28.12.2014 Views

summary - Netherlands Proteomics Centre

summary - Netherlands Proteomics Centre

summary - Netherlands Proteomics Centre

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

netherlands<br />

centre<br />

npc highlights 11 | April 2010<br />

Featuring cutting edge research projects<br />

and enabling technologies of the<br />

<strong>Netherlands</strong> <strong>Proteomics</strong> <strong>Centre</strong>


2<br />

3<br />

4<br />

6<br />

26<br />

28<br />

30<br />

31<br />

34<br />

36<br />

Frontpage<br />

DeltaVision real-time imaging. Shown is a specialized tissue<br />

culture slide mounted on a nanometer-accuracy motorized<br />

stage of a DeltaVision RT microscope. Cells expressing various<br />

fluorescent markers are imaged live at high resolution while<br />

undergoing cell division. Example images are displayed in the<br />

background.<br />

Contents<br />

About<br />

Preface<br />

News headlines<br />

HighLights<br />

6 Geert Kops; Dividing the goods: preventing chromosome segregation errors<br />

10 Rumyana Karlova and Sacco de Vries; The plasma membrane receptor complex perceiving plant steroids<br />

14 Paul Boersema, Vanessa Ding and Albert Heck; In depth profiling of tyrosine phosphorylation in human embryonic<br />

stem cells<br />

18 Arjen Scholten, Reinout Raijmakers and Albert Heck; Finding new targets of small molecules by chemical proteomics<br />

22 Barbara Möller and Dolf Weijers; Cell-cell communication in plant embryos<br />

Interviews: Paola Picotti; <strong>Proteomics</strong> for the analysis of cellular networks<br />

Jonathan Sweedler; Understanding neuropeptide complexity in the brain<br />

Valorisation within the <strong>Netherlands</strong> <strong>Proteomics</strong> <strong>Centre</strong><br />

Bioinformatics<br />

NPC Top Publications<br />

NPC Research Hotels<br />

Martin Schuurmans, Combining the best existing forces<br />

11<br />

| NPC Highlights 11 | April 2010<br />

><br />

About<br />

The <strong>Netherlands</strong> <strong>Proteomics</strong> <strong>Centre</strong> (NPC) is a strategic collaboration of research groups from six<br />

universities, four academic medical centres and several research institutes and biotech companies.<br />

With a scientific program addressing key areas of proteomics in several projects, and specialised<br />

‘research hotels’, the NPC performs high-quality research and knowledge transfer in an<br />

international context. The NPC is part of the <strong>Netherlands</strong> Genomics Initiative.<br />

In NPC Highlights researchers present progress and results from NPC projects of the scientific program<br />

and the research hotels. NPC Highlights is published by the <strong>Netherlands</strong> <strong>Proteomics</strong> <strong>Centre</strong>.<br />

<strong>Netherlands</strong> <strong>Proteomics</strong> <strong>Centre</strong><br />

Coordination and editing<br />

Padualaan 8<br />

Marian van Opstal<br />

NL 3584 CH Utrecht<br />

Bèta Communicaties, The Hague<br />

t +31 30 253 4564<br />

e info@npc.genomics.nl<br />

Lay-out<br />

w www.netherlandsproteomicscentre.nl<br />

Frans Koeman<br />

F.Koeman DTP-Services, Zoetermeer<br />

Editorial Board<br />

Albert Heck, Scientific Director NPC<br />

Photography frontpage<br />

Werner Most, Managing Director NPC<br />

Bas van Breukelen<br />

Bert Poolman, member Executive Board NPC<br />

Hermen Overkleeft, member Executive Board NPC Printing<br />

Rob Liskamp, member Executive Board NPC<br />

Bestenzet BV, Zoetermeer<br />

Martje Ebberink, Communications NPC<br />

To subscribe to NPC Highlights please<br />

send an e-mail with your full name, organisation<br />

© 2010 Copyright <strong>Netherlands</strong> <strong>Proteomics</strong> <strong>Centre</strong> and address to info@npc.genomics.nl


Welcome This edition of NPC Highlights<br />

features a number of selected research<br />

projects from the NPC scientific program.<br />

Some of them were presented at the NPC<br />

Progress Meeting 2010 held on February 16.<br />

The meeting was once again a very well<br />

attended and lively event, and included a<br />

poster session and presentations by the keynote speakers Paola<br />

Picotti, Institute of Molecular Systems Biology, ETH Zurich, and<br />

Jonathan Sweedler, Chemistry Department, University of Illinois.<br />

Interviews with these inspiring scientists as well as information<br />

about the first NPC poster prize winners are included in this<br />

issue.<br />

With the start of the renewed program in 2009, the NPC has also<br />

intensified and extended some complementary initiatives, in<br />

particularly: the bioinformatics-proteomics interface program<br />

in collaboration with NBIC; the valorisation program including<br />

dedicated funding for proof-of-concept studies; personal<br />

coaching and more public-oriented educational activities like our<br />

contributions to the science days in NEMO and the very successful<br />

DNA labs on the road for high schools. More information about<br />

these initiatives is also presented in this issue of NPC Highlights.<br />

Finally, I would like to point out that you are always welcome<br />

at the NPC office for further information or any relevant<br />

contribution to this proteomics community. Please feel free to<br />

visit our website at www.netherlandsproteomicscentre.nl or get<br />

in contact with our public relations officer, Martje Ebberink, at<br />

info@npc.genomics.nl<br />

| <br />

Albert Heck, scientific director NPC<br />

preface


News Headlines<br />

Inspiring NPC Progress<br />

Meeting 2010 well attended<br />

Pink Ribbon and KWF grant<br />

for Arzu Umar<br />

The <strong>Netherlands</strong><br />

<strong>Proteomics</strong><br />

<strong>Centre</strong> (NPC)<br />

held its annual<br />

Progress<br />

Meeting on<br />

February 16.<br />

The event drew<br />

an audience<br />

of around<br />

200, consisting of scientists from inside and outside the NPC<br />

program, as well as delegates from several biotech companies.<br />

The international speakers, Jonathan Sweedler and<br />

Paola Picotti, and the NPC researchers put their marks on<br />

this event by highlighting successes achieved and challenges<br />

still looming. Congratulations to Lara Fornai (FOM Institute<br />

AMOLF), Remon van Geel (Nijmegen <strong>Centre</strong> for Molecular Life<br />

Sciences) and Nikolai Mischerikow (Utrecht University) who<br />

were awarded a travel grant of e 1,000 for the best poster at<br />

the NPC meeting.<br />

| NPC Highlights 11 | April 2010<br />

NPC researcher Arzu Umar and Madelaine<br />

Tilanus-Linthorst (Erasmus MC) have been<br />

awarded a Pink Ribbon grant of € 125,000.<br />

Umar and Tilanus will use their grant to<br />

identify protein markers for the early<br />

detection of breast cancer using a tissue<br />

proteomics approach. The results of this project will aid in the<br />

development of a non-invasive diagnostic test for the early<br />

detection of breast cancer.<br />

NPC researchers Arzu Umar, John Foekens and Theo Luider<br />

(Erasmus MC) have received a grant of € 534,000 from the KWF<br />

Kankerbestrijding (Dutch foundation for cancer research). The<br />

aim of the project is to identify a predictive protein profile for<br />

tamoxifen therapy resistance in patients with advanced breast<br />

cancer. The results of this project will provide a predictive tool<br />

for tamoxifen therapy resistance as well as pinpoint any key proteins<br />

that could serve as targets for new therapy development.<br />

Uncovering stem cell<br />

differentiation<br />

Signal proteins for plant<br />

stem cells discovered<br />

Wageningen biochemist Dolf Weijers and<br />

his German colleagues have discovered<br />

how stem cells in a plant embryo are<br />

formed. The cells communicate with<br />

one another via the transportation of<br />

a protein. The research results have<br />

been recently published in Nature (10<br />

March 2010). Unlike animals, plants<br />

produce new organs — leaves, roots and<br />

flowers — throughout their entire life. This task is undertaken<br />

by the meristems, growth tips in which stem cells are located.<br />

Meristems are located in the young plant embryo. Weijers<br />

studied the formation of root meristems in the embryo of the<br />

model plant Arabidopsis thaliana. The process begins with the<br />

programming of one cell as the ‘hypophysis’ which regulates<br />

stem cells in the roots. It is known that the formation of the<br />

hypophysis is controlled by the gene activator called Monopteros.<br />

However, it was hitherto unknown how this activator regulates<br />

hypophysis formation. Weijers received a VIDI grant in 2006 from<br />

NWO for this research and a NPC grant (awarded to Sacco de<br />

Vries). More information about this research has been described<br />

by Weijers in the highlight article in this issue (see pages 21-24).<br />

Researchers of NPC (Utrecht University, Hubrecht Institute<br />

and Leiden University Medical <strong>Centre</strong>) have reported on a<br />

significant advance in the understanding of stem cell differentiation<br />

using proteomics technology. The results are published<br />

in the high profile journal Cell Stem Cell, shedding light on<br />

how we can manipulate and programme stem cells to specific<br />

fates, creating a source of tissue replacement for regenerative<br />

medicine purposes.<br />

Biomarker discovery for<br />

head and neck cancer<br />

Annually, some 2,500 people in the<br />

<strong>Netherlands</strong> are diagnosed with<br />

head and neck cancer. In 20-40% of<br />

patients, the tumour returns after<br />

surgery. Up to now, it was impossible<br />

to predict among which patients this<br />

could occur. In her PhD study, NPCresearcher<br />

Tieneke Schaaij-Visser<br />

(UU) used proteomics techniques to compare hundreds of<br />

proteins of normal and (pre)cancer tissues of patients. She has<br />

managed to identify a number of proteins (biomarkers), with<br />

which one can effectively predict which treated patients have<br />

a significant risk of a recurring tumour.


awards honours news<br />

Albert Heck visiting<br />

Professor ETH Zurich<br />

ETH Zurich has appointed Albert Heck, scientific director of<br />

the <strong>Netherlands</strong> <strong>Proteomics</strong> <strong>Centre</strong>, as visiting professor of<br />

Systems Biology. Starting 1 March 2010, Heck will for five<br />

months be resident of the Institute for Molecular Systems<br />

Biology, led by Ruedi Aebersold. Heck will use his time in<br />

Zurich to further study the opportunities of this rapidly<br />

evolving scientific field. Heck’s visiting professorship is made<br />

possible by the ‘Distinguished Visiting Scientist Stipend’ of the<br />

<strong>Netherlands</strong> Genomics Initiative (NGI).<br />

Jacques Neefjes wins<br />

ERC Advanced Grant<br />

In its second competition for established<br />

top researchers, the European<br />

Research Council (ERC) has selected<br />

research leaders to perform their<br />

pioneering research throughout<br />

Europe. Grants are awarded to<br />

exceptional research leaders who<br />

have distinguished themselves in<br />

their originality and significance of<br />

their research contributions. NPC<br />

project leader Jacques Neefjes (NKI) has won the prestigious<br />

ERC Advanced Grant (€ 2,1 million).<br />

Neefjes is interested to obtain more insight in how the immune<br />

system is being controlled. Hereto Neefjes focuses on a<br />

certain group of molecules, MHC class II, which are important<br />

for the production of antibodies and stimulation of killer-cells.<br />

They are involved in immune defense against infections and<br />

cancer, but also play a role in auto-immunity upon transplantation.<br />

Neefjes tested many human proteins and identified<br />

about 300 proteins that are someway involved in this system.<br />

Neefjes will use his grant to sort out how these proteins are<br />

interconnected. Moreover he will explore the effects of<br />

various chemical compounds.<br />

DGMS Award 2010 for<br />

Albert Heck<br />

Albert Heck, scientific director of the <strong>Netherlands</strong><br />

<strong>Proteomics</strong> <strong>Centre</strong>, has been awarded the DGMS Award 2010<br />

‘Massenspektrometrie in den Biowissenschaften’ for his<br />

scientific contributions in mass spectrometry. Heck received<br />

his award at the annual (43rd) mass spectrometry conference<br />

in Hall (7-11 March 2010). The DGMS award is an initiative of<br />

the German Mass Spectrometry Society and has been awarded<br />

since 2009 to those who have provided an excellent scientific<br />

contribution to mass spectrometry.<br />

NPC participates in DNA-labs<br />

on the road<br />

The DNA-labs on the road are a great<br />

hit among high schools, teachers and<br />

students. Hundreds of teachers and<br />

thousands of students can talk about<br />

the success of this educational project. But still the DNA-labs<br />

continue in developing educational material and giving insights<br />

in modern genomics research.<br />

On March 12 the <strong>Centre</strong> for Society and Genomics organized<br />

the ‘DNA-lab dag’, a national conference on genomics for high<br />

school teachers and technical assistants. Keynote speakers<br />

were forensic geneticist Prof. Dr. Peter de Knijff (genetics<br />

and human evolution) and Prof. Dr. Jan Hoeijmakers (DNA and<br />

ageing). Also workshops on genomics were organised, including<br />

two workshops of the <strong>Netherlands</strong> <strong>Proteomics</strong> <strong>Centre</strong>.<br />

NPC researchers receive<br />

STW Grant for characterization<br />

of antibodies<br />

Technology Foundation<br />

STW has awarded NPC<br />

researchers Albert Heck<br />

and Esther van Duijn<br />

(Utrecht University) a<br />

STW Grant of € 992,000.<br />

Heck and Van Duijn<br />

will use their grant to characterize antibodies for therapeutic<br />

applications. In collaboration with MS Vision, a unique mass<br />

spectrometer will be built dedicated to the analysis of intact<br />

antibodies. Dutch biotechnology company Merus also takes part<br />

in the project, ensuring access to complex mixtures of antibodies<br />

with improved clinical efficacy.<br />

Geert Kops receives<br />

KWF Grant<br />

NPC theme leader Geert Kops (UMCU)<br />

has received a grant of € 500,000 from<br />

the KWF (Dutch foundation for cancer<br />

research) for his research on a protein<br />

(bubr1) which plays an important role<br />

in chromosome division. Mutations of<br />

this protein causes a rare disease called<br />

‘mosaic variegated aneuploidy’ (MVA). Patients with this<br />

disease have all kinds of developmental disorders, develop<br />

cancer at a young age and also die young. Kops and his<br />

colleagues showed that mutations in the bubr1 gene cause<br />

aneuploidy. They will use the grant to find the precise function<br />

of the protein and how mutations disturb its function.<br />

|


Geert Kops<br />

Dividing the goods:<br />

preventing chromosome<br />

segregation errors<br />

| NPC Highlights 11 | April 2010<br />

Cell division requires duplication of the genome and the<br />

subsequent physical separation of the two duplicates. This<br />

separation occurs during the strictly regulated process of<br />

mitosis. One of the critical enzymatic activities in mitosis is<br />

carried out by the kinase Mps1. Inhibiting Mps1 is sufficient<br />

to kill cells, making it a potential target for novel anti-cancer<br />

strategies. We have generated a unique cellular model<br />

system that allows in-depth examination of the roles of<br />

Mps1 activity in mitosis.<br />

Each and every time a cell divides, the whole complement<br />

of chromosomes needs to be distributed equally amongst the<br />

daughter cells during the process known as mitosis. Errors in<br />

chromosome segregation are devastating to the developing<br />

organism, almost always leading to early embryonic death.<br />

Similar errors in cells in the adult can contribute to tumour<br />

formation in various ways and indeed, whole chromosome<br />

aneuploidy is a hallmark of human cancers. To prevent errors<br />

in chromosome segregation, cells have evolved highly regulated<br />

networks of processes that monitor mitosis at various<br />

stages. The Mps1 kinase is a central player in many of these<br />

processes. Our NPCII-funded research aims at understanding<br />

how Mps1 regulates the chromosome segregation machinery.<br />

Error-prevantion machinery Mitosis is a dramatic process.<br />

In the span of a mere hour, cellular morphology and most<br />

structures in the cell change to accommodate chromosome<br />

segregation. During this time, the chromosomes condense, the<br />

nuclear envelope breaks down, most tubulin is re-assembled<br />

into a highly dynamic mitotic spindle, the cell rounds up and<br />

all chromosomes attach to spindle microtubules migrate to<br />

the cell equator and are subsequently pulled apart. Essential<br />

for error-free chromosome segregation is chromosome<br />

bi-orientation, meaning the attachment of one copy of a pair<br />

of duplicated chromosomes to microtubules from one side of<br />

the cell and attachment of the other copy to microtubules<br />

from the opposing side (see Figure 1).<br />

Two processes ensure that this configuration is achieved for<br />

all chromosomes before the cell starts the segregation process<br />

in mitotic anaphase. The first of these is the attachment<br />

error-correction machinery. In early mitosis, many errors in<br />

chromosome attachment that cannot result in bi-orientation<br />

are made, and the Aurora B kinase corrects such errors [1].<br />

The second process is the mitotic checkpoint. This checkpoint<br />

ensures that the segregation phase is not initiated until<br />

all chromosomes are fully and stably attached [2]. On the<br />

molecular level, the checkpoint is engaged by chromosomes


What this research is about:<br />

Cell division checkpoint possible<br />

target for anti-cancer drug<br />

In order to get from a fertilized egg-cell to a full human being, trillions and trillions of cell<br />

divisions are needed. All of them have to be carried out perfectly, since mistakes in the distribution<br />

of chromosomes between the two daughter cells lead to cell death and miscarriage.<br />

“It is very important to an organism that every cell division is error-free. We want to discover<br />

how that can be possible,” says Geert Kops of the Department of Physiological Chemistry and<br />

Cancer Genomics <strong>Centre</strong> at the University Medical <strong>Centre</strong> Utrecht.<br />

Cell division is a complex and dynamic process that is heavily controlled by molecular checkpoints.<br />

