br />

only the extraordinary beauty of nature, but also touches upon an important environmental issue<br />

affecting life on our planet.<br />

chapter 28: About <strong>CST</strong><br />

“At <strong>CST</strong> we measure<br />

our performance in<br />

the world by practicing<br />

triple bottom line<br />

economics: People,<br />

Planet, and Profit.”<br />

David Comb, Director of<br />

Corporate Social Responsibility<br />

Produce from <strong>CST</strong>’s Employee Garden<br />

Atrium at <strong>CST</strong> Headquarters, Danvers, MA<br />

Trees Red; Estelle Disch<br />

282 For Research Use Only. Not For Use in Diagnostic Procedures.<br />

www.cellsignal.com/cstcommitments<br />

283


29 <strong>CST</strong> Nature Conservancy<br />

Section IV: Additional Information<br />

Sagono<br />

Bamboo Forest<br />

This small patch of bamboo forest,<br />

on the outskirts of Kyoto, is a popular<br />

Japanese tourist destination that<br />

people have visited for thousands<br />

of years because of its sense of<br />

peacefulness and tranquility.<br />

Conservancy Efforts & Resources<br />

Human know-how still cannot replace nature, but we have acquired the means of disrupting natural<br />

processes on a global scale. With this newfound ability comes a responsibility to manage the planet’s<br />

resources wisely. By safeguarding vital ecosystems whose services we depend on, rather than plundering<br />

them for immediate gain, we can ensure that nature flourishes. In so doing, humans will benefit too.<br />

As part of <strong>CST</strong>’s commitment to the environment, each year we support non-profit organizations that are<br />

helping to protect the earth’s magnificent biodiversity and the ecosystems that sustain life on our planet.<br />

chapter 29: <strong>CST</strong> Nature Conservancy<br />

2015<br />

Protected Landscapes<br />

The best way to protect endangered<br />

species is often to protect the land these<br />

creatures call home. Usually, the bigger<br />

the swath of land that can be set aside,<br />

the better. The same general approach<br />

holds for preserving treasured forests,<br />

mountains, waterfalls, rivers, and lakes.<br />

Ecosystems blend into each other, and<br />

nature, accordingly, does not abide<br />

by the artificial boundaries drawn by<br />

humans—even though natural systems<br />

can, of course, be heavily impacted by<br />

societal decisions and activities.<br />

Support:<br />

• Essex County Greenbelt Association<br />

• The Jurassic Coast Trust<br />

• Kunming Institute of Botany<br />

• Wild Bird Society of Japan<br />

Jurassic Coast on the eastern shore of England. © Joaquin Pinho/Getty Images<br />

Red Crowned Crane is<br />

an endangered species<br />

found only in Japan<br />

and China. Famous for<br />

their complex dancing<br />

rituals, this rare species<br />

is most threatened by<br />

habitat destruction.<br />

© Kerstin Hinze/Minden Pictures<br />

2014–13<br />

Magnificent Marine<br />

Environments<br />

Images of Earth by astronauts nearly<br />

four decades ago dramatically proved<br />

what we knew but only dimly appreciated—the<br />

Earth is a water planet. But<br />

the sea-blue dominating those images<br />

concealed as much as it revealed. Unlike<br />

the land’s sculpted landscape, which<br />

suggested diversity and begged for<br />

further exploration, the uniform surface<br />

of the ocean suggested sameness. This<br />

could not be further from the truth as we<br />

continue to find startling new life forms<br />

and are constantly reminded just how<br />

dependent we are on the ocean.<br />

As coastal populations and economic<br />

activity continue to grow, the need for<br />

protecting additional coastal areas will<br />

grow as well.<br />

Support:<br />

• Southern Environmental Association<br />

• Toledo Institute for Development<br />

and Environment<br />

• Ecologic Development Fund<br />

• Large Pelagics Research Center<br />

• Oceana<br />

• Conservation Law Foundation<br />

• Ipswich River Watershed Association<br />

• Nature Conservancy<br />

• New England Aquarium<br />

© Akira Kaede/Getty Images<br />

284 For Research Use Only. Not For Use in Diagnostic Procedures.<br />

Spinyhead Blenny (Acanthemblemaria spinosa) emerges from its coral home to feed at Lighthouse Reef, Belize.<br />

© Brian J. Skerry/National Geographic Creative<br />

www.cellsignal.com/cstcsr<br />

285


Section IV: Additional Information<br />

chapter 29: <strong>CST</strong> Nature Conservancy<br />

2013<br />

Mesoamerican Reef<br />

Extending more than 600 miles from<br />

Isla Contoy, a bird sanctuary near the<br />

Yucatan Peninsula, to the Bay Islands<br />

of Honduras, the Mesoamerican Reef is<br />

the largest barrier reef in the Western<br />

Hemisphere and the second largest<br />

in the world. About one-third of this<br />

natural wonder, and arguably the choicest<br />

stretch of reef, lies off the coast of<br />

Belize. Charles Darwin once described<br />

this complex—consisting of dazzling<br />

corals, crystal clear blue waters, and a<br />

host of aquatic denizens—as “the most<br />

remarkable reef in the West Indies.”<br />

Support:<br />

• Southern Environmental Association<br />

• Toledo Institute for Development<br />

and Environment<br />

• Ecologic Development Fund<br />

• Large Pelagics Research Center<br />

2009–10<br />

Conservation<br />

Without Borders<br />

Nature does not stop at the borders<br />

of a national park or between nations.<br />

Appreciation of that fact has led to a<br />

growing realization among conservationists<br />

that the best, and perhaps only,<br />

way to preserve the environment<br />

and protect wildlife is through large<br />

landscapes that transcend the artificial<br />

boundaries drawn by humans. Animals<br />

need room to rove for any number of<br />

reasons—and these needs cannot<br />

be met within isolated sanctuaries<br />

standing as islands amidst a sea of<br />

development. The home range for a<br />

single grizzly bear, for example, can<br />

cover 1,000 square miles—and wolves<br />

roam even farther. Securing movement<br />

corridors and other “linkages” between<br />

designated wilderness areas has thus<br />

become a cornerstone of an emerging<br />

conservation strategy.<br />

Support:<br />

• Yellowstone to Yukon<br />

• Intl. Gorilla Conservation<br />

• Large Pelagics Research Center<br />

• Gombe School of Environment<br />

and Society<br />

Cushion sea star (Oreaster reticulatus) sitting in sea grass beds (Thalassia), South Water Caye, Belize. © Tony Rath/tonyrath.com<br />

Eastern Gorillas, Volcanoes National Park, Rwanda. © Thomas Marent/Minden Pictures<br />

2011–12<br />

Conservation<br />

Treasures<br />

Since its founding in 1951, The Nature<br />

Conservancy (TNC) has protected about<br />

120 million acres in all 50 American<br />

states and more than 30 countries. In<br />

that time, the organization has focused<br />

its conservation efforts on what it calls<br />

“The Last Great Places”—habitats<br />

deemed special in terms of the plant<br />

and animal species they harbor, as well<br />

as the natural processes they sustain.<br />

Support:<br />

• Appalachian Mountain Club<br />

• Ecologic Development Fund<br />

• Essex County Greenbelt Association<br />

• Gombe School of Environment<br />

and Society<br />

• Ipswich River Watershed Association<br />

• Large Pelagics Research Center<br />

• The Nature Conservancy<br />

• Trout Unlimited, Alaska<br />

2007–08<br />

World’s Fisheries<br />

in Crisis<br />

For most of human history, it seemed<br />

inconceivable that our species could<br />

make an appreciable dent in the ocean’s<br />

bounty. Surely we’d do nothing to<br />

jeopardize the principal source of animal<br />

protein for a billion people. Yet today, with<br />

millions of fishing vessels patrolling the<br />

seas and marine life decimated in their<br />

wake, ocean observers know the opposite<br />

to be true: “In all the seas there are very<br />

few places left with undisturbed fish<br />

stocks”, claims Boris Worm of Dalhousie<br />

University in Halifax, Canada. “The whole<br />

ocean has been transformed.”<br />

Support:<br />

• New England Aquarium<br />

• Conservation Law Foundation<br />

Atlantic Salmon (Salmo salar),<br />

St. Jean River, Quebec<br />

Giant sequoia (Sequoiadendron giganteum) in Yosemite National Park, California. © David Noton/Minden Pictures<br />

Leafy sea dragon (Phycodurus eques), found only in temperate waters around southern<br />

and western Australia. © Paul Sutherland/National Geographic Creative<br />

286 For Research Use Only. Not For Use in Diagnostic Procedures. www.cellsignal.com/cstcsr<br />

© Paul Nicklen<br />

287


Section IV: Additional Information<br />

chapter 29: <strong>CST</strong> Nature Conservancy<br />

2005–06<br />

Spectacular<br />

Migrations<br />

Each year, traveling by land, air, and<br />

sea, animals make time-honored<br />

journeys in pursuit of their destiny. They<br />

follow paths taken by their forebears,<br />

obeying cues they alone perceive and<br />

dare not ignore. Swimming, flying, running,<br />

and crawling, by day and by night,<br />

these creatures venture forth in large<br />

groups or solo. If movement is life, ours<br />

is truly a planet in motion.<br />

Support:<br />

• Operation Migration<br />

• The Atlantic Salmon Federation<br />

2002<br />

Freshwater Wetlands<br />

The occupants of planet Earth are<br />

dependent on an elaborate life-support<br />

system that maintains the air we breathe,<br />

regulates temperature, supplies reserves<br />

of food and water, and shields us from<br />

deadly radiation. This system, provided<br />

by nature free-of-charge, offers a broad<br />

array of critical services: purifying the<br />

air and water, maintaining soil fertility,<br />

decomposing and detoxifying wastes,<br />

recycling essential nutrients, stabilizing<br />

the climate, protecting us from the sun’s<br />

ultraviolet rays, mitigating floods and<br />

droughts, pollinating our crops, and<br />

controlling agricultural pests.<br />

Support:<br />

• Ipswich River Watershed Association<br />

• Massachusetts Chapter of<br />

The Nature Conservancy<br />

Group of caribou (Rangifer tarandus) swimming through an Alaskan river during migration. © Michio Hoshino/Minden Pictures<br />

Underwater landscape of water lilies, Okavango Delta, Botswana. © Frans Lanting/Frans Lanting Stock<br />

2003–04<br />

Plight of the<br />

Pollinators<br />

Ecologists are nowhere close to<br />

documenting all the valuable services<br />

performed by the 100,000 or more<br />

species of pollinators. While dispensing<br />

pollen, bees, bats, birds, and other<br />

animals also add to ecosystem productivity<br />

by facilitating the spread of seeds<br />

and the redistribution of nitrogen-rich<br />

wastes. With their contributions largely<br />

unrecognized, pollinators hold their<br />

place as the unsung heroes of the<br />

natural world.<br />

Support:<br />

• Xerces Society<br />

• Bat Conservation International<br />

2001–02<br />

Nature’s Services<br />

Astronauts on a space station are<br />

dependent on finely-tuned engineered<br />

systems to maintain the air they<br />

breathe, regulate the temperature,<br />

provide food and water, dispose of their<br />

waste products, and protect them from<br />

deadly radiation. Here on Earth, humans<br />

are also dependent on an elaborate<br />

life-support system that sustains the<br />

biosphere which all organisms inhabit.<br />

Support:<br />

• Union of Concerned Scientists<br />

Rafflesia (Rafflesia keithii), 33” wide, 2nd largest specimen found in Borneo, Mount Kinabalu Indonesia. © Frans Lanting/Frans Lanting Stock<br />

Quetzal (Pharomachrus mocinno) in Guatemala. © Steve Winter/National Geographic Stock<br />

