05.01.2013 Views

SUNDAY, DECEMBER 4- Late Abstracts 1 - Molecular Biology of the ...

SUNDAY, DECEMBER 4- Late Abstracts 1 - Molecular Biology of the ...

SUNDAY, DECEMBER 4- Late Abstracts 1 - Molecular Biology of the ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

<strong>SUNDAY</strong>, <strong>DECEMBER</strong> 4- <strong>Late</strong> <strong>Abstracts</strong> 1<br />

Membrane Trafficking<br />

<strong>SUNDAY</strong><br />

1970<br />

A novel SNX3-dependent Retromer pathway is required for Wnt secretion.<br />

I. McGough 1 , M. Harterink 2 , F. Port 3 , M. Lorenowicz 2 , R. Korswagen 2 , P. Cullen 1 ; 1 Biochemistry,<br />

University <strong>of</strong> Bristol, Bristol, England, 2 Hubrecht Institute, Utrecht, Holland, 3 Institute <strong>of</strong><br />

<strong>Molecular</strong> Life Sciences, University <strong>of</strong> Zurich<br />

Wnt proteins play a central role in development. Wnt secretion is mediated by <strong>the</strong> Wnt-binding<br />

protein Wntless (Wls), which transports Wnt from <strong>the</strong> Golgi to <strong>the</strong> cell surface for release. Wnt<br />

secretion requires recycling <strong>of</strong> Wls through a retromer-dependent endosome-to-Golgi trafficking<br />

pathway, but <strong>the</strong> mechanism remains poorly understood. Using C. elegans and Drosophila<br />

genetics, we established that Wls recycling requires an evolutionarily conserved, ‘non-classical’<br />

retromer pathway, that functions independently <strong>of</strong> <strong>the</strong> classical retromer sorting nexins. We<br />

found sorting nexin-3, SNX3, has an evolutionarily conserved function in Wls recycling and Wnt<br />

secretion. SNX3 interacts directly with <strong>the</strong> cargo-selective sub-complex <strong>of</strong> retromer. Live cell<br />

imaging reveals WLS exits SNX3 labelled endosomes through vesicular carriers ra<strong>the</strong>r than<br />

classic retromer decorated tubular pr<strong>of</strong>iles. These results establish SNX3 as part <strong>of</strong> an<br />

alternative retromer pathway that functionally separates retrograde transport <strong>of</strong> Wls from o<strong>the</strong>r<br />

retromer cargo.<br />

1971<br />

The lectin ArtinM binds to and is internalized by hematopoietic cell lines.<br />

P. A. Buranello 1 , V. M. Mazucato 1 , M. R. Pinto 1 , M. C. Barreira 1 , M. C. Jamur 1 , C. Oliver 1 ;<br />

1 Biologia Celular e <strong>Molecular</strong> e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão<br />

Preto, Ribeirão Preto, Brazil<br />

The D-mannose binding lectin ArtinM, extracted from Artocarpus integrifolia, has an important<br />

role in <strong>the</strong> defense against pathogens. Artin M is imunnomodulatory and activates immune cells,<br />

such as mast cells, neutrophils and macrophages. It is known that ArtinM can induce cell death<br />

in various hematopoietic cell lines. The aim <strong>of</strong> <strong>the</strong> present study is to fur<strong>the</strong>r characterize <strong>the</strong><br />

effects <strong>of</strong> Artin M on hematopoetic cell lines. The human hematopoietic cell lines NB4, K562<br />

and U937 were used. Flow cytometric analysis showed that NB4 cells bind 2.4 times less ArtinM<br />

than K562 and U937 cells. By scanning electron microscopy, it was possible to observe that<br />

ArtinM activated <strong>the</strong> various cell lines. After 5 minutes <strong>of</strong> incubation, <strong>the</strong> K562 cells already<br />

presented ruffles on <strong>the</strong>ir surface. Only after 15 minutes <strong>of</strong> incubation was this activation<br />

observed in NB4 and U937 cells. However, <strong>the</strong> ruffling was less intense. After one hour <strong>of</strong><br />

incubation in presence <strong>of</strong> ArtinM, all <strong>of</strong> <strong>the</strong> cell lines exhibited extensive ruffling. This ruffling<br />

was most evident in <strong>the</strong> K562 cells. Artin M internalization was investigated by fluorescence<br />

microscopy using biotinylated Artin M followed by streptavidin-FITC. After 15 minutes <strong>of</strong><br />

incubation <strong>the</strong> K562 and U937 cells had already internalized <strong>the</strong> lectin, but internalized lectin<br />

was seen only after 30 minutes in <strong>the</strong> NB4 cells. One hour after incubation, ArtinM had<br />

accumulated in cytoplasmic vesicles in all <strong>of</strong> <strong>the</strong> cell lines. This internalization was independent<br />

<strong>of</strong> clathrin and caveolin. In addition, <strong>the</strong> cytoplasmic vesicles containing ArtinM did not colocalize<br />

with CD63, a lysosomal marker. Our results show that ArtinM activates and is<br />

internalized by hematopoietic cells and that <strong>the</strong> ArtinM <strong>the</strong>n accumulates in cytoplasmic<br />

vesicles. The binding, activation and internalization <strong>of</strong> ArtinM may all contribute to <strong>the</strong> ArtinM<br />

induced cell death seen in hematopoietic cells.


<strong>SUNDAY</strong><br />

1972<br />

Proteomic Characterization <strong>of</strong> <strong>the</strong> Binding Partners <strong>of</strong> Dopamine Transporter (DAT) and<br />

Functional DAT Mutants.<br />

S. M. Moore 1,2 , A. Rao 2 , A. Sorkin 1 , C. C. Wu 1 ; 1 Cell <strong>Biology</strong> and Physiology, University <strong>of</strong><br />

Pittsburgh, Pittsburgh, PA, 2 Pharmacology, University <strong>of</strong> Colorado Anschutz Medical Campus,<br />

Aurora, CO<br />

Dopamine transporter (DAT) is a 12-transmembrane domain, integral membrane protein that<br />

acts to terminate <strong>the</strong> synaptic transmission <strong>of</strong> <strong>the</strong> neurotransmitter dopamine. DAT is normally<br />

expressed primarily on <strong>the</strong> plasma membrane <strong>of</strong> <strong>the</strong> cell and is internalized through clathrinmediated<br />

endocytosis. It has been shown that with N-terminal deletion, DAT undergoes<br />

increased internalization through augmented levels <strong>of</strong> endocytosis. Conversely, a C-terminal<br />

deletion variant <strong>of</strong> DAT has been shown to reside primarily in <strong>the</strong> endoplasmic reticulum, with<br />

unknown endocytic effects. The main objective <strong>of</strong> this project is to utilize mass spectrometry<br />

pr<strong>of</strong>iling methods to analyze <strong>the</strong> binding partners <strong>of</strong> <strong>the</strong>se different DAT constructs in order to<br />

increase <strong>the</strong> understanding <strong>of</strong> DAT trafficking and endocytosis. Initial studies in mice expressing<br />

a full-length hemagglutinin (HA)-tagged DAT construct combined immunoprecipitation (IP) using<br />

an HA antibody with pr<strong>of</strong>iling mass spectrometry. These studies resulted in <strong>the</strong> identification <strong>of</strong> a<br />

variety <strong>of</strong> potential DAT binding partners. Some <strong>of</strong> <strong>the</strong> identified proteins were previously known<br />

to be DAT binding partners, o<strong>the</strong>rs were known to be involved in endocytosis, and still o<strong>the</strong>rs<br />

were known to be involved in synaptic transmission. These mouse studies inspired fur<strong>the</strong>r<br />

proteomic studies in cell lines containing <strong>the</strong> aforementioned DAT constructs, due to <strong>the</strong><br />

identification <strong>of</strong> endocytic proteins in <strong>the</strong> mouse studies along with <strong>the</strong> difference in endocytic<br />

patterns in <strong>the</strong> DAT construct containing cell lines. Full length (FL), N-terminally deleted (ΔN),<br />

and C-terminally deleted (ΔC) DAT constructs, dually tagged with YFP and HA, were stably<br />

expressed in porcine aortic endo<strong>the</strong>lial (PAE) cells. These constructs <strong>the</strong>n underwent IP using<br />

ei<strong>the</strong>r HA or GFP antibodies. After IP <strong>the</strong> samples were digested with trypsin and analyzed on<br />

ei<strong>the</strong>r an LTQ linear ion trap mass spectrometer, for pr<strong>of</strong>iling experiments, or a TSQ Vantage<br />

triple quadrupole mass spectrometer, for quantitative experiments. In <strong>the</strong>se cell studies, several<br />

proteins were identified that were present in all three samples while o<strong>the</strong>rs were expressed<br />

differentially between <strong>the</strong> samples when compared to <strong>the</strong> parental PAE cell line. Proteins<br />

present in all samples included ubiquitin, calmodulin, and alpha-tubulin. Differentially expressed<br />

proteins included clathrin heavy chain, Rho-GEF 12, and AP2 alpha-1 subunit, among a variety<br />

<strong>of</strong> o<strong>the</strong>rs. This differential expression indicates that <strong>the</strong> endocytic changes that occur with <strong>the</strong>se<br />

deletion mutations are accompanied by changes in <strong>the</strong> proteins that associate with DAT.<br />

1973<br />

Cell adhesion and survival requires integrin rapid recycling.<br />

N. Waxmonsky 1 , S. Conner 1 ; 1 University <strong>of</strong> Minnesota, Minneapolis, MN<br />

Integrins are transmembrane heterodimers composed <strong>of</strong> an α and a β subunit that mediate<br />

attachment between <strong>the</strong> cell and its environment, which is a requirement for cell survival in<br />

many cell types. Cells maintain adhesion by trafficking integrins to <strong>the</strong> cell surface through<br />

endosomal sorting pathways; however, <strong>the</strong> molecular mechanisms governing this process are<br />

undefined. We hypo<strong>the</strong>sized that integrin recycling through <strong>the</strong> endosomal compartment is<br />

important for maintenance <strong>of</strong> cell adhesion. In order to test this, we depleted HeLa cells <strong>of</strong><br />

factors with known roles in long- and short-loop recycling. Depletion <strong>of</strong> a long-loop recycling<br />

factor, EHD1 (Eps15 Homology Domain), did not lead to loss <strong>of</strong> adhesion. However, depletion<br />

<strong>of</strong> factors that have established roles at <strong>the</strong> early endosome, AAK1 (Adaptor-Associated Kinase<br />

1) and EHD3, led to cell rounding and an eventual loss <strong>of</strong> cell adhesion. Defects in integrin<br />

delivery to <strong>the</strong> cell surface disrupts formation <strong>of</strong> focal adhesion complexes. We investigated


<strong>SUNDAY</strong><br />

whe<strong>the</strong>r <strong>the</strong>re were differences in number <strong>of</strong> focal adhesion complexes in cells with short-loop<br />

recycling defects. To do so, we visualized complexes using a vinculin antibody and found a<br />

decrease in number <strong>of</strong> focal adhesion complexes in AAK1 and EHD3 depleted cells. To observe<br />

focal adhesion formation in cells with recycling defects, we used a TIRF live cell imaging<br />

approach to visualize <strong>the</strong> dynamics <strong>of</strong> β3 integrin-GFP positive focal complex formation. The<br />

experiment revealed a reduction in <strong>the</strong> rate <strong>of</strong> focal adhesion complex formation in AAK1 and<br />

EHD3 depleted cells. Commensurate with this observation, we found that β3 integrin total levels<br />

were also reduced in AAK1 and EHD3, but not EHD1, depleted cells. This raised <strong>the</strong> possibility<br />

that <strong>the</strong> observed reduction might result from a biosyn<strong>the</strong>tic defect or a mis-sorting toward <strong>the</strong><br />

degradative pathway. To distinguish between <strong>the</strong>se possibilities, we disrupted <strong>the</strong> degradative<br />

pathway by depleting Tsg101 (Tumor susceptibility gene 101), a factor with a role in <strong>the</strong> ESCRT<br />

complex. In doing so, we found partial restoration <strong>of</strong> β3 integrin total levels and cell attachment<br />

upon depletion <strong>of</strong> AAK1/Tsg101 and EHD3/Tsg101. Depletion <strong>of</strong> factors involved in short-loop<br />

recycling resulted in a loss <strong>of</strong> adhesion, a reduction in focal adhesion formation rate and missorting<br />

to <strong>the</strong> degradative pathway. This experimental evidence indicates that cell adhesion<br />

requires integrin short-loop recycling.<br />

1974<br />

CFTR anion channel modulates expression <strong>of</strong> human transmembrane mucin MUC3<br />

through <strong>the</strong> PDZ protein GOPC.<br />

T. Pelaseyed 1 , G. C. Hansson 1 ; 1 Department <strong>of</strong> Medical Biochemistry, Institute <strong>of</strong> Biomedicine,<br />

University <strong>of</strong> Go<strong>the</strong>nburg, Göteborg, Sweden<br />

The transmembrane mucins in <strong>the</strong> enterocyte are type 1 transmembrane proteins with long and<br />

rigid mucin domains, rich in proline, threonine and serine residues that carry numerous Oglycans.<br />

Three <strong>of</strong> <strong>the</strong>se mucins, MUC3, MUC12 and MUC17 are unique in harboring C-terminal<br />

class I PDZ motifs, making <strong>the</strong>m suitable ligands for PDZ proteins. A screening <strong>of</strong> 123 different<br />

human PDZ domains for binding to MUC3 identified a strong interaction with <strong>the</strong> PDZ protein<br />

GOPC (Golgi-associated PDZ and coiled-coil motif-containing protein). This interaction was<br />

mediated by <strong>the</strong> C-terminal PDZ motif <strong>of</strong> MUC3, binding to <strong>the</strong> single GOPC PDZ domain.<br />

GOPC is also a binding partner for cystic fibrosis transmembrane conductance regulator<br />

(CFTR) that directs CFTR for degradation. Overexpression <strong>of</strong> GOPC downregulated <strong>the</strong> total<br />

levels <strong>of</strong> MUC3, an effect that was reversed by introducing CFTR. The results suggest that<br />

CFTR and MUC3 compete for binding to GOPC, which in turn can regulate levels <strong>of</strong> <strong>the</strong>se two<br />

proteins. For <strong>the</strong> first time a direct coupling between mucins and <strong>the</strong> CFTR channel is<br />

demonstrated, a finding that will shed fur<strong>the</strong>r light on <strong>the</strong> still poorly understood relationship<br />

between cystic fibrosis and <strong>the</strong> mucus phenotype <strong>of</strong> this disease.<br />

1975<br />

For “The Perfect Antagonists” <strong>the</strong> winners are: MyoIC and MyoIE, for <strong>the</strong>ir role in “Actin<br />

shaping for vesicle exocytosis at <strong>the</strong> immune synapse.”<br />

J. M. DIAZ MUNOZ 1,2 , M. I. Yuseff 1 , D. Lankar 1 , P. Pierobon 1 , M. Rosemblatt 3 , A. M. Lennon-<br />

Dumenil 1 ; 1 INSERM U932, Immunity and cancer, Institut Curie, Paris, France, 2 Genetica<br />

<strong>Molecular</strong> y Microbiologia, P. Universidad Catolica de Chile, Santiago de Chile, Chile,<br />

3 Fundacion Ciencia para la Vida, Santiago de Chile, Chile<br />

Engagement <strong>of</strong> <strong>the</strong> B Cell Receptor (BCR) by surface-te<strong>the</strong>red antigens (Ag) leads to formation<br />

<strong>of</strong> a synapse that promotes Ag uptake and presentation onto MHCII molecules. Here, we<br />

highlight <strong>the</strong> membrane trafficking events and associated molecular mechanisms required for<br />

efficient Ag extraction and processing at <strong>the</strong> B cell synapse. We show that MHCII-containing<br />

lysosomes are recruited at <strong>the</strong> synapse and locally undergo exocytosis, a process that relies on