These checkpoints monitor if the duplicated chromosomes are properly connected<br />

before the process of cell division can go into its next phase. If they are not, the cell will<br />

receive more time to achieve proper chromosome connection. To elucidate this mechanism,<br />

Kops also looks at tumour cells. These cells make mistakes during cell division because their<br />

checkpoints do not efficiently see the faulty connections. Kops: “Errors seem a formula for<br />

success for these cells. But there is a limit to the amount of mistakes that can be tolerated<br />

without compromising viability. If we can inactivate the checkpoint by some drug, the tumour<br />

cell will make so many mistakes that it will soon die.”<br />

In this article the author describes the research on the key drugable protein in the checkpoint<br />

machinery. “If this protein is switched off the cell invariably dies. With proteomics we want<br />

to find out how this checkpoint protein exactly works and which other proteins it talks to. For<br />

instance, protein modifications cause the switches in its activity and location,” says Kops. “So<br />

if we understand how this works we can design better strategies for the development of new<br />

anti-cancer treatments aimed at inhibiting this protein. That is the ambition lurking on the<br />

horizon.”<br />

| <br />

NPC Research Theme T1: Cancer <strong>Proteomics</strong><br />

that have made no connections to spindle microtubules. In<br />

such a case, the unattached chromosomes themselves produce<br />

an inhibitor of the anaphase promoting complex/cyclosome<br />

(APC/C), an E3 ubiquitin ligase that directs destruction of<br />

proteins to initiate anaphase. Thus, as long as unattached<br />

chromosomes persist, the checkpoint will not allow cells to<br />

proceed with chromosome segregation.<br />

bi-orientation<br />

checkpoint<br />

It has been proposed that the error-correction and mitotic<br />

checkpoint machineries are dysfunctional in cancers, causing<br />

whole chromosome aneuploidy [3]. In addition, we have shown<br />

that full but not partial inactivation of the mitotic checkpoint<br />

induces cell death by causing massive chromosome segregation<br />

errors [4, 5]. In collaboration with René Medema of the<br />

Laboratory of Experimental Oncology at the University Medical<br />

Center Utrecht, our lab has recently shown that such levels of<br />

chromosome mis-segregations can also be reached by partial<br />

inhibition of multiple pathways in mitosis [5]. Importantly,<br />

tumour cells proved to be more sensitive to such combinatorial<br />

prometaphase<br />

metaphase<br />

Figure 1 | In early mitosis (prometaphase), the mitotic checkpoint prevents<br />

cell-cycle progression while chromosomes attempt to attach with each copy<br />

to opposite poles (bi-orientation). Once productive attachment of every<br />

chromosome is reached and chromosomes have bi-oriented (metaphase), the<br />

checkpoint is satisfied and chromosome segregation is allowed.


accept modified forms of ATP or ATP-like compounds [7]. One<br />

such form, a bulky version of the Src inhibitor PP1, inhibits<br />

these engineered kinases. Another is a bulky APT that carries<br />

a tagged phosphate that, when transferred to a substrate, can<br />

be affinity purified [8]. Figure 3 shows two human cell lines<br />

that express the engineered form of Mps1 as their sole source<br />

of Mps1 protein. Addition of the ATP analog 23dMB-PP1 inhibited<br />

Mps1 activity and caused severe chromosome segregation<br />

errors. Careful timing studies showed that full enzymatic<br />

inhibition can be achieved within 5 minutes of addition of this<br />

ATP analog.<br />

Figure 2 | Mps1 facilitates the correction of faulty chromosome-to-microtubule<br />

attachments by Aurora B kinase. Mps1 directly phosphorylates Borealin<br />

to promote activation of Aurora B thus enhancing error-correction.<br />

targeting than immortalized normal human cells. Organismal<br />

studies are underway to examine if this distinction between<br />

cancerous and non-cancerous cells will allow specific tumour<br />

cell killing in an animal.<br />

In collaboration with Albert Heck and Shabaz Mohammed of<br />

the Biomolecular Mass Spectrometry lab at Utrecht University,<br />

these cell lines are currently being used to find Mps1 substrates<br />

with two approaches (see Figure 3). In the first, phosphor-proteins<br />

in whole cell lysates of Mps1-inhibited cells will<br />

be quantitatively compared to those of uninhibited cells using<br />

stable isotope labelling in cell culture (SILAC) and dimethyl<br />

labelling, depending on the cell line.<br />

In the other approach, recombinant engineered Mps1 protein<br />

will be added to fractioned cell lysates together with tagged<br />

Dual function protein Not surprisingly, many enzymes<br />

partake in the mitotic processes that coordinate and regulate<br />

chromosome bi-orientation and the mitotic checkpoint.<br />

A<br />

B<br />

| NPC Highlights 11 | April 2010 | Geert Kops<br />

Mps1 is a dual specificity kinase that is critical for correcting<br />

improper chromosome-microtubule attachments as well as for<br />

ensuring mitotic checkpoint activity. In addition, Mps1 is one<br />

of the most promising targets in the anti-cancer strategy that<br />

induces the high frequency of chromosome segregation errors.<br />

It is not understood how activity of Mps1 is regulated or how<br />

it orchestrates the various Mps1-dependent processes. Thus<br />

far, only one functionally relevant substrate of Mps1 has been<br />

found. Our lab showed in 2008 that Mps1 phosphorylates<br />

Borealin, an activator of Aurora B, and thereby facilitates<br />

attachment error-correction [6] (see Figure 2). The Mps1-dependent<br />

phosphorylation sites on Borealin were identified by<br />

proteomics of in vitro phosphorylated protein, and inactivating<br />

mutation of these sites led to significant problems in attachment<br />

error-correction. Strikingly, mutation of these sites<br />

to mimic phosphorylation fully rescues Mps1 deficiency in relation<br />

to error-correction and effectively bypassed the need for<br />

Mps1 in this process. These experiments proved that Borealin<br />

is a relevant functional substrate of Mps1 in the error-correction<br />

process. The identity of substrates of Mps1 in the mitotic<br />

checkpoint or other mitotic processes, however, is unknown.<br />

C<br />

Chemical genetics To investigate the role of Mps1 in mitosis<br />

in more detail, our lab generated a cellular system that allows<br />

specific inhibition of Mps1 enzymatic activity and can also be<br />

used to chemically tag direct cellular substrates. This system,<br />

termed chemical genetics, was pioneered by Kevan Shokat<br />

(UCSF) and makes use of engineered kinase mutants that can<br />

Figure 3 | Chemical genetic inhibition of Mps1.<br />

A. Two human cell lines expressing LAP-tagged Mps1 variants, either analogsensitive<br />

(as) or wild-type (WT), instead of endogenous Mps1.<br />

B. Inhibition of Mps1-as activity (pT676) in mitosis (M) by 23dMB-PP1.<br />

C. Two proteomics approaches for identifying cellular substrates of Mps1<br />

using chemical genetics. Green represents acitve kinase, red inactive kinase.


ATP analog. Subsequent affinity purification and proteomic<br />

analyses will reveal potential relevant direct Mps1 substrates.<br />

Uncovering networks Tight spatio-temporal control of the<br />

processes that govern chromosome segregation ensures chromosomal<br />

stability. This control is exerted, amongst others,<br />

by various protein kinases. Our lab aims to understand the<br />

signalling networks that are influenced by a group of kinases<br />

that is activated at unattached kinetochores and is pivotal for<br />

chromosome bi-orientation and the mitotic cell-cycle checkpoint.<br />

A powerful method to uncover these networks is the use<br />

of chemical genetics combined with quantitative phospho-proteomics.<br />

With chemical genetics, a cell line is generated that<br />

lacks expression of endogenous kinase but instead expresses<br />

an engineered form that is amenable to inhibition by bulky,<br />

ATP-like compounds.<br />

Using NPC funding, we have established such cell lines to<br />

study the Mps1 kinase. Our initial results have shown that<br />

Mps1 can be enzymatically inhibited with bulky PP1 analogs<br />

within minutes, leading to absence of all known Mps1<br />

functions in mitosis. Using quantitative phospho-proteomics<br />

methods, these cells will be used to identify Mps1-controlled<br />

pathways and direct Mps1 substrates. The same methods will<br />

be applied to other kinases within this functional subfamily.<br />

Ultimately, these studies will facilitate the description of<br />

the signalling networks that ensure error-free chromosome<br />

segregation.<br />

References<br />

1 Vader, G., Medema, R.H., and Lens, S.M. (2006) The<br />

chromosomal passenger complex: guiding Aurora-B through<br />

mitosis. J Cell Biol 173, 833-837.<br />

2 Musacchio, A., and Salmon, E.D. (2007) The spindle-assembly<br />

checkpoint in space and time. Nat Rev Mol Cell Biol 8,<br />

379-393.<br />

3 Kops, G.J., Weaver, B.A., and Cleveland, D.W. (2005) On<br />

the road to cancer: aneuploidy and the mitotic checkpoint.<br />

Nat Rev Cancer 5, 773-785.<br />

4 Kops, G.J., Foltz, D.R., and Cleveland, D.W. (2004)<br />

Lethality to human cancer cells through massive chromosome<br />

loss by inhibition of the mitotic checkpoint. Proc Natl<br />

Acad Sci USA 101, 8699-8704.<br />

5 Janssen, A., Kops, G.J., and Medema, R.H. (2009) Elevating<br />

the frequency of chromosome missegregation as a strategy<br />

to kill tumor cells. Proc Natl Acad Sci USA 106, 19108-<br />

19113.<br />

6 Jelluma, N. et al. (2008) Mps1 phosphorylates Borealin to<br />

control Aurora B activity and chromosome alignment. Cell<br />

132, 233-246.<br />

7 Bishop, A.C., Buzko, O., and Shokat, K.M. (2001) Magic bullets<br />

for protein kinases. Trends Cell Biol 11, 167-172.<br />

8 Blethrow, J.D. et al. (2008). Covalent capture of kinasespecific<br />

phosphopeptides reveals Cdk1-cyclin B substrates.<br />

Proc Natl Acad Sci USA 105, 1442-1447.<br />

Author<br />

| <br />

Geert Kops<br />

Summary<br />

From conception onwards, many trillions of cell divisions generate<br />

a human individual. Each cell division requires duplication<br />

of the genome and the subsequent physical separation of<br />

the two duplicates. This separation occurs during the process<br />

of mitosis, after the replicated genomes in the form of distinct<br />

chromosomes attach to fibres of the spindle apparatus<br />

coming from opposite directions and align on the cell equator.<br />

Shortly after all chromosomes have aligned in this fashion,<br />

each chromosome copy is dragged to opposite sides of the cell<br />

after which the cell cleaves in the middle. Mitosis is highly<br />

dynamic and strictly regulated in space and time. Amongst<br />

<strong>summary</strong><br />

Contact<br />

Dr. Geert Kops<br />

Department of Physiological Chemistry and<br />

Cancer Genomics <strong>Centre</strong><br />

UMC Utrecht<br />

Universiteitsweg 100<br />

3584 CG Utrecht<br />

T +31 88 755 51 63<br />

g.j.p.l.kops@umcutrecht.nl<br />

http://ruummc.med.uu.nl/research/signalkops.html<br />

the critical enzymatic activities that exert this control is the<br />

Mps1 kinase. Depleting Mps1 protein is sufficient to kill cells<br />

by causing massive chromosome segregation errors. Mps1<br />

has thus been proposed as a potential target in anti-cancer<br />

strategies that aim at elevating the frequency of chromosome<br />

segregation errors. We have generated a cellular model system<br />

that allows specific, penetrant and temporally controlled inhibition<br />

of the enzymatic activity of Mps1. These cells are being<br />

used to uncover the roles of Mps1 in mitosis and to identify its<br />

direct substrates using quantitative phospho-proteomics. They<br />

furthermore present the best cellular model system to date<br />

for investigations into the effects of penetrance and duration<br />

of Mps1 inhibition on cell viability.


Rumyana Karlova and<br />

Sacco de Vries<br />

The plasma membrane<br />

receptor complex perceiving<br />

plant steroids<br />

10 | NPC Highlights 11 | April 2010<br />

Just like in humans, steroids play an important role in<br />

the development of a plant. Large protein complexes<br />

located in the plasma membrane act as receptors for these<br />

steroids. Researchers of Wageningen University have<br />

isolated such a plasma membrane receptor complex by<br />

co-immunoprecipitation and determined the composition<br />

by mass spectrometry. This forms the basis for a model to<br />

explain how steroid signalling in plant cells works.<br />

Animals as well as plants use steroid hormones as important<br />

signalling molecules. Plant steroids or brassinosteroids are<br />

perceived by a hetero-oligomeric plasma membrane receptor<br />

complex. This membrane complex is instrumental in transmitting<br />

the steroid signal to a wide range of different targets that<br />

control cell elongation, cell division, plant fertility and help<br />

defend against plant pathogens. It is not clear how all these<br />

different effects are integrated.<br />

To help find additional components we have isolated the receptor<br />

complex from Arabidopsis seedlings by co-immunoprecipitation<br />

with one of the GFP-tagged receptors. Identification<br />

of the associated proteins was then done by LC-MS. To verify<br />

the interactions several independent methods have been used.<br />

Biochemical interaction using phosphoproteomics was performed<br />

on isolated proteins. Furthermore, genetic interactions<br />

were established using intermediate strength mutant alleles<br />

and cellular interaction was determined using fluorescence<br />

microspectroscopy of differentially tagged proteins. Only<br />

when all these criteria were met an interactor was considered<br />

to be ‘real’.<br />

Our current NPC2 project focuses on one of the receptorinteracting<br />

proteins, surprisingly a transcription regulator of<br />

the MADS-box class. The aim of this project is to determine<br />

the spatial dynamics of this direct membrane-to-nucleus<br />

shortcut and how that operates functionally in plant cells.<br />

Plant hormones Steroid hormones are potent molecules that<br />

are important long-distance regulators in the animal body.<br />

Natural steroids are synthesized starting from cholesterol and<br />

well-known examples are testosterone, estradiol or corticosterone.<br />

They are synthesized in gonads or in adrenal glands,<br />

are lipid-soluble and are perceived in the cytoplasm of target<br />

cells by soluble receptors that act as transcriptional regulators.<br />

Steroids are involved in a large number of processes as<br />

diverse as development, inflammation and fertility.<br />

Similar molecules, called brassinosteroids, exist in the model<br />

plant Arabidopsis Initially they were isolated from Brassica<br />

pollen, hence their name. They are synthesized from the


What this research is about:<br />

How plant hormones can influence<br />

plant development<br />

Sooner or later we all depend on sufficient plant growth. That could be in the form of biomass for<br />

the production of biofuels or for our own nutrition. “That’s why we are interested in the genetic<br />

circuits that determine how a plant develops. Plant hormones, such as steroids, play an important<br />

role in plant growth,” explains Sacco de Vries, professor of Biochemistry in the department of<br />

Agrotechnology & Food sciences at Wageningen University. “By adapting genetic routes you can influence<br />

the height and yield of a crop. Of course there is a limit to what you can change.”<br />

De Vries and his research group study a protein complex of the model plant Arabidopsis that is<br />

involved in growth but also affects the defence system against plant pathogens. “When this protein<br />

complex is triggered by the presence of steroids, it influences both processes. In humans this works<br />

in the same way. When we are busy fighting infections there is no energy left for growth or vice<br />

versa.”<br />

The big question is how you can breed plants that have high resistance to disease and at the same<br />

time produce a large amount of the desired products.<br />

“Simple solutions do not exist,” says De Vries. “If you change one pathway, it could have an effect<br />

on other cell processes. To find the right balance we need to improve our knowledge on a fundamental<br />

level.”<br />

In this article the authors describe their approach to determine the existence, location and function<br />

of their protein complex, using a proteomics-based analysis in combination with biochemical and genetic<br />

techniques. “The combination of techniques is essential. We found proteins we would not have<br />

found by using the classical techniques. Furthermore we even manage to visualise the protein-protein<br />

interactions and location of the protein complex in the membrane by fluorescent labels,” says<br />

De Vries. “Our next project is to elucidate the three-dimensional structure of the protein complex.”<br />

| 11<br />

NPC Research Theme T2: Proteome Biology of Plants<br />

steroid campestanol, are also lipid-soluble and are found to<br />

consist of about 70 different molecules. They occur naturally<br />

in plants, especially in flowers, and constitute a normal element<br />

of our daily diet, although the concentrations are probably<br />

too low to have any physiological effect on animal steroid<br />

perception. In contrast to animal cells, they are not perceived<br />

by nuclear receptors but by a plasma membrane located<br />

receptor called brassinosteroid-insensitive-1 or BRI1 [1].<br />

While the BRI1 receptor was identified in a genetic screen<br />

aimed at identifying mutants insensitive to the ligand, it was<br />

also found as a mutant that behaved in the dark as if it was<br />

in the light, indicating a close connection between brassinosteroids<br />

and light control [2]. Based on the phenotypes of<br />

the plants, roles in male fertility, plant resistance, immunity<br />

and in morphogenesis have been reported [2]. At the cellular<br />

level, the action of brassinosteroids is thought to be primarily<br />

in the control of cell expansion, the reason why strong bri1<br />

mutations result in miniature plants (see Figure 1). An open<br />

question therefore is how this single ligand-receptor combination<br />

is able to control all of these diverse pathways.<br />

Figure 1 | A wild-type (back)<br />

and bri1 mutant (front) plants.