288 For Research Use Only. Not For Use in Diagnostic Procedures.<br />

www.cellsignal.com/cstcsr 289


30<br />

Section IV: Additional Information<br />

Through their ongoing<br />

commitment to innovative<br />

research, <strong>CST</strong> scientists<br />

have contributed to a<br />

wealth of peer-reviewed<br />

publications in cancer<br />

research, proteomicsbased<br />

discovery, and<br />

novel antibody technology.<br />

2014<br />

Gray, V.E. et al. (2014) Signatures of natural selection<br />

on mutations of residues with multiple posttranslational<br />

modifications. Mol. Biol. Evol. 31, 1641–1645.<br />

Oslund, R.C. et al. (2014) A Phosphohistidine<br />

Proteomics Strategy Based on Elucidation of a Unique<br />

Gas-phase Phosphopeptide Fragmentation Mechanism.<br />

J. Am. Chem. Soc. [Epub ahead of print].<br />

Mulherkar, S. et al. (2014) The small GTPases RhoA<br />

and Rac1 regulate cerebellar development by controlling<br />

cell morphogenesis, migration and foliation. Dev.<br />

Biol. [Epub ahead of print].<br />

2013<br />

Miller, R.A. et al. (2013) Biguanides suppress hepatic<br />

glucagon signalling by decreasing production of cyclic<br />

AMP. Nature 494, 256–260.<br />

Ling, H. et al. (2013) Transforming growth factor β<br />

neutralization ameliorates pre-existing hepatic fibrosis<br />

and reduces cholangiocarcinoma in thioacetamidetreated<br />

rats. PLoS ONE 8, e54499.<br />

Mulherkar, S. et al. (2013) The Small GTPase RhoA Is<br />

Required for Proper Locomotor Circuit Assembly. PLoS<br />

ONE 8, e67015.<br />

Israelsen, W.J. et al. (2013) PKM2 Isoform-Specific<br />

Deletion Reveals a Differential Requirement for Pyruvate<br />

Kinase in Tumor Cells. Cell 155, 397–409.<br />

Hitosugi, T. et al. (2013) Tyr26 phosphorylation of<br />

PGAM1 provides a metabolic advantage to tumours<br />

by stabilizing the active conformation. Nat. Commun.<br />

4, 1790.<br />

Morandell, S. et al. (2013) A Reversible Gene-Targeting<br />

Strategy Identifies Synthetic Lethal Interactions<br />

between MK2 and p53 in the DNA Damage Response<br />

In Vivo. Cell Rep. 5, 868–877.<br />

2012<br />

Hornbeck, P.V. et al. (2012) PhosphoSitePlus: a<br />

comprehensive resource for investigating the structure<br />

and function of experimentally determined posttranslational<br />

modifications in man and mouse. Nucleic<br />

Acids Res. 40, 261–270.<br />

Katayama, R. et al. (2012) Mechanisms of acquired<br />

crizotinib resistance in ALK-rearranged lung Cancers.<br />

Sci. Transl. Med. 4, 120ra17.<br />

Feng, H. et al. (2012) Phosphorylation of dedicator<br />

of cytokinesis 1 (Dock180) at tyrosine residue<br />

Y722 by Src family kinases mediates EGFRvIII-driven<br />

glioblastoma tumorigenesis. Proc. Natl. Acad. Sci. USA<br />

109, 3018–3023.<br />

Garnett, M.J. et al. (2012) Systematic identification of<br />

genomic markers of drug sensitivity in cancer cells.<br />

Nature 483, 570–575.<br />

Cheung, W.C. et al. (2012) A proteomics approach for<br />

the identification and cloning of monoclonal antibodies<br />

from serum. Nat. Biotechnol. 30, 447–452.<br />

290 For Research Use Only. Not For Use in Diagnostic Procedures.<br />

Publications<br />

by <strong>CST</strong> Scientists<br />

Stokes, M.P. et al. (2012) PTMScan direct: identification<br />

and quantification of peptides from critical signaling<br />

proteins by immunoaffinity enrichment coupled<br />

with LC-MS/MS. Mol. Cell Proteomics 11, 187–201.<br />

Ren, H. et al. (2012) Identification of anaplastic<br />

lymphoma kinase as a potential therapeutic target in<br />

ovarian cancer. Cancer Res. 72, 3312–3323.<br />

Rimkunas, V.M. et al. (2012) Analysis of Receptor<br />

Tyrosine Kinase ROS1-Positive Tumors in Non-Small<br />

Cell Lung Cancer: Identification of a FIG-ROS1 Fusion.<br />

Clin. Cancer Res. 18, 4449–4457.<br />

Xu, W. et al. (2012) Dynamic tyrosine phosphorylation<br />

modulates cycling of the HSP90-P50(CDC37)-AHA1<br />

chaperone machine. Mol. Cell 47, 434–443.<br />

Hardee, M.E. et al. (2012) Resistance of glioblastomainitiating<br />

cells to radiation mediated by the tumor<br />

microenvironment can be abolished by inhibiting transforming<br />

growth factor-β. Cancer Res. 72, 4119–4129.<br />

Hezel, A.F. et al. (2012) TGF-β and αvβ6 integrin act<br />

in a common pathway to suppress pancreatic cancer<br />

progression. Cancer Res. 72, 4840–4845.<br />

Hitosugi, T. et al. (2012) Phosphoglycerate mutase 1<br />

coordinates glycolysis and biosynthesis to promote<br />

tumor growth. Cancer Cell 22, 585–600.<br />

Sato, S. et al. (2012) Proteomics-directed cloning of<br />

circulating antiviral human monoclonal antibodies.<br />

Nat. Biotechnol. 30, 1039–1043.<br />

Buckley, S.M. et al. (2012) Regulation of pluripotency<br />

and cellular reprogramming by the ubiquitin-proteasome<br />

system. Cell Stem Cell 11, 783–798.<br />

Stokes, M.P. et al. (2012) Quantitative Profiling of DNA<br />

Damage and Apoptotic Pathways in UV Damaged Cells<br />

Using PTMScan Direct. Int J. Mol. Sci. 14, 286–307.<br />

Foulkes-Murzycki, J.E. et al. (2012) Cooperative Effects<br />

of Drug-Resistance Mutations in the Flap Region<br />

of HIV-1 Protease. ACS Chem. Biol. 8, 513–518.<br />

Li, Z. et al. (2012) Gr-1+CD11b+ cells are responsible<br />

for tumor promoting effect of TGF-β in breast cancer<br />

progression. Int. J. Cancer 131, 2584–2595.<br />

2011<br />

Hülper, P. et al. (2011) Tumor localization of an<br />

anti-TGF-β antibody and its effects on gliomas.<br />

Int. J. Oncol. 38, 51–59.<br />

Gu, T.L. et al. (2011) Survey of tyrosine kinase signaling<br />

reveals ROS kinase fusions in human cholangiocarcinoma.<br />

PLoS ONE 6, e15640.<br />

Kettenbach, A.N. et al. (2011) Absolute quantification<br />

of protein and post-translational modification abundance<br />

with stable isotope-labeled synthetic peptides.<br />

Nat. Protoc. 6, 175–186.<br />

Verstraeten, V.L. et al. (2011) Protein farnesylation<br />

inhibitors cause donut-shaped cell nuclei attributable<br />

to a centrosome separation defect. Proc. Natl. Acad.<br />

Sci. USA 108, 4997–5002.<br />

Dammer, E.B. et al. (2011) Polyubiquitin linkage<br />

profiles in three models of proteolytic stress suggest<br />

the etiology of Alzheimer disease. J. Biol. Chem. 286,<br />

10457–10465.<br />

Brave, S.R. et al. (2011) Assessing the activity of cediranib,<br />

a VEGFR-2/3 tyrosine kinase inhibitor, against<br />

VEGFR-1 and members of the structurally related<br />

PDGFR family. Mol. Cancer Ther. 10, 861–873.<br />

Pighi, C. et al. (2011) Phospho-proteomic analysis of<br />

mantle cell lymphoma cells suggests a pro-survival<br />

role of B-cell receptor signaling. Cell. Oncol. (Dordr)<br />

34, 141–153.<br />

Gu, T.L. et al. (2011) Survey of activated FLT3<br />

signaling in leukemia. PLoS ONE 6, e19169.<br />

Yadav, H. et al. (2011) Protection from obesity and<br />

diabetes by blockade of TGF-β/Smad3 signaling.<br />

Cell Metab. 14, 67–79.<br />

Kremer, K.N. et al. (2011) Stromal Cell-Derived<br />

Factor-1 Signaling via the CXCR4-TCR Heterodimer<br />

Requires Phospholipase C-{beta}3 and Phospholipase<br />

C-{gamma}1 for Distinct Cellular Responses.<br />

J. Immunol. 187, 1440–1447.<br />

Zhang, Q. et al. (2011) TGF-β regulates DNA methyltransferase<br />

expression in prostate cancer, correlates<br />

with aggressive capabilities, and predicts disease<br />

recurrence. PLoS ONE 6, e25168.<br />

Kim, W. et al. (2011) Systematic and quantitative<br />

assessment of the ubiquitin-modified proteome.<br />

Mol. Cell 44, 325–340.<br />

Emanuele, M.J. et al. (2011) Global Identification of<br />

Modular Cullin-RING Ligase Substrates. Cell 147,<br />

459–474.<br />

Feng, H. et al. (2011) Activation of Rac1 by Src-dependent<br />

phosphorylation of Dock180Y1811 mediates<br />

PDGFRα-stimulated glioma tumorigenesis in mice and<br />

humans. J. Clin. Invest. 121, 4670–4684.<br />

Biswas, S. et al. (2011) Anti-transforming growth<br />

factor ß antibody treatment rescues bone loss and<br />

prevents breast cancer metastasis to bone. PLoS ONE<br />

6, e27090.<br />

Bouquet, F. et al. (2011) TGFβ1 inhibition increases<br />

the radiosensitivity of breast cancer cells in vitro and<br />

promotes tumor control by radiation in vivo. Clin.<br />

Cancer Res. 17, 6754–6765.<br />

Fan, J. et al. (2011) Tyrosine Phosphorylation of<br />

Lactate Dehydrogenase A Is Important for NADH/<br />

NAD+ Redox Homeostasis in Cancer Cells. Mol. Cell.<br />

Biol. 31, 4938–4950.<br />

Hitosugi, T. et al. (2011) Tyrosine phosphorylation<br />

of mitochondrial pyruvate dehydrogenase kinase 1<br />

is important for cancer metabolism. Mol. Cell 44,<br />

864–877.<br />

Lee, K.A. et al. (2011) Ubiquitin ligase substrate<br />

identification through quantitative proteomics at both<br />

the protein and peptide levels. J. Biol. Chem. 286,<br />

41530–41538.<br />

Lonning, S. et al. (2011) Antibody targeting of TGF-β<br />

in cancer patients. Curr. Pharm. Biotechnol. 12,<br />