<strong>SUNDAY</strong><br />

<strong>the</strong> SNARE protein Vamp-7. Lysosome secretion allows <strong>the</strong> extracellular release <strong>of</strong> proteases,<br />

whose activities promote <strong>the</strong> extraction <strong>of</strong> <strong>the</strong> immobilized Ag. We fur<strong>the</strong>r show that local reorganization<br />

<strong>of</strong> cortical actin by type I Myosins, which link <strong>the</strong> cortex to <strong>the</strong> plasma membrane,<br />

is required for lysosome exocytosis and Ag extraction. Remarkably, while <strong>the</strong> short-tail Myosin<br />

IC is recruited at <strong>the</strong> synapse and facilitates vesicle secretion and Ag uptake, <strong>the</strong> long-tail<br />

Myosin IE negatively regulates both processes. These results suggest that <strong>the</strong> two class I<br />

Myosins play antagonistic roles in <strong>the</strong> local reorganization <strong>of</strong> cortical actin for vesicle secretion<br />

and Ag uptake. The B cell synapse <strong>the</strong>refore emerges as a highly specialized site where tightly<br />

regulated exocytic and endocytic events take place thanks to <strong>the</strong> local shaping <strong>of</strong> <strong>the</strong><br />

membrane-cytoskeleton interface by class I Myosins<br />

1976<br />

Determination <strong>of</strong> <strong>the</strong> Subcellular, Surface, and Extracellular Localization <strong>of</strong> Hsp70s in<br />

Mammalian Cells.<br />

R. Medina* 1 , M. Rashedan* 1 , N. Nikolaidis 1 ; 1 Biological Science, California State University,<br />

Fullerton, Fullerton, CA<br />

70-kD Heat shock proteins (Hsp70s) are a family <strong>of</strong> molecular chaperones that play essential<br />

roles in stress response by promoting protein homeostasis. Members <strong>of</strong> this family are primarily<br />

localized in different subcellular compartments, including <strong>the</strong> cytosol, <strong>the</strong> endoplasmic reticulum,<br />

and <strong>the</strong> mitochondria. Apart from <strong>the</strong>ir primary location in specific parts <strong>of</strong> <strong>the</strong> cell, different<br />

Hsp70s have also been found in o<strong>the</strong>r places within <strong>the</strong> cell, at <strong>the</strong> cellular membrane, and at<br />

<strong>the</strong> extracellular milieu. Although a few stresses and pathophysiological conditions, like cancer,<br />

have been related with <strong>the</strong> re-localization <strong>of</strong> Hsp70s within <strong>the</strong> cell, <strong>the</strong>ir translocation to <strong>the</strong><br />

membrane, and <strong>the</strong>ir secretion from viable cells, <strong>the</strong> majority <strong>of</strong> <strong>the</strong>se conditions remain<br />

unknown. Additionally, <strong>the</strong> signaling mechanisms involved in <strong>the</strong> trafficking <strong>of</strong> intracellular<br />

Hsp70s remain elusive. To this end we studied <strong>the</strong> intra-, membrane- and extra-cellular<br />

localization <strong>of</strong> four members <strong>of</strong> <strong>the</strong> Hsp70 family. Specifically, we determined <strong>the</strong> re-localization<br />

<strong>of</strong> HSPA1A and HSPA8, primarily localized in <strong>the</strong> cytosol, HSPA5, primarily localized in <strong>the</strong><br />

endoplasmic reticulum, and HSPA9, primarily localized in <strong>the</strong> mitochondria. Human embryonic<br />

cells treated with different stressors, including heat and ethanol, or untreated were subjected to<br />

sub-cellular fractionation and <strong>the</strong> presence <strong>of</strong> Hsp70s was determined by Western. The amount<br />

<strong>of</strong> Hsp70s in <strong>the</strong> different fractions was quantified and normalized for equal loads using fractionspecific<br />

antibodies, e.g., beta-actin for <strong>the</strong> cytosolic fraction. These experiments revealed that<br />

under normal growth conditions all four proteins were present in <strong>the</strong> nuclear, cytosolic,<br />

mitochondrial, and membrane fractions, as well as in <strong>the</strong> extracellular medium. In heat-shocked<br />

cells, although all proteins were still present in all fractions <strong>the</strong>ir amounts in each fraction were<br />

significantly different from <strong>the</strong> untreated cells. These results strongly suggest that after stress<br />

<strong>the</strong> different Hsp70s are being re-localized within <strong>the</strong> cell, are anchored at <strong>the</strong> cellular<br />

membrane, and are secreted from <strong>the</strong> cell. Additional experiments using pharmacological<br />

manipulation <strong>of</strong> intracellular trafficking pathways are currently being performed to identify <strong>the</strong><br />

molecular mechanism used by Hsp70s to achieve <strong>the</strong>ir translocation and subsequent secretion.<br />

This study is <strong>the</strong> first step towards elucidating <strong>the</strong> biological importance <strong>of</strong> <strong>the</strong> relocalization and<br />

membrane occurrence <strong>of</strong> <strong>the</strong>se chaperones.<br />

*equal contribution


<strong>SUNDAY</strong><br />

1977<br />

Fab1/PIKfyve Is Required in Multiple Organs, and is <strong>the</strong> Major Pathway for Production <strong>of</strong><br />

PI(3,5)P2 and PI5P in Mammals.<br />

S. N. Zolov 1 , D. Bridges 1 , Y. Zhang 2 , R. Verma 1 , W-W. Lee 1 , G. M. Lenk 3 , K. Converso-Baran 4 ,<br />

R. Albin 5 , A. Saltiel 1,6 , M. Meisler 3 , M. Russell 4 , L. S. Weisman 2 ; 1 Life Sciences Institute,<br />

University <strong>of</strong> Michigan, Ann Arbor, MI, 2 Department <strong>of</strong> Cell and Developmental <strong>Biology</strong> and Life<br />

Sciences Institute, University <strong>of</strong> Michigan, Ann Arbor, MI, 3 Department <strong>of</strong> Human Genetics,<br />

University <strong>of</strong> Michigan, Ann Arbor, MI, 4 Department <strong>of</strong> Pediatrics and Communicable Diseases,<br />

University <strong>of</strong> Michigan, Ann Arbor, MI, 5 Department <strong>of</strong> Neurology, University <strong>of</strong> Michigan, Ann<br />

Arbor, MI, 6 Departments <strong>of</strong> Internal Medicine, University <strong>of</strong> Michigan, Ann Arbor, MI<br />

Mutations that cause defects in <strong>the</strong> levels <strong>of</strong> <strong>the</strong> signaling lipids PI(3,5)P2 and PI5P lead to<br />

pr<strong>of</strong>ound neurodegeneration in mice. Moreover mutations in human FIG4 predicted to lower<br />

PI(3,5)P2 and PI5P levels underlie some cases <strong>of</strong> Charcot-Marie-Tooth syndrome and<br />

Amyotrophic <strong>Late</strong>ral Sclerosis. In mammals, PI(3,5)P2 is generated by a protein complex that<br />

includes <strong>the</strong> lipid kinase Fab1/PIKfyve, <strong>the</strong> scaffolding protein Vac14, and <strong>the</strong> lipid phosphatase<br />

Fig4. Fibroblasts cultured from Vac14 -/- mutant mice have a 50% reduction in <strong>the</strong> levels <strong>of</strong><br />

PI(3,5)P2 and PI5P, which leaves open <strong>the</strong> question <strong>of</strong> whe<strong>the</strong>r <strong>the</strong>re is a second pathway to<br />

generate <strong>the</strong>se lipids. To address this question we characterize a Fab1 gene-trap mouse<br />

(Fab1 β-geo/β-geo ). The homozygous Fab1 �β-geo/β-geo mouse is a hypomorph that expresses 10% <strong>of</strong><br />

<strong>the</strong> normal levels <strong>of</strong> Fab1 and produces 50% <strong>of</strong> <strong>the</strong> normal levels <strong>of</strong> PI(3,5)P2. When we used<br />

shRNA silencing in Fab1 β-geo/β-geo fibroblasts to knock-down <strong>the</strong> remaining Fab1, we observed a<br />

loss <strong>of</strong> detectable PI(3,5)P2 and an 85% reduction in PI5P. These fibroblasts also have a<br />

substantial increase in <strong>the</strong> amount <strong>of</strong> PI3P, <strong>the</strong> substrate for Fab1. Thus, similar to yeast,<br />

conversion <strong>of</strong> PI3P to PI(3,5)P2 in mammals requires <strong>the</strong> Fab1 pathway. Surprisingly, much <strong>of</strong><br />

<strong>the</strong> PI5P pool also requires <strong>the</strong> Fab1 pathway. Similar to Vac14 -/- and Fig4 -/- mice, <strong>the</strong> Fab1<br />

hypomorphic mouse exhibits neurodegeneration in <strong>the</strong> brain and in peripheral sensory neurons.<br />

However, <strong>the</strong>se mice also have pr<strong>of</strong>ound defects in <strong>the</strong> heart, lung, kidney, thymus, spleen and<br />

intestine. Importantly, we examined <strong>the</strong> heart in Vac14 -/- and Fig4 -/- mouse mutants and found<br />

similar defects to those observed in <strong>the</strong> Fab1 β-geo/β-geo mouse. Thus PI(3,5)P2 and possibly PI5P<br />

have major roles in multiple organs.<br />

1978<br />

Characterization <strong>of</strong> Trak1, a novel protein implicated in hypertonia and epilepsy.<br />

C. A. Lee 1 , L. Li 1 , L-S. Chin 1 ; 1 Pharmacology, Emory University, Atlanta, GA<br />

Hypertonia—a pathophysiological condition characterized by postural abnormalities, jerky<br />

movements, and tremor—is associated with a number <strong>of</strong> neurological disorders, including<br />

cerebral palsy, dystonia, Parkinson's disease, stroke, and epilepsy. The pathogenic<br />

mechanisms that trigger hypertonia remain elusive. Interestingly, a homozygous truncation<br />

mutation in <strong>the</strong> trafficking protein, kinesin-binding 1 (Trak1) protein causes a recessively<br />

transmitted form <strong>of</strong> hypertonia in mice. Moreover, recent genome wide high-density SNP-based<br />

linkage analysis has linked variants in Trak1 to childhood absence epilepsy in humans. Despite<br />

<strong>the</strong> critical importance <strong>of</strong> Trak1 in maintaining neuronal function, <strong>the</strong> mechanism and sites <strong>of</strong><br />

Trak1 action remain unclear and <strong>the</strong> pathogenic mechanism by which Trak1 mutation causes<br />

hypertonia is unknown. We have generated a highly specific anti-Trak1 antibody and shown that<br />

endogenous Trak1 protein is expressed in multiple tissues, including <strong>the</strong> brain.<br />

Immun<strong>of</strong>luorescence confocal microscopic studies show that endogenous Trak1 protein is<br />

localized in axons, dendrites, and cell bodies <strong>of</strong> mouse cortical neurons, supporting a functional<br />

role <strong>of</strong> Trak1 in neuronal physiology. We found that endogenous Trak1 protein is associated<br />

with both early endosomes and mitochondria in cortical neurons. Our data suggest that Trak1


<strong>SUNDAY</strong><br />

protein may play a critical role in maintaining neuronal homeostasis by regulating endosomal<br />

trafficking and mitochondrial transport. Fur<strong>the</strong>r characterization <strong>of</strong> <strong>the</strong> mechanisms <strong>of</strong> Trak1<br />

action and its regulation will provide a better understanding <strong>of</strong> <strong>the</strong> role <strong>of</strong> Trak1 in hypertonia<br />

and epilepsy.<br />

1979<br />

Post-Fusion Actin Coating <strong>of</strong> Secretory Vesicles is required for Active Content Extrusion<br />

in Alveolar Type II Cells.<br />

P. Miklavc 1 , E. Hecht 1 , N. Hobi 2 , O. H. Wittekindt 1 , T. Haller 2 , P. Dietl 1 , E. Felder 1 , C. Kranz 1 , M.<br />

Frick 1 ; 1 University <strong>of</strong> Ulm, Ulm, Germany, 2 Innsbruck Medical University, Innsbruck, Austria<br />

Exocytosis <strong>of</strong> secretory vesicles is a fundamental cellular process allowing regulated secretion<br />

<strong>of</strong> vesicle contents in <strong>the</strong> extracellular space. Growing evidence suggests that regulation <strong>of</strong><br />

fusion pore dilation during <strong>the</strong> post-fusion stage <strong>of</strong> exocytosis plays an important role in release<br />

<strong>of</strong> secretory vesicle contents. However, dependent on <strong>the</strong> nature <strong>of</strong> <strong>the</strong> cargo additional<br />

mechanisms might be essential to facilitate effective release. We have recently described in<br />

alveolar type II cells that surfactant-storing secretory vesicles (lamellar bodies) are coated with<br />

actin following fusion with <strong>the</strong> plasma membrane. Surfactant, a lipoprotein-like substance, does<br />

not readily diffuse out <strong>of</strong> fused lamellar bodies following opening and dilation <strong>of</strong> <strong>the</strong> fusion pore.<br />

Using fluorescence microscopy, atomic force microscopy and biochemical assays we present<br />

evidence that actin coating and subsequent compression <strong>of</strong> <strong>the</strong> actin coat is essential to<br />

facilitate surfactant secretion. Simultaneous imaging <strong>of</strong> <strong>the</strong> vesicle membrane and <strong>the</strong> actin coat<br />

revealed that contraction <strong>of</strong> <strong>the</strong> actin coat compresses <strong>the</strong> vesicle following fusion. This leads to<br />

active extrusion <strong>of</strong> vesicle contents. Preventing actin coating <strong>of</strong> fused LBs with latrunculin A<br />

inhibits surfactant secretion almost completely. Subsequent compression <strong>of</strong> <strong>the</strong> actin coat is<br />

modulated by myosin II. In summary our data suggest that fusion pore opening and dilation is<br />

not sufficient for “passive” release <strong>of</strong> bulky vesicle cargo in alveolar type II cells and that active<br />

extrusion mechanisms are required.<br />

1980<br />

Synaptotagmin-7 - <strong>the</strong> link between fusion-activated Ca 2+ -entry and regulation <strong>of</strong> fusion<br />

pore expansion?<br />

N. Sharma 1 , P. Miklavc 1 , K. Thompson 1 , O. H. Wittekindt 1 , E. Felder 1 , P. Dietl 1 , M. Frick 1 ;<br />

1 Institute <strong>of</strong> General Physiology, University <strong>of</strong> Ulm, Ulm, Germany<br />

Ca 2+ is <strong>the</strong> key element in regulated exocytosis controlling multiple steps during <strong>the</strong> exocytic<br />

pre- and post-fusion stages. We have recently described a “fusion-activated” Ca 2+ -entry (FACE)<br />

via vesicular P2X4 receptors during <strong>the</strong> post-fusion stage <strong>of</strong> lamellar body exocytosis in alveolar<br />

type II (ATII) cells. FACE regulates fusion pore expansion and vesicle content release during<br />

<strong>the</strong> post-fusion phase <strong>of</strong> exocytosis (PNAS, 2011 Aug 30;108(35):14503-8). Yet, <strong>the</strong> question<br />

remained how this locally restricted Ca 2+ signal is translated into mechanical force promoting<br />

fusion pore expansion. In this study we tested whe<strong>the</strong>r synaptotagmins could form a molecular<br />

link between FACE and fusion pore expansion. Members <strong>of</strong> <strong>the</strong> synaptotagmin family that<br />

localize to secretory vesicles have been found to promote fusion pore expansion via Ca 2+<br />

binding to C2 domains. Using RT-PCR and western-blotting we identified synaptotagmin-7 as<br />