Figure 2 | FRET-FLIM employing BRI1-CFP and SERK3-YFP using a transient<br />

assay in protoplasts. A τ of less than 2.1 (green in the false-colour image)<br />

indicates that the two proteins are in close proximity to form a PM-located<br />

hetero-oligomer.<br />

12 | NPC Highlights 11 | April 2010 | Sacco de Vries<br />

Research from several groups including ours [3, 4] has indicated<br />

that the BRI1 receptor is part of a complex that also<br />

contains members of another class of PM-located receptors,<br />

the Somatic Embryogenesis Receptor-like Kinases (SERKs),<br />

that as BRI1 itself are LRR type single pass TM, dual specificity<br />

type of receptor kinases capable of transphosphorylating both<br />

Ser/Thr as well as Tyr residues [5, 6]. Most likely the SERKs<br />

do not bind ligands themselves and are therefore more likely<br />

co-receptors. There is some evidence that the active configuration<br />

is a tetramer [4]. We have used a proteomics approach<br />

to isolate the membrane receptor complex that is involved in<br />

brassinolide perception starting from tagged versions of the<br />

SERK receptors.<br />

Here we will describe how we used additional lines of<br />

evidence using genetics, biochemistry and direct interaction<br />

studies using fluorescence microscopy to verify<br />

the configuration of the initially determined receptor<br />

complex. The main research question in our group is what<br />

the precise biochemical role is of the non-ligand-binding<br />

co-receptors. Based on genetic and biochemical evidence<br />

it was recently proposed that they act as ‘signalling output<br />

enhancers’ by increasing the phosphorylation status<br />

after activation of the main BRI1 receptor. There is also<br />

some evidence that suggests that the precise combination<br />

of co-receptors with the main receptor serves to increase<br />

specificity or to link brassinosteroid pathways with other<br />

pathways [7].<br />

Members of the family The expression pattern of the SERK1-<br />

GFP gene assessed in fluorescent Arabidopsis plants appeared<br />

highly complex [8]. The SERK1 protein complex was isolated<br />

by using this GFP-tagged version under the control of its<br />

own promoter after re-introduction into a wild type plant<br />

[4]. We could identify the third member of the SERK family,<br />

SERK3 or BAK1, identified as a partner of BRI1 by others. BRI1<br />

was also detected, strengthening the idea that SERK1 was<br />

indeed a novel co-receptor in the main brassinolide pathway.<br />

Biochemical interactions using pull-down and in vitro transphosphorylation<br />

assays confirmed the interactions between<br />

SERK1, SERK3 and BRI1 [5]. However, null mutations of the<br />

SERK1 gene yielded completely wild-type looking plants with<br />

no obvious defects. When combining an intermediate strength<br />

mutant allele of BRI1 with alleles of SERK3 and SERK1 the BRI1<br />

phenotype became much more pronounced [9], providing formal<br />

genetic proof for the interactions. Finally, we could show<br />

that fluorescently-tagged SERK1 and BRI1 receptors indeed<br />

co-localize at the PM and by using FLIM (see Figure 2). FRET<br />

was shown between both proteins [10]. This way, biochemical,<br />

genetic and cellular interaction indeed all confirmed<br />

the original observation made of the membrane complex [4].<br />

Current work is directed in trying to quantify the proportion of<br />

the receptors that are in complex in vivo using a variety of different<br />

approaches and to establish the structures of the SERK<br />

receptors (see Figure 3).<br />

Our new project within the NPCII program is directed towards<br />

a third interacting protein of the SERK1 receptor complex, the<br />

transcriptional regulator AGL15. This protein is a member of<br />

the large family of MADS box proteins under study in the group<br />

of Gerco Angenent. We have also verified the interactions<br />

at both biochemical, genetic and cellular level and are in<br />

the process of looking at the precise cell types in the intact<br />

plant where this interaction is taking place because in plant<br />

research it is quite unusual to find a transcriptional regulator<br />

directly interacting with a PM receptor kinase in this project.<br />

Investigating mutants The main conclusion from the work is<br />

that plant membrane receptor complexes can be isolated<br />

Figure 3 | Comparative modelling of the SERK1 kinase domain is helpful in<br />

elucidating its final structure using X-ray diffraction. Red: ATP binding site,<br />

yellow: activation loop and blue: C-terminal tail.<br />

Model drawn by Matije van den Toorn (Wageningen UR),


using a single-step affinity purification and in sufficient<br />

quantity to enable identification of its components by mass<br />

spectrometry. More recently we have modified the original<br />

procedure fairly extensively to the point where we can start<br />

to compare the receptor complexes associated with different<br />

members of the gene family and also to investigate how<br />

receptor complexes look like from mutant backgrounds that<br />

miss one or two of the complex components.<br />

This work should provide a basis for answering basic questions<br />

concerning the replacement of the now missing proteins by<br />

close homologs and/or the stability of the remaining complexes.<br />

In a model species such as Arabidopsis, where research is<br />

dominated by forward and reverse genetics as the main tools<br />

for identifying interactors, the isolation of protein complexes<br />

offers an alternative approach to identify proteins that would<br />

otherwise have been missed. It is now being incorporated in<br />

the research activities of many laboratories.<br />

One thing that has become clear from our work is that other<br />

lines of evidence such as from genetic and cell-biological<br />

experiments are required to confirm the protein interactions<br />

found.<br />

8 Kwaaitaal, M. and de Vries, S.C. (2007) The SERK1 gene is<br />

expressed in procambium and immature vascular cells. J<br />

Exp Bot 58, 2887-2896.<br />

9 Albrecht, C. et al. (2008) The Arabidopsis SERK proteins<br />

serve brassinolide-dependent and independent signaling<br />

pathways. Plant Physiol 148: 611-619.<br />

10 Hink, M.A. et al. (2008) Fluorescence Fluctuation Analysis<br />

of AtSERK1 Oligomerization. Biophysical J 1052-1062.<br />

Research team<br />

1 2 3<br />

4<br />

5<br />

References<br />

1 Li, J. and Chory, J. (1997) A putative leucine-rich repeat<br />

receptor kinase involved in brassinosteroid signal transduction.<br />

Cell 90, 929–938.<br />

2 Clouse, S.D. and Sasse, J.M. (1998) BRASSINOSTEROIDS:<br />

Essential Regulators of Plant Growth and Development.<br />

Annu Rev Plant Physiol Plant Mol Biol 49, 427-451.<br />

3 Nam, K.H. and Li, J. (2002) BRI1/BAK1, a receptor kinase<br />

pair mediating brassinosteroid signaling. Cell 110, 203-212.<br />

4 Karlova, R. et al. (2006) The Arabidopsis Somatic<br />

Embryogenesis Receptor-like Kinase 1 protein complex includes<br />

Brassinosteroid-insensitive1. Plant Cell 18, 626-638.<br />

5 Karlova, R. et al. (2009) Identification of in vitro phosphorylation<br />

sites in the Arabidopsis thaliana Somatic<br />

Embryogenesis Receptor-like Kinase family. <strong>Proteomics</strong> 8,<br />

368-379.<br />

6 Wang, X. et al. (2008) Sequential transphosphorylation<br />

of the BRI1/BAK1 receptor kinase complex impacts early<br />

events in brassinosteroid signaling. Dev. Cell 15, 220–235.<br />

7 Chinchilla, D. et al. (2009) One for all: the receptor-associated<br />

kinase BAK1. Trends Plant Sci 14: 535-541.<br />

Summary<br />

Plant steroids have an important role in plant development<br />

and are perceived by a plasma membrane receptor complex.<br />

This complex consists of a main, ligand-binding receptor involved<br />

in the entire spectrum of cellular processes affected by<br />

steroids and a number of co-receptors that are thought to be<br />

non ligand-binding. We have isolated such a plasma membrane<br />

receptor complex by co-immunoprecipitation using one of the<br />

co-receptors fused to GFP. The composition of the complex<br />

<strong>summary</strong><br />

6 7 8<br />

Esther van Loenen (1), Cathy Albrecht (2), Jan Willem Borst (3),<br />

Na Li (4), Rumyana Karlova (5),<br />

Sjef Boeren (6), Walter van Dongen (7) and Sacco de Vries (8).<br />

Contact<br />

Prof. Sacco C. de Vries<br />

Laboratory for Biochemistry<br />

Wageningen University and Research centre<br />

Dreijenlaan 3<br />

6703 HA Wageningen<br />

T +31 317 483 866<br />

sacco.devries@wur.nl<br />

was then determined by mass spectrometry, followed by<br />

extensive biochemical, genetic and cell biological experiments<br />

for confirmation.<br />

Apart from other related plasma membrane receptors,<br />

proteins acting as adaptors such as a member of the 14-3-3<br />

family were found. The most surprising result however was<br />

the presence of the transcriptional regulator AGL15 as part of<br />

the membrane receptor complex. Current work is therefore<br />

aimed to establish a comprehensive cellular model to explain<br />

how steroid signalling in plant cells uses a plasma membrane<br />

receptor directly activating a transcription factor.<br />

| 13


Paul Boersema, Vanessa Ding<br />

and Albert Heck<br />

In depth profiling of tyrosine<br />

phosphorylation in human<br />

embryonic stem cells<br />

14 | NPC Highlights 11 | April 2010<br />

Human stem cells are inclined to differentiate easily<br />

during culturing; thus losing their ability to transform into<br />

other cell types. Fibroblast growth factor 2 plays a role<br />

in preventing this process by activating a receptor that<br />

transmits the signal by phosphorylating tyrosine residues of<br />

proteins. But the events following signal transmission that<br />

lead to the maintenance of the pluripotency of stem cells<br />

remain unclear. Using a very sensitive proteomics set-up,<br />

the tyrosine phosphorylation sites were studied in detail<br />

and a comprehensive picture of tyrosine phosphorylation<br />

was obtained.<br />

tency. Culturing conditions have been optimized by the use of<br />

additives to induce self-renewal rather than differentiation, but<br />

how exactly the added factors are involved in the maintenance<br />

of the pluripotency state remains unclear.<br />

Stem cells are used more and more to study embryonic development<br />

in vitro. Also, these pluripotent cells promise therapeutic<br />

applications for diseases that are currently difficult to treat.<br />

Human ESCs can be derived from an early stage of an embryo.<br />

These cells are isolated and can then be grown and expanded<br />

for more efficient use and to limit the number of embryos that<br />

are required.<br />

However, embryonic stem cells show a tendency to proliferate<br />

and differentiate easily during growth. Their culturing therefore<br />

requires special attention. Any stimulation, such as stress,<br />

may result in the development of these stem cells into specific<br />

cell types after which they will largely lose their ability to<br />

transform into other cell types. It requires precise fine tuning<br />

of growth conditions to keep these cells in a state of pluripo-<br />

Several cellular signalling pathways have been shown to be<br />

implicated with hESC self-renewal. Fibroblast growth factor 2<br />

(FGF-2) is a crucial growth factor in the maintenance of the<br />

pluripotent state of human embryonic stem cells (hESCs). FGFs<br />

bind receptor tyrosine kinases, i.e. they phosphorylate tyrosine<br />

residues in proteins. Their main task is the transmission of the<br />

signal from the stimulus by phosphorylating tyrosine residues<br />

on specific down-stream proteins. Inhibition of the FGF receptor<br />

causes the down-regulation of several proteins that are<br />

considered markers of pluripotency. FGF-2, on the other hand,<br />

is known to activate FGF receptors, which have been shown<br />

to turn on several signalling pathways. Therefore, the growth<br />

factor FGF-2 is widely used for the long-term culture of hESCs.<br />

However, the downstream signalling events following FGF-2<br />

stimulation and its link to hESC self-renewal and the maintenance<br />

of pluripotency remain to be determined.


What this research is about:<br />

Preventing stem cells specialising<br />

too early<br />

Stem cells have the ability to differentiate into a range of specialized cell types. They promise therapeutic<br />

applications for diseases that are currently difficult to treat such as Alzheimer’s disease because of this<br />

property. Human stem cells are isolated from embryos and then cultured and grown. During the culturing,<br />

however, the stem cells have a tendency to differentiate into specialized cells prematurely, after which<br />

they lose their ability to transform into other cell types.<br />

The growth factor FGF-2 is added during the culturing of stem cells, explains researcher Paul Boersema. “If<br />

you leave this factor out, the stem cells start to specialize. But we do not exactly understand how FGF‐2<br />

operates in this process.” The growth factor stimulates a receptor on the cell membrane, which in turn<br />

starts a signalling process in the cell. The signal is transmitted through phosphorylation on a tyrosine residue<br />

of certain proteins. “Some of the proteins which are phosphorylated by the FGF-2 receptor are known,”<br />

explains Boersema, “but most of these are present in such a low concentration that they are hard to study.”<br />

Boersema has therefore designed a process to isolate these phosphorylated tyrosine peptides by binding<br />

them to a bead containing an antibody. Subsequently the beads are separated, the peptides eluted and<br />

analyzed using LC-MS. Boersema cooperated with researchers from the Singapore Bioprocessing Technology<br />

Institute who showed him the details of the enrichment of tyrosine phosphorylated peptides, while the<br />

Utrecht group instructed the Singaporeans on the LC-MS methods, including stable isotope labelling<br />

techniques to implement quantitative module.<br />

The cooperation resulted in the discovery of many proteins that were formerly not known to play a role<br />

in FGF receptor signalling. Several of these proteins are known to have a role in cytoskeletal dependent<br />

processes in adult cells. “This may mean that they also play a role in the differentiation of stem cells,” says<br />

Boersema. “Eventually it may enable us to find a suitable growth factor to regulate specialization of stem<br />

cells.”<br />

| 15<br />

NPC Enabling Technologies E3: New Mass Spectrometric Tools in <strong>Proteomics</strong><br />

In this collaborative study, the effect of the growth factor FGF‐2<br />

on hESCs was investigated. NPC researchers at the Utrecht<br />

University collaborated with researchers from the Bioprocessing<br />

Technology Institute, Biopolis in Singapore, to study the effect<br />

of FGF-2 on hESCs by profiling tyrosine phosphorylation upon<br />

FGF-2 stimulation. The experience of the Utrecht group with<br />

sensitive LC-MS and stable isotope dimethyl labelling for quantitative<br />

proteomics was combined with the know-how of the<br />

Singapore group about culturing hESCs and performing<br />

immunoprecipitation of tyrosine phosphorylated peptides.<br />

Figure 1 | Immunoprecipitation with immobilized antibodies against tyrosine<br />

phosphorylation can be performed before digestion at the protein level or<br />

after digestion at the peptide level.<br />

Isolating phosphorylated peptides We focused on the profiling<br />

of tyrosine phosphorylation upon FGF-2 stimulation. As the<br />

target proteins that are involved in this signalling are largely<br />

unknown, we set out to identify them by an LC-MS based<br />

proteomics approach. The large scale analysis of cellular phosphorylation<br />

— and particularly, tyrosine phosphorylation — by<br />

LC-MS is challenging because of the relatively low abundance of<br />

phosphorylation. The overwhelming background of non-modified<br />

peptides causes phosphopeptides to remain undetected when<br />

no proper precautions are taken.


New discoveries To quantitatively profile to what extent individual<br />

tyrosine sites are phosphorylated upon FGF-2, we had to<br />

incorporate a stable isotope labelling method in the workflow.<br />

The milligrams of sample required for an efficient immunoprecipitation<br />

of tyrosine phosphorylated peptides make most<br />

labelling approaches cost-prohibitive. For that reason, we chose<br />

to implement stable isotope dimethyl labelling as it uses rather<br />

cheap reagents and can thus be used for the labelling of large<br />

amounts of sample. hESCs were stimulated with FGF-2 for 0,<br />

5 or 15 minutes respectively. The proteins from these samples<br />

were digested and differentially labelled with the dimethyl<br />

labels (see Figure 2). From the mixture of the three different<br />

samples, tyrosine phosphorylated peptides were selectively<br />

enriched by immunoprecipitation. LC-MS analysis on these<br />

enriched peptides then allowed us to identify and quantify 316<br />

unique tyrosine phosphorylated peptides [2].<br />

Of these peptides, 138 showed different levels of increased<br />

tyrosine phosphorylation (Figure 3), while for most of the<br />

other peptides the phosphorylation levels did not significantly<br />

change. An increase in tyrosine phosphorylation was, not<br />

surprisingly, found on peptides from FGF receptors and some of<br />

their known downstream targets. Furthermore, an increase in<br />

Figure 2 | The proteomics workflow to study the effect of FGF-2 on the tyrosine phosphorylation was found on many proteins that are<br />

tyrosine phosphoproteome of hESCs. hESCs are stimulated with FGF-2 for not known to be involved in FGF signalling. These proteins may<br />

0, 5 or 15 min after which the cells are lysed and the proteins digested.<br />

be new players in the FGF pathway or may shed some light on<br />

The different samples are differentially labelled with stable isotope<br />

new effectors of FGF-2. For example, several receptor tyrosine<br />

dimethyl labels after which the mixture of these peptides is enrich for<br />

tyrosine phosphorylated peptides by immobilized antibodies against tyrosine kinases other than FGF receptors were found to be increased<br />

phosphorylation. The enriched peptides are then analyzed by nano-LC-MS. in tyrosine phosphorylation. As FGF-2 has no known specificity<br />

for these receptors, transactivation of these receptors or some<br />

16 | NPC Highlights 11 | April 2010 | Paul Boersema other effect may be the cause of their activation. To confirm<br />

the phosphorylation of these receptor tyrosine kinases we used<br />

Various techniques have been developed to enrich for phosphopeptides<br />

prior to LC-MS analysis, for example, approaches based<br />

a human phospho-RTK array. In this array, antibodies against<br />

on titanium dioxide (TiO 2 ) or immobilized metal affinity chromatography<br />

(IMAC). TiO 2 and IMAC are specifically suited for the<br />

analysis of serine and threonine phosphorylation. The even<br />

lower abundance of tyrosine phosphorylation requires a different<br />

approach. Antibodies have been generated that specifically<br />

bind phosphorylated tyrosine residues. By immobilizing these<br />

antibodies on agarose beads, tyrosine phosphorylated peptides<br />

or proteins can be immunoprecipitated and a high degree of<br />

enrichment can be obtained. Performing the precipitation<br />

step at peptide level can be advantageous. As the end product<br />

that is analyzed by LC-MS is a peptide mixture, performing the<br />

precipitation at peptide level reduces the number of nonmodified<br />

peptides that can arise from non-modified stretches<br />

of the proteins or from non-modified proteins that happen to<br />

bind tyrosine phosphorylated proteins [1] (see Figure 1). The<br />

precipitated tyrosine phosphorylated peptides can be identified<br />

by LC-MS.<br />

We have been utilizing a very sensitive set-up in which relatively<br />

narrow (i.e. 50 mm inner diameter) but long (up to 40 cm)<br />

LC columns allow for efficient separation of the compounds also<br />

when longer gradients of up to three hours are used. Using this<br />

approach, we could generate a library of 735 unique tyrosine<br />

phosphorylation sites from hESCs that were stimulated with<br />

FGF-2 for 0, 1, 5, 15 and 60 minutes, respectively [2]. These<br />

Figure 3 | Clustering of different tyrosine phosphorylation profiles upon<br />

sites only partially overlapped with sites that were detected FGF‐2 stimulation. All tyrosine phosphorylated peptides from the FGF receptors<br />

and several of their known substrates are found in cluster 3 and before and that have been deposited in public repositories.<br />

4.