2176–2189.<br />

2010<br />

Moritz, A. et al. (2010) Akt-RSK-S6 kinase signaling<br />

networks activated by oncogenic receptor tyrosine<br />

kinases. Sci. Signal. 3, ra64.<br />

van Vugt, M.A. et al. (2010) A mitotic phosphorylation<br />

feedback network connects Cdk1, Plk1, 53BP1, and<br />

Chk2 to inactivate the G(2)/M DNA damage checkpoint.<br />

PLoS Biol. 8, e1000287.<br />

Gu, T.L. et al. (2010) Identification of activated<br />

Tnk1 kinase in Hodgkin’s lymphoma. Leukemia 24,<br />

861–865.<br />

Pecquet, C. et al. (2010) Induction of myeloproliferative<br />

disorder and myelofibrosis by thrombopoietin receptor<br />

W515 mutants is mediated by cytosolic tyrosine 112<br />

of the receptor. Blood 115, 1037–1048.<br />

Nishimura, E.K. et al. (2010) Key roles for transforming<br />

growth factor beta in melanocyte stem cell maintenance.<br />

Cell Stem Cell 6, 130–140.<br />

Hudon, V. et al. (2010) Renal tumour suppressor<br />

function of the Birt-Hogg-Dubé syndrome gene<br />

product folliculin. J. Med. Genet. 47, 182–189.<br />

Hirschey, M.D. et al. (2010) SIRT3 regulates mitochondrial<br />

fatty-acid oxidation by reversible enzyme<br />

deacetylation. Nature 464, 121–125.<br />

Schoeberl, B. et al. (2010) An ErbB3 antibody,<br />

MM-121, is active in cancers with ligand-dependent<br />

activation. Cancer Res. 70, 2485–2494.<br />

Ghosh, R. et al. (2010) Phellodendron amurense<br />

bark extract prevents progression of prostate tumors<br />

in transgenic adenocarcinoma of mouse prostate:<br />

potential for prostate cancer management. Anticancer<br />

Res. 30, 857–865.<br />

Kang, S. et al. (2010) p90 ribosomal S6 kinase 2<br />

promotes invasion and metastasis of human head and<br />

neck squamous cell carcinoma cells. J. Clin. Invest.<br />

120, 1165–1177.<br />

Ganapathy, V. et al. (2010) Targeting the Transforming<br />

Growth Factor-beta pathway inhibits human basal-like<br />

breast cancer metastasis. Mol. Cancer 9, 122.<br />

Takaku, S. et al. (2010) Blockade of TGF-beta<br />

enhances tumor vaccine efficacy mediated by CD8(+)<br />

T cells. Int. J. Cancer 126, 1666–1674.<br />

Bonnette, P.C. et al. (2010) Phosphoproteomic characterization<br />

of PYK2 signaling pathways involved in<br />

osteogenesis. J. Proteomics 73, 1306–1320.<br />

Aguiar, M. et al. (2010) Gas-Phase Rearrangements<br />

Do Not Affect Site Localization Reliability in<br />

Phosphoproteomics Data Sets. J. Proteome Res. 9,<br />

3103–3107.<br />

Carretero, J. et al. (2010) Integrative genomic and<br />

proteomic analyses identify targets for Lkb1-deficient<br />

metastatic lung tumors. Cancer Cell 17, 547–559.<br />

Andersen, J.N. et al. (2010) Pathway-based identification<br />

of biomarkers for targeted therapeutics: personalized<br />

oncology with PI3K pathway inhibitors. Sci. Transl.<br />

Med. 2, 43ra55.<br />

Buel, G.R. et al. (2010) Fyn promotes phosphorylation<br />

of collapsin response mediator protein 1 at tyrosine<br />

504, a novel, isoform-specific regulatory site. J. Cell<br />

Biochem. 111, 20–28.<br />

Knowlton, M.L. et al. (2010) Profiling Y561-dependent<br />

and -independent substrates of CSF-1R in epithelial<br />

cells. PLoS ONE 5, e13587.<br />

Edwards, J.R. et al. (2010) Inhibition of TGF-β<br />

signaling by 1D11 antibody treatment increases bone<br />

mass and quality in vivo. J. Bone Miner. Res. 25,<br />

2419–2426.<br />

chapter 30: publications by cst scientists<br />

www.cellsignal.com/cstpubs 291


Section IV: Additional Information<br />

chapter 30: publications by cst scientists<br />

Publications by<br />

<strong>CST</strong> Scientists<br />

Please visit our website to view<br />

publications prior to 2003.<br />

www.cellsignal.com/cstpubs<br />

Heo, J.M. et al. (2010) A stress-responsive system<br />

for mitochondrial protein degradation. Mol. Cell 40,<br />

465–480.<br />

Demir, E. et al. (2010) The BioPAX community<br />

standard for pathway data sharing. Nat. Biotechnol.<br />

28, 935–942.<br />

Bennett, E.J. et al. (2010) Dynamics of cullin-RING<br />

ubiquitin ligase network revealed by systematic quantitative<br />

proteomics. Cell 143, 951–965.<br />

2009<br />

Vitsky, A. et al. (2009) Homeostatic role of transforming<br />

growth factor-beta in the oral cavity and esophagus<br />

of mice and its expression by mast cells in these<br />

tissues. Am. J. Pathol. 174, 2137–2149.<br />

Terabe, M. et al. (2009) Synergistic enhancement<br />

of CD8+ T cell-mediated tumor vaccine efficacy by<br />

an anti-transforming growth factor-beta monoclonal<br />

antibody. Clin. Cancer Res. 15, 6560–6569.<br />

Ueda, R. et al. (2009) Systemic inhibition of transforming<br />

growth factor-beta in glioma-bearing mice<br />

improves the therapeutic efficacy of glioma-associated<br />

antigen peptide vaccines. Clin. Cancer Res. 15,<br />

6551–6559.<br />

Gordus, A. et al. (2009) Linear combinations of docking<br />

affinities explain quantitative differences in RTK<br />

signaling. Mol. Syst. Biol. 5, 235.<br />

Hitosugi, T. et al. (2009) Tyrosine phosphorylation inhibits<br />

PKM2 to promote the Warburg effect and tumor<br />

growth. Sci. Signal. 2, ra73.<br />

Harrington, M.G. et al. (2009) The morphology and<br />

biochemistry of nanostructures provide evidence for<br />

synthesis and signaling functions in human cerebrospinal<br />

fluid. Cerebrospinal Fluid Res. 6, 10.<br />

Pan, S. et al. (2009) Mass spectrometry based targeted<br />

protein quantification: methods and applications.<br />

J. Proteome Res. 8, 787–797.<br />

Boccalatte, F.E. et al. (2009) The enzymatic activity<br />

of 5-aminoimidazole-4-carboxamide ribonucleotide<br />

formyltransferase/IMP cyclohydrolase is enhanced by<br />

NPM-ALK: new insights in ALK-mediated pathogenesis<br />

and the treatment of ALCL. Blood 113, 2776–2790.<br />

Kang, S. et al. (2009) Fibroblast growth factor receptor<br />

3 associates with and tyrosine phosphorylates<br />

p90 RSK2, leading to RSK2 activation that mediates<br />

hematopoietic transformation. Mol. Cell. Biol. 29,<br />

2105–2117.<br />

ten Klooster, J.P. et al. (2009) Mst4 and Ezrin induce<br />

brush borders downstream of the Lkb1/Strad/Mo25<br />

polarization complex. Dev. Cell 16, 551–562.<br />

Xu, P. et al. (2009) Quantitative proteomics reveals<br />

the function of unconventional ubiquitin chains in<br />

proteasomal degradation. Cell 137, 133–145.<br />

Yu, J. et al. (2009) Mutation-specific antibodies for<br />

the detection of EGFR mutations in non-small-cell lung<br />

cancer. Clin. Cancer Res. 15, 3023–3028.<br />

Haack, H. et al. (2009) Diagnosis of NUT midline<br />

carcinoma using a NUT-specific monoclonal antibody.<br />

Am. J. Surg. Pathol. 33, 984–991.<br />

Yu, Y. et al. (2009) A site-specific, multiplexed kinase<br />

activity assay using stable-isotope dilution and highresolution<br />

mass spectrometry. Proc. Natl. Acad. Sci.<br />

USA 106, 11606–11611.<br />

292 For Research Use Only. Not For Use in Diagnostic Procedures.<br />

Kasyapa, C. et al. (2009) Phosphorylation of the<br />

SSBP2 and ABL proteins by the ZNF198-FGFR1 fusion<br />

kinase seen in atypical myeloproliferative disorders as<br />

revealed by phosphopeptide-specific MS. Proteomics<br />

9, 3979–3988.<br />

Schwer, B. et al. (2009) Calorie restriction alters mitochondrial<br />

protein acetylation. Aging Cell 8, 604–606.<br />

Kubota, K. et al. (2009) Sensitive multiplexed analysis<br />

of kinase activities and activity-based kinase identification.<br />

Nat. Biotechnol. 27, 933–940.<br />

Ji, H. et al. (2009) EGF-induced ERK activation promotes<br />

CK2-mediated disassociation of alpha-Catenin<br />

from beta-Catenin and transactivation of beta-Catenin.<br />

Mol. Cell 36, 547–559.<br />

Lechtreck, K.F. et al. (2009) The Chlamydomonas<br />

reinhardtii BBSome is an IFT cargo required for export<br />

of specific signaling proteins from flagella. J. Cell Biol.<br />

187, 1117–1132.<br />

2008<br />

Fattouh, R. et al. (2008) Transforming growth factorbeta<br />

regulates house dust mite-induced allergic airway<br />

inflammation but not airway remodeling. Am. J. Respir.<br />

Crit. Care Med. 177, 593–603.<br />

Perry, K. et al. (2008) Treatment of transforming<br />

growth factor-beta-insensitive mouse Renca tumor by<br />

transforming growth factor-beta elimination. Urology<br />

72, 225–229.<br />

Nam, J.S. et al. (2008) An anti-transforming growth<br />

factor beta antibody suppresses metastasis via cooperative<br />

effects on multiple cell compartments. Cancer<br />

Res. 68, 3835–3843.<br />

Nam, J.S. et al. (2008) Transforming growth factor<br />

beta subverts the immune system into directly promoting<br />

tumor growth through interleukin-17. Cancer Res.<br />

68, 3915–3923.<br />

Frantz, S. et al. (2008) Transforming growth factor<br />

beta inhibition increases mortality and left ventricular<br />

dilatation after myocardial infarction. Basic Res.<br />

Cardiol. 103, 485–492.<br />

Guo, A. et al. (2008) Signaling networks assembled by<br />