<strong>the</strong> Ca 2+ -binding is<strong>of</strong>orm predominantly expressed in primary ATII cells. Immun<strong>of</strong>luorescence<br />

confirmed for <strong>the</strong> first time that synaptotagmin-7 is localised on lamellar bodies. We next<br />

performed functional studies over-expressing wt synaptotagmin-7 or mutants abolishing Ca 2+ -<br />

binding to ei<strong>the</strong>r <strong>the</strong> C2A, C2B or C2A and C2B domains. We measured diffusion rates <strong>of</strong><br />

fluorescent dyes through <strong>the</strong> fusion pore to directly assess dynamic changes in fusion pore<br />

diameters. Our results show that overexpression <strong>of</strong> nei<strong>the</strong>r wt nor mutant synaptotagmin-7


<strong>SUNDAY</strong><br />

affects fusion pore expansion in <strong>the</strong> absence <strong>of</strong> FACE. However, when fusions were followed by<br />

FACE fusion pore expansion was modulated by over-expression <strong>of</strong> wt or mutant synaptotagmin-<br />

7.<br />

In summary <strong>the</strong>se data confirm that FACE constitutes an “autoregulatory” mechanism,<br />

modulating <strong>the</strong> post-fusion fate <strong>of</strong> individual vesicles. In this model synaptotagmin-7 constitutes<br />

<strong>the</strong> Ca 2+ sensor for FACE providing <strong>the</strong> link between FACE and fusion pore expansion in<br />

lamellar body exocytosis.<br />

1981<br />

Analysis <strong>of</strong> GGA null mice demonstrates a non-redundant role for mammalian GGA2<br />

during development.<br />

B. Doray 1 , J. Govero 1 , S. Kornfeld 1 ; 1 Washington University Sch Med, St Louis, MO<br />

The mammalian GGAs (Golgi-localized, gamma-ear containing, ADP-ribosylation factor- binding<br />

proteins) comprise three homologous proteins that function as monomeric clathrin adaptors.<br />

Numerous studies using cultured mammalian cells have shown that <strong>the</strong> three GGAs function in<br />

<strong>the</strong> transport <strong>of</strong> cargo proteins between <strong>the</strong> trans-Golgi network and endosomes. However, <strong>the</strong><br />

in vivo role(s) <strong>of</strong> <strong>the</strong>se adaptor proteins and <strong>the</strong>ir possible functional redundancy has not been<br />

analyzed. To address this issue, gene ablation studies were performed in mice using insertional<br />

mutagenesis. Our data demonstrate that loss <strong>of</strong> GGA1 or GGA3 alone is well tolerated whereas<br />

<strong>the</strong> absence <strong>of</strong> GGA2 results in embryonic lethality (C57BL/6J and 129/Ola mixed parentage),<br />

or neonatal lethality (C57BL/6NJ background). Out <strong>of</strong> 169 pups from Gga2 heterozygous<br />

crosses in <strong>the</strong> C57BL/6NJ genetic background, a single Gga2 -/- mouse escaped lethality,<br />

remains alive and is fertile when crossed with a Gga2 +/- male. This result indicates that GGA2<br />

mediates some vital function during embryogenesis and <strong>the</strong> neonatal period that cannot be<br />

compensated for by GGA1 and/or GGA3 but beyond this period, GGA2 may be dispensable.<br />

This result is also consistent with our expression studies showing that <strong>the</strong> level <strong>of</strong> GGA2 is<br />

highest in <strong>the</strong> brain during embryonic development and early life but decreases substantially two<br />

weeks after birth. In contrast, brain expression <strong>of</strong> GGA1 and GGA3 remains consistently high<br />

from late embryogenesis through <strong>the</strong> adult stage. The fact that all three GGAs are expressed at<br />

comparable levels in <strong>the</strong> brain during embryonic development excludes <strong>the</strong> possibility that<br />

GGA2 is <strong>the</strong> sole GGA present during this phase, at least in <strong>the</strong> brain. The combined loss <strong>of</strong><br />

GGA1 and GGA3 also results in a high incidence <strong>of</strong> neonatal mortality (58% mortality within <strong>the</strong><br />

first 3 weeks) but in this case <strong>the</strong> expression level <strong>of</strong> GGA2 may be inadequate to compensate<br />

for <strong>the</strong> loss <strong>of</strong> <strong>the</strong> o<strong>the</strong>r two GGAs. We conclude that <strong>the</strong> three mammalian GGAs are essential<br />

proteins that are not fully redundant.<br />

1982<br />

Severing <strong>of</strong> actin filaments by c<strong>of</strong>ilin is required for both disassembly and assembly <strong>of</strong><br />

<strong>the</strong> actin patch in fission yeast.<br />

Q. Chen 1 , T. Pollard 1,2 ; 1 Department <strong>of</strong> <strong>Molecular</strong>, Cell and Developmental <strong>Biology</strong>, Yale<br />

University, New Haven, CT, 2 Department <strong>of</strong> <strong>Molecular</strong> Biophysics and Biochemistry, Yale<br />

University, New Haven, CT<br />

We use mutant c<strong>of</strong>ilins to test <strong>the</strong> proposal that c<strong>of</strong>ilin severs actin filaments during endocytosis<br />

in fission yeast. We replaced <strong>the</strong> endogenous protein with a mutant c<strong>of</strong>ilin that is defective in<br />

severing actin filaments. We used quantitative fluorescence microscopy to study <strong>the</strong> endocytic<br />

actin patches in <strong>the</strong>se mutant cells. We tracked GFP tagged patch markers in <strong>the</strong> patches,<br />

including early endocytic adaptor proteins, activators <strong>of</strong> Arp2/3 complex and actin filaments.<br />

Consistent with <strong>the</strong> hypo<strong>the</strong>sis, <strong>the</strong> actin patches disassembled far slower during <strong>the</strong>ir inward<br />

movement than in wild type cells. Abundant actin filaments accumulated at endocytic sites, even


<strong>SUNDAY</strong><br />

after <strong>the</strong> membrane adaptor proteins dissociated from <strong>the</strong> patches. We were surprised to find<br />

that actin patches also assembled slowly in <strong>the</strong>se c<strong>of</strong>ilin mutant cells. Adaptor proteins such as<br />

End4p and Pan1p accumulated and persisted at <strong>the</strong> endocytic sites more than 10 times longer<br />

than in wild type cells, followed by delayed put persistent recruitment <strong>of</strong> activators <strong>of</strong> Arp2/3<br />

complex, including WASP and myosin I. We propose that severing by c<strong>of</strong>ilin normally produces<br />

short actin filaments that diffuse out <strong>of</strong> actin patches and stimulate Arp2/3 complex in adjacent<br />

patches by serving as <strong>the</strong> mo<strong>the</strong>r filaments that initiate <strong>the</strong> autocatalytic branching reaction.<br />

Additional feedback mechanisms seem to prolong <strong>the</strong> early steps in <strong>the</strong> pathway until <strong>the</strong> later<br />

steps are executed.<br />

1983<br />

Structural and functional characterization <strong>of</strong> cargo-binding sites <strong>of</strong> <strong>the</strong> µ4-subunit <strong>of</strong><br />

adaptor protein complex 4.<br />

B. H. Ross 1 , E. A. Corales 1 , Y. Lin 1 , J. H. Hurley 2 , J. S. Bonifacino 3 , P. V. Burgos 1 , G. A.<br />

Mardones 1 ; 1 Universidad Austral de Chile Sch Med, Valdivia, Chile, 2 LMB, National Institute <strong>of</strong><br />

Diabetes and Digestive and Kidney Diseases, NIH, Be<strong>the</strong>sda, MD, 3 CBMP, National Institute <strong>of</strong><br />

Child Health and Human Development, NIH, Be<strong>the</strong>sda, MD<br />

Adaptor protein (AP) complexes assist protein trafficking by playing key roles in <strong>the</strong> selection <strong>of</strong><br />

cargo molecules to be sorted in post-Golgi compartments. The medium-sized subunit (µ1-µ4) <strong>of</strong><br />

<strong>the</strong> four heterotetrameric AP complexes recognizes YXXØ-sequences (Ø is a bulky hydrophobic<br />

residue), which are sorting signals in transmembrane proteins. A conserved region, <strong>the</strong> µ2binding<br />

site, mediates recognition <strong>of</strong> YXXØ-signals. Recently we found that a non-canonical<br />

YXXØ-signal binds to a distinct µ4-binding site <strong>of</strong> <strong>the</strong> AP-4 complex. In this study we aimed to<br />

determine <strong>the</strong> functionality <strong>of</strong> both binding sites on <strong>the</strong> recognition <strong>of</strong> this non-canonical YXXØsignal.<br />

We used site-directed mutagenesis, yeast-two hybrid (Y2H) analyses, iso<strong>the</strong>rmal titration<br />

calorimetry (ITC), and X-ray crystallography. Substitutions in ei<strong>the</strong>r <strong>of</strong> both binding sites on µ4<br />

abrogated binding to <strong>the</strong> APP-tail in Y2H experiments. Fur<strong>the</strong>r characterization by ITC showed<br />

no binding only with <strong>the</strong> R283D substitution at <strong>the</strong> µ4-binding site, in contrast with a decrease in<br />

binding affinity with <strong>the</strong> substitution D190A at <strong>the</strong> µ2-binding site. We solved <strong>the</strong> crystal<br />

structure <strong>of</strong> <strong>the</strong> C-terminal domain <strong>of</strong> <strong>the</strong> D190A mutant <strong>of</strong> <strong>the</strong> µ4 subunit bound to <strong>the</strong> noncanonical<br />

YXXØ-signal. This structure showed no significant difference compared to that <strong>of</strong><br />

wild-type µ4 subunit. Our mutational, biochemical and structural analyses established <strong>the</strong> role <strong>of</strong><br />

<strong>the</strong> µ4-binding site for <strong>the</strong> non-canonical YXXØ-signal.<br />

FONDECYT 1100896<br />

1984<br />

Regulation <strong>of</strong> <strong>the</strong> RalGAP Complex by Akt-Catalyzed Phosphorylation.<br />

D. Leto 1,2 , X-W. Chen 1,2 , A. Burk 1,2 , T. Xiong 1,3 , G. Yu 1,2 , A. Saltiel 1,2 ; 1 University <strong>of</strong> Michigan,<br />

Ann Arbor, MI, 2 Life Sciences Institute, University <strong>of</strong> Michigan, Ann Arbor, MI, 3 Department <strong>of</strong><br />

<strong>Molecular</strong> and Integrative Physiology, University <strong>of</strong> Michigan, Ann Arbor, MI<br />

In response to feeding, <strong>the</strong> anabolic hormone insulin increases glucose uptake into adipocytes<br />

by stimulating exocytosis <strong>of</strong> vesicles containing <strong>the</strong> facilitative glucose transporter, Glut4. The<br />

small vesicle-localized G protein, RalA, is rapidly and transiently activated by insulin in an Aktdependent<br />

manner. When bound to GTP, RalA facilitates recognition <strong>of</strong> Glut4 vesicles at <strong>the</strong><br />

plasma membrane by interacting with <strong>the</strong> exocyst, an 8-subunit te<strong>the</strong>ring complex. We have<br />

previously shown that in <strong>the</strong> absence <strong>of</strong> insulin, RalA is retained in a largely inactive state by a<br />

RalGAP Complex comprised <strong>of</strong> a regulatory subunit, RGC1, and a catalytic subunit, RGC2, that<br />

contains a GAP domain with specific activity toward RalA. Activation <strong>of</strong> this GTPase by insulin<br />

requires inhibition <strong>of</strong> <strong>the</strong> RalGAP complex. Here, we show that insulin stimulates Akt-catalyzed


<strong>SUNDAY</strong><br />

phosphorylation <strong>of</strong> RGC1/2 on at least three residues, and that phosphorylation inhibits <strong>the</strong><br />

complex without directly inactivating its catalytic activity. We have identified a novel interaction<br />

between 14-3-3 and RGC1/2. This interaction is dependent on phosphorylation <strong>of</strong> RGC2 on a<br />

residue that was previously identified as an Akt target site. 14-3-3 binding does not interrupt <strong>the</strong><br />

interaction between RGC1 and RGC2 or <strong>the</strong> catalytic activity <strong>of</strong> <strong>the</strong> complex. Thus, 14-3-3 may<br />

regulate RGC1/2 by altering its interaction with RalA, as has been reported for o<strong>the</strong>r GAP<br />

complexes.<br />

1985<br />

The Rab GAP Msb3/Gyp3 regulates dose-dependent Rab5/Vps21 signaling.<br />

D. Nickerson 1 , A. Merz 1 ; 1 Biochemistry, University <strong>of</strong> Washington, Seattle, WA<br />

Membrane transport into and out <strong>of</strong> endosomes in yeast requires <strong>the</strong> function <strong>of</strong> a trio <strong>of</strong> Rab5<br />

orthologs: Vps21, Ypt52 and Ypt53. While loss <strong>of</strong> Vps21 causes obvious trafficking phenotypes,<br />

<strong>the</strong> roles <strong>of</strong> its paralogs Ypt52 and Ypt53 are mostly unexplored and <strong>the</strong> regulatory logic <strong>of</strong> <strong>the</strong><br />

trio is unclear. We report that Ypt52 and Ypt53 cooperate with Vps21 to mediate biosyn<strong>the</strong>tic<br />

transport <strong>of</strong> CPY and multivesicular body cargoes to <strong>the</strong> vacuole. Cells deficient in multiple<br />

Rab5 paralogs display syn<strong>the</strong>tic defects in both <strong>the</strong>se processes and in calcium resistance.<br />

Ypt53 functions as a calcineurin-dependent calcium stress response factor. We also find that<br />

conditions <strong>of</strong> Rab5 signaling deficiency result in significant upregulation <strong>of</strong> YPT53, suggesting<br />

that Ypt53 functions as a positive modulator <strong>of</strong> Rab5 signal in response to both environmental<br />

stresses and trafficking malfunctions. We fur<strong>the</strong>r report that Msb3/Gyp3 functions as <strong>the</strong><br />

principle GAP for Vps21 in vivo. Loss <strong>of</strong> Gyp3 activity results in both greater accumulation <strong>of</strong><br />

GTP-bound Vps21 on endosomes and mislocalization <strong>of</strong> Vps21 to vacuoles, suggesting that<br />

Gyp3 serves <strong>the</strong> dual purposes <strong>of</strong> negatively modulating <strong>the</strong> degree <strong>of</strong> Rab5 signaling and<br />

spatially restricting Vps21 to endosomes.<br />

1986<br />

Glycosaminoglycan attachment affects <strong>the</strong> intracellular transport <strong>of</strong> <strong>the</strong> amyloid<br />

precursor protein.<br />

D. Mihov 1 , M. Spiess 1 ; 1 Biozentrum, University <strong>of</strong> Basel, Basel, Switzerland<br />

Amyloid precursor protein (APP) is a type I transmembrane protein implicated in <strong>the</strong><br />

pathophysiology <strong>of</strong> Alzheimer disease by releasing <strong>the</strong> amyloidogenic Aβ peptide. The<br />

intracellular distribution <strong>of</strong> <strong>the</strong> different secretases and <strong>the</strong> sorting and transport <strong>of</strong> APP define<br />

<strong>the</strong> extents <strong>of</strong> amyloidogenic and non-amyloidogenic processing <strong>of</strong> <strong>the</strong> protein. APP splice<br />

variants lacking exon 15 contain a glycosaminoglycan (GAG) attachment site and are modified<br />

with a single chondroitin sulfate (ChS) chain. We have previously shown that GAG attachment<br />

affects exocytic and endocytic traffic <strong>of</strong> model proteins. Therefore, we investigated <strong>the</strong> effect <strong>of</strong><br />