42 of the human RTKs are immobilized. After proteins from the<br />

sample are bound to the antibodies, a phosphotyrosine antibody<br />

that is conjugated to horseradish peroxidase is used to detect<br />

tyrosine phosphorylation levels. In this way, the activation of<br />

insulin receptor, insulin growth factor 1 receptor, ephrin type A<br />

receptors 1 and 2 and vascular endothelial growth factor receptor<br />

2 were confirmed.<br />

Amongst the other proteins on which tyrosine phosphorylation<br />

was shown to be increased were Src family kinase proteins and<br />

other proteins that, in adult cells, are known to be involved in<br />

cytoskeletal depending processes. Interestingly, changes to cytoskeletal<br />

dependent processes have been known to play major<br />

roles during hESC differentiation.<br />

References<br />

1 Boersema, P. J. et al. (2009) In depth qualitative and quantitative<br />

profiling of tyrosine phosphorylation using a combination<br />

of phosphopeptide immuno-affinity purification<br />

and stable isotope dimethyl labeling. Mol Cell <strong>Proteomics</strong><br />

Jan;9(1):84-99<br />

2 Ding, V.M.Y et al. (2010) Tyrosine phosphorylation profiling<br />

in FGF-2 stimulated human embryonic stem cells.<br />

Manuscript in preparation<br />

Successful approach The combination of stable isotope<br />

dimethyl labelling with selective immunoprecipitation of tyrosine<br />

phosphorylated peptides and sensitive LC-MS allows for a<br />

comprehensive analysis of the immediate tyrosine phosphorylation<br />

events following the stimulation of receptor tyrosine kinases<br />

such as the EGF and FGF receptors [1,2]. We have been able<br />

to identify relatively high numbers of tyrosine phosphorylation<br />

sites in single LC-MS runs. The immunoprecipitation of tyrosine<br />

phosphorylated peptides has thereby increased the specificity<br />

of the approach, while the (nano-) LC-MS approach enabled the<br />

sensitive detection of also lower abundant phosphopeptides.<br />

Finally, stable isotope dimethyl labelling allowed MS based<br />

quantitation while working with large amounts of sample.<br />

Research team<br />

In the study of the effect of the growth factor FGF-2 on hESCs<br />

we successfully applied the new approach. Several known substrates<br />

of FGF receptors were shown to be increased in tyrosine<br />

phosphorylation upon FGF-2 stimulation, but also proteins that<br />

have not been associated with FGF receptor signalling were<br />

found with increased tyrosine phosphorylation. A significant<br />

amount of these proteins have been known, in adult cells, to<br />

be involved in cytoskeletal dependent processes. These processes<br />

are important in the differentiation of embryonic cells.<br />

Therefore, the current data would suggest that FGF-2 is one of<br />

the regulators of these processes which, in turn, may be the<br />

reason for FGF-2 being required for long-term culture of hESC.<br />

Summary<br />

To keep stem cells in their pluripotent state, specific growth<br />

conditions are required. One of the additives often used is the<br />

growth factor FGF-2. In this collaborative study between NPC<br />

researchers in Utrecht and researchers of the Bioprocessing<br />

Technology Institute in Singapore, the role of FGF-2 in the<br />

maintenance of the pluripotency state of human embryonic<br />

stem cells was investigated. FGF-2 stimulates FGF receptors<br />

that, in turn, phosphorylate several proteins at specific tyrosine<br />

residues. Using a combination of stable isotope dimethyl<br />

labelling for MS based quantitation, immunoprecipitation<br />

with antibodies against tyrosine phosphorylation to enrich<br />

<strong>summary</strong><br />

From left to right: Simone Lemeer, Vanessa Ding, Paul<br />

Boersema, Leong Yan Foong<br />

Contact<br />

Prof. Albert Heck<br />

Biomolecular Mass Spectrometry and <strong>Proteomics</strong> Group<br />

Utrecht University<br />

Padualaan 8,<br />

3584 CH Utrecht, The <strong>Netherlands</strong>.<br />

T +31 30 253 6797<br />

a.j.r.heck@uu.nl<br />

http://bioms.chem.uu.nl<br />

for tyrosine phosphorylated peptides, and sensitive LC-MS<br />

analysis we have been able to identify relatively high numbers<br />

of tyrosine phosphorylation sites and thereby obtain a rather<br />

comprehensive picture of tyrosine phosphorylation upon a<br />

stimulus. Several hundreds of tyrosine phosphorylation events<br />

upon FGF-2 stimulation could thus be monitored. Several<br />

known targets of FGF receptors were identified, but also some<br />

proteins that have not been associated with FGF receptor<br />

signalling showed an increase in tyrosine phosphorylation.<br />

Several of these proteins are known to have a role in cytoskeletal<br />

dependent processes in adult cells. Furthermore, several<br />

other receptors were shown to be activated upon FGF-2,<br />

which could be confirmed by another phospho-array. Exactly<br />

how these receptors are activated remains to be elucidated.<br />

| 17


Arjen Scholten, Reinout<br />

Raijmakers and Albert Heck<br />

Finding new targets of small<br />

molecules by chemical<br />

proteomics<br />

18 | NPC Highlights 11 | April 2010<br />

Chemical proteomics allows the study of proteins in tissue<br />

or cells by utilizing their affinity for small molecules such as<br />

signalling molecules or drugs. Using a combination of pull-downs<br />

with immobilized compounds and the use of modified<br />

molecules and quantitative mass spectrometric approaches,<br />

a detailed overview can be generated of the specificity<br />

certain proteins have for such molecules. This may lead to<br />

new screening tools or drug target discovery strategies.<br />

In every aspect of life, small molecules, like metabolites and<br />

signalling molecules but also many of the drugs that are used in<br />

the clinic, exert their effects by interacting with proteins. They<br />

can act as substrates, agonists, antagonists or as co-factors. The<br />

interaction of small molecules of both natural and synthetic origin<br />

can be studied by ‘chemical proteomics’; a multidisciplinary<br />

research area that integrates biochemistry and cell biology with<br />

organic synthesis and mass spectrometry [1].<br />

In chemical proteomics, the small molecules of interest are<br />

often engineered to be used as a ‘bait’ to specifically enrich<br />

their protein targets from a complex proteome such as cell or<br />

tissue lysates. This approach is useful in determining which<br />

proteins are targeted by these small molecules. The technique<br />

has proven its worth in the elucidation of drug targets as well<br />

as targets of endogenous signalling molecules. Here we show<br />

an example of both of these to highlight the unique strengths<br />

of chemical proteomics.<br />

In the first example we use immobilized cAMP analogues to<br />

screen the distinct intracellular localization profiles of type I<br />

and type II cAMP-dependent protein kinase through probing<br />

their differential interaction with A-kinase anchoring proteins.<br />

In the second example we used immobilized versions of the<br />

phosphodiesterase-5 inhibitor sildenafil, the active compound<br />

of Viagra (see Figure 1). Chemical proteomics methods<br />

allowed the identification of novel protein targets of sildenafil<br />

outside the family of phosphodiesterases and optimization of<br />

these interactions by using close analogues of sildenafil.<br />

Endogenous signalling molecules The second messenger<br />

cAMP (3’-5’-cyclic-adenosine monophosphate) is a ubiquitous<br />

signalling molecule involved in the translation of extracellular<br />

signals into intracellular responses. It is implicated in a large<br />

number of key cellular processes. The main target of cAMP<br />

is the cAMP-dependent protein kinase (PKA), a hetero-tetrameric<br />

protein that consists of two regulatory (PKA-R) and two


What this research is about:<br />

Fishing for proteins with small<br />

molecule bait<br />

Hundreds of proteins have been isolated, characterized and sequenced using proteomics techniques.<br />

But lately, researchers are reaching the limit: the concentration in which new, uncharacterized<br />

proteins are present in cells or tissue is too low to identify them using standard proteomics methods.<br />

Chemical proteomics is a new research area that solves this problem. Small molecules having an<br />

affinity for the proteins to be studied, are coupled to a solid carrier, mostly small beads. Cell or tissue<br />

lysate is added to this immobilized ‘bait’ and specific proteins bind to it.<br />

These proteins can then be easily separated from the solution with a so-called pull-down experiment.<br />

In this way, the bound proteins are isolated and may then be characterized by, for example, high<br />

resolution mass spectrometry. “The beauty of this method is that you can study proteins that are<br />

present in (very) low concentrations in tissue or cells without modifying them. This offers great new<br />

possibilities,” says Arjen Scholten, who developed this technology for the signalling molecule cAMP<br />

during his PhD research.<br />

In one of the studies described here, Scholten and colleagues used cAMP and its derivatives to study a<br />

large and diverse family of kinase anchoring proteins in rat heart tissue. It is not well known how and<br />

to which type of kinases these AKAPs bind. “We showed that, using the pull-down method, it is possible<br />

to screen the binding properties of many of these AKAPs directly in their natural environment,” says<br />

Scholten.<br />

The same method was used to study, in collaboration with Pfizer, the binding characteristics of<br />

derivates of the well known drug sildenafil, or Viagra. Using the pull-down method, Scholten and<br />

Raijmakers discovered the drug not only has an affinity for phosphodiesterase-5, its target, but also for<br />

another protein. The researchers showed that certain derivatives of sildenafil can even have a higher<br />

affinity for this second protein. “In this way, sildenafil can become a starting point for the development<br />

of drugs against novel targets.”<br />

| 19<br />

NPC Enabling Technologies E2: Chemical Approaches to Proteome Biology<br />

catalytic subunits (PKA-C). To accommodate all the different<br />

tasks of cAMP and PKA within a single cell efficient spatial and<br />

temporal regulation of its activity is required.<br />

Figure 1 | Sildenafil is the active compound of the drug Viagra, most often<br />

used to treat erectile dysfunction. Sildenafil is an inhibitor of phosphodiesterase-5.<br />

http://en.wikipedia.org<br />

This is acquired through interaction with the large and diverse<br />

family of A-kinase anchoring proteins (AKAPs). AKAPs tether<br />

PKA, together with other signalling proteins (phosphatases,<br />

phosphodiesterases etc.), to distinct loci within the cell close<br />

to PKA’s substrates to ensure tight control over the required<br />

signalling paths. There are two main isoforms of PKA-R, type I<br />

and type II. Both have specific preferences for different<br />

AKAPs. For many AKAPs their preference is currently unknown<br />

and the existence of type I specific AKAPs is predicted,<br />

however not established [2]. Furthermore, due to their low<br />

concentrations in cells and tissues, it is a challenge to study<br />

AKAPs using traditional proteomics methods.


NH<br />

NH<br />

NH 2<br />

N<br />

N<br />

NH<br />

NH 2<br />

NH<br />

HN<br />

HN<br />

N N<br />

N N<br />

O<br />

O<br />

O<br />

O<br />

OH O P OH<br />

O O P OH<br />

H 3C<br />

O<br />

O<br />

8-AHA-2’OMe-cAMP (C8-OCH3) 8AHA-cAMP (C8)<br />

Protein Isolation<br />

C<br />

C<br />

N<br />

N<br />

Protein Digestion<br />

Light labeling<br />

Heavy labeling<br />

LC-MS/MS<br />

PKA RIIα<br />

PKA RIα<br />

m/z<br />

m/z<br />

Figure 2 | Experimental setup of the comparison of the cellular protein<br />

interactors of two cAMP analogues by stable isotope labelling and<br />

quantitative mass spectrometry.<br />

On the basis of these observed ratios, six AKAPs behaved as<br />

perfectly PKA-RII interacting proteins, while the other six have<br />

ratios that indicate interaction with both PKA-RI and PKA-RII.<br />

Of these, AKAP1 and AKAP10 are well described so-called dual<br />

specific AKAPs, explaining their intermediate ratios. These<br />

analyses confirmed the known specificity of several AKAPs<br />

and the specificity of AKAP14, AKAP2 and AKAP12 could be<br />

established.<br />

Interactions of sildenafil As described above, the same<br />

technology can be used to determine which proteins interact<br />

with specific drugs, like sildenafil. Sildenafil — sold under the<br />

brand name Viagra — is an inhibitor of phosphodiesterase-5<br />

(PDE-5). PDE proteins form a family of enzymes that hydrolyze<br />

cyclic nucleotides, with PDE-5 specifically targeting cGMP.<br />

PDE-5 is an interesting pharmaceutical target as its inhibition<br />

enhances the activity of the nitric oxide-cGMP pathway, which<br />

is involved in, amongst others, penile erection. Several potent<br />

PDE-5 inhibitors, including sildenafil, are now widely used for<br />

the treatment of erectile dysfunction. These inhibitors have a<br />

track record of being safe and well-tolerated and are considered<br />

to have a very good specificity for their target protein,<br />

PDE-5. Whether these inhibitors can interact with any other<br />

cellular protein targets, is largely unknown.<br />

To identify other potential targets of sildenafil, we generated<br />

several compounds that are structurally related to sildenafil<br />

20 | NPC Highlights 11 | April 2010<br />

| Arjen Scholten<br />

Figure 3 | Analysis of the interaction of different proteins (including PDE-5,<br />

Screening binding specificities We recently developed a<br />

chemical proteomics method based on immobilized cAMP to<br />

isolate and identify a large set of cAMP binding proteins, such<br />

as several PKA-Rs, PDEs, EPACs and more than ten different<br />

AKAPs [3]. In the present study, this method was adapted to<br />

screen a large variety of AKAPs for their binding preference<br />

towards PKA-RI and/or PKA-RII, in cell and tissue extracts [4].<br />

To do so, two immobilized cAMP analogues were used in<br />

parallel for the isolation of target proteins: 8-AHA-cAMP and<br />

2’-OMe-8AHA-cAMP (see Figure 2).<br />

The former interacts equally well with PKA-RI and PKA-RII,<br />

whereas the latter has a three- to four-fold higher affinity<br />

for PKA-RI. As the 2’-OMe beads will effectively isolate more<br />

PKA-RI, also more of its associated AKAPs will present in the<br />

corresponding pull-downs. By comparing the protein enrichment<br />

patterns of both cAMP analogues, using dimethyl stable<br />

isotope labelling and quantitative mass spectrometry, the<br />

binding specificities of AKAPs can be elucidated by comparing<br />

their enrichment behaviour to that of the different PKA<br />

isoforms.<br />

As a proof of concept, we applied this screening method to<br />

two different cell lysates (HEK293 and RCC10) and two different<br />

rat tissues (lung and testis), from which we isolated<br />

and quantified twelve different AKAPs. As expected, the<br />

enrichment pattern of PKA-RI and PKA-RII is distinguishable by<br />

their relative efficiency of recovery from the two pull-downs.<br />

PEBP-2) with several immobilized analogues of the drug sildenafil by label<br />

free quantitative mass spectrometry and confirmation by western blotting.