oncogenic EGFR and c-Met. Proc. Natl. Acad. Sci. USA<br />

105, 692–697.<br />

Pan, S. et al. (2008) Application of targeted quantitative<br />

proteomics analysis in human cerebrospinal fluid using<br />

a liquid chromatography matrix-assisted laser desorption/ionization<br />

time-of-flight tandem mass spectrometer<br />

(LC MALDI TOF/TOF) platform. J. Proteome Res. 7,<br />

720–730.<br />

Kang, S. et al. (2008) Epidermal growth factor<br />

stimulates RSK2 activation through activation of<br />

the MEK/ERK pathway and src-dependent tyrosine<br />

phosphorylation of RSK2 at Tyr-529. J. Biol. Chem.<br />

283, 4652–4657.<br />

Li, K. et al. (2008) Regulation of WRN protein cellular<br />

localization and enzymatic activities by SIRT1-mediated<br />

deacetylation. J. Biol. Chem. 283, 7590–7598.<br />

Dentin, R. et al. (2008) Hepatic glucose sensing<br />

via the CREB coactivator CRTC2. Science 319,<br />

1402–1405.<br />

Taylor, E.B. et al. (2008) Discovery of TBC1D1 as an<br />

insulin-, AICAR-, and contraction-stimulated signaling<br />

nexus in mouse skeletal muscle. J. Biol. Chem. 283,<br />

9787–9796.<br />

Miyake, Z. et al. (2007) Activation of MTK1/MEKK4<br />

by GADD45 through induced N-C dissociation and<br />

dimerization-mediated trans autophosphorylation<br />

of the MTK1 kinase domain. Mol. Cell. Biol. 27,<br />

2765–2776.<br />

Li, Y., et al. (2008) HDAC6 is required for epidermal<br />

growth factor-induced beta-catenin nuclear localization.<br />

J. Biol. Chem. 283, 12686–12690.<br />

Kaushansky, A. et al. (2008) A quantitative study of<br />

the recruitment potential of all intracellular tyrosine<br />

residues on EGFR, FGFR1 and IGF1R. Mol. Biosyst. 4,<br />

643–653.<br />

Kaushansky, A. et al. (2008) System-wide investigation<br />

of ErbB4 reveals 19 sites of Tyr phosphorylation that<br />

are unusually selective in their recruitment properties.<br />

Chem. Biol. 15, 808–817.<br />

Pan, W. et al. (2008) Wnt3a-mediated formation of<br />

phosphatidylinositol 4,5-bisphosphate regulates LRP6<br />

phosphorylation. Science 321, 1350–1353.<br />

Altarejos, J.Y. et al. (2008) The Creb1 coactivator<br />

Crtc1 is required for energy balance and fertility. Nat.<br />

Med. 14, 1112–1117.<br />

Stratford, A.L. et al. (2008) Y-box binding protein-1<br />

serine 102 is a downstream target of p90 ribosomal<br />

S6 kinase in basal-like breast cancer cells. Breast<br />

Cancer Res. 10, R99.<br />

Stokes, M.P. et al. (2007) Profiling of UV-induced ATM/<br />

ATR signaling pathways. Proc. Natl. Acad. Sci. USA<br />

104, 19855–19860.<br />

Engelman, J.A. et al. (2008) Effective use of PI3K<br />

and MEK inhibitors to treat mutant Kras G12D and<br />

PIK3CA H1047R murine lung cancers. Nat. Med. 14,<br />

1351–1356.<br />

2007<br />

Liu, V.C. et al. (2007) Tumor evasion of the immune<br />

system by converting CD4+CD25- T cells into<br />

CD4+CD25+ T regulatory cells: role of tumor-derived<br />

TGF-beta. J. Immunol. 178, 2883–2892.<br />

Nakanishi, H. et al. (2007) TGF-beta-neutralizing<br />

antibodies improve pulmonary alveologenesis and<br />

vasculogenesis in the injured newborn lung. Am. J.<br />

Physiol. Lung Cell Mol. Physiol. 293, 151–161.<br />

Gerber, S.A. et al. (2007) The absolute quantification<br />

strategy: application to phosphorylation profiling of<br />

human separase serine 1126. Meth. Mol. Biol. 359,<br />

71–86.<br />

Gu, T.L. et al. (2007) Phosphoproteomic analysis<br />

identifies the M0-91 cell line as a cellular model for<br />

the study of TEL-TRKC fusion-associated leukemia.<br />

Leukemia 21, 563–566.<br />

Li, X. et al. (2007) Akt/PKB regulates hepatic metabolism<br />

by directly inhibiting PGC-1alpha transcription<br />

coactivator. Nature 447, 1012–1016.<br />

Dong, S. et al. (2007) 14-3-3 Integrates prosurvival<br />

signals mediated by the AKT and MAPK pathways<br />

in ZNF198-FGFR1-transformed hematopoietic cells.<br />

Blood 110, 360–369.<br />

Gu, T.L. et al. (2007) A novel fusion of RBM6 to CSF1R<br />

in acute megakaryoblastic leukemia. Blood 110,<br />

323–333.<br />

Kang, S. et al. (2007) FGFR3 activates RSK2 to mediate<br />

hematopoietic transformation through tyrosine<br />

phosphorylation of RSK2 and activation of the MEK/ERK<br />

pathway. Cancer Cell 12, 201–214.<br />

Kass, E.M. et al. (2007) Stability of checkpoint kinase<br />

2 is regulated via phosphorylation at serine 456.<br />

J. Biol. Chem. 282, 30311–30321.<br />

Rikova, K. et al. (2007) Global survey of phosphotyrosine<br />

signaling identifies oncogenic kinases in lung<br />

cancer. Cell 131, 1190–1203.<br />

2006<br />

Nam, J.S. et al. (2006) Bone sialoprotein mediates the<br />

tumor cell-targeted prometastatic activity of transforming<br />

growth factor beta in a mouse model of breast<br />

cancer. Cancer Res. 66, 6327–6335.<br />

Steen, H. et al. (2006) Phosphorylation analysis by<br />

mass spectrometry: myths, facts, and the consequences<br />

for qualitative and quantitative measurements.<br />

Mol. Cell Proteomics 5, 172–181.<br />

Shalizi, A. et al. (2006) A calcium-regulated MEF2<br />

sumoylation switch controls postsynaptic differentiation.<br />

Science 311, 1012–1017.<br />

Kling, D.E. et al. (2006) Distribution of ERK1/2 and<br />

ERK3 during normal rat fetal lung development. Anat.<br />

Embryol. 211, 139–153.<br />

Goss, V.L. et al. (2006) A common phosphotyrosine<br />

signature for the Bcr-Abl kinase. Blood 107,<br />

4888–4897.<br />

Cheng, D. et al. (2006) Relative and absolute quantification<br />

of postsynaptic density proteome isolated from<br />

rat forebrain and cerebellum. Mol. Cell Proteomics 5,<br />

1158–1170.<br />

Kirkpatrick, D.S. et al. (2006) Quantitative analysis of<br />

in vitro ubiquitinated cyclin B1 reveals complex chain<br />

topology. Nat. Cell Biol. 8, 700–710.<br />

Walters, D.K. et al. (2006) Activating alleles of JAK3<br />

in acute megakaryoblastic leukemia. Cancer Cell 10,<br />

65–75.<br />

Griswold, I.J. et al. (2006) Kinase domain mutants of<br />

Bcr-Abl exhibit altered transformation potency, kinase<br />

activity, and substrate utilization, irrespective of sensitivity<br />

to imatinib. Mol. Cell. Biol. 26, 6082–6093.<br />

Walters, D.K. et al. (2006) Phosphoproteomic analysis<br />

of AML cell lines identifies leukemic oncogenes. Leuk.<br />

Res. 30, 1097–1104.<br />

Mercher, T. et al. (2006) JAK2T875N is a novel<br />

activating mutation that results in myeloproliferative<br />

disease with features of megakaryoblastic leukemia in<br />

a murine bone marrow transplantation model. Blood<br />

108, 2770–2779.<br />

Beausoleil, S.A. et al. (2006) A probability-based<br />

approach for high-throughput protein phosphorylation<br />

analysis and site localization. Nat. Biotechnol. 24,<br />

1285–1292.<br />

Gu, T.L. et al. (2006) Phosphotyrosine profiling identifies<br />

the KG-1 cell line as a model for the study of<br />

FGFR1 fusions in acute myeloid leukemia. Blood 108,<br />

4202–4204.<br />

www.cellsignal.com/cstpubs<br />

293


31<br />

Section IV: Additional Information<br />

Index<br />

by Pathways, Protocols, and targets<br />

Pathway Diagrams and Tables<br />

Adherens Junction Dynamics 110<br />

Akt Binding Partners 65<br />

Akt Substrates 55<br />

AMPK Signaling 146<br />

AMPK Substrates 147<br />

Amyloid Plaque and Neurofibrillary Tangle 190<br />

Formation in Alzheimer’s Disease<br />

Angiogenesis Signaling in<br />

171<br />

Tumor Neovascularization<br />

Autophagy Signaling 97<br />

B Cell Receptor Signaling 178<br />

Calcium, cAMP, and Lipid Signaling<br />

84<br />

Kinase-Disease Associations<br />

Cell Cycle Control: G1/S Checkpoint 104<br />

Cell Cycle Control: G2/M DNA Damage 105<br />

Checkpoint<br />

Cell Cycle/Checkpoint Control<br />

102<br />

Kinase-Disease Associations<br />

Death Receptor Signaling 92<br />

Deubiquitinase Table 135<br />

Dopamine Signaling in Parkinson’s Disease 191<br />

ErbB/HER Signaling 74<br />

ESC Pluripotency Differentiation 165<br />

Examples of Crosstalk Between<br />

31<br />

Post-Translational Modifications<br />

G-Protein Coupled Receptor<br />

79<br />

Signaling to MAP Kinase/Erk<br />

G-Protein Coupled Receptor Signaling: Overview 78<br />

Hedgehog Signaling 163<br />

Hippo Signaling 162<br />

Histone Lysine Methylation 27<br />

Histone Modifications Table 28<br />

Inhibition of Apoptosis 91<br />

Protocols and Troubleshooting <strong>Guide</strong>s<br />

ChIP General Protocol 196<br />

ChIP Troubleshooting <strong>Guide</strong> 200<br />

Cultured Cells (Immunocytochemistry, IF-IC) 210<br />

Protocol<br />

Flow Cytometry Alternate Protocol 204<br />

Flow Cytometry General Protocol 204<br />

Frozen/Cryostat Tissue Sections (IF-F) Protocol 211<br />

IHC Frozen Protocol<br />

217<br />

(using SignalStain ® Boost Detection Reagent)<br />

IHC Paraffin Protocol<br />

216<br />

(using SignalStain ® Boost Detection Reagent)<br />

IHC Troubleshooting <strong>Guide</strong> 219<br />

Insulin Receptor Signaling 144<br />