GAG attachment on intracellular traffic and processing <strong>of</strong> APP. HeLa cells were transiently<br />

transfected with <strong>the</strong> neuronal-specific splice variant APP-695 or with its corresponding<br />

proteoglycan is<strong>of</strong>orm APP-677, lacking exon 15. Since APP is naturally tyrosine sulfated, we<br />

used radioactive sulfate to metabolically label transfected cells and to simultaneously detect <strong>the</strong><br />

GAG-containing and GAG-free forms <strong>of</strong> APP. About 50% <strong>of</strong> APP-677 was GAG-modified,<br />

containing ChS chains. Pulse-chase experiments showed that GAGs accelerated <strong>the</strong><br />

biosyn<strong>the</strong>tic exocytosis <strong>of</strong> <strong>the</strong> APP-677 proteoglycan in comparison with APP-695 and APP-677<br />

lacking ChS. APP contains well characterized signals for endocytosis from <strong>the</strong> plasma<br />

membrane. While GAG-free APP was internalized at a rate <strong>of</strong> about 6% per min, GAG<br />

attachment resulted in a significant reduction <strong>of</strong> endocytosis <strong>of</strong> at least threefold. The rates <strong>of</strong><br />

recycling, however, were not affected. Our results indicate that exon-15 splicing and subsequent


<strong>SUNDAY</strong><br />

GAG attachment alters APP intracellular transport, potentially influencing its processing and Aβ<br />

generation.<br />

1987<br />

Methyl esterification <strong>of</strong> retinal proteins is essential for rod-mediated vision.<br />

J. R. Christiansen 1 , S. Kolandaivelu 1 , M. O. Bergo 2 , S. G. Young 3 , V. Ramamurthy 1,4 ;<br />

1 Ophthalmology, West Virginia University, Morgantown, WV, 2 Cancer Center Sahlgrenska,<br />

University <strong>of</strong> Go<strong>the</strong>nburg, 3 eDepartments <strong>of</strong> Medicine and Human Genetics, David Geffen<br />

School <strong>of</strong> Medicine University <strong>of</strong> California, Los Angeles, 4 Biochemistry, West Virginia University<br />

Background: Proteins ending in a “CAAX” box are first prenylated at <strong>the</strong>ir c-terminal cysteine<br />

and <strong>the</strong>n RAS-converting enzyme 1 (RCE1) cleaves <strong>the</strong> final three amino acids before<br />

isoprenylcysteine methyltransferase (ICMT) catalyzes <strong>the</strong> methyl esterification <strong>of</strong> <strong>the</strong> newly<br />

prenylated cysteine residue. Prenylation is crucial for a protein’s ability to interact with<br />

membrane domains. The contribution <strong>of</strong> <strong>the</strong> final two steps to a protein’s localization and<br />

function are variable and depends on <strong>the</strong> protein being studied. We recently demonstrated <strong>the</strong><br />

importance <strong>of</strong> RCE1-mediated proteolysis in phototransduction. In <strong>the</strong> absence <strong>of</strong> Rce1,<br />

photoreceptors do not function and rapidly degenerate. To determine if <strong>the</strong> effects seen in <strong>the</strong><br />

retina conditional knockout <strong>of</strong> Rce1 were related to <strong>the</strong> lack <strong>of</strong> proteolysis, lack <strong>of</strong> carboxyl<br />

methylation, or both we analyzed <strong>the</strong> retina <strong>of</strong> mice with reduced levels <strong>of</strong> Icmt.<br />

Methods: We utilized <strong>the</strong> hypomorphic Icmt fl allele as a tool to generate animals expressing<br />

various levels <strong>of</strong> Icmt in <strong>the</strong> retina. Electroretinogram (ERG) recordings were used to analyze<br />

visual function <strong>of</strong> littermate animals with a range <strong>of</strong> Icmt levels. Morphology and<br />

phototransduction protein expression pr<strong>of</strong>iles were investigated by immun<strong>of</strong>luorescence and<br />

immunoblotting respectively.<br />

Results: ERG recordings displayed delayed photoreceptor cell responses and reduced<br />

downstream electrical responses to light stimuli. The delayed photoreceptor cell response<br />

correlates to changes in Icmt message levels in <strong>the</strong> retina. In agreement with delayed<br />

photoreceptor cell responses, critical phototransduction protein levels were also altered. To<br />

investigate <strong>the</strong> cause <strong>of</strong> <strong>the</strong> reduced downstream neuronal responses, we are currently verifying<br />

<strong>the</strong> development and proper synaptic stratification <strong>of</strong> inner retinal neurons.<br />

Conclusion: Efficient coupling <strong>of</strong> signal transduction in photoreceptor neurons requires methyl<br />

esterification <strong>of</strong> a phototransduction protein. In comparison with Rce1 CKO, we did not observe<br />

rapid retinal degeneration or loss <strong>of</strong> visual function. At low light levels, <strong>the</strong> visual response was<br />

reduced corresponding to reductions in Icmt. In agreement with <strong>the</strong> loss <strong>of</strong> visual response<br />

phototransduction protein turnover was also increased.<br />

1988<br />

Turnover <strong>of</strong> amyloid precursor protein carboxy terminal fragment beta (C99) in lysosomal<br />

compartments.<br />

A. Rivera-Dictter 1 , H. Bustamante 1 , V. Muñoz 1 , V. Cavieres 1 , G. A. Mardones 1 , J. S. Bonifacino 2 ,<br />

P. V. Burgos 1 ; 1 Universidad Austral de Chile Sch Med, Valdivia, Chile, 2 CBMP, National Institute<br />

<strong>of</strong> Child Health and Human Development, NIH, Be<strong>the</strong>sda, MD<br />

Proteolytic processing <strong>of</strong> <strong>the</strong> amyloid precursor protein by β-secretase generates C99, which<br />

subsequently is cleaved by γ-secretase, yielding <strong>the</strong> amyloid β peptide (Aβ). C99 contains<br />

within its cytosolic tail key signal motifs for its delivery to lysosomal compartments. Our aim was<br />

to investigate lysosomal turnover <strong>of</strong> C99 in a mechanism independent <strong>of</strong> γ-secretase cleavage.<br />

H4 neuroglioma cells stably expressing C99-EGFP-WT were analyzed in pulse-chase<br />

experiments with cycloheximide, in <strong>the</strong> absence or presence <strong>of</strong> γ-secretase inhibitors testing<br />

several conditions in order to: 1) block delivery to lysosomes and 2) disrupt lysosomal function.


<strong>SUNDAY</strong><br />

Blocking delivery <strong>of</strong> C99 to lysosomes or disrupting lysosomal function caused an enhancement<br />

in its proteolytic processing by γ-secretase. Similar conditions but in <strong>the</strong> presence γ-secretase<br />

inhibitors showed a significant delay in C99 turnover with a strong accumulations in lysosomal<br />

compartments. Our results show that lysosomes play an important role in <strong>the</strong> turnover <strong>of</strong> C99<br />

suggesting that dysfunction <strong>of</strong> <strong>the</strong>se organelles might have key implications in Aβ production<br />

and Alzheimer.<br />

FONDECYT 1100027 and DID-UACH<br />

1989<br />

Ciliary targeting <strong>of</strong> sensory receptors is coordinated by Arf and Rab GTPases and <strong>the</strong>ir<br />

effectors.<br />

D. Deretic 1 , J. Wang 1 ; 1 University <strong>of</strong> New Mexico, Albuqueruqe, NM<br />

Ciliopathies encompass a wide range <strong>of</strong> human diseases caused by dysfunctional primary cilia,<br />

however trafficking <strong>of</strong> signaling receptors to <strong>the</strong> ciliary membrane remains poorly understood. A<br />

recently described ciliary targeting complex organized by Arf4 binds at <strong>the</strong> trans-Golgi network<br />

(TGN) to <strong>the</strong> VxPx motif present in membrane proteins targeted to primary cilia, including<br />

rhodopsin. We now show that <strong>the</strong> ciliary targeting is initiated at <strong>the</strong> TGN, by <strong>the</strong> formation <strong>of</strong> a<br />

tripartite complex between rhodopsin, Arf4GTP and <strong>the</strong> Arf-GAP ASAP1, which each recognize<br />

one <strong>of</strong> <strong>the</strong> two ciliary targeting motifs <strong>of</strong> rhodopsin, VxPx and FR, respectively. Subsequently,<br />

ASAP1 recruits Rab11a and <strong>the</strong> Arf/Rab11 effector FIP3 displaces rhodopsin from ASAP1. On<br />

ciliary transport carriers, ASAP1 provides an activation platform for <strong>the</strong> Rab11a-Rabin8-Rab8<br />

ciliary destination module. Ciliary localization <strong>of</strong> rhodopsin-GFP-VxPx expressed in epi<strong>the</strong>lial<br />

cells is lost upon ablation <strong>of</strong> ASAP1 that causes formation <strong>of</strong> actin-rich periciliary membrane<br />

projections, to which ciliary cargo is redirected. Fur<strong>the</strong>rmore, [FR-AA]rhodopsin-GFP-VxPx that<br />

is defective in ASAP1 binding fails to engage Rab8 and translocate across <strong>the</strong> periciliary<br />

diffusion barrier. As <strong>the</strong> VxPx and FR targeting motifs are present in o<strong>the</strong>r sensory receptors,<br />

comparable coupling <strong>of</strong> cargo recognition with cargo destination likely confers directionality to<br />

ciliary membrane transport.<br />

Organelles and Membrane <strong>Biology</strong><br />

1990<br />

Determinants <strong>of</strong> ciliary identity in Giardia intestinalis.<br />

A. Wiedmann 1 , K. Hagen 1 , S. C. Dawson 1 ; 1 Department <strong>of</strong> Microbiology, University <strong>of</strong> California,<br />

Davis, Davis, CA<br />

Flagella (or cilia) are essential for <strong>the</strong> function <strong>of</strong> many cell types yet little is known about how<br />

different cell types, given <strong>the</strong> same genetic information, can build flagella <strong>of</strong> different size,<br />

protein composition and function. The unicellular intestinal parasite Giardia intestinalis forms<br />

four pairs <strong>of</strong> flagella distinct in length, behavior and protein composition (1). Identifying factors<br />

that allow for <strong>the</strong> formation <strong>of</strong> <strong>the</strong>se flagella could inform research on human diseases caused<br />

by defects in flagellar growth, such as polycystic kidney disease, as well as well as lead to<br />

better treatment <strong>of</strong> <strong>the</strong> disease caused by this parasite. To determine how flagellar identities are<br />

established in Giardia we are (1) developing a novel whole-genome overexpresision anti-sense<br />

screen, and (2) in a candidate approach, are investigating <strong>the</strong> role <strong>of</strong> three putative NEK<br />

kinases in ciliogenesis.<br />

Giardia is a binucleate organism and thus random mutagenesis is not a feasible approach for<br />

identifying ciliogenesis factors. We are using T7 polymerase to direct overexpression <strong>of</strong> a library<br />

<strong>of</strong> small genomic DNA fragments to allow for a whole genome screen for factors important for


<strong>SUNDAY</strong><br />

<strong>the</strong> establishment <strong>of</strong> ciliary identity in Giardia intestinalis. Our candidate approach focuses on<br />

three NEK kinases. NEK kinases were first identified for <strong>the</strong>ir role in mitosis but have been<br />

shown to play a role in ciliogenesis and flagellar length determination in model systems such as<br />

Tetrahymena (2,3). Similar to <strong>the</strong> Tetrahymena genome, <strong>the</strong> giardial genome encodes an<br />

unusually large number <strong>of</strong> <strong>the</strong>se kinases, suggesting that some may have non-mitotic,<br />

regulatory roles. Consistent with this hypo<strong>the</strong>sis, we have identified several putative NEK<br />

kinases associated with <strong>the</strong> giardial ventral disk and flagella. Most <strong>of</strong> <strong>the</strong>se are predicted to be<br />

catalytically inactive and may play structural or kinase activity independent regulatory roles.<br />

Three <strong>of</strong> <strong>the</strong> kinases, GL50803_11311, _8445, and _95593, however, contain a canonical<br />

kinase active site. We are using morpholino knockdown as well as overexpression <strong>of</strong> kinaseinactive<br />

and constitutively active variants to assess whe<strong>the</strong>r <strong>the</strong>se putative kinases contribute to<br />

Giardia motility and/or ciliogenesis.<br />

References:<br />

1. Dawson SC and House SA (2010) Methods Cell Biol.<br />

2. Quarmby LM and Mahjoub MR (2005) J Cell Sci.<br />

3. Wloga D, et al (2006) Mol Biol Cell.<br />

1991<br />

Trophic accumulation <strong>of</strong> <strong>the</strong> antidepressant sertraline/Zol<strong>of</strong>t in fungal secretory<br />

membranes.<br />

E. Perlstein 1 , J. Chen 1 , D. Korostyshevsky 1 ; 1 Lewis-Sigler Institute/Princeton University,<br />

Princeton, NJ<br />

Many antidepressants are cationic amphipaths, which spontaneously accumulate in cellular<br />

membranes in <strong>the</strong> absence <strong>of</strong> <strong>the</strong>ir protein targets. However, <strong>the</strong> clinical relevance <strong>of</strong><br />

progressive cellular membrane infiltration by antidepressants in humans is poorly understood.<br />

Here we take an “evolutionary pharmacology” approach to studying <strong>the</strong> effects <strong>of</strong> <strong>the</strong> selectiveserotonin<br />

reuptake inhibitor sertraline/Zol<strong>of</strong>t on cellular membrane homeostasis in <strong>the</strong> simple<br />

eukaryote Saccharomyces cerevisiae, which lacks a serotonin transporter entirely. We<br />

biochemically characterized <strong>the</strong> association <strong>of</strong> tritiated sertraline with <strong>the</strong> membranes <strong>of</strong> <strong>the</strong><br />

fungal secretory pathway, and in parallel performed a quantitative ultrastructural analysis <strong>of</strong><br />

membrane quality control in untreated vs. sertraline-treated cells. These experiments have<br />

revealed that sertraline enters yeast cells and <strong>the</strong>n reshapes <strong>the</strong> secretory pathway by a<br />

complex process. Internalization <strong>of</strong> <strong>the</strong> neutral species proceeds by simple diffusion amplified<br />

by lysosomotropism, but is counteracted by ATP-dependent xenobiotic efflux. At equilibrium, a<br />

small fraction (10-15%) <strong>of</strong> spontaneously reprotonated sertraline is soluble while <strong>the</strong> remainder<br />

partitions into secretory pathway membranes by both adsorption and intercalation. Asymmetric<br />

accumulation <strong>of</strong> sertraline results in concentration- and time-dependent local membrane<br />

curvature stresses, which in turn trigger an adaptive autophagy-dependent relief response. Our<br />

model appears to support <strong>the</strong> notion <strong>of</strong> a serotonin transporter-independent, “amphipath<br />

accumulation” component <strong>of</strong> antidepressant pharmacology in humans.<br />

1992<br />

The UBX-domain-containing protein Ubx2/ Ubxd8 regulates lipid droplet homeostasis.<br />

C-W. Wang 1 , S-C. Lee 1 ; 1 Institute <strong>of</strong> Plant and Microbial <strong>Biology</strong>, Academia Sinica, Nankang,<br />

Taipei, Taiwan<br />

Lipid droplets (LDs) are central organelles for maintaining lipid homeostasis. However, how cells<br />

control <strong>the</strong> size and number <strong>of</strong> LDs remains largely unknown. Herein, we report that Ubx2, a<br />

protein involved in endoplasmic reticulum (ER)-associated degradation (ERAD), is crucial for LD