and that could be immobilized on beads. Using these compounds,<br />

we then performed pull-downs from various rat tissue<br />

samples, followed by (semi-) quantitative LC-MS/MS to identify<br />

the interacting proteins and to compare their affinity for<br />

the different sildenafil analogues. Using this method, we were<br />

able to identify several proteins that could interact directly<br />

with the immobilized sildenafil, albeit with lower affinity than<br />

the main target, PDE-5 (see Figure 3) [5].<br />

Some of these proteins were other members of the PDE<br />

family, but also the family of phosphatidylethanolamine binding<br />

proteins (PEBPs), which includes the abundantly present<br />

Raf-kinase inhibitory protein (RKIP), was found to interact<br />

with sildenafil analogues [6]. Interestingly, some of the<br />

sildenafil analogues that were used in this experiment had a<br />

very different affinity for the various identified proteins, with<br />

one of them showing significantly increased specificity for the<br />

PEBP family of proteins (see Figure 3). This shows that chemical<br />

proteomics, particularly in combination with (small) drug<br />

libraries can be used to identify novel targets for (derivatives<br />

of) existing drugs [7].<br />

3 Scholten, A. et al. (2006) Analysis of the cGMP/cAMP<br />

interactome using a chemical proteomics approach in<br />

mammalian heart tissue validates sphingosine kinase type<br />

1-interacting protein as a genuine and highly abundant<br />

AKAP. J Proteome Res 5, 1435-1447.<br />

4 Aye, T. T. et al. (2009) Selectivity in enrichment of cAMPdependent<br />

protein kinase regulatory subunits type I and<br />

type II and their interactors using modified cAMP affinity<br />

resins. Mol Cell <strong>Proteomics</strong> 8, 1016-1028.<br />

5 Dadvar, P. et al. (2009) A chemical proteomics based enrichment<br />

technique targeting the interactome of the PDE5<br />

inhibitor PF-4540124. Mol Biosyst 5, 472-482.<br />

6 Dadvar, P. et al. (2009) Phosphatidylethanolamine-binding<br />

proteins, including RKIP, exhibit affinity for phosphodiesterase-5<br />

inhibitors. Chembiochem 10, 2654-2662.<br />

7 Dadvar, P. (2009) Probing the drug interactome by chemical<br />

proteomics, PhD thesis.<br />

Research team<br />

Important challenge We have shown that chemical proteomics<br />

is a versatile tool to study the interactions of small molecules<br />

with a proteome that can be applied in many facets of biology.<br />

We applied the technology both to small signalling molecules<br />

like cAMP and to clinically relevant drugs like sildenafil to<br />

identify target proteins. To gain momentum in drug target<br />

discovery and other fields, the development of more chemical<br />

proteomic tools targeting a wider range of protein classes will<br />

be an important challenge for the coming years.<br />

| 21<br />

References<br />

1 Bantscheff, M., Scholten, A., Heck, A. J. (2009) Revealing<br />

promiscuous drug-target interactions by chemical proteomics.<br />

Drug Discov Today 14, 1021-1029.<br />

2 Scholten, A., Aye, T. T., Heck, A. J. (2008) A multi-angular<br />

mass spectrometric view at cyclic nucleotide dependent<br />

protein kinases: in vivo characterization and structure/<br />

function relationships. Mass Spectrom Rev 27, 331-353.<br />

Summary<br />

Chemical proteomics is an emerging research field that allows<br />

the characterization of all proteins interacting with small<br />

molecules using a combination of pull-downs with immobilized<br />

molecules and LC-MS/MS for identification. As we show here,<br />

this can be applied both to small signalling molecules like<br />

cAMP to reveal their cellular targets, and to clinically relevant<br />

drugs like sildenafil to identify targets and potential off-targets.<br />

By extending this strategy to make use of modified molecules<br />

<strong>summary</strong><br />

From left to right: Reinout Raijmakers, Albert Heck, Arjen<br />

Scholten.<br />

Contact<br />

Prof. Albert Heck<br />

Biomolecular Mass Spectrometry and <strong>Proteomics</strong> Group<br />

Utrecht University<br />

Padualaan 8,<br />

3584 CH Utrecht, The <strong>Netherlands</strong>.<br />

T +31 30 253 6797<br />

a.j.r.heck@uu.nl<br />

http://bioms.chem.uu.nl<br />

and quantitative mass spectrometric approaches, a detailed<br />

overview can be generated of the specificity certain proteins<br />

have for such molecules. This yields valuable information on<br />

drug specificity and on how abundantly present small molecules<br />

in cells can be part of different pathways.<br />

The work was performed at the Biomolecular Mass<br />

Spectrometry and <strong>Proteomics</strong> group at Utrecht University, in<br />

close collaboration with Pfizer Pharma Therapeutics, Sandwich<br />

Laboratories, United Kingdom (sildenafil) and the Medical<br />

Physiology department of the University Medical Hospital in<br />

Utrecht (cAMP).


Barbara Möller and<br />

Dolf Weijers<br />

Cell-cell communication<br />

in plant embryos<br />

22 | NPC Highlights 11 | April 2010<br />

The production of leaves, roots and flowers by plants<br />

throughout their entire life is carried out by the so-called<br />

meristems, growth tips in which stem cells are first formed.<br />

Meristems are located in the young plant embryo. We<br />

studied the formation of the root meristems in the embryo<br />

of the model plant Arabidopsis thaliana. The process<br />

begins with the programming of one cell as the ‘hypophysis’<br />

which contols stem cells in the roots. It is known that<br />

the formation of the hypophysis is controlled by the gene<br />

activator called MONOPTEROS. However, it was hitherto not<br />

known how this activator regulates hypophysis formation.<br />

Plants start their lives simple, as seedlings with one or two<br />

leaves, a small stem and a root. Later however, the plant body<br />

is further elaborated by the formation of new leaves, stems,<br />

branches, flowers and roots. Hence, the mature plant often<br />

does not even remotely resemble the seedling. Plants owe this<br />

remarkable capacity to generate many new organs to the activity<br />

of meristems, growth tips harbouring the plant equivalent<br />

of stem cells. These stem cells can be active over many<br />

years, and are prevented from differentiation by organizer<br />

cells. In our research group, we are interested in the problem<br />

of how meristems (stem cells and organizer) are first formed<br />

in the young plant embryo.<br />

Like animals plants derive from a single cell, the zygote,<br />

and all cell types and organs, including the meristems,<br />

are formed early during embryogenesis. The meristem<br />

that will give rise to the entire root system (root meristem)<br />

is formed when the embryo of the model plant<br />

Arabidopsis thaliana is approximately 50 cells in size (see<br />

Figure 1). We have previously shown that the transcription<br />

factor MONOPTEROS (MP), upon its activation by the<br />

plant hormone auxin, promotes the formation of the root<br />

meristem organizer (hypophysis). Intriguingly however,<br />

MP does not act in this cell, but promotes its specification<br />

from the adjacent cells. Hence, the formation of<br />

the first meristem involves cell-cell communication.<br />

The signal that activates MP, auxin, is itself transported<br />

from the MP-expressing cells to the hypophysis cell, but<br />

this is alone not sufficient for specification of the cell<br />

as organizer. We therefore postulated the existence of a<br />

hypothetical second MP-dependent signal that is transported<br />

to the hypophysis [1].


What this research is about:<br />

Discovery of essential protein for<br />

early root formation in plants<br />

How do plant embryonic cells know which organs they should develop into Dolf Weijers,<br />

assistant professor at Wageningen University and his group, together with his German<br />

colleagues from the University of Tübingen and the Max Planck Institute, studied how the<br />

early formation of root tissue is controlled. They discovered a mobile transcription factor<br />

which is essential in the process. The results were recently published in Nature.<br />

In contrast to animals, plants produce new organs, in other words stems, leaves, roots<br />

and flowers, throughout their lifetime. Stem cells are located in the growing tips of roots<br />

and are responsible for the growth of new roots. “Relatively little is known about early<br />

embryogenesis in plants and how one cell is programmed to become for example a root<br />

founder cell,” says Weijers.<br />

In earlier studies on embryonic cells in the model organism, Arabidopsis thaliana,<br />

Weijers and his colleagues discovered that for a cell to become the root founder cell, the<br />

plant hormone auxin is required. This hormone is sent by neighboring embryo cells. The<br />

transcription factor MONOPTEROS (MP) promotes the transport of the hormone to the<br />

neighboring cell.<br />

“However, this is not sufficient on its own,” says Weijers, “so we looked for other genes<br />

which are expressed exclusively in the presence of MP.” In a comprehensive survey of all<br />

the genes activated by MP, the researchers identified the gene TMO7. “We discovered that<br />

the protein formed by the TMO7 gene migrates from the location of its synthesis in the<br />

embryo to the root founder cell. So both auxin and TMO7 are mobile factors activated by MP<br />

and required as signals for root formation in the embryo.” The detective work in the plant<br />

researchers’ genetics laboratory does not end here, however. “We will investigate which<br />

other factors are involved and unravel more of the regulatory network that controls plant<br />

embryogenesis,” says Weijers.<br />

| 23<br />

NPC Research Theme T2: Proteome Biology of Plants<br />

Microarray-based approach In a study that was recently<br />

published [2], we have taken a microarray-based approach<br />

in order to find genes, pathways and eventually intercellular<br />

signals that operate downstream of MP in root meristem<br />

formation. Affymetrix genechips, featuring 22,000 of<br />

the approximately 26,000 Arabidopsis genes, were hybridized<br />

with mRNA isolated from normal seedlings, seedlings<br />

lacking MP altogether, or seedlings in which MP had been<br />

inhibited during 1 hour. After statistical analysis, we obtained<br />

a set of about 100 genes that is robustly downregulated<br />

in the absence of MP activity. When analyzing<br />

gene ontology of these 100 genes, we found that genes<br />

encoding transcription factors were strongly over-represented.<br />

This suggests that MP primarily regulates other<br />

transcription factors, and sits at the summit of a transcriptional<br />

network.<br />

To determine which of these genes are controlled by MP during<br />

early embryogenesis, we analyzed the expression patterns of<br />

a subset (including most transcription factor genes) in normal<br />

embryos and in embryos lacking MP. We found 4 genes that<br />

we named TARGET OF MONOPTEROS (TMO), to be expressed<br />

in the sub-domain of the embryo where MP acts. As expected,<br />

their expression depends on MP activity.<br />

Supported by funding from the NPC (Grant no. NPC4.11<br />

awarded to Sacco de Vries), and helped by the group of Gerco<br />

Angenent, we used Chromatin Immunoprecipitation (ChIP) to<br />

show that MP binds in vivo to the promoters of 3 of these 4<br />

TMO genes. Furthermore, functional genetic investigations<br />

showed that TMO5 and TMO7 both mediate MP-dependent<br />

root formation, and that elimination of TMO7 leads to rootless<br />

defects similar to those caused by the loss of MP.


Auxin<br />

Stem cell divisions<br />

TMO5<br />

MP<br />

Auxin<br />

TMO7<br />

Auxin TMO7<br />

Hypophysis fate<br />

Figure 1 | Model for embryonic root initiation.<br />

Left: Arabidopsis thaliana embryo at globular stage consists of a sphere of<br />

embryonic cells and a filament of extra-embryonic cells. The root meristem<br />

forms at the junction of both and involves inner cells of the embryo (pink)<br />

and the uppermost extra-embryonic cell (yellow).<br />

Right: MP is activated by auxin in the embryonic cells (pink, top), and<br />

activates expression of TMO5 and TMO7 genes. TMO5 remains in these cells<br />

and contributes to stem cell divisions. TMO7 protein moves to the adjacent<br />

extra-embryonic cell (yellow, bottom), where it contributes to specifying<br />

this cell as the organizer (hypophysis). To achieve this, MP also promotes<br />

transport of auxin to the future hypophysis.<br />

24 | NPC Highlights 11 | April 2010<br />

TMO5 and TMO7 encode proteins belonging to the same family<br />

of basic Helix-Loop-Helix (bHLH) transcription factors. This<br />

family mediates cell fate specification in other developmental<br />

processes in plants [3], but also in animals [4]. To determine<br />

how these two bHLH factors contribute to MP-dependent root<br />

formation, we analyzed the localization of both proteins.<br />

We made the remarkable discovery that, while TMO5 protein<br />

remains in the cells where it is activated by MP, TMO7 protein<br />

moves to the hypophysis cell (see Figure 1). This transport<br />

depends on the size of the protein, since increasing its size by<br />

adding 3xGFP impairs movement. Therefore, TMO7 represents<br />

a novel intercellular signal molecule in embryonic root<br />

formation. Even though the mechanism of action of this novel<br />

intercellular signal is not known, it appears that its movement<br />

to the hypophysis is critical for its function in root formation.<br />

considered necessary for DNA binding [4]. The protein is localized<br />

both in cytoplasm and nucleus of the cells it is produced<br />

in, and is mostly nuclear in the cells it is transported to [2].<br />

Several scenarios could explain this localization, among which<br />

are either passive diffusion or active transport and blocking<br />

of further movement by nuclear trapping. No matter what the<br />

mechanisms are, we expect that protein-protein interactions<br />

will be critical to achieve limited directional transport. We are<br />

currently in the process of identifying proteins that interact<br />

with a mobile, or with a non-mobile form of Green Fluorescent<br />

Protein (GFP) – tagged TMO7 protein using immunoprecipitation<br />

(IP) followed by nano-Liquid Chromatography-tandem<br />

Mass Spectrometry (nLC-MS/MS). We have previously used this<br />

method, established by the group of Sacco de Vries, to identify<br />

in vivo interactors of- and phosphorylation sites on plant<br />

proteins [5,6]. From this analysis we expect to identify factors<br />

involved in TMO7 transport. Since bHLH transcription factors<br />

likely act as dimers [4], we also expect to find the transcription<br />

factor with which TMO7 interacts in the hypophysis.<br />

2. How is root formation spatially coordinated through the<br />

TMO transcription factors<br />

TMO5 and TMO7 are both direct MP target genes, and activated<br />

in precisely the same cells. Both are required for root formation,<br />

but in different cells. TMO5 is active in the future stem<br />

cells and TMO7 acts in the future organizer cell (hypophysis).<br />

Certainly, different genes will be controlled in each of these<br />

cell types. We now use these factors to determine which genes<br />

and cellular pathways are controlled in both of these cell types<br />

in order to generate a functional meristem (stem cells and<br />

organizer). We will again employ microarray technology to<br />

identify target genes of the TMO5 and TMO7 bHLH transcription<br />

factors.<br />

3. What controls MP activity<br />

In addition to its role in promoting root formation in the embryo,<br />

MP has many other functions. In the embryo, MP controls<br />

formation of the vascular tissues, of embryonic leaves, and<br />

has several other, post-embryonic functions [7]. To accommodate<br />

all these activities, MP is expressed in a rather broad pattern<br />

(see Figure 2). The TMO genes however, are expressed in<br />

much narrower domains (FIG). A crucial question is how MP ac-<br />

| Dolf Weijers<br />

Figure 2 | Expression patterns of MP, TMO5 and TMO7.<br />

Specific questions The identification of a set of genes that is<br />

directly controlled by the key regulatory transcription factor<br />

MP, and the demonstration that TMO7 is a mobile transcription<br />

factor, now allow asking a number of specific questions.<br />

These are all being addressed in our research group, and will<br />

be discussed below.<br />

1. What is the mechanism of transport and action of the<br />

mobile TMO7 transcription factor<br />

TMO7 is a very small transcription factor, only 11 kDa in<br />

size, and lacks the basic region of the bHLH domain that is<br />

MP TMO5 TMO7<br />

During the heart stage of embryogenesis, MP is expressed in a wide pattern<br />

(left). While some the direct target gene TMO5 (middle) is expressed in a<br />

similar pattern, TMO7 (right) is expressed in a small subdomain that corresponds<br />

to the root stem cells. Schemes represent an interpretation of MP<br />

protein and TMO5 and TMO7 mRNA accumulation patterns.


tivates one target gene in a broad pattern, while it activates<br />

another in a very restricted domain. Our working hypothesis<br />

is that MP assembles into different transcription complexes to<br />

activate distinct sets of genes in different cells.<br />

To determine MP transcription complexes, we have adapted<br />

an immunoprecipitation and nLC-MS/MS procedure for isolating<br />

nuclear proteins and their interacting proteins. We now<br />

use this method to isolate MP-GFP protein from embryos and<br />

identify its co-factors.<br />

6 Smith, M. et al. (2009) Cyclophilin40 is required for miRNA activity<br />

in Arabidopsis. Proc Natl Acad Sci USA 106, 5424-5429.<br />

7 Hardtke C.S. et al. (2004) Overlapping and non-redundant<br />

functions of the Arabidopsis auxin response factors<br />

MONOPTEROS and NONPHOTOTROPIC HYPOCOTYL 4.<br />

Development 131, 1089-1100.<br />

8 Remington D.L. et al. (2004) Contrasting modes of diversification<br />

in the Aux/IAA and ARF gene families. Plant Physiol<br />

135, 1738-1752.<br />

A large gene family In our recent work we have defined a<br />

cell-cell signaling process as being critical for the formation of<br />

the first meristem in the plant embryo, and we have identified<br />

an intercellular communication signal. Interestingly, MP is a<br />

member of a large gene family of AUXIN RESPONSE FACTORs<br />

(ARFs), transcription factors that control gene expression in<br />

response to auxin. Because auxin is itself transported to the<br />

hypophysis, we anticipate that other ARF transcription factors<br />

act in the hypophysis and perhaps in other pattern formation<br />

processes in the plant embryo. We are currently investigating<br />

the developmental role of the other members of the ARF<br />

family in plant embryo development, and hope to contribute<br />

to understanding the molecular basis for the ability of auxin to<br />

trigger many different responses in plant development.<br />

Research team<br />

1 2 3<br />

4<br />

5<br />

6 7 8<br />

References<br />

1 Weijers, D. et al. (2006) Auxin triggers transient, local signalling<br />

for cell specification in Arabidopsis embryogenesis.<br />

Dev Cell 10, 265-270.<br />

2 Schlereth, A.S et al. (2010). MONOPTEROS controls embryonic<br />

root formation by regulating a mobile trancription<br />

factor. Nature, Advance Online Publication doi: :10.1038/<br />

nature08836.<br />

3 Pillitteri, L.J. and Torii, K.U. (2007) Breaking the silence:<br />

three bHLH proteins direct cell-fate decisions during stomatal<br />

development. Bioessays 29, 861-870.<br />

4 Massari, M. E. and Murre, C. (2000) Helix-loop-helix proteins:<br />

regulators of transcription in eukaryotic organisms.<br />

Mol Cell Biol 20, 429–440.<br />

5 Michniewicz, M. et al. (2007) Antagonistic regulation of PIN<br />

phosphorylation by PP2A and PINOID directs auxin flux. Cell<br />

130, 1044-1056.<br />

Summary<br />

Plants make new organs throughout their lives due to the<br />

activity of stem cells located in meristems. Our group is<br />

interested in the problem of how stem cells and meristems are<br />

initated in the young plant embryo. We have previously shown<br />

that root meristem initiation in the plant embryo requires<br />

cell-cell communication between adjacent embryonic and<br />

extra-embryonic cells. Recently, through identifying direct tar<strong>summary</strong><br />