Jak and Cytokine Receptor Mutants 176<br />

Jak/Stat Signaling: IL-6 Receptor Family 181<br />

Jak/Stat Utilization 177<br />

MAPK/Erk in Growth and Differentiation 46<br />

Mitochondrial Control of Apoptosis 93<br />

mTOR Signaling 54<br />

NF-κB Signaling 182<br />

Notch Signaling 161<br />

Nuclear Receptors Signaling 41<br />

PI3 Kinase/Akt Signaling 53<br />

Protein Acetylation 26<br />

Regulation of Actin Dynamics 115<br />

Regulation of Apoptosis Overview 90<br />

Regulation of Microtubule Dynamics 116<br />

SAPK/JNK Signaling 48<br />

Signaling Pathways Activating p38 MAP Kinase 47<br />

T Cell Receptor Signaling 179<br />

TGF-β Signaling 164<br />

Toll-like Receptor Signaling 180<br />

Translational Control: Overview 36<br />

Translational Control: Regulation of elF2 38<br />

Translational Control: Regulation of elF4E 37<br />

and p70 S6K<br />

Tumor Immunology 183<br />

Tyrosine Kinases Kinase-Disease Associations 71<br />

Ubiquitin Ligases 125<br />

Ubiquitin/Proteasome 124<br />

Vesicle Trafficking in Presynaptic Neurons: 189<br />

Synchronous Release<br />

Warburg Effect 145<br />

Wnt/β-Catenin Signaling 160<br />

In-Cell Western Protocol 213<br />

IP Denatured Protein Protocol 237<br />

IP Native Protein Protocol 235<br />

Paraffin Tissue Sections (IF-P) Protocol 212<br />

PathScan ® Sandwich ELISA<br />

224<br />

Antibody Pair Protocol<br />

PathScan ® Sandwich ELISA<br />

223<br />

Chemiluminescent Protocol<br />

PathScan ® Sandwich ELISA Colorimetric Protocol 221<br />

WB Fluorescent Protocol 230<br />

WB General Protocol 228<br />

WB Troubleshooting <strong>Guide</strong> 232<br />

Targets<br />

14-3-3 113<br />

4E-BP 35<br />

4EHP 35<br />

4E-T 35<br />

4F2hc/CD98 142<br />

5-HTR1A 187<br />

5-HTR4 187<br />

53BP1 101<br />

A1/Bfl-1 88<br />

A20/TNFAIP3 174<br />

A2B5 187<br />

ABCC4 142<br />

ABCG2 142<br />

ABIN-1 174<br />

c-Abl 44<br />

ACAD 142<br />

ACAT2 142<br />

ACE2 119<br />

AceCS1 142<br />

Acetyl-CoA Carboxylase 142<br />

ACF1 24<br />

Acinus 88<br />

Ack1 113<br />

Actin 113<br />

ACO2 142<br />

ACPP 69<br />

ACSL1 142<br />

α-Actinin 113<br />

ACVR1 158<br />

ADAM 109<br />

ADAMTS1 109, 170<br />

ADAP 174<br />

ADAR1 35<br />

ADH1 142<br />

Adiponectin 142<br />

β-Adrenergic Receptor 77, 83<br />

ADRM1 123<br />

Afadin 109, 113<br />

AFP 158<br />

AGR2 158<br />

AhR 39<br />

AID 174<br />

AIF 88<br />

AIM2 174<br />

Aiolos 174<br />

Ajuba 109<br />

AKAP 77, 83<br />

AKR1C2 142<br />

Akt 51<br />

ALDH1A1 142<br />

Aldolase A 142<br />

Alix 88<br />

ALK 69<br />

ALPP 158<br />

AMACR 142<br />

Ambra1 96<br />

AMFR 123<br />

AML1 174<br />

AMPA Receptor 187<br />

AMPK 142<br />

Amylase 142<br />

β-Amyloid 187<br />

Androgen Receptor 39<br />

Angiopoietin-2 158, 170<br />

Annexin 83, 113<br />

ANT2/SLC25A5 142<br />

AP-2 88<br />

AP2M1 88, 113<br />

Apaf-1 88<br />

APBA2 187<br />

APC 100, 123, 158<br />

Ape1 100<br />

ApoA 83<br />

ApoE 187<br />

ApoM 83<br />

APP/β-Amyloid 187<br />

APPL1 119<br />

APS 44<br />

AQP2 142<br />

Arf6 119<br />

Arginase-1 142<br />

Argonaute 35<br />

ARID1A/BAF250A 24<br />

ARK5 142<br />

Aromatase 39<br />

ARP 113<br />

Arrestin 44, 77, 187<br />

Artemis 100<br />

AS160 142<br />

ASCT2 142<br />

ASF1 24<br />

ASH2L 24<br />

ASK1 44<br />

ASM 83<br />

Ataxin-1 187<br />

ATF 44, 142<br />

Atg 96<br />

ATGL 142<br />

Atlastin-1 113<br />

ATM 100<br />

ATP Citrate Lyase 142<br />

ATP2A/SERCA 83<br />

ATP6V1B1/2 51<br />

ATPIF1 142<br />

ATR 100<br />

ATRIP 100<br />

AUF1/hnRNP D 35<br />

Aurora 100<br />

Aven 88<br />

Axin 158<br />

Axl 69<br />

BACE 187<br />

BACH 100, 174<br />

Bad 88<br />

BAFF 174<br />

Bag1 119<br />

BAG6 119<br />

Bak1 88<br />

BAP1 123<br />

BAP31 88<br />

Basigin/EMMPRIN 174<br />

Bassoon 187<br />

BATF 174<br />

Bax 88<br />

BCAT 142<br />

Bcl-2 88<br />

BCL2L10 88<br />

BCL6 174<br />

Bcl10 174<br />

Bcl-11B 158<br />

Bcl-w 88<br />

Bcl-xL 88<br />

Bcr 44<br />

Bcr-Abl 44<br />

Beclin-1 96<br />

BID 88<br />

Bif-1 96<br />

Bik 88<br />

Bim 88<br />

BiP 119<br />

BIRC6 88<br />

Bit1 88<br />

Blimp-1/PRDI-BF1 174<br />

Blk 174<br />

BLM 100<br />

BLNK 174<br />

BMAL 187<br />

Bmf 88<br />

Bmi1 24<br />

BMP 158<br />

BMPR2 158<br />

BNIP3 96<br />

BNIP3L/Nix 96<br />

Bora 100<br />

BORIS 24<br />

Brachyury 158<br />

BRCA1 100<br />

BRD 24<br />

BrdU 100<br />

chapter 31: Index<br />

BRE 100<br />

BRF1/2 35<br />

Brg1 24<br />

BRM 24<br />

Brn2/POU3F2 187<br />

BRSK 187<br />

BSP II 109<br />

BTAF1 24<br />

Btk 174<br />

Bub 100<br />

C1QBP 142<br />

CA 142, 170<br />

CABIN1 24<br />

CACYBP 158<br />

CAD 142<br />

Cadherin 109, 170<br />

Calbindin 187<br />

Calcineurin A 83<br />

Caldesmon-1 113<br />

Calmodulin 83<br />

Calnexin 119<br />

Calpastatin 119<br />

Calumenin 83<br />

CaMKI 187<br />

CAND1 123<br />

CARD 174<br />

CASK 187<br />

Caspases 88<br />

Caspr2 187<br />

Catalase 142<br />

Catenin 109, 113, 158<br />

Cathepsin B 187<br />

Caveolin-1 113, 119<br />

CBARA1/MICU1 83<br />

CBFβ 174<br />

Cbl-b 123<br />

c-Cbl 123<br />

CBP 24, 170<br />

CCR2 174<br />

CCT2 119<br />

CCTα 142<br />

CD (Cell Surface<br />

Markers)<br />

109, 113, 170,<br />

174, 187<br />

CD2AP 113<br />

Cdc2 100<br />

Cdc6 100<br />

Cdc7 100<br />

CDC20 100<br />

cdc25 100<br />

CDC37 100, 113, 119<br />

Cdc42 113<br />

Cdc45 100<br />

CDC73 100<br />

294 For Research Use Only. Not For Use in Diagnostic Procedures.<br />

295


Section IV: Additional Information<br />

chapter 31: Index<br />

CDCP1 158<br />

CdGAP 113<br />

CDK 24, 100, 187<br />

CDT1 100<br />

CDX2 158<br />

CEA/CD66e 109<br />

CEACAM1 158<br />

C/EBP 142<br />

CEND1 187<br />

CENP-A 24<br />

CENP-T 100<br />

Centrin-2 113<br />

CFTR 83<br />

CHAF1A 24<br />

CHD 24<br />

CHFR 100<br />

β2-Chimerin 113<br />

CHIP 123<br />

Chk 100<br />

Choline Kinase 83<br />

CHOP 119<br />

Chronophin/PDXP 113<br />

CIITA 174<br />

CIN85 113<br />

CIRBP 187<br />

CISH 174<br />

CK1 100, 158, 187<br />

Claspin 100<br />

Clathrin Heavy Chain 119<br />

Claudin-1 109, 113<br />

CLCN3 187<br />

CLIC4 142<br />

CLIP1/CLIP170 113<br />

CLK3 35<br />

CLOCK 24<br />

CNOT 35<br />

CNPase 187<br />

Cofilin 113<br />

Coilin 35<br />

Complexin 187<br />

Connexin 43 109<br />

Cool/Pix 113<br />

COPS5 123<br />

Cortactin 113<br />

COUP-TF 39<br />

COX IV 142<br />

Cox 174<br />

CP110 100<br />

CPEB1 35<br />

C-Peptide 143<br />

cPLA2 83<br />

CPT1A 142<br />

CRABP1 142<br />

CRADD/RAIDD 88<br />

CREB 187<br />

Cripto 158, 170<br />

CrkII 113<br />

CrkL 174<br />

CRMP-2 187<br />

CRYAB 119<br />

Csk 44<br />

CtBP 158<br />

CTCF 24<br />

CTDSPL2 24<br />

CtIP 100<br />

CTMP 51<br />

CTR1/SLC31A1 142<br />

CTR9 24<br />

CUEDC2 100<br />

CUL 123<br />

CXXC1 24<br />

Cyclic AMP 83<br />

Cyclin 101<br />

Cyclin T1 24<br />

Cyclophilin A 174<br />

CYLD 123<br />

CYP3A4 142<br />

CYP11A1 142<br />

CYR61 109, 170<br />

Cytochrome c 88<br />

Cytokine Receptor 174<br />

Common β-Chain<br />

Dab 113, 187<br />

DAG Lipase 83, 187<br />

DAP 88<br />

DAP12 174<br />

DAPK1 88<br />

DAPK3/ZIPK 88<br />

DARPP-32 187<br />

DAX1 158<br />

Daxx 88<br />

DAZL 158<br />

DBC1 24<br />

DC-SIGN 174<br />

DCBLD2 187<br />

DCP1B 35<br />

Cleaved Drosophila 88<br />

Dcp-1 (Asp216)<br />

DcR 88<br />

DDB 123<br />

DDC 187<br />

DDR 69<br />

DDX4 35, 158<br />

Dectin-1 174<br />

Deltex-2 158<br />

DEPTOR/DEPDC6 51<br />

Derlin-1 119<br />

Desmin 113<br />

Dexras1 44<br />

DFF45/DFF35 88<br />

DGCR8 35<br />

DHCR24/Seladin-1 142<br />

DHX29 35<br />

Diap 113<br />

Dicer1 35<br />

DIDO1 89<br />

DJ-1 51, 187<br />

DKK 158<br />

DLK1 158<br />

DLL 158, 170<br />

DLST 142<br />

DMAP1 24<br />

DNA Polymerase 101<br />

DNA-PK 101<br />

DNAJC2/MPP11 119<br />

DNMT 24<br />

DOCK180 113<br />

Dok 44<br />

Dopamine ß-Hydroxylase<br />

187<br />

(DBH)<br />

Doublecortin 187<br />

DPYD 142<br />

DR 24, 89<br />

DRAK2 89<br />

Drebrin 187<br />

Drosha 35<br />

DRP1 113<br />

DSG2 113<br />

DUSP 44<br />

Dvl 158<br />

Dynamin 119<br />

DYRK1 101, 187<br />

Dysbindin 187<br />

E2-25K/Hip2 123<br />

E2A 174<br />

E2F-1 101<br />

EAAT 187<br />

EAPP 101<br />

EB-1 113<br />

EDC4/Ge-1 35<br />

EEA1 119<br />

eEF 35<br />

Eg5 101<br />

EGF Receptor 69<br />

EGR 187<br />

eIF 35<br />

eIF4G2/p97 35, 89<br />

ELAVL1/HuR 35<br />

Elk-1 44<br />

ELP 24<br />

Emerin 113<br />

EML4 113<br />

Endoglin 158<br />

Endonuclease G 89<br />

Enolase 142<br />

eNOS 51, 170<br />

ENPP1 142<br />

ENSA 101<br />

EOMES 158<br />

EPAC 113<br />

EpCAM 109, 113<br />

Eph 69, 170<br />

EPLIN 113<br />

Eps8 69<br />

Eps15 119<br />

ERC1 174<br />

ERCC1 101<br />

EREG 69<br />

Erk1/2 (p44/42 MAPK) 44<br />

Erk5 44<br />

Erlin 113<br />

Ero1-Lα 119<br />

ERp44 119<br />

ERp57 119<br />

ERp72 119<br />

ERRα 39<br />

ESET 24<br />

Estrogen Receptor 39<br />

Etk/BMX 69<br />

ETO 174<br />