<strong>SUNDAY</strong><br />

maintenance. Ubx2 redistributes from ER to LDs when LDs start to form and enlarge during<br />

diauxic shift and in <strong>the</strong> stationary phase. ubx2Δ cells contain abnormal number and reduced<br />

size <strong>of</strong> LDs and <strong>the</strong>ir triacylglycerol (TAG) is reduced to 50% <strong>of</strong> <strong>the</strong> normal level. Deletion <strong>of</strong><br />

ei<strong>the</strong>r UBX or UBA domain in Ubx2 has no effect, but deletion <strong>of</strong> both causes LD phenotypes<br />

similar to that in ubx2Δ. The reduced TAG in ubx2Δ is likely due to mislocalization <strong>of</strong> Lro1, one<br />

<strong>of</strong> <strong>the</strong> acyltransferases required for TAG syn<strong>the</strong>sis, that normally resides at <strong>the</strong> putative LD<br />

assembly sites abutted LDs. The mammalian Ubxd8 expressed in yeast complements <strong>the</strong><br />

defect <strong>of</strong> ubx2Δ, implying a functional conservation for <strong>the</strong>se UBX-domain-containing proteins in<br />

lipid homeostasis.<br />

1993<br />

Mitochondrial Cardiolipin and Phosphatidylethanolamine have Overlapping Functions in<br />

Mitochondrial Fusion in Saccharomyces cerevisiae.<br />

A. S. Joshi 1 , M. N. Thompson 1 , M. Hüttemann 2 , M. L. Greenberg 1 ; 1 Department <strong>of</strong> Biological<br />

Sciences, Wayne State University, Detroit, MI, 2 Center for <strong>Molecular</strong> Medicine and Genetics,<br />

Wayne State University, Detroit, MI<br />

The two non-bilayer forming mitochondrial phospholipids cardiolipin (CL) and<br />

phosphatidylethanolamine (PE) play crucial roles in maintaining mitochondrial morphology. We<br />

have previously shown that <strong>the</strong>se phospholipids have overlapping functions and <strong>the</strong> loss <strong>of</strong> both<br />

is syn<strong>the</strong>tically lethal. Recent in vitro studies have shown that CL stimulates <strong>the</strong> GTPase activity<br />

<strong>of</strong> <strong>the</strong> fusion protein Mgm1p and is required for assembly <strong>of</strong> its is<strong>of</strong>orms. Because <strong>the</strong> lack <strong>of</strong> CL<br />

does not lead to defects in <strong>the</strong> mitochondrial network, we hypo<strong>the</strong>sized that PE may<br />

compensate for CL for maintenance <strong>of</strong> mitochondrial tubular morphology and fusion. To test this<br />

hypo<strong>the</strong>sis, we constructed a conditional double mutant crd1�psd1� containing null alleles <strong>of</strong><br />

CRD1 (lacking CL) and PSD1 (lacking mitochondrial PE), in which <strong>the</strong> wild type CRD1 gene is<br />

present on a plasmid under control <strong>of</strong> <strong>the</strong> TET OFF promoter. Our in vivo studies demonstrate<br />

for <strong>the</strong> first time that mitochondrial fusion and maintenance <strong>of</strong> a mitochondrial tubular network<br />

require <strong>the</strong> presence <strong>of</strong> CL and mitochondrial PE. The crd1�psd1� mutant in <strong>the</strong> presence <strong>of</strong><br />

tetracycline exhibited highly fragmented mitochondria, a phenotype similar to that observed in<br />

fusion mutants. The crd1�psd1� cells fur<strong>the</strong>r exhibited loss <strong>of</strong> mtDNA and reduced membrane<br />

potential, characteristic <strong>of</strong> cells devoid <strong>of</strong> mitochondrial fusion. Deletion <strong>of</strong> FIS1 or DNM1, which<br />

are required for mitochondrial fission, restored <strong>the</strong> tubular mitochondrial morphology in<br />

crd1�psd1� cells. Loss <strong>of</strong> CL and mitochondrial PE led to reduced levels <strong>of</strong> small and large<br />

is<strong>of</strong>orms <strong>of</strong> <strong>the</strong> fusion protein Mgm1p, possibly accounting for <strong>the</strong> fusion defect. Taken toge<strong>the</strong>r,<br />

<strong>the</strong>se data suggest that mitochondrial phospholipids CL and PE are required to maintain tubular<br />

mitochondrial morphology and have overlapping functions in mitochondrial fusion. Defects in<br />

mitochondrial fusion may underlie <strong>the</strong> mitochondrial morphological variation observed in Barth<br />

syndrome (BTHS) lymphoblasts and in cardiac and skeletal muscles <strong>of</strong> <strong>the</strong> mouse model <strong>of</strong><br />

BTHS.<br />

1994<br />

CPSAR1, a protein involved in vesicular trafficking inside <strong>the</strong> chloroplast.<br />

N. Z. Khan 1 , E. Lindquist 1 , S. Karim 1 , H. Aronsson1 1 ; 1 Plant and Environmental Sciences,<br />

University <strong>of</strong> Go<strong>the</strong>nburg, Go<strong>the</strong>nburg, Sweden<br />

COPII coated vesicles are involved in <strong>the</strong> secretory pathway transporting proteins and lipids<br />

from <strong>the</strong> endoplasmic reticulum (ER) to <strong>the</strong> Golgi. On <strong>the</strong> basis <strong>of</strong> Arabidopsis genome<br />

sequences and web prediction tools some important components <strong>of</strong> COPII i.e. <strong>the</strong> Sec23-Sec24<br />

complex, <strong>the</strong> Sec13-Sec31 complex, and <strong>the</strong> small GTPase SAR1 homologues were predicted<br />

to be chloroplast localized. It has recently been verified that CPSAR1 is involved in a vesicular


<strong>SUNDAY</strong><br />

transport system inside <strong>the</strong> chloroplast similar to <strong>the</strong> transport mechanism that exists between<br />

<strong>the</strong> ER and <strong>the</strong> Golgi. CPSAR1 has a role in thylakoid biogenesis and in vesicle budding from<br />

<strong>the</strong> inner envelope membrane for fusion with <strong>the</strong> thylakoids. As this mechanism is similar to <strong>the</strong><br />

cytosolic vesicular transport, a putative model is proposed in which <strong>the</strong> CPSAR1 need o<strong>the</strong>r<br />

protein partners for <strong>the</strong> vesicular formation and transport. For this purpose we are using coimmunoprecipitation<br />

and yeast-2-hybrid for identification <strong>of</strong> protein-protein interaction involving<br />

CPSAR1. Ongoing bioinformatic studies <strong>of</strong> common motifs important for interactions and cargos<br />

<strong>of</strong> <strong>the</strong> secretory pathway gives that <strong>the</strong>y also exist for chloroplast localized proteins i.e. possible<br />

protein interactors to <strong>the</strong> chloroplast vesicle traffic system. In addition, we are studying <strong>the</strong><br />

intraplastidial role <strong>of</strong> CPSAR1 from isolated chloroplast envelopes in vitro.<br />

1995<br />

A Novel Function <strong>of</strong> AWP1/ZFAND6: Regulation <strong>of</strong> Pex5p Export by Interacting with Cysmonoubiquitinated<br />

Pex5p and AAA ATPase, Pex6p.<br />

N. Miyata 1 , K. Okumoto 1,2 , S. Mukai 1 , M. Noguchi 2 , Y. Fujiki 1,3 ; 1 Department <strong>of</strong> <strong>Biology</strong>, Faculty<br />

<strong>of</strong> Sciences, Kyushu University, Fukuoka, Japan, 2 Graduate School <strong>of</strong> Systems Life Sciences,<br />

Kyushu University, Fukuoka, Japan, 3 JST, CREST, Tokyo, Japan<br />

In biogenesis <strong>of</strong> peroxisome, a subcellular organelle, Pex5p is <strong>the</strong> shuttling receptor for<br />

peroxisomal matrix proteins harboring peroxisome-targeting signal 1 (PTS1). Several peroxins<br />

are involved in <strong>the</strong> Pex5p shuttling between <strong>the</strong> cytosol and peroxisomes. However, <strong>the</strong> precise<br />

mechanism underlying <strong>the</strong> PTS1 receptor shuttling remains elusive. We herein suggest that<br />

liver cytosol contains at least two distinct factors involved in <strong>the</strong> Pex5p export. We isolate one <strong>of</strong><br />

<strong>the</strong> factors by biochemical fractionation and in vitro Pex5p export assay and identify it as<br />

AWP1/ZFAND6 (termed p40), a ubiquitin-binding NF-κB modulator. In in vitro Pex5p export<br />

assay, recombinant p40, stimulates Pex5p export, whilst anti-p40 antibody interferes with Pex5p<br />

export. p40 interacts with AAA ATPase Pex6p, but not Pex1p-Pex6p complex. p40 binds Cysubiquitinated<br />

form <strong>of</strong> Pex5p more preferentially than unmodified Pex5p, apparently via its A20<br />

zinc-finger domain. RNA interference for p40 significantly affects <strong>the</strong> PTS1 protein import into<br />

peroxisomes. Fur<strong>the</strong>rmore, in <strong>the</strong> p40 knocked-down cells Pex5p is unstable, as in fibroblasts<br />

from patients each defective in Pex1p, Pex6p, and Pex26p, all prerequisite to <strong>the</strong> Pex5p export.<br />

Taken toge<strong>the</strong>r, p40 is a novel c<strong>of</strong>actor <strong>of</strong> Pex6p involved in <strong>the</strong> regulation <strong>of</strong> Pex5p export in<br />

peroxisome biogenesis.<br />

1996<br />

Docosahexaenoic acid is required for peroxisomal elongation, a prerequisite for division<br />

<strong>of</strong> peroxisomes.<br />

A. Itoyama 1 , M. Honsho 2 , Y. Abe 2 , Y. Yoshida 2 , Y. Fujiki 2,3 ; 1 Graduate School <strong>of</strong> Systems Life<br />

Sciences, Kyushu University, Fukuoka, Japan, 2 Department <strong>of</strong> <strong>Biology</strong>, Faculty <strong>of</strong> Sciences,<br />

Kyushu University, Fukuoka, Japan, 3 CREST, JST, Tokyo, Japan<br />

Peroxisomal division is strictly regulated by several fission factors and cellular environments. In<br />

<strong>the</strong> past decade, metabolic control <strong>of</strong> peroxisomal morphogenesis is postulated but remains<br />

largely undefined. We herein identify docosahexaenoic acid (DHA, C22:6n-3) as <strong>the</strong> regulator <strong>of</strong><br />

<strong>the</strong> peroxisomal morphogenesis. Peroxisomes are much less abundant in fibroblasts from<br />

patients defective in peroxisomal fatty-acid β-oxidation. Supplementation <strong>of</strong> DHA to such<br />

fibroblasts induces proliferation <strong>of</strong> peroxisomes up to <strong>the</strong> same level in normal fibroblasts. DHAinducible<br />

peroxisomal proliferation is abrogated upon treatment with dynamin-like protein 1<br />

siRNA, suggesting <strong>the</strong> DHA-induced division <strong>of</strong> peroxisomes. DHA-induced peroxisomal<br />

division is initiated by elongation <strong>of</strong> peroxisomes in a Pex11p-dependent manner. Fur<strong>the</strong>rmore,<br />

DHA augments hyper-oligomerization <strong>of</strong> Pex11pβ, giving rise to Pex11pβ-enriched regions on


<strong>SUNDAY</strong><br />

<strong>the</strong> elongated peroxisome membrane. Collectively, <strong>the</strong>se findings suggest that DHA is<br />

responsible for <strong>the</strong> elongation step <strong>of</strong> peroxisomes, <strong>the</strong> prerequisite stage for <strong>the</strong> subsequent<br />

fission <strong>of</strong> peroxisomes.<br />

1997<br />

<strong>Molecular</strong> Basis for Targeting <strong>of</strong> C-tail-anchored Proteins to Peroxisomes.<br />

Y. Yagita 1 , Y. Fujiki 2,3 ; 1 Graduate School <strong>of</strong> Systems Life Sciences, Kyushu University, Fukuoka,<br />

Japan, 2 Department <strong>of</strong> <strong>Biology</strong>, Faculty <strong>of</strong> Sciences, Kyushu University, Fukuoka, Japan,<br />

3 CREST, JST, Tokyo, Japan<br />

C-tail anchored (TA) proteins are a distinct class <strong>of</strong> membrane proteins that harbor a single<br />

transmembrane domain at <strong>the</strong> extreme C-terminal region and expose <strong>the</strong>ir N-terminal functional<br />

domains to <strong>the</strong> cytosol. Although TA proteins are found in all <strong>of</strong> subcellular membranes facing<br />

<strong>the</strong> cytosol and play pivotal roles in various biological processes, <strong>the</strong> pathways by which <strong>the</strong>y<br />

are targeted to and inserted into specific organellar membranes, including peroxisomal<br />

membranes, are not fully defined. We herein show that knockdown <strong>of</strong> Pex19p, a predominantly<br />

cytosolic protein that functions as a chaperon and/or soluble receptor for newly syn<strong>the</strong>sized<br />

peroxisomal membrane proteins (PMPs), eliminates <strong>the</strong> import <strong>of</strong> peroxisomal TA proteins (P-<br />

TAs) in vivo. Pex19p forms complexes with P-TAs in <strong>the</strong> cytosol. These results indicate that<br />

Pex19p is involved in <strong>the</strong> import <strong>of</strong> P-TAs as well as non-TA-type PMPs. We also show that P-<br />

TAs, which form complexes with Pex19p, are specifically targeted to peroxisomes, onto Pex3p,<br />

even under ATP-depleted condition in an import assay using semi-intact mammalian cells. The<br />

targeting <strong>of</strong> P-TAs to peroxisomes is driven by Pex19p-Pex3p interaction. Collectively, <strong>the</strong>se<br />

results strongly suggest that P-TAs are, like most PMPs, targeted to peroxisomes in a Pex19p-<br />

and Pex3p-dependent manner. Thus, P-TAs share <strong>the</strong> import pathway with non-TA-type PMPs,<br />

in contrast to <strong>the</strong> TA proteins directed to <strong>the</strong> ER and mitochondrial outer membrane that do not<br />

share <strong>the</strong> import pathways with non-TA proteins.<br />

1998<br />

Novel Function <strong>of</strong> Peroxisome Targeting Signal Type-1 Receptor Pex5p in Pex14p<br />

Stability: Study Using a Newly Isolated Peroxisome-Deficient CHO Cell Mutant, ZPEG101.<br />

R. Natsuyama 1 , K. Okumoto 1,2 , Y. Fujiki 1,3 ; 1 Grad. Sch. <strong>of</strong> Sys. Life Sci. <strong>of</strong> Kyushu University.,<br />

Fukuoka, Japan, 2 Dept. <strong>of</strong> Biol., Grad. Sch. <strong>of</strong> Sci., <strong>of</strong> Kyushu University., 3 JST, CREST<br />

Pex5p is one <strong>of</strong> <strong>the</strong> peroxins required for peroxisome biogenesis and plays an important role in<br />

peroxisomal matrix protein import. Majority <strong>of</strong> peroxisomal matrix proteins possess peroxisome<br />

targeting signal type-1 (PTS1) at <strong>the</strong> C-terminus. Pex5p recognizes PTS1 in cytosol via its Cterminal<br />

region consisting <strong>of</strong> seven tetratricopeptide repeats and also interacts with peroxisome<br />

membrane peroxins Pex14p and Pex13p via its N-terminal region. Toge<strong>the</strong>r with extensive<br />

studies in yeasts and mammals, Pex5p is now proposed as a shuttling receptor between <strong>the</strong><br />

cytosol and peroxisomes. Here, we found a novel function <strong>of</strong> Pex5p in stabilizing Pex14p by<br />

making use <strong>of</strong> a newly isolated ZPEG101, a PEX5-deficient CHO cell mutant, showing typical<br />

mutant phenotypes with defects in import <strong>of</strong> both PTS1 and PTS2 proteins. No Pex5p was<br />

detected in ZPEG101, indicating ZPEG101 is <strong>the</strong> first pex5 CHO cell mutant completely lacking<br />

Pex5p. Interestingly, <strong>the</strong> expressed level <strong>of</strong> Pex14p was significantly decreased in ZPEG101, as<br />

compared to that in wild-type CHO-K1 and o<strong>the</strong>r previously isolated pex5 mutants. More<br />

detailed analyses <strong>of</strong> ZPEG101 are in progress.