Dolf Weijers (1), Barbara Möller (2), Bert de Rybel (3),<br />

Eike Rademacher (4), Annemarie Lokerse (5), Cristina Llavata<br />

Peris (6), Willy van den Berg (7), Giulinano Giuliani (8)<br />

Contact<br />

Dr. Dolf Weijers<br />

Laboratory of Biochemistry<br />

Wageningen University<br />

Dreijenlaan 3<br />

6703 HA Wageningen<br />

the <strong>Netherlands</strong><br />

T +31 317 482 866<br />

dolf.weijers@wur.nl<br />

www.bic.wur.nl/UK/research/Plant+Development/<br />

get genes of the critical transcription factor MONOPTEROS, we<br />

have discovered a novel intercellular signaling molecule. This<br />

molecule, the small transcription factor TMO7, is transported<br />

from the embryonic to the extra-embryonic cells to initiate<br />

the stem cell organizer. Here we discuss these findings and describe<br />

our current and future work aimed at understanding the<br />

cell-cell communication mechanism and the transcriptional<br />

networks underlying the formation of a functional stem cell<br />

population in the plant embryo.<br />

| 25


<strong>Proteomics</strong> for the analysis of<br />

cellular networks<br />

Interview with keynote speakers<br />

Increasingly more mathematical modelling and simulation is<br />

used to explain and predict behaviour of cellular networks<br />

under different circumstances. “Mathematical modelling<br />

requires good quantitative data and these are not easy to<br />

obtain from complex protein mixtures,” says Paola Picotti. She<br />

therefore designed assays based on a targeted approach to<br />

obtain quantitative data from the yeast proteome.<br />

“Proteins are fascinating structures,” says Paola Picotti, PhD.<br />

“It’s intriguing to find out how they fold and how they perform<br />

their function.” For almost ten years Picotti has been involved<br />

in studying proteins using different spectroscopic and mass<br />

spectrometric techniques. “I started studying single proteins<br />

and I am now involved in studying complete systems of interacting<br />

molecules.” Picotti joined Prof. Ruedi Aebersold’s group<br />

at ETH Zurich in 2006 as a post-doctoral fellow to learn more<br />

about advanced mass spectrometric techniques. She focuses<br />

on the development of targeted proteomic techniques based<br />

on selected reaction monitoring.<br />

26 | NPC Highlights 11 | April 2010<br />

Targeted approach<br />

Obtaining quantitative data from cellular networks, e.g. metabolic<br />

or signalling networks, is difficult because the proteins<br />

are present in very different concentrations and some of<br />

them have a high sequence similarity. The classical method of<br />

shotgun proteomics is not always efficient enough to monitor<br />

all the proteins of interest. It means the digestion of protein<br />

mixtures, separation of proteins by liquid chromatography and<br />

identification of these proteins by tandem mass spectrometry.<br />

Picotti and her colleagues therefore decided to try a targeted<br />

approach. They created a list from all the known proteins<br />

in the network under investigation. They selected proteotypic<br />

peptides (PTPs), peptides that are unique to a protein<br />

from each protein. They then developed highly sensitive and<br />

quantitative MS assays for each PTP based on selected reaction<br />

monitoring (SRM). “It means that you allow only specified<br />

peptides to be measured, based on MS coordinates that you<br />

have selected to be specific to the peptide,” explains Picotti.<br />

The technique works well; proteins with abundances between<br />

1.3 million and less than 50 copies per cell could be detected<br />

in total yeast proteome digests, without the need for fractionation<br />

or enrichment.<br />

Synthetic peptides<br />

The proof-of-principle of the SRM assay was delivered by the<br />

analysis of a real network, yeast carbon metabolism. “It was<br />

really nice to see that we were able to measure varying concentrations<br />

of metabolic proteins within the entire network in<br />

one hour. However the development and validation of all the<br />

Paola Picotti (IMSB, ETH Zurich) is specialised in the field of spectroscopic<br />

and mass spectrometric techniques. She gave a keynote lecture during the<br />

NPC Progress Meeting 2010 in Utrecht on 16 February.<br />

different SRM assays was tedious and very time-consuming,”<br />

remarks Picotti. “Therefore we developed SRM assays on the<br />

basis of crude synthetic proteotypic peptide libraries. Since<br />

the composition of a synthetic library is known exactly and the<br />

concentrations of the peptides are similar, the confidence in<br />

the synthetic-peptide-based SRM assays is much higher. It<br />

allows us to develop assays much faster than before: we<br />

needed 8 hours for 150 different kinases and phosphatases.”<br />

Picotti and her colleagues have now also developed assays for<br />

the complete yeast proteome, a total of 6,500 assays (for all<br />

the open reading frames). The assays will be put onto a public<br />

website, the MRMAtlas (www.mrmatlas.org), so that anyone<br />

can use them for their own applications. They can also be<br />

“Proteins are fascinating structures.<br />

It’s intriguing to find out how they fold and how<br />

they perform their function”<br />

used for trying to measure proteins so far undetected in different<br />

conditions. About 2,000 proteins which have not been<br />

detected yet are coded by the yeast genome.<br />

Challenging<br />

According to Picotti, it’s now time to have fun with SRM: “We<br />

can focus more on the biological instead of the technological<br />

development, which took a lot of time.”<br />

The next project is the human proteome which is very challenging.<br />

“It may be more difficult to find unique peptides and<br />

the dynamic range of protein abundance will be bigger. We<br />

will focus on the first 9,000 proteins already observed by other<br />

researchers.”<br />

Lilian Vermeer


NPC Progress Meeting 2010<br />

Understanding neuropeptide<br />

complexity in the brain<br />

Unravelling the chemistry occurring in the brain that<br />

influences behaviour is a topic that greatly intrigues Jonathan<br />

Sweedler. It is part of his fascination with neuroscience and<br />

more specifically, in understanding the functioning of the<br />

brain. His research group develops and uses new analytical<br />

tools for nanoscale study of neuronal networks to help<br />

understand cell to cell signalling in the brain.<br />

“Neuropeptides are essential molecules which play an important<br />

role in behaviour, learning and memory,” says Jonathan<br />

Sweedler, professor of chemistry at the University of Illinois,<br />

USA. “However neuropeptides are hard to study because they<br />

are heterogeneously distributed, contain a large number of<br />

post-translational modifications, and are active over a wide<br />

range of physiological concentrations. Furthermore they can<br />

quickly degrade after cellular release.”<br />

Fortunately today the sensitivity of detection methods has improved<br />

in order of magnitude. Sweedler: “Whereas in the past<br />

an ‘entire slaughter house’ was needed to isolate enough purified<br />

peptide material to characterize it, today much smaller<br />

quantities are sufficient.” Sweedler’s group has contributed to<br />

the improvement of detection methods by developing many<br />

new peptidomic approaches for assaying nanoliter volume<br />

samples. They have also developed optimal preparation protocols<br />

for small-volume samples. These methods enabled them<br />

to characterize signalling molecules in samples ranging from<br />

single cells to entire brain regions.<br />

Well conserved<br />

“The nice thing about neurotransmitters and neuromodulators<br />

is that they are remarkably well conserved across many different<br />

species,” says Sweedler. “Therefore we can use a range<br />

of animal models to study their neuronal networks, choosing<br />

the most appropriate animal depending on our research<br />

question. We also use several models to develop and test new<br />

techniques.” Consequently Sweedler’s lab is like a zoo: species<br />

ranging from rats, mice, birds to honey bees and sea slugs are<br />

their neuropeptide research models.<br />

“Neuropeptides are essential molecules<br />

which play an important role in<br />

behaviour, learning and memory”<br />

One of Sweedler’s ‘favourite’ animals is the sea slug, Aplysia<br />

californica. “This animal is frequently used in neuroscience<br />

because it has a simple nervous system, consisting of just a<br />

few thousand large, easily-identified neurons. Moreover, this<br />

animal is able to learn and remember what it learns.<br />

Jonathan Sweedler (University of Illinois, USA) gave a keynote lecture<br />

during the NPC Progress Meeting 2010, in Utrecht on 16 February.<br />

Eric Kandel, with critical help from Aplysia, received a Nobel<br />

Prize in 2000 for boosting our understanding of the biological<br />

basis of learning and memory.” Sweedler’s group is involved in<br />

the annotation of neuropeptides from Aplysia and they have<br />

characterized hundreds of unique peptides from whole brains<br />

to single cells in Aplysia.<br />

Molecular mechanisms<br />

To help understand the molecular mechanisms behind an<br />

animal’s circadian rhythm, Sweedler’s group characterized<br />

endogenous neuropeptides from a specific region of the rat<br />

brain by mass spectrometry. This region, the suprachiasmatic<br />

nucleus (SCN), is known to control circadian rhythm, the timekeeping<br />

system responsible for our daily rhythms. In addition<br />

to established circadian neuropeptides, they found peptides<br />

with unknown roles. One of them, called little SAAS, induces a<br />

phase delay in the circadian system after exogenous application.<br />

Therefore, the MS-based discovery of a peptide has led<br />

to new insights into how we maintain our circadian rhythm.<br />

The Sweedler group identified hundreds of peptides in the<br />

honey bee brain, and is now trying to determine which ones<br />

are bioactive. By quantifying peptide level changes during<br />

food selection and foraging, they found that some peptides<br />

were clearly more abundant in the brains of bees after they<br />

had collected either nectar or pollen. “Bees appear predisposed<br />

to collect a specific type of food and this predisposition<br />

is observable in their brain peptide signature,” says Sweedler.<br />

Future<br />

Sweedler realizes that advances in mass spectrometer instrumentation<br />

enable the discovery of unexpected neuropeptides<br />

at an increasing rate. “However this has also created many<br />

new questions. So who knows what we will discover in the<br />

future!”<br />

Lilian Vermeer<br />

| 27


Valorisation within the<br />

Dear NPC Highlights readers,<br />

Recently we introduced the NPC Valorisation Voucher as a new NPC instrument to enable<br />

proof-of-concept studies for valorisation. There appears to be a great need for this funding,<br />

as we have witnessed many applications upon going public. Two NPC Valorisation Vouchers<br />

have now been granted by the board. Apart from the application from Huib Ovaa (NKI) the<br />

board recently approved an application from Bobby Florea (Leiden University). Both awardees<br />

will present their business plans elsewhere on these pages.<br />

I would like to bring to your attention again that the NPC Valorisation Voucher is truly<br />

meant for valorisation plans with a clear market potential. One could for example think of<br />

beginning a start-up business or supporting the out-licensing of patent rights and obtaining<br />

Bas Nagelkerken<br />

the last proof-of-concept to close a deal. This implies that projects that are in a too early<br />

NPC Valorisation Manager<br />

stage cannot yet be eligible for funding from this source. If you have valorisation plans please<br />

contact me to discuss your ideas. Together we can decide whether your plans are suitable for<br />

submitting for a Voucher or if there might be other ways to promote your initiative.<br />

I’m looking forward to receiving many of your interesting proposals this year!<br />

Contact<br />

All the best!<br />

<strong>Netherlands</strong> <strong>Proteomics</strong> <strong>Centre</strong><br />

+31 30 253 4564<br />

Bas Nagelkerken<br />

nagelkerken@npc.genomics.nl<br />

NPC Valorisation Manager<br />

www.netherlandsproteomicscentre.nl<br />

28 | NPC Highlights 11 | April 2010 | Valorisation Horizon Valorisation Grant<br />

Life Sciences Pre-Seed Grant<br />

The Life Sciences Pre-Seed Grant offers a great opportunity<br />

for researchers associated with a Dutch university or research<br />

institution. Worth up to € 250,000, the Pre-Seed Grant<br />

offers superb prospects for those involved in applied research<br />

and who are looking to exploit their fundamental research<br />

commercially by starting up a new business. The Life Science<br />

Pre-Seed grant can be positioned as a next step to a NPC<br />

Valorisation Voucher. Deadline upcoming call: 5 October 2010.<br />

More information: www.preseedgrant.nl<br />

New Venture Business Plan<br />

Competition<br />

New Venture is aimed at anyone who wants to enterprise<br />

with an innovative idea. An idea is innovative if it provides<br />

satisfaction to a new or an existing need in a new way, and<br />

if it clearly adds something compared to existing products,<br />

services or technologies. Each round, participants can win<br />

prize money with their idea, feasibility study or business plan.<br />

These winnings rise to three prizes of € 25,000, in the third<br />

round. Moreover, the national press devotes considerable<br />

attention to the winners of New Venture. Each round ends<br />

with a festive ceremony.<br />

More information: www.newventure.nl<br />

The Horizon Valorisation Project Grant is a new valorisation<br />

tool available for researchers that have already been awarded<br />

a Horizon Breakthrough project to further support valorisation<br />

of their research results. The grant may be used to validate<br />

research, or perform pre-clinical research or (further) clinical<br />

research to improve the valorisation potential of the research<br />

as performed in the Horizon Breakthrough project. The<br />

maximum grant per Horizon Valorisation project is € 50,000.<br />

The proposals may be submitted throughout the year, but within<br />

three months after the ending of the corresponding Horizon<br />

Breakthrough project. Before applying, the candidate should<br />

contact NGI to receive a personal link to the ProjectNet system.<br />

For detailed information on the programme, visit the websites<br />

of ZonMw (www.zonmw.nl) or NGI (www.genomics.nl) and click<br />

on the Horizon page.<br />

Patent Workshop 2010<br />

The <strong>Netherlands</strong> Genomics Initiative, the NL Patent Office<br />

and the Programme Office IOP Genomics kindly invite you<br />

to participate in the Patent Workshop 2010. This workshop<br />

will discuss basic patent facts, as well as look at knowledge<br />

protection from a genomics/Life Science point of view.<br />

If you are a PhD student, postdoc or project leader from either<br />

academia or a company wanting to know more about patents,<br />

this is the workshop for you! The workshop takes place on<br />

11 May 2010 at the Patent Office in Rijswijk and is free of charge.<br />

For more information or registration, please contact<br />

Ms. Anjali Raghoenath anjali.raghoenath@agentschapnl.nl


npc valorisation<br />

<strong>Netherlands</strong> <strong>Proteomics</strong> <strong>Centre</strong><br />

Two NPC Valorisation Vouchers have now been granted<br />

UbiQ: commercial development<br />

of synthetic Nedd8 and SUMO<br />

conjugates<br />

The start-up company UbiQ originates from research<br />

developed in the Chemical Biology group at the <strong>Netherlands</strong><br />

Cancer Institute. The UbiQ team is pleased to be the first<br />

recipient of the <strong>Netherlands</strong> <strong>Proteomics</strong> <strong>Centre</strong> Valorisation<br />

Voucher for their project ‘Commercial development of<br />

synthetic Nedd8 and SUMO conjugates’.<br />

The expertise of UbiQ is the development of research<br />

tools and assay reagents based on Ubiquitin and Ubiquitinlike<br />

proteins. These are small proteins that act as posttranslational<br />

modifiers and are involved in regulating various<br />

biologically important processes. To date, the development<br />

of these types of reagents has been hampered by the<br />

complexity of their biological modification. UbiQ has solved<br />

this problem by developing a highly efficient chemical ligation<br />

technology that gives access to these well-defined protein<br />

conjugates. With this proprietary technology they have<br />

developed reagents for profiling the substrate specificity of<br />

deubiquitinating enzymes and the high-throughput screening<br />

of deubiquitinating enzyme inhibitors.<br />

With the help of the NPC Valorisation Voucher UbiQ will<br />

develop (assay) reagents based on the Ubiquitin-like proteins<br />

Chemical <strong>Proteomics</strong> Reagents<br />

Chemical <strong>Proteomics</strong> Reagents (CPR) is an entrepreneurial<br />

initiative from the Bio-organic synthesis group at the Leiden<br />

Institute for Chemistry that received an NPC valorisation<br />

voucher of 85,000 Euros. This grant gives the CPR team the<br />

opportunity to perform a market feasibility study in the next<br />

18 months.<br />

CPR has profound expertise in design and synthesis of<br />

activity-based probes (ABP) for functional proteomics studies.<br />

These probes contain an enzyme reactive group, a targeting<br />

sequence and a reporter group. The technique has successfully<br />

been use for profiling the proteasome [1] and cathepsin<br />

protein hydrolases [2] in vitro, in living cells and animals.<br />

During the start-up period, CPR will focus on serving the<br />

scientific community by marketing novel ABPs and improving<br />

functional and chemical proteomics work flows for profiling<br />

protein hydrolases. Interesting enzymatic activities such as<br />

glucosidases and kinases are in the pipe-line.<br />

CPR starts with a team of three: Bobby Florea as executive<br />

officer for assay development, business administration,<br />

marketing and sales, Rian van den Nieuwendijk as technology<br />

officer for synthesis, Q&A, distribution and logistics and<br />

Hermen Overkleeft for advisory, marketing and PR tasks.<br />

The founders expect a turnover of 20-50 k€ in the first 2 years<br />

of operation followed by robust growth rates provided by full<br />

The UbiQ team (from left): Alfred Nijkerk (CEO), Farid El Oualid (COO) and<br />

Huib Ovaa (CSO).<br />

Nedd8 and SUMO. Since they are potentially new drug<br />

targets, there has been an increasing demand for well-defined<br />

Nedd8 and SUMO based (assay) reagents and a large market<br />

potential. It is interesting to draw a comparison with research<br />

into kinases, which has been one of the prime targets of<br />

pharmaceutical industry over the past decade. For a long<br />

time, this field has had the luxury of an array of techniques<br />

based on the availability of phosphorylated peptides. UbiQ will<br />

fill this commercial space for the Ubiquitin, Nedd8 and SUMO<br />

research market.<br />

Contact<br />

Huib Ovaa & Farid El Oualid<br />

+31 20 512 1979; h.ovaa@nki.nl<br />

The CPR team consist of (from left): Bobby Florea, Hermen Overkleeft and<br />

Rian van den Nieuwendijk<br />

spectrum chemical proteomics services for preclinical assays,<br />

target identification, validation and activity quantification.<br />

References<br />

1 Verdoes et al. (2006) A fluorescent broad-spectrum proteasome inhibitor for<br />

labelling proteasomes in vitro and in vivo. Chem & Biol 13, 1217-26.<br />

2 Hillaert et al. (2009) Receptor-mediated targeting of cathepsins in professional<br />

antigen presenting cells. Angew Chem Int Ed Engl 48, 1629-32.<br />

Contact<br />

Bogdan Florea<br />

+31 71 527 4355; b.florea@chem.leidenuniv.nl<br />

| 29


Bioinformatics<br />

High speed light path network<br />

In 2009 SURFnet and NWO successfully organized the<br />

‘Enlighten your research’ competition for the second<br />

time, a competition on dynamic light paths. The goal<br />

was to implement high speed and low latency response<br />

time-dedicated internet connections in the existing<br />

research infrastructure via optical fibres.<br />

Some of the bottlenecks in mass spectrometry based proteomics<br />

are the large datasets and ever increasing size of the<br />

data files. This becomes even more apparent when these data<br />

files need to be transferred from one cluster to the other,<br />

requiring large amounts of time for data transfer (I/O) when<br />

using the current network technologies. Other limitations are<br />

the large numbers of small jobs, where low response time can<br />

result in big delays in processing time. Optical fibres allow<br />

for high speed connections (currently 10 gigabit per second)<br />

and quick response times and therefore would be ideal in this<br />

setup. With the ‘Enlighten your research’ grant it is now possible<br />

to use the so-called dynamic light paths in this network.<br />

Dynamic light paths are not fixed connections between one or<br />

more clusters nodes, but can however be set up ‘on demand’<br />

to provide exclusive fast connections with quick response<br />

30 | NPC Highlights 11 | April 2010 | Bioinformatics<br />

NPC theme leaders Péter Horvatovitch (University of Groningen) and Bas van<br />

Breukelen (Utrecht University) were together one of the three winners of the<br />