Evi-1 174<br />

EVL 113<br />

EWS 24<br />

Exportin 5 35<br />

Ezh2 24<br />

Ezrin/Moesin/Radixin 113<br />

FAAH1 142<br />

FABP 142, 187<br />

FADD 89<br />

FAF1 89<br />

FAIM 89<br />

FAK 109<br />

FAM129B 44<br />

Fas 89<br />

Fas Ligand 89<br />

Fascin 113<br />

Fatty Acid Synthase 142<br />

FE65 187<br />

FEN-1 101<br />

Fer 113<br />

Fes 113<br />

Fetuin A 142<br />

Acidic FGF 158, 170<br />

Basic FGF 158<br />

FGF Receptor 69, 170<br />

Fgr 174<br />

FHIT 142<br />

Fibrillarin 113<br />

FIH 158, 170<br />

Filamin 113<br />

FIP200 96<br />

FKBP4 51, 119<br />

FLCN 51<br />

FLI1 158<br />

Flightless-I 158<br />

FLIP 89<br />

Flotillin 113<br />

FLT3 69<br />

FMRP 35<br />

α-Fodrin 89<br />

FosB 44<br />

c-Fos 44<br />

Delta FosB 187<br />

FoxA2/HNF3β 142<br />

FoxC 142, 158<br />

FoxD 158<br />

FoxK 158<br />

FOXM1 101<br />

FoxO 51<br />

FoxP 158, 174<br />

FRA1 44<br />

Fragilis 158<br />

Frizzled 158<br />

FTH1 142<br />

Fumarase 142<br />

FUS/TLS 35<br />

FXR 35<br />

Fyn 174<br />

FYVE-CENT 113<br />

G9a/EHMT2 24<br />

Gab 51<br />

GABA(B)R 77, 187<br />

GABARAP 96<br />

GABARAPL2 96<br />

GAD 187<br />

GADD45 α 101<br />

Galectin/LGALS 174<br />

GAP43 187<br />

GAPDH 142<br />

GATA 142, 158<br />

GCK 45<br />

GCN2 119<br />

GCN5L2 24<br />

GCNF/NR6A1 158<br />

GEF-H1 113<br />

Gelsolin 83, 113<br />

Geminin 101<br />

GFAP 187<br />

GFAT 142<br />

GFI1b 158<br />

GGA3 187<br />

GIMAP5 174<br />

GIT 109, 113<br />

GKAP 187<br />

GLDC 142<br />

GLI 158<br />

Glucagon 142<br />

Glucocorticoid Receptor 39<br />

Glucose-6-Phosphate 142<br />

Dehydrogenase<br />

Glut4 142<br />

Glutamate<br />

187<br />

Dehydrogenase 1/2<br />

Glycogen Synthase 142<br />

GM130 113, 119<br />

GNB3 77, 187<br />

Golgin-97 113<br />

GOPC 119<br />

GP130 174<br />

GPR50 187<br />

GPX1 142<br />

GRAF1 187<br />

Granzyme 89<br />

Grb2 45<br />

Grb10 45<br />

Gremlin 158, 170<br />

GRK2 187<br />

GRK6 174<br />

Grp75 119<br />

Grp94 119<br />

GSK-3 51<br />

GSTP1 187<br />

GUCY1A2 142<br />

Gα 77, 83<br />

GβL 51<br />

Hamartin/TSC1 52<br />

HAUSP 123<br />

HDAC 24<br />

HECTH9 123<br />

HEF1/NEDD9 113<br />

Helios 174<br />

HELLS 25<br />

HER/ErbB 69, 70<br />

Heregulin 158<br />

hERG1α 142<br />

HES1 158<br />

HEXIM1 25<br />

Hexokinase 142<br />

HGK 45<br />

HGS 51<br />

Hic-5 109<br />

HIF-1 158, 170<br />

HIF-2 158<br />

Hip 119<br />

HIPK2 89<br />

HIRA 25<br />

Histones 24<br />

HMG (High Mobility 25<br />

Group)<br />

HMOX2/HO-2 142<br />

HNF 142<br />

hnRNP 35<br />

HO-1 142, 170<br />

Homer1 187<br />

Hop 119<br />

HP1 25<br />

HPK1 174<br />

HR6A/HR6B 101<br />

HRS 119<br />

HS1 174<br />

HSF1 119<br />

HSL 142<br />

HSP 119<br />

HtrA2/Omi 89<br />

Huntingtin 187<br />

HYOU1 119<br />

c-IAP 88<br />

Cleaved Drosophila 89<br />

ICE (drICE) (Asp230)<br />

ID 158<br />

IDH 142<br />

IDO 175<br />

IFI16 174<br />

IFIT1 174<br />

IFITM 174<br />

IFN 174<br />

IGBP1 174<br />

IGF-I Receptor 142, 143<br />

IGF-II Receptor/CI-M6PR 119<br />

IGFBP 143<br />

βIG-H3 109<br />

Ikaros 174<br />

IκB 174<br />

IKK 174, 175<br />

IL (Interleukins) 175<br />

ILK1 109<br />

IMP1 35<br />

Importin β1 113<br />

INCENP 101<br />

iNOS 175<br />

INPP4b 83<br />

Insulin 143<br />

Insulin Receptor 143<br />

Integrin 109, 113, 170<br />

Interleukins 175<br />

INTS9 25<br />

IP3 Receptor 83<br />

IQGAP 113<br />

IRAK 175<br />

IRAP 143<br />

IRE1α 119<br />

IRF 175<br />

IRS 143<br />

ISG15 123<br />

ITCH 123<br />

Itk 175<br />

IWS1 35<br />

IκB 174, 175<br />

Jagged 158, 170<br />

Jak 175<br />

JARID 25<br />

JIP4/SPAG9 45<br />

JMJD 25<br />

JNK/SAPK 45<br />

Jun 45<br />

K48-linkage Specific 123<br />

Polyubiquitin<br />

K63-linkage Specific 123<br />

Polyubiquitin<br />

KEAP1 123<br />

Keratin 113<br />

KHSRP 35<br />

Ki-67 101<br />

KIBRA 158<br />

KIF3A 113, 158<br />

KIFC1 113<br />

Kinectin 1 113<br />

KISS1R 187<br />

c-Kit 70<br />

KLF4 158<br />

KLHL12 123<br />

KSR1 45<br />

Ku70 101<br />

Ku80 101<br />

La Antigen 35<br />

Lamin 89, 113<br />

LAMP1 113<br />

LAMTOR 51<br />

Langerin 175<br />

LAP2a 89<br />

LASP1 113<br />

LAT 175<br />

LAT1 143<br />

LATS 101<br />

LC3 96<br />

Lck 175<br />

LCMT1 25<br />

LCP1 113<br />

LDH-A 143<br />

LEDGF 25<br />

LEF1 158<br />

Lefty1 158<br />

LGP2 175<br />

LIMD1 158<br />

LIMK 113<br />

LIN28 158<br />

Lipin 1 143<br />

5-Lipoxygenase 175<br />

LIS1 187<br />

LITAF 175<br />

Livin 89<br />

296 For Research Use Only. Not For Use in Diagnostic Procedures.<br />

297


Section IV: Additional Information<br />

chapter 31: Index<br />

LKB1 143<br />

LLGL1 113<br />

LMAN1 113<br />

Lmx1B 158<br />

LPP 109<br />

LRF/Pokemon 175<br />

LRIG1 70<br />

LRP 158<br />

LRRK2 187<br />

LSD1 25<br />

LSm2 35<br />

Lsp1 175<br />

LXR-β 143<br />

Lyn 175<br />

Lyric/Metadherin 109<br />

LysRS 35<br />

M-CSF Receptor 70<br />

M-RIP 113<br />

MacroH2A1 24<br />

Mad-1 89<br />

MAD2L1 101<br />

MAG 187<br />

Malic Enzyme 143<br />

MALT1 175<br />

MAML 158<br />

Mannose Receptor 175<br />

MAP2 187<br />

MAP4K3 51<br />

MAPBPIP/ROBLD3/p14 35<br />

MAPKAPK 45<br />

MAPKSP1/MP1 35<br />

MARCKS 83<br />

MARK 113<br />

Maspin 89, 109, 170<br />

MASTL 101<br />

MAVS 175<br />

Max 89<br />

MBD3 25<br />

MBTPS2 119<br />

MCF2/Dbl 113<br />

Mcl-1 89<br />

MCM 101<br />

MCP-1 175<br />

MDA-5 175<br />

MDH2 143<br />

MDR1/ABCB1 143<br />

MeCP2 25<br />

MED 25<br />

MEF2 45<br />

MEIS1/2 175<br />

MEK 45<br />

MEKK3 45<br />

MELK 187<br />

Mena 187<br />

Menin 25<br />

MEP50 25<br />

Mer 70<br />

MERIT40 101<br />

Merlin 187<br />

MESD2 158<br />

Mesothelin 109<br />

Met 70<br />

MetAP2 35<br />

mGluR1 77, 187<br />

MGMT 101<br />

MIB1 123<br />

Mic-1 158<br />

Microcephalin-1/BRIT1 101<br />

β2-microglobulin 174<br />

Mig6 45<br />

Mili 35<br />

Mios 51<br />

MIS-R2 158<br />

MITF 158<br />

Mitofusin 143<br />

Miwi 35<br />

Miz-1 175<br />

MKK 45<br />

MKP 44<br />

MLH1 101<br />

MLK 45<br />

MLKL 89<br />

MLLT1/ENL 25<br />

MMP 109, 170<br />

MNDA 175<br />

Mnk1 35<br />

MO25α/CAB39 143<br />

MOB1 158<br />

Moesin 113<br />

MORF4L1/MRG15 25<br />

Mre11 101<br />

MRP/ABCC 143<br />

MRPL11 35<br />

MSH 101<br />

MSK 45<br />

Mst 89<br />

Msx1 158<br />

MTA1 25<br />

MTAP 143<br />

MTMR 96<br />

mTOR 51<br />

MTSS1 113<br />

MUC1 109<br />

Munc18-1 187<br />

Musashi 187<br />

c-Myb 158<br />

Myc 89<br />

MyD88 175<br />

Myelin Basic Protein 187<br />

Myeloperoxidase 175<br />

MYH 101<br />

MyoD1 158<br />

Myosin 113<br />

Myosin Light Chain 113, 114<br />

MYPT1 114<br />

Myt1 101<br />

Na Channel β1 Subunit 187<br />

Na,K-ATPase 114<br />

NAC1 158<br />

NAE1/APPBP1 123<br />

Naked 158<br />

NALP1 175<br />

Nanog 158<br />

NBR1 96<br />

NCAM (CD56) 109<br />

NCAPD3 101<br />

NCBP1/CBP80 35<br />

NCK1 114<br />

NCoR1 25<br />

NCS1 187<br />

NDP52 175<br />

NDRG 158, 170<br />

NEDD 123<br />

NEK7 101<br />

Nestin 187<br />

NeuN 187<br />

NeuroD 187<br />

Neurofilament 187<br />

Neurogenin 2 187<br />

Neuropeptide Y 187<br />

Neuropilin 170, 187<br />

NF-κB 175<br />

NFAT 175<br />

NFI-C 25<br />

NG2 187<br />

NGF 187<br />

NHERF 187<br />

Nicastrin 187<br />

NIK 175<br />

NIPA 101<br />

NIPSNAP1 83<br />

NKCC1 187<br />

NKX2 158<br />

NLRC4 175<br />

NLRP3 175<br />

NLRX1 175<br />

NMDAR 187, 188<br />

NME1/NDKA 143<br />

Nna1 188<br />

Nod1 175<br />

Nogo-A 188<br />

NOS 175, 188<br />

Notch 158, 170<br />

NPC1L1 143<br />

NPL4 123<br />

NPM 101<br />

NQO1 143<br />

NRBF-2 39<br />

NRF1 25, 35, 143<br />

NSF 119<br />

nSMase1 83<br />

NT5E/CD73 170, 188<br />

NTAL/LAB 175<br />

NTF2 114<br />

Nucleolin 25<br />

Nucleomethylin 25<br />

Nucleostemin 158<br />

NuMA 101<br />

Numb 158<br />

NUP88 114<br />

NUP98 114<br />

Nur77 39<br />

NUT1 25<br />

NXF1 35<br />

O-GlcNAc 143<br />

OCRL1 119<br />

OGDH 143<br />

OGT 143<br />

Oligophrenin-1 188<br />

Opioid Receptor 77, 188<br />

ORC 101<br />

OS-9 119<br />

OSR1 114<br />

OTUB1 123<br />

OTULIN 123<br />

OTX2 158<br />

p115 RhoGEF 114<br />

p14 ARF 101<br />

p18 INK4C 101<br />

p190 RhoGAP 114<br />

p21 Waf1/Cip1 101<br />

p27 Kip1 101<br />

p35/25 188<br />

p38 MAPK 45<br />

p39 188<br />

p44/42 MAPK (Erk1/2) 44<br />

p47phox 175<br />

p48 Primase 101<br />

p53 101<br />

p57 Kip2 101<br />

p58 Primase 101<br />

p58IPK 119<br />

p63-α 101<br />

p67phox 175<br />

p70 S6 Kinase 51<br />

p73 101<br />

p75NTR 188<br />

p90RSK1 45<br />

p95/NBS1 101<br />

PA28 123<br />

PABP 35<br />

PACT 35<br />

PAF1 25<br />

Paip2A 35<br />

PAK 114<br />

PANK 143<br />

PAR-4 89<br />

PAR2 114<br />

PARK9 188<br />

Parkin 188<br />

α-Parvin 109<br />

PARN 35<br />

PARP 89<br />

PASK 143<br />

PAX 158<br />

Paxillin 109<br />

PBK/TOPK 101<br />

Pbx1 175<br />

PC1/3 188<br />

PC2 188<br />

PCAF 25<br />

PCK 143<br />

PCM-1 114<br />

PCNA 101<br />