1999<br />

P7BP1 E3 ubiquitin ligase controls <strong>the</strong> quality <strong>of</strong> PTS2 receptor, Pex7p.<br />

Y. Miyauchi 1 , Y. Fujiki 1 ; 1 Kyushu University, Fukuoka, Japan<br />

<strong>SUNDAY</strong><br />

Peroxisomes are ubiquitous single membrane-bound organelles that contain about 100 different<br />

enzymes catalyzing various metabolic pathways such as fatty-acid ƒÀ-oxidation and<br />

e<strong>the</strong>rglycerolipid syn<strong>the</strong>sis. Peroxisomes are formed by growth and division <strong>of</strong> preexisting<br />

peroxisomes after posttranslational import <strong>of</strong> newly syn<strong>the</strong>sized peroxisomal matrix and<br />

membrane proteins. Two distinct signals, peroxisomal targeting signal type 1 (PTS1) and PTS2,<br />

direct proteins to <strong>the</strong> peroxisomal matrix. PEX5 and PEX7 encode <strong>the</strong> cytosolic receptors for<br />

PTS1 and PTS2, respectively. Mono-Ubiquitination <strong>of</strong> Pex5p is required for its export from<br />

peroxisomal membrane to <strong>the</strong> cytosol in yeast to mammals. However, regulation <strong>of</strong> <strong>the</strong> function<br />

and transport <strong>of</strong> Pex7p remains undefined.<br />

We first investigated whe<strong>the</strong>r or not Pex7p is ubiquitinated and degraded by proteasomes in<br />

cells. Addition <strong>of</strong> a proteasome inhibitor, MG132 to cell culture delayed <strong>the</strong> turnover rate <strong>of</strong><br />

Pex7p. We also attempted to isolate any potential Pex7p-binding proteins potentially involved in<br />

<strong>the</strong> degradation pathway <strong>of</strong> Pex7p. From a cell line stably expressing FLAG-Pex7p, we isolated<br />

several Pex7p-binding proteins, termed P7BPs: Pex7p binding proteins. Suppression <strong>of</strong> P7BP1<br />

by RNAi resulted in a delay <strong>of</strong> Pex7p turnover rate, as observed in <strong>the</strong> MG132-treated cells,<br />

hence implying that P7BP1 plays a role in Pex7p ubiquitination. Fur<strong>the</strong>rmore, we also show that<br />

<strong>the</strong> degradation <strong>of</strong> dysfunctional Pex7p mediated by P7BP1 is crucially required for <strong>the</strong><br />

maintenance <strong>of</strong> normal PTS2 import. Our findings may define a mechanism underlying Pex7p<br />

degradation and its importance in regulating PTS2 import.<br />

2000<br />

A System to Quantify <strong>the</strong> Differential Import <strong>of</strong> Peroxisomal Matrix Protein by Measuring<br />

Fluorescence Intensity.<br />

M. Noguchi 1 , Y. Fujiki 1,2 ; 1 Graduate School <strong>of</strong> Systems Life Sciences, Kyushu University,<br />

Fukuoka, Japan, 2 CREST, JST<br />

Fourteen distinct peroxins are identified as essential factors for peroxisome biogenesis in<br />

mammals, <strong>of</strong> which ten are involved in peroxisomal import <strong>of</strong> matrix proteins. Peroxisomal<br />

matrix protein import is regulated by various cellular factors. However, mechanisms underlying<br />

such regulations are poorly understood, partly, if not all, due to <strong>the</strong> lack <strong>of</strong> quantitative detection<br />

method with high resolution. Here, we developed a monitoring system that utilizes <strong>the</strong><br />

differences in <strong>the</strong> stability <strong>of</strong> fluorescent reporters localized between peroxisomes and <strong>the</strong><br />

cytosol. An FKBP12 variant, termed destruction domain (DD), is rapidly and constitutively<br />

degraded by proteasomes when expressed in mammalian cells. Degradation <strong>of</strong> DD is reversibly<br />

protected by addition <strong>of</strong> a specific syn<strong>the</strong>tic ligand. In <strong>the</strong> absence <strong>of</strong> <strong>the</strong> ligand, a reporter DD-<br />

EGFP-PTS1, EGFP fused with <strong>the</strong> DD and peroxisomal targeting signal 1, is largely degraded<br />

in <strong>the</strong> cytosol prior to entering peroxisomes in wild-type cells. In contrast, in <strong>the</strong> presence <strong>of</strong> <strong>the</strong><br />

ligand <strong>the</strong> reporter becomes stable and translocates into peroxisomes. Upon withdrawal <strong>of</strong> <strong>the</strong><br />

ligand, <strong>the</strong> reporter in peroxisomes remains intact, whilst that in <strong>the</strong> cytosol is rapidly degraded.<br />

Thus, peroxisomal protein import can be readily quantified by simply measuring <strong>the</strong><br />

fluorescence intensity <strong>of</strong> whole cells.


<strong>SUNDAY</strong><br />

2001<br />

<strong>Molecular</strong> architecture <strong>of</strong> OPA1, <strong>the</strong> dynamin-related GTPase involved in mitochondrial<br />

fusion.<br />

A. M. Fontainhas 1 , J. A. Heymann 1 , S. Fang 1 , J. E. Hinshaw 1 ; 1 LCBB, National Institute <strong>of</strong><br />

Diabetes and Digestive and Kidney Diseases, NIH, Be<strong>the</strong>sda, MD<br />

Mitochondria, <strong>the</strong> powerhouse <strong>of</strong> <strong>the</strong> cell, continually undergo fission and fusion. OPA1 (Optic<br />

atrophy 1) - a dynamin-related GTPase is essential for <strong>the</strong> fusion <strong>of</strong> <strong>the</strong> inner mitochondrial<br />

membrane and is involved in cristae maintenance. Once OPA1 is transported to <strong>the</strong><br />

mitochondria, OPA1 is proteolytically processed into a long and a short form. The long form <strong>of</strong><br />

OPA1 is anchored to <strong>the</strong> inner membrane <strong>of</strong> <strong>the</strong> mitochondria through a N-terminal<br />

transmembrane domain, while <strong>the</strong> short form <strong>of</strong> OPA1 (OPA1s) is found in <strong>the</strong> inner<br />

mitochondrial space. Both forms are required for mitochondrial membrane fusion in vivo. Over<br />

200 mutations <strong>of</strong> OPA1 have been identified that lead to vision loss, mainly through retinal<br />

ganglion cells. Ban, et al. (2010) showed that OPA1s preferentially tubulates and clusters large,<br />

negatively charged liposomes. However, <strong>the</strong> short form alone was unable to mediate membrane<br />

fusion. It has been proposed that tubulation may be <strong>the</strong> mechanism by which fusion <strong>of</strong><br />

mitochondrial membranes occur. To elucidate OPA1’s 3D structure and how it interacts with<br />

lipids we used cryo-electron microscopy to visualize and analyze <strong>the</strong> topological arrangement <strong>of</strong><br />

OPA1 on <strong>the</strong>se protein-lipid tubules. We show that OPA1s wraps around <strong>the</strong> perimeter <strong>of</strong> <strong>the</strong>se<br />

tubes in a helical fashion. We observed that tubulation is not GTP dependent. However,<br />

GMPPCP stabilizes <strong>the</strong> chevron like pattern. OPA1s also forms an organized 2D protein lattice,<br />

indicating that specific intermolecular interactions are at play. Tubes, under all conditions, range<br />

in diameter from 50 to 150 nm with a majority <strong>of</strong> <strong>the</strong>m falling in <strong>the</strong> 60-70 nm range.<br />

Understanding how OPA1’s interactions with <strong>the</strong> membrane leads to fusion <strong>of</strong> inner<br />

mitochondrial membrane will give us a better understanding <strong>of</strong> how mutations in <strong>the</strong> protein<br />

affect its function and give rise to vision loss.<br />

Reference:<br />

Ban, T., Heymann, J. A., Song, Z., Hinshaw, J. E., Chan, D. C.: OPA1 disease alleles causing<br />

dominant optic atrophy have defects in cardiolipin-stimulated GTP hydrolysis and membrane<br />

tubulation. Hum. Mol. Genet. 2010, 19: 2113-22.<br />

2002<br />

Probing <strong>the</strong> dynamics <strong>of</strong> single lipids in supported lipid bilayers and living cells by<br />

fluorescence correlation spectroscopy and confocal microscopy.<br />

C-F. Hsieh 1 , L-L. Yang 1 , Y-L. Lin 2 , C-F. Chou 1 ; 1 Institute <strong>of</strong> Physics, Academia Sinica, Taipei,<br />

Taiwan, 2 National Taiwan University, Institute <strong>of</strong> Chemistry, Taiwan<br />

Lipid raft served as a platform for recruiting signaling components <strong>of</strong> effective signal<br />

transduction involving various cellular activities, such as cell proliferation, differentiation,<br />

migration, and apoptosis in living cells. However, <strong>the</strong> single-lipid dynamics <strong>of</strong> lipid raft or nonlipid<br />

raft domain in living cells are not well elucidated.<br />

We apply fluorescence correlation spectroscopy (FCS) to probe <strong>the</strong> mobility <strong>of</strong> single lipids. We<br />

observed <strong>the</strong> behaviors <strong>of</strong> non-raft lipids and raft lipids in supported lipid bilayers and found that<br />

<strong>the</strong> diffusion constant <strong>of</strong> raft lipids (~2.4 × 10 -8 cm 2 /s) is much smaller than non-raft lipids (~8.1 ×<br />

10 -8 cm 2 /s) and spans a wide range. These results suggest that <strong>the</strong> raft lipids are in a trend to<br />

aggregate in supported bilayers while non-raft lipids are not.<br />

Confocal microscopy was used to monitor <strong>the</strong> dynamics <strong>of</strong> raft lipids induced by ligand-receptor<br />

binding in living cells. The RGD-coated quantum dots (Qdots) were recognized and bound to


<strong>SUNDAY</strong><br />

integrin to trigger <strong>the</strong> aggregation <strong>of</strong> lipid raft. The duration <strong>of</strong> lipid-raft aggregation was<br />

extended from 10 minutes (induced by ligand-coated micro-bead) to 2 hours (induced by ligandcoated<br />

Qdot), and finally <strong>the</strong> aggregate reached a diameter around 5 micrometers. In our<br />

studies, RGD-coated Qdot bound to a small number <strong>of</strong> integrin proteins may provide an<br />

effective means to observe <strong>the</strong> early events <strong>of</strong> lipid raft aggregation in a living cell.<br />

2003<br />

<strong>Molecular</strong> Mechanism <strong>of</strong> F-BAR Protein Pacsin2 in Caveolae Biogenesis.<br />

Y. Senju 1 , S. Suetsugu 1 ; 1 Institute <strong>of</strong> <strong>Molecular</strong> and Cellular Biosciences, The University <strong>of</strong><br />

Tokyo, Tokyo, Japan<br />

Protein kinase C (PKC) and casein kinase substrate in neurons 2 (Pacsin2) belongs to <strong>the</strong><br />

EFC/F-BAR domain protein family and deforms membrane. The N-terminal F-BAR domain <strong>of</strong><br />

pacsin2 forms a crescent-shaped dimer with a positively charged surface. The C-terminal SH3<br />

domain associates with dynamin2, a GTPase implicated in vesicle endocytosis. Therefore,<br />

pacsin2 is involved in membrane invaginations including caveolae (Senju et al., 2011). Previous<br />

study has demonstrated that <strong>the</strong> cells overexpressing pacsin2 showed membrane invaginations.<br />

However, <strong>the</strong> number <strong>of</strong> membrane invagination was larger in <strong>the</strong> cells overexpressing pacsin2<br />

F-BAR domain than in <strong>the</strong> cells overexpressing pacsin2 full length, which was interpreted as<br />

autoinhibition <strong>of</strong> pacsin2 resulted from <strong>the</strong> intramolecular interaction between F-BAR domain<br />

and SH3 domain. Because pacsin2 has been identified as a PKC substrate, we investigated <strong>the</strong><br />

autoinhibition mechanism based on <strong>the</strong> phosphorylation <strong>of</strong> pacsin2 by PKC. First, we performed<br />

an in vitro kinase assay, and confirmed that PKC actually phosphorylated pacsin2. Then, we<br />

generated two different pacsin2 mutants by substitution with ei<strong>the</strong>r alanine or glutamic acid, and<br />

identified <strong>the</strong> phosphorylation site in pacsin2 by in vitro kinase assay. Next, we generated a<br />

phospho-specific antibody, and confirmed that endogenous pacsin2 was phosphorylated after<br />

<strong>the</strong> PKC activator PMA treatment using immun<strong>of</strong>luorescence microscopy and western blotting<br />

analysis. The number <strong>of</strong> membrane invaginations induced by phospho-mimic mutant <strong>of</strong> pacsin2<br />

was lesser than that induced by wild-type pacsin2. After <strong>the</strong> PMA treatment, <strong>the</strong> number <strong>of</strong><br />

membrane invagination was decreased in <strong>the</strong> cells expressing wild-type pacsin2, but <strong>the</strong>re was<br />

no significant effect in <strong>the</strong> cells expressing ei<strong>the</strong>r phospho-mimic, phospho-deficient mutant, or<br />

F-BAR domain <strong>of</strong> pacsin2. Therefore, <strong>the</strong>se data suggest that phosphorylated pacsin2 prefers<br />

an open protein conformation that permits interaction with its partners such as dynamin2. The<br />

interaction <strong>of</strong> pacsin2 with dynamin2 probably caused dynmin2-induced scission <strong>of</strong> membrane<br />

invaginations, which in turn decreased <strong>the</strong> number <strong>of</strong> pacsin2-induced membrane invaginations.<br />

Fur<strong>the</strong>r experimentation will be required to test this hypo<strong>the</strong>sis and reveal <strong>the</strong> physiological role<br />

<strong>of</strong> pacsin2 in caveolae biogenesis.<br />

2004<br />

Identification and characterization <strong>of</strong> atypical FYVE Domains for specific interaction with<br />

phospholipids.<br />

J-E. Gil 1 , I-S. Kim 2 , B. Ku 1 , W. Park 1 , B-H. Oh 1 , S. Ryu 2 , W. Heo 1 ; 1 Korea Advanced Institute <strong>of</strong><br />

Science and Technology, Daejeon, Korea, 2 Pohang University <strong>of</strong> Science and Technology,<br />

Korea<br />

Interactions <strong>of</strong> lipid binding domains with intracellular lipids play a significant role in regulation <strong>of</strong><br />

numerous signaling and cellular mechanisms. To characterize interactions <strong>of</strong> lipid binding<br />

domains with phospholipids, five classes <strong>of</strong> lipid binding domains were screened by fluorescent<br />

imaging and classified based on <strong>the</strong>ir subcellular localizations. Six <strong>of</strong> over one hundred kinds <strong>of</strong><br />

lipid binding domains were localized in <strong>the</strong> plasma membrane where a number <strong>of</strong> signaling<br />

cascades are generally initiated. Interestingly, we found FYVE domain <strong>of</strong> ZFYVE27 which is


<strong>SUNDAY</strong><br />

localized in <strong>the</strong> plasma membrane unlike typical FYVE domains which are localized in <strong>the</strong><br />

endosomes. Conventionally, it has been known that lipid binding pocket <strong>of</strong> FYVE domains has<br />