‘Enlighten your research competition’. They received the award from Minister<br />

Plasterk at the prize giving ceremony, which was part of the launch event for<br />

GigaPort 3 on February 18, 2010. They also received free access to the light<br />

path network and e 20,000 to integrate light paths in their research.<br />

and tested with the developed tools. At the moment the bioinformatics<br />

group in Groningen, under the supervision of Peter<br />

Horvatovich, are implementing the first high-throughput data<br />

processing services for time alignment of multiple LC-MS chromatograms<br />

using the large computation power of the Dutch<br />

Life Science Cluster (SARA) and the fast connection provided<br />

by the dynamic light paths.<br />

With the help of the dynamic light paths and the Dutch Life<br />

Science Grid we hope and expect to provide high-throughput<br />

and stable data processing services to the Dutch proteomics<br />

community, based on the tools developed in different Dutch<br />

and international bioinformatics laboratories.<br />

times not shared with other users. This in turn allows for an<br />

efficient usage of the available computational and storage<br />

resources where it is most needed.<br />

Implementation<br />

In the coming months the light path connections will be implemented<br />

between five clusters of the Dutch Life Science Grid<br />

located at the Universities of Groningen, Utrecht, Wageningen<br />

and Amsterdam and the Erasmus Medical <strong>Centre</strong> in Rotterdam<br />

ErasmusMC grid cluster<br />

and data storage<br />

UvA grid cluster<br />

and data storage<br />

Lightpath<br />

Lightpaths<br />

UU grid cluster<br />

and data storage<br />

Lightpath<br />

Lightpaths<br />

RUG grid cluster<br />

and data storage<br />

Lightpath<br />

Lightpath<br />

WUR grid cluster<br />

and data storage<br />

Contact<br />

Dr. Ir. Bas van Breukelen<br />

Biomolecular Mass Spectrometry and <strong>Proteomics</strong> Group<br />

T +31 30 253 9761<br />

M +31 6 422 225 20<br />

b.vanbreukelen@uu.nl<br />

Task force<br />

The <strong>Netherlands</strong> <strong>Proteomics</strong> <strong>Centre</strong> (NPC) and the <strong>Netherlands</strong> Bioinformatics<br />

<strong>Centre</strong> (NBIC) have joined forces in setting up a large task force in<br />

the Gaining Momentum Theme. This task force aims to build a platform<br />

for proteomics based on bioinformatics and to provide tools, workflows<br />

and high-throughput data processing services for the proteomics community.<br />

Several groups throughout the <strong>Netherlands</strong> contribute to this platform by<br />

creating new (bioinformatics) tools as well as adapting existing tools so<br />

that they can be used as workflow management tools. The idea here is<br />

that all the tools are developed so that they can make use of existing data<br />

standards and be coupled to each other without the need to develop a<br />

plethora of data conversion algorithms. Moreover all the tools will be able<br />

to run as a ‘web service’ which enables other bioinformaticians to couple<br />

these tools without needing to run them locally.<br />

One important part of this platform is the infrastructure which is provided<br />

by NBIC. All the major universities and medical centres have a server (a<br />

cluster node) with multiple CPU’s and storage capacity to host the web<br />

services and which form a grid (BIG GRID and SARA) together.<br />

Impression of the cluster network and light path connections


Top publications with NPC contribution<br />

In this NPC HighLights we provide a short list of<br />

papers that appeared recently in some of the top<br />

journals and to which NPC participants contributed.<br />

With the guarantee of being by far not comprehensive<br />

, this overview shows some elegant<br />

ground-breaking research.<br />

MONOPTEROS controls embryonic root<br />

initiation by regulating a mobile transcription<br />

factor<br />

Schlereth, A., Moller, B., Liu, W., Kientz, M., Flipse, J.,<br />

Rademacher, E.H., Schmid, M., Juergens, G., Weijers D.<br />

Nature (2010) Mar 10<br />

Entwicklungsgenetik, Zentrum für Molekularbiologie der<br />

Pflanzen (ZMBP), Universität Tübingen, Auf der Morgenstelle<br />

3, 72076 Tübingen, Germany. Present address: Syngenta Crop<br />

Protection, CH-4332 Stein, Switzerland.<br />

Acquisition of cell identity in plants relies strongly on<br />

PMID: 20220754<br />

positional information, hence cell–cell communication and<br />

inductive signalling are instrumental for developmental<br />

patterning. During Arabidopsis embryogenesis, an extra-embryonic<br />

cell is specified to become the founder cell of the<br />

primary root meristem, hypophysis, in response to signals from<br />

adjacent embryonic cells. The auxin-dependent transcription<br />

factorMONOPTEROS (MP) drives hypophysis specification by<br />

promoting transport of the hormone auxin from the embryo<br />

to the hypophysis precursor. However, auxin accumulation<br />

is not sufficient for hypophysis specification, indicating that<br />

additional MP-dependent signals are required3. Here we<br />

describe the microarray-based isolation ofMPtarget genes<br />

that mediate signalling fromembryo to hypophysis. Of three<br />

direct transcriptional target genes, TARGET OFMP5 (TMO5)<br />

andTMO7 encode basic helix–loop–helix (bHLH) transcription<br />

factors that are expressed in the hypophysis-adjacent embryo<br />

cells, and are required and partially sufficient for MP-dependent<br />

root initiation. Importantly, the small TMO7 transcription<br />

factor moves from its site of synthesis in the embryo to the<br />

hypophysis precursor, thus representing a novel MP-dependent<br />

intercellular signal in embryonic root specification.<br />

Histone chaperones ASF1 and NAP1<br />

differentially modulate removal of active<br />

histone marks by LID-RPD3 complexes during<br />

NOTCH silencing<br />

Moshkin, Y.M., Kan, T.W., Goodfellow, H., Bezstarosti, K.,<br />

Maeda, R.K., Pilyugin, M., Karch, F., Bray,S.J., Demmers, J.A.,<br />

Verrijzer, C.P.<br />

Mol Cell (2009) Sep 24;35(6):782-93.<br />

Department of Biochemistry, Center for Biomedical Genetics,<br />

Erasmus University Medical Center, P.O.<br />

Box 1738, 3000 DR Rotterdam, The <strong>Netherlands</strong><br />

PMID: 19782028<br />

Histone chaperones are involved in a variety of chromatin<br />

transactions. By a proteomics survey, we identified the interaction<br />

networks of histone chaperones ASF1, CAF1, HIRA, and<br />

NAP1. Here, we analyzed the cooperation of H3/H4 chaperone<br />

ASF1 and H2A/H2B chaperone NAP1 with two closely related<br />

silencing complexes: LAF and RLAF. NAP1 binds RPD3 and<br />

LID-associated factors (RLAF) comprising histone deacetylase<br />

RPD3, histone H3K4 demethylase LID/KDM5, SIN3A, PF1,<br />

EMSY, and MRG15. ASF1 binds LAF, a similar complex lacking<br />

RPD3. ASF1 and NAP1 link, respectively, LAF and RLAF to<br />

the DNA-binding Su(H)/Hairless complex, which targets the<br />

E(spl) NOTCH-regulated genes. ASF1 facilitates gene-selective<br />

removal of the H3K4me3 mark by LAF but has no effect on<br />

H3 deacetylation. NAP1 directs high nucleosome density near<br />

E(spl) control elements and mediates both H3 deacetylation<br />

and H3K4me3 demethylation by RLAF. We conclude that histone<br />

chaperones ASF1 and NAP1 differentially modulate local<br />

chromatin structure during gene-selective silencing.<br />

Recombination-induced tag exchange to track<br />

old and new proteins<br />

Verzijlbergen, K.F., Menendez-Benito, V., van Welsem, T., van<br />

Deventer, S.J., Lindstrom, D.L., Ovaa, H., Neefjes, J., Gottschling,<br />

D.E., van Leeuwen, F.<br />

Proc Natl Acad Sci U S A (2010) Jan 5;107(1):64-8.<br />

Division of Gene Regulation, <strong>Netherlands</strong> Cancer Institute, 1066 CX<br />

Amsterdam, The <strong>Netherlands</strong>.<br />

PMID: 20018668<br />

The dynamic behavior of proteins is critical for cellular<br />

homeostasis. However, analyzing dynamics of proteins and<br />

protein complexes in vivo has been difficult. Here we<br />

describe recombination-induced tag exchange (RITE), a<br />

genetic method that induces a permanent epitope-tag switch<br />

in the coding sequence after a hormone-induced activation<br />

of Cre recombinase. The time-controlled tag switch provides<br />

a unique ability to detect and separate old and new proteins<br />

in time and space, which opens up opportunities to investigate<br />

the dynamic behavior of proteins. We validated the<br />

technology by determining exchange of endogenous histones<br />

in chromatin by biochemical methods and by visualizing and<br />

quantifying replacement of old by new proteasomes in single<br />

cells by microscopy. RITE is widely applicable and allows<br />

probing spatiotemporal changes in protein properties by<br />

multiple methods.<br />

| 31


Nucleotide excision repair-induced H2A ubiquitination<br />

is dependent on MDC1 and RNF8<br />

and reveals a universal DNA damage response<br />

Marteijn, J.A., Bekker-Jensen, S., Mailand, N., Lans, H.,<br />

Schwertman, P., Gourdin, A.M., Dantuma, N.P., Lukas, J.,<br />

32 | NPC Highlights 11 | April 2010 | Abstracts<br />

Vermeulen, W.<br />

J Cell Biol (2009) Sep 21;186(6):835-47.<br />

Department of Genetics, Center for Biomedical Genetics,<br />

Erasmus Medical Center, 3015 GE Rotterdam, <strong>Netherlands</strong>.<br />

PMID: 19797077<br />

Chromatin modifications are an important component of the of<br />

DNA damage response (DDR) network that safeguard genomic<br />

integrity. Recently, we demonstrated nucleotide excision<br />

repair (NER)-dependent histone H2A ubiquitination at sites<br />

of ultraviolet (UV)-induced DNA damage. In this study, we<br />

show a sustained H2A ubiquitination at damaged DNA, which<br />

requires dynamic ubiquitination by Ubc13 and RNF8. Depletion<br />

of these enzymes causes UV hypersensitivity without affecting<br />

NER, which is indicative of a function for Ubc13 and RNF8 in<br />

the downstream UV-DDR. RNF8 is targeted to damaged DNA<br />

through an interaction with the double-strand break (DSB)-DDR<br />

scaffold protein MDC1, establishing a novel function for MDC1.<br />

RNF8 is recruited to sites of UV damage in a cell cycle-independent<br />

fashion that requires NER-generated, single-stranded<br />

repair intermediates and ataxia telangiectasia-mutated and<br />

Rad3-related protein. Our results reveal a conserved pathway<br />

of DNA damage-induced H2A ubiquitination for both DSBs and<br />

UV lesions, including the recruitment of 53BP1 and Brca1.<br />

Although both lesions are processed by independent repair<br />

pathways and trigger signaling responses by distinct kinases,<br />

they eventually generate the same epigenetic mark, possibly<br />

functioning in DNA damage signal amplification.<br />

Accumulation of ubiquitin conjugates in<br />

a polyglutamine disease model occurs<br />

without global ubiquitin-proteasome system<br />

impairment<br />

Maynard, C.J., Böttcher, C., Ortega, Z., Smith, R., Florea, B.I.,<br />

Díaz-Hernández, M., Brundin, P., Overkleeft, H.S., Li, J.,<br />

Lucas, J.J., Dantuma, N.P.<br />

Proc Natl Acad Sci U S A (2009) Aug 18;106(33):13986-91.<br />

Department of Cell and Molecular Biology, Karolinska<br />

Institutet, S-17177 Stockholm, Sweden<br />

PMID: 19666572<br />

Aggregation-prone proteins have been suggested to overwhelm<br />

and impair the ubiquitin/proteasome system (UPS) in polyglutamine<br />

(polyQ) disorders, such as Huntington’s disease (HD).<br />

Overexpression of an N-terminal fragment of mutant huntingtin<br />

(N-mutHtt), an aggregation-prone polyQ protein responsible for<br />

HD, obstructs the UPS in cellular models. Furthermore, based on<br />

the accumulation of polyubiquitin conjugates in brains of R6/2<br />

mice, which express human N-mutHtt and are one of the most<br />

severe polyQ disorder models, it has been proposed that UPS<br />

dysfunction is a consistent feature of this pathology, occurring in<br />

both in vitro and in vivo models. Here, we have exploited transgenic<br />

mice that ubiquitously express a ubiquitin fusion degradation<br />

proteasome substrate to directly assess the functionality of<br />

the UPS in R6/2 mice or the slower onset R6/1 mice. Although<br />

expression of N-mutHtt caused a general inhibition of the UPS in<br />

PC12 cells, we did not observe an increase in the levels of proteasome<br />

reporter substrate in the brains of R6/2 and R6/1 mice.<br />

We show that the increase in ubiquitin conjugates in R6/2 mice<br />

can be primarily attributed to an accumulation of large ubiquitin<br />

conjugates that are different from the conjugates observed upon<br />

UPS inhibition. Together our data show that polyubiquitylated<br />

proteins accumulate in R6/2 brain despite a largely operative<br />

UPS, and suggest that neurons are able to avoid or compensate<br />

for the inhibitory effects of N-mutHtt.<br />

A comprehensive framework of E2-RING<br />

E3 interactions of the human ubiquitinproteasome<br />

system<br />

van Wijk, S.J., de Vries, S.J., Kemmeren, P., Huang, A., Boelens, R.,<br />

Bonvin, A.M., Timmers, H.T.M.<br />

Mol Syst Biol (2009) 5:295.<br />

Division of Biomedical Genetics, Department of Physiological<br />

Chemistry, University Medical Center Utrecht, Utrecht, The<br />

<strong>Netherlands</strong>.<br />

PMID: 19690564<br />

Covalent attachment of ubiquitin to substrates is crucial to<br />

protein degradation, transcription regulation and cell signalling.<br />

Highly specific interactions between ubiquitin-conjugating<br />

enzymes (E2) and ubiquitin protein E3 ligases fulfil essential roles<br />

in this process. We performed a global yeast-two hybrid screen to<br />

study the specificity of interactions between catalytic domains of<br />

the 35 human E2s with 250 RING-type E3s. Our analysis showed<br />

over 300 high-quality interactions, uncovering a large fraction of<br />

new E2-E3 pairs. Both within the E2 and the E3 cohorts, several<br />

members were identified that are more versatile in their interaction<br />

behaviour than others. We also found that the physical<br />

interactions of our screen compare well with reported functional<br />

E2-E3 pairs in in vitro ubiquitination experiments. For validation<br />

we confirmed the interaction of several versatile E2s with E3s<br />

in in vitro protein interaction assays and we used mutagenesis<br />

to alter the E3 interactions of the E2 specific for K63 linkages,<br />

UBE2N(Ubc13), towards the K48-specific UBE2D2(UbcH5B). Our<br />

data provide a detailed, genome-wide overview of binary E2-E3<br />

interactions of the human ubiquitination system.<br />

Other highlighted publications<br />

Muñoz, J., Heck, A.J.<br />

Snapshots of kinase activities<br />

Nat Biotechnol (2009) Oct;27(10):912-3.<br />

PMID: 19816446


abstracts<br />

npc<br />

Albers, H.M.G., Dong, A., van Meeteren, L.A., Egan, D.A.,<br />

Sunkara, M., van Tilburg, E.W., Schuurman, K., van Tellingen,<br />

O., Morris, A.J., Smyth, S.S., Moolenaar, W.H., Ovaa, H.<br />

A boronic acid-based inhibitor of autotaxin reveals rapid<br />

turnover of LPA in the circulation<br />

Proc Natl Acad Sci U S A (2010) Apr 1<br />

Wiederhold, E., Veenhoff, L.M., Poolman, B., Slotboom, D.J.<br />

<strong>Proteomics</strong> of Saccharomyces cerevisiae organelles<br />

Mol Cell <strong>Proteomics</strong> (2010) Mar;9(3):431-45.<br />

PMID: 19955081<br />

Mahmoudi, T., Li, V.S., Ng, S.S., Taouatas, N., Vries, R.G.,<br />

Mohammed, S., Heck, A.J., Clevers H.<br />

The kinase TNIK is an essential activator of Wnt target genes<br />

EMBO J (2009) 28; 3329-3340<br />

PMID: 19816403<br />

Witte, M.D., Florea, B.I., Verdoes, M., Adeyanju, O., van der<br />

Marel, G.A., Overkleeft, H.S.<br />

O-GlcNAc peptide epoxyketones are recognized by mammalian<br />

proteasomes<br />

J Am Chem Soc (2009) Sep 2;131(34):12064-5.<br />

PMID: 19658393<br />

Ramadurai, S., Holt, A., Krasnikov, V., van den Bogaart, G.,<br />

Killian, J.A., Poolman, B.<br />

Lateral diffusion of membrane proteins<br />

J Am Chem Soc (2009) Sep 9;131(35):12650-6.<br />

PMID: 19673517<br />

van Wijk S.J., Timmers H.T.M.<br />

The family of ubiquitin-conjugating enzymes (E2s): deciding<br />

between life and death of proteins<br />

FASEB J (2009); Apr;24(4):981-93.<br />

PMID: 19940261<br />

Frederiks, F., Heynen, G.J., van Deventer, S.J., Janssen, H.,<br />

van Leeuwen, F.<br />

Two Dot1 isoforms in Saccharomyces cerevisiae as a result of<br />

leaky scanning by the ribosome<br />

Nucleic Acids Res (2009)37(21):7047-58.<br />

PMID: 19778927<br />

Schaaij-Visser, T.B., Graveland, A.P., Gauci, S., Braakhuis,<br />

B.J., Buijze, M., Heck, A.J., Kuik, D.J., Bloemena, E.,<br />

Leemans, C.R., Slijper, M., Brakenhoff, R.H.<br />

Differential <strong>Proteomics</strong> Identifies Protein Biomarkers That<br />