PCTAIRE 1 101<br />

PD-L1 175<br />

PDAP1 158<br />

PDCD4 89<br />

PDE5 83<br />

PDGF Receptor 70, 170<br />

PDHK1 143<br />

PDI 119<br />

PDK1 51<br />

PDLIM2 114<br />

Pdx1 143<br />

PEA-15 89<br />

PELO 101<br />

PEN2 188<br />

Perforin 89<br />

Perilipin 143<br />

PERK 35, 119<br />

PFKFB 143<br />

PFKL 143<br />

PFKP 143<br />

PGAM1 143<br />

PGC1α 143<br />

PGD 143<br />

PGRMC1 143<br />

PHB1 39, 101<br />

PHC1 25<br />

PHD-2/Egln1 158, 170<br />

PHF 25<br />

PHGDH 143<br />

PHLDA3 89<br />

Phospholamban 83<br />

PI3 Kinase 51, 52<br />

PI3 Kinase Class II 52<br />

PI3 Kinase Class III 52<br />

PI4 Kinase 52<br />

PIAS 175<br />

PICH 101<br />

Pim 175<br />

Pin1 101<br />

PINK1 188<br />

PIP4K2 83<br />

PIP5K1 83<br />

Pirin 175<br />

PiT1/SLC20A1 143<br />

PITSLRE/CDK11 100<br />

PKA 83<br />

PKC 83<br />

PKD/PKCµ 83<br />

PKG-1 114<br />

PKM 143<br />

PKR 35, 119<br />

PLA2G1B 143<br />

PLC 83<br />

PLD 83<br />

Plectin-1 114<br />

Plexin A 188<br />

PLK 101<br />

PNK 101<br />

PNUTS 52<br />

Podoplanin 114<br />

POLR3A 25<br />

Pontin/RUVBL1 25<br />

PP1α 102<br />

PP2A 102<br />

PP2C 45<br />

PP5 102<br />

PPARγ 39<br />

PPIG 35<br />

PPP1CB 102<br />

PPP2R 102<br />

PRAS40 52<br />

PRC1 114<br />

Prdx1 143<br />

Presenilin 188<br />

PREX1 114<br />

PRK2 83<br />

PRMT 25<br />

Profilin-1 114<br />

Progesterone Receptor 39<br />

Prolactin Receptor 175<br />

Prostate Specific<br />

119<br />

Membrane Antigen<br />

Protor2 52<br />

PRP4K 35<br />

PSA/KLK3 109<br />

PSD93 188<br />

PSD95 188<br />

PSMA 123<br />

PSMB 123<br />

PSMC 123<br />

PSMD 123<br />

PTBP1 35<br />

PTCH 158<br />

PTEN 52<br />

PTK7 70<br />

PTP1B 143<br />

PTP4A3 114<br />

PTPA/PPP2R4 102<br />

PTPN14 158<br />

PTPN18 158<br />

PTPN22 175<br />

PU.1 175<br />

Puma 89<br />

Pumilio 35<br />

PVR/CD155 114<br />

Pyk2 45<br />

Pyruvate Dehydrogenase 143<br />

Rab 119<br />

Rabex-5 119<br />

Rac1/Cdc42 114<br />

Rac1/Rac2/Rac3 114<br />

RACK1 114<br />

Rad 102<br />

RAD21 25, 102<br />

Rad23B 123<br />

Radixin 114<br />

Raf 45<br />

RAG1 175<br />

Rag 52<br />

RAGE 175<br />

RAIG1 77, 158<br />

Ral 114<br />

RalBP1 114<br />

Ran 114<br />

RanBP1 114<br />

RANK 175<br />

RANK Ligand 175<br />

RANTES 175<br />

Rap1 114<br />

Raptor 52<br />

RAR 39<br />

Ras 45, 114, 158<br />

Ras-GRF1 188<br />

RasGRP3 114<br />

Rb 102<br />

Rb-like 1 102<br />

RBAP46 25<br />

RBBP5 25<br />

RBPSUH 158<br />

RBX1 119, 123<br />

RCAS1 119<br />

RCC 102, 114<br />

RCHY1 123<br />

RECK 109, 170<br />

RecQ4 102<br />

RecQL 102<br />

REDD1 52<br />

RelB 175<br />

Renin 109<br />

REPS1 119<br />

Reptin/RuvBL2 25<br />

Ret 70<br />

Rev-Erba 39<br />

RGS4 77, 188<br />

Rheb 52<br />

Rho 114<br />

Rhodopsin 188<br />

Ribosomal Protein 35<br />

Rictor 52<br />

Rig-I 175<br />

RING1 25<br />

RIP 89, 175<br />

RKIP 45<br />

RMP 35<br />

RNF 25<br />

ROCK 114<br />

Ron 70, 170<br />

ROR 70<br />

ROS1 70<br />

RPA 102<br />

Rpb1 CTD 25, 102<br />

RPL11 35<br />

RRM1 102<br />

RSK 45<br />

Rubicon 96<br />

RUNX 158<br />

RXR 39<br />

RyR1 83<br />

S5a/PSMD4 123<br />

S6 Ribosomal Protein 35<br />

S100 83<br />

Sall4 158<br />

SAM68 35<br />

SAMHD1 175<br />

SAP102 188<br />

Sara 158<br />

SARM1 175<br />

SATB1 25<br />

SAV1 158<br />

SCAI 114<br />

SCAP 143<br />

SCD1 143<br />

SCF 158<br />

Scribble 158<br />

SDF1 175<br />

SDHA 143<br />

298 For Research Use Only. Not For Use in Diagnostic Procedures.<br />

299


Section IV: Additional Information<br />

chapter 31: Index<br />

Sec23 114<br />

Sec24 114<br />

Sec31 114<br />

Secretagogin 188<br />

Securin 102<br />

SEK1/MKK4 45<br />

Semaphorin 188<br />

SENP 123<br />

Sestrin-2 102<br />

SET 25<br />

SF2/ASF 35<br />

SF3B1 35<br />

SFRP1 158<br />

SGK 52<br />

SGLT1 143<br />

SGTA 119<br />

SH2D1A 175<br />

SHANK2 188<br />

Sharpin 123<br />

Shc 45<br />

Shh/Ihh 158, 159<br />

SHIP 45, 175<br />

SHMT 143<br />

Shootin1 188<br />

SHP 45, 175<br />

SIK2 143<br />

Sin1 52<br />

SIN3A 25<br />

SINTBAD 175<br />

SirT 25<br />

Siva-1 89<br />

SIX1 159<br />

SKAR 35<br />

Skp 123<br />

SLBP 102<br />

SLC1A4 188<br />

SLC4A4/NBC1 143<br />

SLC7A11/xCT 143<br />

SLP76 175<br />

Slug 159<br />

Smac/Diablo 89<br />

Smad 159<br />

SMARCA 25<br />

SMARCC 25<br />

SMC 102<br />

SMG-1 102<br />

SMN1 35<br />

Smurf 159<br />

SMYD 25<br />

Snail 159<br />

SNAP25 188<br />

SNARK/NUAK2 143<br />

SNF2H 25<br />

SNF5 25<br />

SNIP/p140Cap 119<br />

SnoN 159<br />

SOCS 175<br />

SOD 143, 188<br />

SOS1 45<br />

Sox 159<br />

SP1 25<br />

SPAK 114<br />

SPARC 159<br />

Spartin 102<br />

SPHK1 83<br />

SPINK3 123<br />

Spinophilin 188<br />

Spry1 70, 170<br />

SPT 25<br />

SQSTM1/p62 96<br />

Src 45<br />

SRC 25<br />

SREBP-1c 143<br />

SRF 45<br />

SSEA 159<br />

SSH1 114<br />

SSRP1 25<br />

SSTR1 188<br />

SSU72 25<br />

STAG2 102<br />

STAM1 119<br />

STAMBP 123<br />

StAR 143<br />

Stargazin 188<br />

Stat 175, 176<br />

Stathmin 114<br />

STEP 188<br />

STF-1 39<br />

STIM 83<br />

STING 176<br />

STOP 187<br />

Succinyl-CoA Synthetase 143<br />

SUFU 159<br />

SUMO 123<br />

Survivin 89<br />

SUV39H1 25<br />

SUZ12 25<br />

Syk 176<br />

Symplekin 35<br />

Synapsin 188<br />

Synaptophysin 188<br />

Synaptotagmin 188<br />

SynGAP 188<br />

Synip 143<br />

Syntaxin 119, 188<br />

α-Synuclein 188<br />

SYVN1 123<br />

T-bet/TBX21 (V365) 176<br />

TAB 45<br />

TACC3 102<br />

TACE 159<br />

TAF 25<br />

TAK1 45<br />

TAL1 176<br />

Talin-1 109, 114<br />

TANK 89<br />

TAP 176<br />

Tau 188<br />

TAX1BP1 89<br />

TAZ 159<br />

TBC1D1 143<br />

TBK1/NAK 176<br />

TBP 25<br />

TCEB3/Elongin A 25<br />

TCF 159<br />

TCL1 52<br />

TCTP 102, 114<br />

TDP43 188<br />

TEAD 159<br />

Tec 176<br />

TECPR1 96<br />

Tenascin C 170, 188<br />

Tensin 2 114<br />

TERF2IP 102<br />

TESK1 114<br />

TFAM 188<br />

TFEB 35<br />

TFF1/pS2 159<br />

TFII 25<br />

Pro-TGF-α 159<br />

TGF-β 159<br />

TGF-β Receptor 159, 170<br />

TGM2 83<br />

TH1L 25<br />

Thap11/Ronin 159<br />

THEMIS 176<br />

THEX1 35<br />

Thioredoxin 143<br />

THOC4/ALY 35<br />

ThPOK 176<br />

Thy1 188<br />

Thymidine Kinase 1 143<br />

Thymidylate Synthase 143<br />

Thyroid Transcription 159<br />

Factor 1 (TTF-1)<br />

TIAR 35<br />

Tid-1 119<br />

Tie2 70, 170<br />

TIF1β 102<br />

TIMP1 109, 170<br />

Tip60 25<br />

TIRAP 176<br />

TLE1/2/3/4 159<br />

TLK1 102<br />

TMEM49/VMP1 96<br />

TMP21 188<br />

TMS1 89<br />

TNF-α 176<br />

TNF-R 89, 176<br />

Tnk1 45<br />

Toll-like Receptor 176<br />

Tollip 176<br />

Topoisomerase IIα 25<br />

TORC/CRTC 143<br />

Torsin A 188<br />

TP/ECGF1 89<br />

TPH-1 188<br />

TPOR 176<br />

TPX2 102<br />

TRA-1-60 159<br />

TRA-1-81 159<br />

TRA-2-54 159<br />

TRADD 89<br />

TRAF 89<br />

TRAIL 89<br />

Transketolase 143<br />

TRAP1/HSP75 119<br />

β-Trcp 123<br />

TREX1 176<br />

Trf 102<br />

TRIAD1 123<br />

TRIB2 159<br />

TRIF 176<br />

TRIM 25, 123, 159<br />

Trk 188<br />

Tropomyosin 114<br />

Troponin 114<br />

TRPV3 83<br />

TRRAP 25<br />

TRXR 143<br />

TSPO 83<br />

TTK 102<br />

TTF1 159<br />

Tuberin/TSC2 52<br />

Tubulin 114, 188<br />

Tug 143<br />

TWEAK 176<br />

TWEAK Receptor/Fn14 176<br />

Twinfilin-1 114<br />

Tyk2 176<br />

Tyro3 70<br />

Tyrosinase 143<br />

Tyrosine Hydroxylase 188<br />

U2AF1 35<br />

UBA2 123<br />

Ubc 123<br />

UBE 123<br />

Ubiquitin 123<br />

K48-linkage Specific 123<br />

Polyubiquitin<br />

K63-linkage Specific 123<br />

Polyubiquitin<br />

Ubiquityl-PCNA (Lys164) 101<br />

UBLE1A/SAE1 123<br />

UBR5 123<br />

UCHL 123<br />

UGT 143<br />

UHRF1 25<br />

ULK1 96<br />

UNC5B 188<br />

uPAR 109, 170<br />

Upf 35<br />

USP 123<br />

UTF1 159<br />

UVRAG 96<br />

VAMP 119, 188<br />

VASP 114<br />

Vav 114<br />

VCAM1 176<br />

VCP 102, 123<br />

VDAC 89<br />

VEGF 159<br />

VEGF Receptor 70, 170<br />

VGLUT 188<br />

VHL 123, 170<br />

Villin-1 114<br />

Vimentin 114<br />

Vinculin 109<br />

Vitamin D3 Receptor 39<br />

VMS1 143<br />

VPRBP 123<br />

VRK 102<br />

Vti1a 188<br />

WASP 114<br />

WAVE 114<br />

Trademarks, Terms and Conditions<br />

WBP2 159<br />

WDR5 25<br />

Wee1 102<br />

WFS1 83<br />

WIF1 159<br />

WIP1 96, 102<br />

WIPI 96<br />

WNK 52<br />

Wnt 159<br />

WRN 102<br />

WSTF 25<br />

WTX 159<br />

WWOX 89<br />

XAF1 89<br />

XBP-1s 35<br />

XIAP 89<br />

XLF 102<br />

XPB 25, 102<br />

XPC 102<br />

XPD 25, 102<br />

XPF 102<br />

XRCC1 102<br />

XRN2 35<br />

YAP 52, 159<br />

YB1 52<br />

Yes 176<br />

YY1 25<br />

ZAP70 176<br />

ZFX 159<br />

ZO 109, 114<br />

ZPR1 35<br />

Zyxin 109<br />

The following are trademarks of Cell Signaling Technology, Inc.:<br />

AcetylScan, AcetylSignature, eXceptional Monoclonal Technology, eXceptional Performance, Cell Signaling Technology logo, Cell Signaling Technology, <strong>CST</strong>,<br />