WXXD, R+HHC+XCG and RVC regions through which FYVE domains bind to PI(3)P. However,<br />

from analysis <strong>of</strong> amino acid sequences and model <strong>of</strong> 3D protein structure, we found that<br />

ZFYVE27-FYVE domain did not contain WXXD, R+HHC+XCG and RVC regions, reflecting it<br />

may have unconventional property in lipid interaction <strong>of</strong> FYVE domain. It was demonstrated that<br />

ZFYVE27-FYVE domain specifically interacts with plasma membrane phospholipids by using<br />

inducible strategies in which phospholipids are depleted by translocating phosphatase to <strong>the</strong><br />

plasma membrane and specific inhibitor treatment. We also observed that growth factor-induced<br />

ZFYVE27 translocation was not induced when this interaction was ruined.<br />

2005<br />

Plin 2 and Plin 3 exhibit characteristics <strong>of</strong> functionally diverse C-terminal domains.<br />

B. Chong 1,2 , L. Cicchini 1,2 , P. Reigan 3 , J. McManaman 1 ; 1 OB/GYN, University <strong>of</strong> Colorado<br />

Denver, Anschutz Medical Campus, Aurora, CO, 2 Graduate Program in <strong>Molecular</strong> <strong>Biology</strong>,<br />

University <strong>of</strong> Colorado Denver, Anschutz Medical Campus, Aurora, CO, 3 Pharmaceutical<br />

Science, University <strong>of</strong> Colorado Denver, Anschutz Medical Campus, Aurora, CO<br />

Perilipin family members, adipophilin (ADPH/Plin2) and TIP47 (Plin3) have <strong>the</strong> highest degree<br />

<strong>of</strong> sequence similarity within <strong>the</strong> perilipin family, with nearly 60% similarity, and are thought to<br />

be redundant lipid droplet associated proteins. Our laboratory previously demonstrated that <strong>the</strong><br />

N-terminal half <strong>of</strong> ADPH is required for association with <strong>the</strong> CLD monolayer and this interaction<br />

protects <strong>the</strong> CLD core from lipolysis. More recent studies indicate that <strong>the</strong> C-terminal half <strong>of</strong><br />

ADPH is a lipid membrane association domain that is required for milk lipid secretion. TIP47<br />

was originally identified as a vessicle trafficking protein but evidence supporting this is<br />

controversial. Additionally, less is known about <strong>the</strong> unique functional domains within TIP47. In<br />

mammary epi<strong>the</strong>lial cells, <strong>the</strong> two family members have distinct cellular localizations and<br />

differential expression patterns over <strong>the</strong> course <strong>of</strong> pregnancy and lactation, which suggest that<br />

<strong>the</strong> two proteins are not functionally redundant. We took a structural and biochemical approach<br />

to understanding how <strong>the</strong>se two proteins are functionally distinct. ADPH and TIP47 share 43%<br />

amino acid similarity in <strong>the</strong>ir C-terminal domain and are hypo<strong>the</strong>sized to be structural homologs.<br />

The C-terminus <strong>of</strong> TIP47 has been crystallized and is known to form a four-helix bundle motif<br />

with a centrally located hydrophobic cleft motif. We constructed a homology model for <strong>the</strong> Cterminus<br />

<strong>of</strong> ADPH, which shows that ADPH forms a putative four-helix bundle and hydrophobic<br />

cleft, similar to that <strong>of</strong> TIP47. Closer examination <strong>of</strong> <strong>the</strong>se structures shows that <strong>the</strong>se two<br />

proteins have noticeable differences in overall shape, with TIP47 being more compact and<br />

ADPH having a slightly more extended in structure. We expressed and purified recombinant<br />

ADPH and TIP47 C-termini from E. coli and analytical size exclusion chromatography (SEC)<br />

shows that <strong>the</strong> two proteins have different retention times, suggestive <strong>of</strong> differences in overall<br />

shape. Additionally, native PAGE comparison shows differences in migration between <strong>the</strong> two<br />

proteins and unique sensitivies to urea denaturation. Analytical ultracentrifugation sedimentation<br />

velocity and SEC suggest that ADPH forms higher order structures and TIP47 does not. We<br />

provide <strong>the</strong> first side-by-side comparison <strong>of</strong> ADPH and TIP47 and show that <strong>the</strong>y are not<br />

structurally identical. Fur<strong>the</strong>r work is needed to determine how <strong>the</strong>se structures account for<br />

functional diversity.


<strong>SUNDAY</strong><br />

2006<br />

Perilipin, Moesin and Microtubule Dependence <strong>of</strong> Lipid Droplet Clustering and<br />

Dispersion.<br />

D. Orlicky 1 , A. Stefanski 2 , B. Chong 2 , J. Monks 2 , C. Monks 3 , J. L. McManaman 2 ; 1 Pathology, UC<br />

Denver, Anschutz Medical Campus, Aurora, CO, 2 Ob/Gyn, UC Denver, Anschutz Medical<br />

Campus, Aurora, CO, 3 Intelligent Imaging Innovations, Inc., Denver, CO<br />

Cytoplasmic lipid droplets (CLD) function in storage, trafficking and secretion <strong>of</strong> lipids as well as<br />

a depot for fatty acid signaling precursors. Perilipin 1, <strong>the</strong> primary CLD binding protein in<br />

adipocytes, is thought to contribute to CLD trafficking by directing fragmentation <strong>of</strong> CLD into<br />

smaller droplets, making <strong>the</strong> lipids more enzymatically available. To study <strong>the</strong> role <strong>of</strong> perilipins in<br />

mediating intracellular CLD trafficking we used stable ectopic expression <strong>of</strong> perilipin 1, or<br />

truncated perilipin 1, in HEK293 cells. Expression <strong>of</strong> ei<strong>the</strong>r full-length or C-term perilipin 1<br />

(amino acids 198-517) initiated formation <strong>of</strong> single, dense CLD clusters that localize near <strong>the</strong><br />

microtubule organizing center (MTOC). Using live-cell imaging, we showed <strong>the</strong>se clustered CLD<br />

became fragmented and moved to <strong>the</strong> periphery <strong>of</strong> <strong>the</strong> cell in response to isoproterenol or<br />

forskolin activation <strong>of</strong> adenylate cyclase- which has previously been shown to regulate PKAmediated<br />

phosphorylation <strong>of</strong> perlipin 1. We fur<strong>the</strong>r show that nocodozole disruption <strong>of</strong><br />

microtubules could inhibit this process, while staurosporine blockage <strong>of</strong> PKA activity disrupted<br />

dispersion, but not <strong>the</strong> initial fragmentation <strong>of</strong> <strong>the</strong> CLD cluster. Finally, siRNA knockdown <strong>of</strong> <strong>the</strong><br />

cortical actin-binding protein, moesin, prevented dispersed CLD from reforming dense clusters<br />

when isoproterenol was removed. These data suggest a model in which CLD clustering and<br />

dispersion are mediated by both microtubules and actin, and trafficking is controlled by PKAphosphorylation<br />

<strong>of</strong> perilipin 1.<br />

2007<br />

Netrin-1-induced axon branching <strong>of</strong> cerebral cortical neurons requires DCC and lipid raft<br />

integrity.<br />

H. Matsumoto 1 , M. Nagashima 1 ; 1 Department <strong>of</strong> Anatomy, Faculty <strong>of</strong> Medicine, Saitama Medical<br />

University, Saitama, Japan<br />

A multifunctional axon guidance cue netrin-1 is known to induce axon branching in developing<br />

cerebral cortical neurons <strong>of</strong> mammals. In this study we investigated whe<strong>the</strong>r netrin-1-induced<br />

cortical axon branching involves DCC (deleted in colorectal cancer; one <strong>of</strong> known receptors for<br />

netrin-1) and lipid rafts (cholesterol- and sphingolipid-enriched membrane microdomains),<br />

employing dissociated cortical neurons prepared from neonatal Syrian golden hamsters.<br />

Stimulation <strong>of</strong> neurons by netrin-1 caused an increase in <strong>the</strong> number <strong>of</strong> branch points along <strong>the</strong><br />

primary axons, which is in consistent with previous reports. We <strong>the</strong>n observed that this increase<br />

was attenuated by application <strong>of</strong> a neutralizing antibody against DCC to <strong>the</strong> culture. Attenuation<br />

<strong>of</strong> netrin-1-induced axon branching was also caused by disruption <strong>of</strong> lipid rafts, while<br />

association <strong>of</strong> DCC to lipid rafts was revealed by Western blot analysis. These data indicate a<br />

significant contribution <strong>of</strong> DCC to, and requirement <strong>of</strong> lipid raft integrity for, <strong>the</strong> promotion <strong>of</strong><br />

cortical axon branching caused by netrin-1. Fur<strong>the</strong>rmore, <strong>the</strong> data suggest <strong>the</strong> possibility that<br />

association <strong>of</strong> DCC with lipid rafts is involved in netrin-1-induced axon branching. (Supported by<br />

Grant-in-Aid for Scientific Research (C) No. 23590225 from Japan Society for <strong>the</strong> Promotion <strong>of</strong><br />

Science.)


<strong>SUNDAY</strong><br />

2008<br />

HGSNAT mono-multimer formation revealed by live-cell two-photon FRET-FLIM<br />

microscopy.<br />

F. Majer 1 , J. Sikora 1 , J. Lukas 1 , D. Musalkova 1 , O. Chernyavskiy 2 , M. Hrebicek 1 ; 1 Inst Inherited<br />

Metabolic Disorders, Charles University, Prague 2, Czech Republic, 2 Department <strong>of</strong><br />

Bioma<strong>the</strong>matics, Institute <strong>of</strong> Physiology, Academy <strong>of</strong> Sciences <strong>of</strong> <strong>the</strong> Czech Republic v.v.i.,<br />

Prague 2, Czech Republic<br />

Human HGSNAT (Acetyl-Coenzyme A: Alpha-Glucosaminide N-Acetyltransferase) is a<br />

membrane enzyme localized in endosomal/lysosomal microdomains catalyzing transmembrane<br />

acetylation <strong>of</strong> heparan sulfate. This syn<strong>the</strong>tic modification is mandatory prerequisite for<br />

subsequent intralysosomal degradation <strong>of</strong> heparan sulfate. Mutations in HGSNAT gene cause<br />

rare but clinically serious lysosomal storage disorder - mucopolysaccharidosis IIIC (Sanfilippo C<br />

syndrome).<br />

Our aim was to find intracellular membrane protein partners <strong>of</strong> HGSNAT.<br />

We have prepared plasmids for transient expression <strong>of</strong> HGSNAT with two alternative starting<br />

ATG codons as well as plasmids coding potentially interacting protein partners <strong>of</strong> HGSNAT. To<br />

detect in vivo protein-protein interactions, we used live two-photon excitation FRET-FLIM time<br />

correlated single photon counting technique with EGFP/mCherry donor-acceptor pairs.<br />

Additionally, we also prepared fraction <strong>of</strong> enriched lysosomal membranes using density gradient<br />

ultracentrifugation and by 2D electrophoresis (Blue Native / SDS PAGE) we identified region <strong>of</strong><br />

HGSNAT protein electrophoretic mobility and controlled protein integrity.<br />

Basic co-localizing experiments implied LAMP1, LAMP2 and Rab7 <strong>the</strong>ir intracellular cooccurrence<br />

with HGSNAT by resolution limited light microscopy. Never<strong>the</strong>less two-photon<br />

FRET-FLIM methodology revealed only HGSNAT-EGFP and HGSNAT-mCherry as potential<br />

interacting partners suggesting mono-multimer formation within <strong>the</strong> endosomal/lysosomal<br />

membrane.<br />

This study was funded by <strong>the</strong> research project MSM0021620806 from <strong>the</strong> Ministry <strong>of</strong> Education,<br />

Youth and Sports <strong>of</strong> <strong>the</strong> Czech Republic and by <strong>the</strong> Internal Grant Agency <strong>of</strong> <strong>the</strong> Ministry <strong>of</strong><br />

Health <strong>of</strong> <strong>the</strong> Czech Republic, Project IGA MZ NS / 10342 - 3 / 2009.<br />

2009<br />

Mechanism underlying <strong>the</strong> expression <strong>of</strong> Na-H Exchanger is<strong>of</strong>orms 1 and 3 in SHR rat<br />

colon and ileum in hypertension.<br />

A. A. Ghefreh 1 , I. Khan 2 , A. A. Ghefreh 1 ; 1 Biochemistry, Kuwait University, Kuwait, 2 Kuwait<br />

University, Kuwait<br />

Ileum and colon play an important role in regulating water and electrolyte homeostasis<br />

mechanisms which are deregulated in hypertension. Role <strong>of</strong> colonic and ileal Na-H exchanger<br />

(NHE) is<strong>of</strong>orms-1 and -3 which regulate water and electrolyte absorption from GI-tract and<br />

kidneys is poorly investigated in hypertension. In this study we investigated a putative role <strong>of</strong><br />

colonic and ileal NHE-1 and -3 is<strong>of</strong>orms in hypertension using a 12-week old spontaneously<br />

hypertensive rat (SHR) model. A group <strong>of</strong> SHR animals was treated with captopril (300 mg/L) ad<br />

libitum starting week 4 post birth and followed till week 12. Age matched genetic counterparts,<br />

WKY animals served as controls in this study. Fur<strong>the</strong>r to elucidate <strong>the</strong> underlying mechanism,<br />

expression <strong>of</strong> NHERF-1 and cGKII which regulate activity <strong>of</strong> NHE was also examined. SHR<br />

animals showed a significant increase in kidney and heart hypertrophy and proteinuria, while<br />

decrease in urine output as compared to WKY controls. These changes were reversed by<br />

captopril treatment. There was significant elevation in myeloperoxidase activity in SHR ileum<br />

and colon as compared to WKY controls. These changes were reversed by captopril treatment.<br />

Expression <strong>of</strong> NHE-1, NHE-3 and NHERF-1 proteins was significantly decreased, while that <strong>of</strong>


<strong>SUNDAY</strong><br />

cGKII was increased in SHR ileum and colon as compared to WKY controls. Captopril treatment<br />

<strong>of</strong> SHR caused a partial reversal <strong>of</strong> <strong>the</strong>se changes, and a significant reversal <strong>of</strong> NHERF-1.<br />

These changes do not seem to be due to any difference in <strong>the</strong> microsomal protein yield. We<br />

demonstrate evidence <strong>of</strong> mild inflammation in SHR colon and ileum due to hypertension. Our<br />

findings showed decreased expression <strong>of</strong> NHE-1, -3 and NHERF-1, which toge<strong>the</strong>r would<br />

suppress uptake <strong>of</strong> water and NaCl from <strong>the</strong> GI-tract and hence may be a counteracting<br />

response in <strong>the</strong> development <strong>of</strong> hypertension in this model.<br />

Acknowledgments: Kuwait University Research administration (grant YM 06/2010) and College<br />

<strong>of</strong> Graduate Studies for financial support and Shared Core Facility, Health Science Center,<br />

Kuwait University for allowing to use <strong>the</strong> instrumentation.<br />

2010<br />

Gustatory receptors not G protein coupled receptors are required for <strong>the</strong> response to Lcanavanine.<br />

Y. Lee 1 , M. Kang 2 , J. Shim 3 , C. Cheong 2 , S. Moon 2 , C. Montell 1 ; 1 Department <strong>of</strong> Biological<br />

Chemistry and Neuroscience, Center for Sensory <strong>Biology</strong>, The Johns Hopkins University School<br />

<strong>of</strong> Medicine, 2 Department <strong>of</strong> Oral <strong>Biology</strong>, Brain Korea21 project, Yonsei University College <strong>of</strong><br />

Dentistry, Seoul, Korea, 3 Department <strong>of</strong> Phamacology, Yonsei University College <strong>of</strong> Medicine,<br />