Predict Local Relapse of Head and Neck Squamous Cell<br />

Carcinomas<br />

Clin Cancer Res (2009) Dec 15;15(24):7666-7675.<br />

PMID: 19996216<br />

Ng, S.S., Mahmoudi, T., Danenberg, E., Bejaoui, I., de Lau, W.,<br />

Korswagen, H.C., Schutte, M., Clevers, H.<br />

Phosphatidylinositol 3-kinase (PI3K) signaling does not activate<br />

the Wnt cascade<br />

J Biol Chem (2009) 284: 35308-35313<br />

PMID: 19850932<br />

Van Hoof, D., Dormeyer, W., Braam, S.R., Passier, R.,<br />

Monshouwer-Kloots, J., Ward-van Oostwaard, D., Heck, A.J.,<br />

Krijgsveld, J., Mummery, C.L.<br />

Identification of cell surface proteins for antibody-based<br />

selection of human embryonic stem cell-derived cardiomyocytes<br />

J Proteome Res (2010) Mar 5; 9 (3):1610-8<br />

PMID: 20088484<br />

Christin, C., Hoefsloot, H.C., Smilde, A.K., Suits, F., Bischoff,<br />

R., Horvatovich, P.L.<br />

Time Alignment Algorithms Based on Selected Mass Traces for<br />

Complex LC-MS Data<br />

J Proteome Res (2010) Mar 5; 9 (3):1483-95<br />

PMID: 20070124<br />

Mischerikow, N., Spedale, G., Altelaar, M., Timmers, H.T.M.,<br />

Pijnappel, P.W.W.M., Heck, A.J.R.<br />

In-depth profiling of post-translational modifications on the<br />

related transcription factor complexes TFIID and SAGA<br />

J.Proteome Res. (2009)11:5020-5030<br />

PMID: 19731963<br />

Raijmakers, R., Kraiczek, K., de Jong, A., Mohammed, S.,<br />

Heck, A.J.<br />

Exploring the human leukocyte phosphoproteome using a<br />

microfluidic RP-TiO2-RP HPLC Phosphochip coupled to a Q-ToF<br />

mass spectrometer<br />

Anal. Chem. (2010) Feb 1;82(3):824-32<br />

Schaaij-Visser, T.B., Brakenhoff, R.H., Jansen, J.W.,<br />

O’Flaherty, M.C., Smeets, S.J., Heck, A.J., Slijper, M.<br />

Comparative proteome analysis to explore p53 pathway<br />

disruption in head and neck carcinogenesis<br />

J <strong>Proteomics</strong>. (2009) Jul 21;72(5):803-14.<br />

PMID: 19446051<br />

van Breukelen, B., Georgiou, A., Drugan, M., Taouatas, N.,<br />

Mohammed, S., Heck, A.J.<br />

LysNDeNovo: An algorithm enabling de novo sequencing of Lys-N<br />

generated peptides fragmented by electron transfer dissociation<br />

<strong>Proteomics</strong> (2010) Mar;10(6):1196-201<br />

PMID: 20077410<br />

Helbig, A.O., de Groot, M.J., van Gestel, R.A., Mohammed, S.,<br />

de Hulster, E.A., Luttik, MA, Daran-Lapujade P, Pronk JT,<br />

Heck AJ, Slijper M.<br />

A three-way proteomics strategy allows differential analysis<br />

of yeast mitochondrial membrane protein complexes under<br />

anaerobic and aerobic conditions<br />

<strong>Proteomics</strong> (2009) Oct;9(20):4787-98.<br />

PMID: 19750512<br />

Jaworski, J. Kapitein, L.C., Gouveia, S.M., Dortland, B.R.,<br />

Wulf, P.S., Grigoriev, I., Camera, P., Spangler, S.A.,<br />

Di Stefano, P., Demmers, J., Krugers, H., Defilippi, P.,<br />

Akhmanova, A., Hoogenraad, C.C.<br />

Dynamic microtubules regulate dendritic spine morphology<br />

and synaptic plasticity<br />

Neuron (2009) Jan 15;61(1):85-100<br />

PMID: 19146815<br />

| 33


NPC Research Hotels<br />

The NPC offers the newest proteomics technologies and facilities in specialised Research Hotels for joint projects with external<br />

scientists. This overview gives you an impression of our expertise and facilities. If you have any specific questions about our<br />

technologies you can directly contact the respective Hotel Manager. If you have more general questions about our Research<br />

Hotels and the conditions for joint projects, please contact the NPC office at info@npc.genomics.nl.<br />

Cell Sorting Hotel Utrecht<br />

Utrecht University<br />

Many protein complexes differ between cell types. The Cell<br />

Sorting Hotel Utrecht offers equipment and expertise to<br />

isolate specific cell types from plant tissues. The goal is to<br />

extract proteins from sorted cells and subject them to<br />

proteomic analysis. Cell-specific proteomics is a technical<br />

goal in itself and as soon as this procedure has been optimized<br />

for Arabidopsis all plant NPCII groups will have access<br />

to the facility using Arabidopsis, tomato and potato cells.<br />

Hotel Manager<br />

Prof. dr. Ben J.G. Scheres<br />

University Utrecht<br />

Department Biology/Molecular Genetics<br />

Padualaan 8<br />

3584 CH Utrecht<br />

T: +31 30 253 3133<br />

E: b.scheres@uu.nl<br />

34 | NPC Highlights 11 | April 2010<br />

Analytical Hotel Delft<br />

Delft University of Technology<br />

Analytical Hotel Delft provides up to date Peptidomics in all<br />

of its aspects: both qualitative and quantitative analysis of<br />

the subset of the proteome designated as the peptidome;<br />

naturally occurring (native, endogenous) peptides with<br />

important biological (regulatory, signal, (neuro)hormonal)<br />

functions. Focus will be on various peptide chemical<br />

identification strategies, including de novo<br />

sequencing, peptide bioinformatics, the<br />

study of bioactive peptide location, as well<br />

as their (physiological/pharmacological)<br />

activities.<br />

Hotel Managers<br />

Dr. Martijn Pinkse &<br />

Prof. dr. Peter Verhaert<br />

Delft University of Technology<br />

Kluyver Laboratory<br />

Julianalaan 67<br />

2628 BC Delft<br />

T: +31 15 278 2344<br />

E: m.w.h.pinkse@tudelft.nl<br />

Analytical Hotel Utrecht<br />

Utrecht University<br />

The main task of the Utrecht Analytical Hotel is to provide<br />

an expert centre for state of the art proteomics analysis.<br />

In the Hotel, innovative mass spectrometric methods are<br />

developed and implemented for the efficient and detailed<br />

characterization of biomolecules in their relation to their<br />

biological function. The scientific focus ranges from single<br />

protein identifications to the analysis of complex, whole<br />

cell lysate samples and determination of posttranslational<br />

modifications.<br />

Analytical Hotel Rotterdam<br />

Erasmus MC<br />

The Erasmus MC Research Hotel is embedded within the<br />

Erasmus MC <strong>Proteomics</strong> Center (part of the Department of<br />

Biochemistry) and offers proteomic and mass spectrometric<br />

services for the Dutch research academic community as well<br />

as for the local scientific community. Projects undertaken<br />

will be on a collaborative basis and consultation regarding<br />

study design; data interpretation is provided by people<br />

from the Research Hotel. The Research Hotel develops mass<br />

spectrometry based proteomics methodologies and protein<br />

separation techniques for the qualitative and quantitative<br />

analysis of (sub) proteomes.<br />

Hotel Managers<br />

Dr. Maarten Altelaar &<br />

Prof. dr. Albert Heck<br />

Utrecht University<br />

Biomolecular Mass Spectrometry<br />

and <strong>Proteomics</strong> Group<br />

Padualaan 8,<br />

3584 CH Utrecht<br />

T: +31 30 253 9554<br />

E: m.altelaar@uu.nl<br />

Hotel Manager<br />

Dr. Jeroen Demmers<br />

<strong>Proteomics</strong> Center, Erasmus MC<br />

Room Ee679<br />

Dr. Molenwaterplein 50<br />

3015 GE Rotterdam<br />

T: +31 10 703 8124<br />

E: j.demmers@erasmusmc.nl


Analytical Hotel Wageningen<br />

Plant Research International/<br />

Wageningen University<br />

Analytical Hotel Groningen<br />

University of Groningen<br />

hotel managers<br />

The Wageningen Research Hotel is strongly linked to activities<br />

within the <strong>Centre</strong> for Biosystems Genomics (CBSG),<br />

which aims to decipher the genetic basis of plant specific<br />

processes and to improve plant performance and product<br />

quality. Traits of interest from both scientific and applied<br />

point of view are: disease resistance against pathogens;<br />

metabolic pathways that contribute to flavour production;<br />

consumer-driven qualities of food products (e.g. tomato)<br />

and developmental aspects of plant growth. The NPCII<br />

plant projects are focused on plant development and signal<br />

transduction in plant meristems. <strong>Proteomics</strong> technologies<br />

will be used in the Hotel to perform protein<br />

profiling for comparative proteomics and to<br />

unravel signalling pathways and the involved<br />

protein complexes.<br />

Hotel Managers<br />

Dr. Twan America &<br />

Prof. dr. Gerco Angenent<br />

Wageningen UR<br />

Plant Research International<br />

Droevendaalsesteeg 1<br />

6708 PB Wageningen<br />

T: +31 317 480 953<br />

E: Angenent@wur.nl<br />

Bioinformatics Hotel Utrecht<br />

Utrecht University<br />

The Bioinformatics Hotel Utrecht mainly focusses on the<br />

development of bioinformatics software and algorithms that<br />

facilitate proteomics research. This can be subdivided in three<br />

research lines.<br />

1. (Raw) data (pre) processing: development and implementation<br />

of algorithms for noise filtering, FDR calculations,<br />

database search software and de novo sequence algorithms.<br />

2. Data storage and dissemination: storage of raw and<br />

processed data into public and local repositories (or LIMS<br />

systems).<br />

3. Knowledge extraction: extract biological information from<br />

pre-processed proteomics data.<br />

Hotel Manager<br />

Dr. Bas van Breukelen<br />

Utrecht University<br />

Biomolecular Mass Spectrometry and<br />

<strong>Proteomics</strong> Group<br />

Padualaan 8<br />

3584 CH Utrecht<br />

T: +31 30 253 9761<br />

E: b.vanbreukelen@uu.nl<br />

The Analytical Hotel Groningen provides expertise and assistance<br />

in experiments requiring the purification, separation<br />

and high resolution analysis of proteins employing<br />

proteomics tools as well as biochemical and biophysical techniques.<br />

These include membrane and organelle isolation,<br />

ultracentrifugation (analytical and preparative), gel- and<br />

solution (chromatography)-based separation of proteins and<br />

peptides (1- and 2-dimensional) coupled to mass spectrometry.<br />

The Hotel offers collaboration and knowledge transfer<br />

to address questions regarding MS-based identification of<br />

proteins in highly complex mixtures and the biophysical<br />

characterization of individual membrane<br />

proteins, protein complexes and their modifications.<br />

Hotel Managers<br />

Dr. Fabrizia Fusetti &<br />

Prof. dr. Bert Poolman<br />

University of Groningen<br />

Groningen Biomolecular Sciences and<br />

Biotechnology Institute & <strong>Centre</strong> for<br />

Synthetic Biology<br />

Nijenborgh 4<br />

9747 AH Groningen<br />

T: +31 50 363 4190<br />

E: b.poolman@rug.nl<br />

Cell Culture Hotel Utrecht<br />

UMC Utrecht<br />

The scientific focus of the Cell Culture Hotel Utrecht is to<br />

provide key technologies for targeted proteomic analyses of<br />

embryonic stem cells (ESCs), induced pluripotent stem cells<br />

(iPS) and differentiated cells. This includes selective tagging<br />

of proteins using bacterial artificial chromosomes (BACs) and<br />

viral-mediated gene manipulation. The Hotel offers a variety<br />

of different epitopes for efficient tagging and purification of<br />

target proteins expressed at endogenous levels. This enables<br />

isolation of protein complexes for proteomic<br />

and genomic analyses.<br />

Hotel Managers<br />

Dr. Pim Pijnappel &<br />

Prof. dr. Marc Timmers<br />

University Medical <strong>Centre</strong> Utrecht<br />

Department of Physiological Chemistry<br />

Division Biomedical Genetics<br />

Universiteitsweg 100<br />

3584 CG Utrecht<br />

T: +31 88 756 8981<br />

E: h.t.m.timmers@umcutrecht.nl<br />

| 35


upcoming events<br />

23-27 May 2010<br />

27 June - 1 July<br />

19-24 September 2010<br />

23-27 October 2010<br />

23 November 2010<br />

|<br />

|<br />

|<br />

|<br />

|<br />

ASMS, Salt Lake City, U.S.A.<br />

Metabolomics 2010, Amsterdam, The <strong>Netherlands</strong><br />

9th HUPO World Congress, Sydney, Australia<br />

EuPa, Estoril, Portugal<br />

NGI Life Sciences Momentum, Utrecht, The <strong>Netherlands</strong><br />

Combining the best<br />

existing forces<br />

Only innovation on a European level will allow us to keep pace<br />

in the world economy. Are we innovative enough No. Do we<br />

have the basis Yes. We do have excellent research, education<br />

and business centres but knowledge and ideas sometimes still<br />

tend to become clogged up in dead ends. Here is where we<br />

clearly need to improve. Four components to achieve this goal<br />

are people, focus, leadership and entrepreneurship.<br />

Dr. Martin Schuurmans<br />

Chairman - European<br />

Institute of Innovation and<br />

Technology (EIT)<br />

http://eit.europa.eu<br />

People, because they are the ones carrying the knowledge<br />

and drive ideas forward. We need more creative and<br />

entrepreneurial people. Focus and leadership are essential,<br />

because efforts and resources must be effectively<br />

and efficiently concentrated on clear goals. Finally,<br />

entrepreneurship is needed to achieve the goals in terms of<br />

new products, services, business and job creation.<br />

© Photo EIT<br />

With the European Institute of Innovation and Technology<br />

(EIT) we contribute to a structural change in the innovation<br />

ecosystem, based on the full integration of the knowledge<br />

triangle of higher education, research and business/innovation.<br />

To unleash Europe’s innovation potential, we will break up<br />

borders between academia and business, between teaching<br />

and research. At the same time, successful integration requires<br />

excellent partners. We need to continue to strengthen our<br />

capacities and invest more in R&D and higher education.<br />

In particular higher education is crucial because we need<br />

people with the right multi-disciplinary skills and an open,<br />

entrepreneurial mindset driving innovation processes.<br />

The EIT’s Knowledge and Innovation Communities (KICs) are<br />

test beds for this new kind of collaborative, entrepreneurial<br />

approach. While building on strong existing partners and<br />

cooperations, the KICs will have a totally new quality in the way<br />

the three sides of the triangle interact. People in the triangle<br />

will work together face-to-face in integrated teams in a number<br />

of co-location innovation centres in the KIC. They will operate<br />

towards explicit goals and under clear leadership of the KIC,<br />

exploiting every avenue of entrepreneurship. Working together<br />

across co-location centres will also contribute to healthier<br />

brain motion over Europe. For now, the three first KICs focus on<br />

climate change adaptation and mitigation, sustainable energy,<br />

and the future information and communication society. Others<br />

may follow in the future, for example in the field of health.<br />

Maybe proteomics will contribute as well!<br />

11

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!