<strong>CST</strong> BIOSCIENCES, <strong>CST</strong> LIFESCIENCES, D4D6, DF53, E1L3N, HTScan, Kinomepath, KinomeView, MethylScan, NG-XMT, PathScan, PhosphoPlus, PhosphoScan,<br />

PhosphoSignature, PhosphoSite logo, PhosphoSite, PhosphoSitePlus, Phosphotrol, Phototope, PTMScan, SignalFire, SignalKine, SignalSilence, SignalSlide,<br />

SignalStain, SimpleChIP, StemLight, UbiScan, XMT, XP<br />

The following are registered trademarks and trademarks are the property of their respective owners:<br />

Acumen is a registered trademark of TTP Labtech. • Aerius is a registered trademark of LI-COR Biosciences. • Alexa Fluor is a registered trademark of Life<br />

Technologies Corporation. • AlphaScreen is a registered trademark of PerkinElmer, Inc. • BackDrop is a registered trademark of Molecular Probes, Inc. • Bio-Plex<br />

is a registered trademark of Bio-Rad Laboratories. • BODIPY is a registered trademark of Molecular Probes, Inc. • DELFIA is a registered trademark of PerkinElmer,<br />

Inc. • DRAQ5 is a registered trademark of Biostatus Limited. • DRAQ7 is a trademark of Biostatus Limited. • DyLight is a trademark of Thermo Fisher Scientific<br />

Inc. and its subsidiaries. • EnVision is a trademark of Dako. • ER-Tracker is a trademark of Molecular Probes, Inc. • Gallios is a trademark of Beckman Coulter, Inc.<br />

• HTRF is a registered trademark of CISbio international. • iBlot is a trademark of Invitrogen. • In-Cell Western is a trademark of LI-COR Biosciences. • LANCE is<br />

a registered trademark of PerkinElmer, Inc. • LanthaScreen is a trademark of Invitrogen Corporation. • LI-COR is a registered trademark of LI-COR Biosciences. •<br />

LumiGLO is a registered trademark of Kirkegaard & Perry Laboratories. • Luminex is a registered trademark of Luminex Corporation. • LysoTracker is a registered<br />

trademark of Molecular Probes, Inc. • MitoTracker is a registered trademark of Life Technologies Corporation. • MULTI-ARRAY is a registered trademark of Meso<br />

Scale Discovery. • New England Biolabs is a registered trademark of New England Biolabs, Inc. • NovaRED is a trademark of Vector Laboratories. • Odyssey is<br />

a registered trademark of LI-COR Biosciences. • Pacific Blue is a trademark of Molecular Probes, Inc. • ProLong is a registered trademark of Life Technologies<br />

Corporation. • Q-View Plus, Q-View, Quansys Biosciences logo are registered trademarks of Quansys Biosciences. • SenseAware is a servicemark of FedEx. •<br />

Sepharose is a registered trademark of GE Healthcare. • SNAPi.d. is a trademark of Millipore. • Stratalinker is a registered trademark of Stratagene. • SYBR Green<br />

is a registered trademark of Molecular Probes, Inc. • Triton is a trademark of The Dow Chemical Company or an affiliated company of Dow. • Trizma is a registered<br />

trademark of Sigma-Aldrich Co. LLC. • Tween is a registered trademark of ICI Americas, Inc. • UniProt is a trademark of European Molecular Biology Laboratory.<br />

Please visit www.cellsignal.com/tm for a current listing of applicable trademark information.<br />

Use of <strong>CST</strong> Motif Antibodies within certain methods (e.g., U.S. Patent No.’s 7,198,896 and 7,300,753) may require a license from <strong>CST</strong>. For information regarding<br />

academic licensing terms please have your technology transfer office contact <strong>CST</strong> Legal Department at <strong>CST</strong>_ip@cellsignal.com. For information regarding commercial<br />

licensing terms please contact <strong>CST</strong> Pharma Services at marketing@cellsignal.com.<br />

The use of reagents from Cell Signaling Technology in conjunction with the technologies identified throughout this guide may require a license to third party<br />

intellectual property rights.<br />

Alexa Fluor dye antibody conjugates are sold under license from Life Technologies Corporation, for research use only for immunocytometry, immunohistochemistry,<br />

high content screening (HCS) analysis, or flow cytometry applications, and excluding use in combination with DNA microarrays and high content screening (HCS).<br />

300 For Research Use Only. Not For Use in Diagnostic Procedures.<br />

301


32<br />

Section IV: Additional Information<br />

Diagram & Table Keys<br />

and additional resources<br />

Applications Key<br />

While all of our antibodies are rigorously tested in a number of relevant applications, some products are more suitable<br />

for a specific application. This information is summarized in various lists and tables found throughout this guide.<br />

chapter 32: Diagram & Table Keys<br />

Pathway Diagram Key<br />

The pathway diagrams found in this guide and on our website have been assembled by <strong>CST</strong> scientists and outside<br />

experts to provide succinct and current overviews of selected signaling pathways.<br />

Direct Stimulatory Modification<br />

Direct Inhibitory Modification<br />

Deacetylase<br />

Ribosomal subunit<br />

WB Western Blotting<br />

IP Immunoprecipitation<br />

IHC Immunohistochemistry<br />

IF Immunofluorescence<br />

F Flow Cytometry<br />

ChIP Chromatin Immunoprecipitation<br />

-IC Immunocytochemistry<br />

-P Paraffin<br />

-F Frozen<br />

E-P Peptide ELISA<br />

Multistep Stimulatory Modification<br />

Multistep Inhibitory Modification<br />

Tentative Stimulatory Modification<br />

Tentative Inhibitory Modification<br />

Separation of Subunits<br />

or Cleavage Products<br />

Joining of Subunits<br />

Translocation<br />

Transcriptional Stimulatory Modification<br />

Transcriptional Inhibitory Modification<br />

Kinase<br />

Phosphatase<br />

Transcription Factor<br />

Caspase<br />

Receptor<br />

Enzyme<br />

pro-apoptotic<br />

pro-survival<br />

GAP/GEF<br />

GTPase<br />

G-protein<br />

Acetylase<br />

TIM-3<br />

Galectin-9<br />

B7-H3<br />

B7-H4<br />

CTLA-4<br />

CD80, 86<br />

PD-1<br />

PD-L1<br />

TCR<br />

MHC<br />

ICOS<br />

ICOSL<br />

OX40<br />

OX40L<br />

CD40<br />

CD40L<br />

CD27<br />

CD70<br />

CD137<br />

CD137L<br />

CD28<br />

Reactivity Key<br />

H human<br />

M mouse<br />

R rat<br />

Hm hamster<br />

Mk monkey<br />

C chicken<br />

Mi mink<br />

Dm D. melanogaster<br />

X Xenopus<br />

Z zebra fish<br />

Amino Acid Properties Table<br />

B bovine<br />

Dg dog<br />

Pg pig<br />

Sc S. cerevisiae<br />

Name<br />

Single<br />

Abbreviation letter code MW pKa Codons<br />

Side chain<br />

characteristics Modifications<br />

Glycine Gly G 57.05 na G-G-(UCAG) Nonpolar<br />

Alanine Ala A 71.09 na G-C-(UCAG) Nonpolar<br />

Valine Val V 99.14 na G-U-(UCAG) Nonpolar<br />

Leucine Leu L 113.16 na C-U-(UCAG), U-U-(AG) Nonpolar<br />

Isoleucine Ile I 113.16 na A-U-(UCA) Nonpolar<br />

Phenylalanine Phe F 147.18 U-U-(UC) Nonpolar<br />

Proline Pro P 97.12 na C-C-(UCAG) Nonpolar Racemization<br />

Serine Ser S 87.08 16 A-G-(UC), U-C-(UCAG) Hydroxyl group Phospho-, Glycosyl-,<br />

Eliminyl-<br />

Threonine Thr T 101.11 7.6 A-C-(UCAG) Hydroxyl group Phospho-, Glycosyl-,<br />

Eliminyl-<br />

Tyrosine Tyr Y 163.18 na U-A-(UC) Hydroxyl group Adenyl-, Phospho-,<br />

Sulfo-<br />

Cysteine Cys C 103.15 8.35 U-G-(UC) Contains sulfur Eliminyl-<br />

Methionine Met M 131.19 na A-U-G Contains sulfur<br />

Asparagine Asn N 114.11 na A-A-(UC) Contains nonbasic<br />

nitrogen<br />

Glutamine Gln Q 128.14 na C-A-(AG) Contains nonbasic<br />

nitrogen<br />

Tryptophan Trp W 186.21 na U-G-G Contains nonbasic<br />

nitrogen<br />

Aspartic Acid Asp D 115.09 3.9 G-A-(UC) Acidic<br />

Glutamic Acid Glu E 129.12 4.07 G-A-(AG) Acidic<br />

All all species<br />

expected<br />

( ) 100% sequence<br />

homology<br />

Glycosyl-, Deamidation<br />

Deamidation<br />

Lysine Lys K 128.17 10.79 A-A-(AG) Basic Acetyl-, Adenyl-,<br />

Carbamyl-, Methyl-<br />

Ubiquitin, SUMO, ISG,<br />

Neddy, UFM1<br />

Arginine Arg R 156.19 12.48 A-G-(AG), C-G-(UCAG) Basic Methyl-, Citrulline<br />

Histidine His H 137.14 8.04 C-A-(UC) Basic Adenyl-, Phospho-<br />

302 For Research Use Only. Not For Use in Diagnostic Procedures.<br />

303


33<br />

Section IV: Additional Information<br />

Contact Information<br />

Global Headquarters<br />

3 Trask Lane, Danvers, MA 01923 USA<br />

Phone: 978-867-2300 | Fax: 978-867-2400 | www.cellsignal.com<br />

Stay Informed!<br />

Request monthly updates on the latest <strong>CST</strong> products and scientific resources. www.cellsignal.com/subscribe<br />

Ordering and Customer Service<br />

Phone: 877-616-2355 [US & Canada toll free] or 978-867-2388<br />

Email: orders@cellsignal.com | Fax: 978-867-2488 | Order Online: www.cellsignal.com<br />

Find Your Local Distributor: www.cellsignl.com/contactus<br />

Technical Support<br />

At <strong>CST</strong>, the same Product Scientists who produce and validate our products are available to help you with your<br />

research needs, openly sharing their expertise and data.<br />

Online support request form: www.cellsignal.com/support<br />

Phone: 877-678-8324 [toll-free in US, Canada] or 978-867-2388 | Fax: 978-867-2400<br />

Request a custom formulation visit: www.cellsignal.com/custom<br />

Product scientists are available 9:00 am – 6:00 pm (EST), Monday–Friday<br />

304 For Research Use Only. Not For Use in Diagnostic Procedures.<br />

305


“…the time has come for us... to puzzle out,<br />

one protein at a time, how signals are really<br />

processed inside cells to create the marvelously<br />

functioning apparatus – the eukaryotic cell.”<br />

Dr. Robert A. Weinberg<br />

Daniel K. Ludwig Professor for Cancer Research at MIT<br />

GDE2015ENG_00<br />

www.cellsignal.com<br />

Printed in the USA on 100% recycled paper (30% post-consumer) waste fiber. Cover Image: Vesicle Trafficking Cellular Landscape, see pg for more information 248.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!