Seoul, Korea<br />

Insects respond to <strong>the</strong>ir surroundings through sensing chemical cues and <strong>the</strong>n decide <strong>the</strong>ir<br />

behaviors. Gustatory sense is one <strong>of</strong> <strong>the</strong> most important chemical sensory systems. There are<br />

68 gustatory receptors (GRs) encoded by 60 gustatory receptor genes. Several gustatory<br />

receptors are known to be required for sensing <strong>of</strong> sugars or bitter chemicals, such as caffeine.<br />

However, <strong>the</strong> functions <strong>of</strong> most GRs are unknown. Here we show that Gr8a and Gr66a are<br />

required for sensing insecticide, L-canavainine, which is produced by plants. Gr8a is expressed<br />

in a subset <strong>of</strong> Gr66a expressing gustatory receptor neurons (GRNs). The action potentials to Lcanavanine<br />

are detected only in <strong>the</strong> Gr8a expressing neuron. The consistence <strong>of</strong> Gr8a<br />

expression pattern and electrophysiological response to L-canavanine suggests <strong>the</strong> possibility<br />

that Gr8a is <strong>the</strong> specific receptor to L-canavanine, while Gr66a is <strong>the</strong> broad receptor to bitter<br />

chemicals. In contrast, DmXR, one <strong>of</strong> <strong>the</strong> G-protein coupled receptors (GPCRs), has been<br />

reported to be required for detecting L-canavanine. However, DmXR missing flies show <strong>the</strong><br />

avoidance to L-canavanine and action potentials to L-canavanine as wild type flies do in our<br />

results. These results imply that Gr8a and Gr66a are indispensible for detection <strong>of</strong> Lcanavanine,<br />

but not DmXR.<br />

2011<br />

NHERF- 1 Modulates Intestinal NaPi transporter NaPi-2b expression in Apical Microvilli.<br />

H. Giral 1 , Y. Caldas 1 , L. Lanzano 2 , E. Gratton 2 , M. Levi 1 ; 1 University <strong>of</strong> Colorado Denver, Aurora,<br />

CO, 2 University <strong>of</strong> California Irvine, CA<br />

The regulation <strong>of</strong> phosphate (Pi) homeostasis is maintained by <strong>the</strong> coordinated function <strong>of</strong> <strong>the</strong> renal and<br />

intestinal phosphate transporters. Several PDZ (PSD-95/discs large/ZO-1 homologous) domain<br />

proteins, including NHERF-1, PDZK1, ShanK2, and PIST play an important role in <strong>the</strong> regulation <strong>of</strong> <strong>the</strong><br />

renal sodium-phosphate (NaPi) co-transporters (NaPi-2a and NaPi-2c). The main mediator <strong>of</strong> intestinal<br />

sodium dependent transcellular Pi transport, NaPi-2b, also contains a PDZ-binding motif consensus in<br />

<strong>the</strong> C-terminal region. However, interactions <strong>of</strong> <strong>the</strong> transporter NaPi-2b with PDZ proteins have been<br />

not described and <strong>the</strong>ir potential role in regulation <strong>of</strong> <strong>the</strong> intestinal transporter is not known. For this<br />

purpose we performed studies with knock-out (KO) mice models and cell culture to determine a<br />

potential role for NHERF-1 and PDZK1 in <strong>the</strong> regulation <strong>of</strong> NaPi-2b.


<strong>SUNDAY</strong><br />

To study <strong>the</strong> putative interaction between NaPi-2b and PDZ proteins we determined <strong>the</strong> Forster<br />

Resonance Energy Transference (FRET) by using Fluorescence Lifetime Imaging Microscopy (FLIM). OK<br />

cells, an extensively used proximal tubule model, and CaCo-2BBE cells, an enterocyte cell model, were<br />

used to perform this technique. First, expression <strong>of</strong> EGFP-NaPi-2b was confirmed in <strong>the</strong> microvilli <strong>of</strong> both<br />

cell types, and images along a single microvillus were obtained with <strong>the</strong> novel Modulation Tracking (MT)<br />

method. Cells co-expressing EGFP-NaPi-2b and mCherry-NHERF-1 were analyzed by FLIM-FRET<br />

technique revealing significant FRET between NaPi-2b and NHERF-1. Parallel studies between <strong>the</strong> pair<br />

NaPi-2b and PDZK1 proteins resulted in non occurrence <strong>of</strong> FRET. To evaluate <strong>the</strong> functional significance<br />

<strong>of</strong> <strong>the</strong>se results we study NaPi-2b expression and activity in NHERF1 KO and PDZK1 KO mice models,<br />

where we found that adaptation to a low Pi diet <strong>of</strong> NaPi2b was markedly impaired in <strong>the</strong> NHERF-1 KO<br />

mice but not in <strong>the</strong> PDZK1 KO.<br />

Our results <strong>the</strong>refore suggest an important role <strong>of</strong> NHERF1 in modulation <strong>of</strong> NaPi-2b expression or<br />

stability in <strong>the</strong> microvilli <strong>of</strong> <strong>the</strong> mouse intestine.<br />

This research was supported by NIH R01 DK066029 to YC, HG, ML, EG and LL; NIH445<br />

P41R03155 to EG and LL; and <strong>the</strong> R01 DK-080769 to BD.<br />

2012<br />

Cytoskeleton modulates Ca2+ sensor Stim1.<br />

C-M. Li 1 , M. Joensuu 1 , E. Jokitalo 1 ; 1 Electron Microscopy Unit, Institute <strong>of</strong> Biotechnology,<br />

University <strong>of</strong> Helsinki, Helsinki, Finland<br />

Stim1 is a 685 amino acids, single-pass transmembrane protein with N-terminal EF hand<br />

located in <strong>the</strong> endoplasmic reticulum (ER) lumen and C-terminal coiled-coil domains facing cell<br />

cytosol. As a sensor Stim1 plays essential role in Ca2+ signalling, Store-operated Ca2+ entry<br />

(SOCE) channel activation, and influx <strong>of</strong> Ca2+ in response to depletion <strong>of</strong> ER luminal Ca2+.<br />

Depletion <strong>of</strong> intracellular Ca2+ stores results in dissociation <strong>of</strong> Ca2+ from EF-hand, thus<br />

activating store-operated channels, and trigger translocation <strong>of</strong> STIM1 proteins to <strong>the</strong> plasma<br />

membrane [1].<br />

Rosado and his colleagues reported in <strong>the</strong>ir recent study that: cytoskeleton regulates <strong>the</strong><br />

interaction between Stim1 and <strong>the</strong> plasma membrane SOCE channel components such as<br />

Orai1 and TRPCs. In Human Embryonic Kidney (HEK-293) cells, disruption <strong>of</strong> microtubules<br />

enhanced both <strong>the</strong> activation <strong>of</strong> SOCE and <strong>the</strong> association between STIM1 and Orai1 or<br />

TRPC1 induced by thapsigargin (TG). Conversely, stabilization <strong>of</strong> microtubules or actin<br />

filaments attenuated both TG evoked activation <strong>of</strong> SOCE and <strong>the</strong> interaction between STIM1<br />

and <strong>the</strong> channel components. However, disruption <strong>of</strong> <strong>the</strong> actin filament network did not affect<br />

TG-evoked association between STIM1 and Orai1 or TRPC1 but enhanced TG stimulated<br />

SOCE [2]. They concluded that <strong>the</strong> microtubules act as a negative regulator <strong>of</strong> SOCE.<br />

We study <strong>the</strong> location <strong>of</strong> Stim1 in association with disruption/stabilization <strong>of</strong> microtubule/actin at<br />

immun<strong>of</strong>luorescence microscopy and immuno electron microscopy level with HEK-293 cells and<br />

hepatocarcinoma Huh-7 cells. We show that Stim1 proteins translocate and aggregate on <strong>the</strong><br />

plasma membrane upon Calcium depletion by TG treatment.<br />

References:<br />

Zhang SL, Yu Y, Roos J, Kozak JA, Deerinck TJ, Ellisman MH, et al. STIM1 is a Ca2+ sensor<br />

that activates CRAC channels and migrates from <strong>the</strong> Ca2+ store to <strong>the</strong> plasma membrane.<br />

Nature 2005;437:902–5.


<strong>SUNDAY</strong><br />

Galán C, Dionisio N, Smani T, Salido GM, Rosado JA. The cytoskeleton plays a modulatory role<br />

in <strong>the</strong> association between STIM1 and <strong>the</strong> Ca2+ channel subunits Orai1 and TRPC1. Biochem<br />

Pharmacol. 2011;82:400-10.<br />

Nuclear Structure and Function<br />

2013<br />

Proteostatic control <strong>of</strong> nuclear targeting pathways by <strong>the</strong> nuclear pore complex protein,<br />

Nup42.<br />

L-K. LIU 1 , C-C. LIN 1 , D. S. Goldfarb 1 ; 1 <strong>Biology</strong>, University <strong>of</strong> Rochester, Rochester, NY<br />

Multiple transport receptors ferry nuclear localization signal (NLS)-bearing cargos across <strong>the</strong><br />

nuclear pore complex (NPC). The FG-domains <strong>of</strong> nucleoporins in <strong>the</strong> NPC are thought to be<br />

required in specific combinations for efficient transport receptor-mediated transports. A model <strong>of</strong><br />

multiple independent NPC translocation routes exist for different transport receptors was<br />

presented. Based on this model, direct competition for <strong>the</strong> same translocation pathway shared<br />

by multiple transport receptors can be expected. In this study, we have found that Nup42<br />

governs a unique stress response that differentially controls import mediated by Kap95/60 and<br />

Kap104. In vivo kinetic analyses show that Kap95/60 and Kap104 compete for limiting docking<br />

sites at Nup42. Binding to Nup42 is <strong>the</strong> rate-limiting step in import, since <strong>the</strong> import rates <strong>of</strong><br />

Kap95/60 and Kap104 NLS-cargos are faster in nup42 DFG cells. Nup42 collaborates with <strong>the</strong><br />

Hsp70s Ssa1/Ssa2 (but not Ssa3/Ssa4) and <strong>the</strong> co-chaperone Swa2 to differentially control<br />

nuclear targeting by Kap95/60 and Kap104. Specifically, Kap95/60 import is favored over<br />

Kap104 import when Ssa1/Ssa2 levels are high, such as in response to heat shock or nutrient<br />

depletion. In contrast, Kap104 import is favored when <strong>the</strong> free Hsp70 pool is depleted, for<br />

example, by sequestration into Huntingtin polyQ protein aggregates. Kap123-mediated import<br />

rates are unaffected by this regulatory circuit. Here we present <strong>the</strong> first evidence that two<br />

specific transport receptors, Kap95/60 and Kap104, share and compete for similar translocation<br />

route in <strong>the</strong> NPC. We also conclude that Nup42 is a Kap-specific gatekeeper that controls<br />

import rates in response to proteostatic stress.<br />

2014<br />

Evidence <strong>of</strong> asymmetric assembly pathway <strong>of</strong> Nuclear Pore Complex.<br />

P. Colombi 1 , C. Vogel 1 , P. Lusk 1 ; 1 Department <strong>of</strong> Cell <strong>Biology</strong>, Yale School <strong>of</strong> Medicine, New<br />

Haven, CT<br />

In an effort to investigate regulatory mechanisms governing nuclear pore complex (NPC)<br />

assembly, we investigated alterations in <strong>the</strong> subcellular distribution <strong>of</strong> multiple nucleoporins<br />

(Nups) through <strong>the</strong> cell cycle in <strong>the</strong> budding yeast S. cerevisiae. Consistent with <strong>the</strong> idea that<br />

NPC assembly is under <strong>the</strong> control <strong>of</strong> cell cycle regulators, we observe that several specific<br />

Nups accumulate in <strong>the</strong> cytoplasm <strong>of</strong> cells arrested in S-phase but not in G1. Interestingly,<br />

<strong>the</strong>se likely NPC intermediates accumulate asymmetrically between <strong>the</strong> mo<strong>the</strong>r and bud and<br />

appear to be strikingly biased towards <strong>the</strong> bud, <strong>of</strong>ten accumulating at <strong>the</strong> bud cortex. These<br />

intermediates can <strong>of</strong>ten be observed integrating into <strong>the</strong> daughter cell nuclear envelope during<br />

anaphase supporting <strong>the</strong> idea that <strong>the</strong>re is a pathway in which Nups are specifically targeted to<br />

<strong>the</strong> bud to be incorporated in <strong>the</strong> NPC <strong>of</strong> <strong>the</strong> daughter cell. Perturbing this pathway by<br />

conditionally sequestering specific newly syn<strong>the</strong>sized Nups results in a decrease in <strong>the</strong> size <strong>of</strong><br />

daughter nuclei. Most strikingly, <strong>the</strong>se daughter cells are no longer able to progress through <strong>the</strong><br />

cell cycle, whereas <strong>the</strong> mo<strong>the</strong>r cells continue to divide. Toge<strong>the</strong>r <strong>the</strong>se results support <strong>the</strong>


<strong>SUNDAY</strong><br />

existence <strong>of</strong> a pathway critical for continued daughter cell proliferation linked to <strong>the</strong> assembly <strong>of</strong><br />

NPC.<br />

2015<br />

Temporal Control <strong>of</strong> Nuclear Envelope Assembly by Phosphorylation <strong>of</strong> Lamin B<br />

Receptor.<br />

L-C. Tseng 1 , R-H. Chen 1 ; 1 Institute <strong>of</strong> <strong>Molecular</strong> <strong>Biology</strong>, Academia Sinica, Taipei, Taiwan<br />

The nuclear envelope <strong>of</strong> metazoans disassembles during mitosis and reforms in late anaphase<br />

after sister chromatids have well separated. The coordination <strong>of</strong> <strong>the</strong>se mitotic events is<br />

important for genome stability, yet <strong>the</strong> temporal control <strong>of</strong> nuclear envelope reassembly is<br />

unknown. Although <strong>the</strong> steps <strong>of</strong> nuclear formation have been extensively studied in vitro using<br />

<strong>the</strong> reconstitution system from egg extracts, <strong>the</strong> temporal control can only be studied in vivo.<br />

Herein, we use time-lapse microscopy to investigate this process in living HeLa cells. We<br />

demonstrate that Cdk1 activity prevents premature nuclear envelope assembly and that<br />

phosphorylation <strong>of</strong> <strong>the</strong> inner nuclear membrane protein lamin B receptor (LBR) by Cdk1<br />

contributes to <strong>the</strong> temporal control. We fur<strong>the</strong>r identify a region in <strong>the</strong> nucleoplasmic domain <strong>of</strong><br />

LBR that inhibits premature chromatin binding <strong>of</strong> <strong>the</strong> protein. We propose that this inhibitory<br />

effect is partly mediated by Cdk1 phosphorylation. Fur<strong>the</strong>rmore, we show that <strong>the</strong> reduced<br />

chromatin-binding ability <strong>of</strong> LBR toge<strong>the</strong>r with Aurora B activity contribute to nuclear envelope<br />

breakdown. Our studies reveal for <strong>the</strong> first time a mechanism that controls <strong>the</strong> timing <strong>of</strong> nuclear<br />

envelope reassembly through modification <strong>of</strong> an integral nuclear membrane protein.<br />

2016<br />

Amphiregulin, an EGF family protein localized to <strong>the</strong> nuclear envelope, activates<br />

directional cell migration via histone H3K9 methylation.<br />

M. Hieda 1 , Y. Yokoyama 1 , Y. Nishioka 1 , H. Tanaka 1 , S. Higashiyama 2 , N. Matsuura 1 , S.<br />

Matsuura 1 ; 1 Graduate School <strong>of</strong> Medicine, Osaka University, Suita, Japan, 2 Proteo Medicine<br />

Research Center, Ehime University, Toon, Japan<br />

EGF family members are expressed on <strong>the</strong> cell surface in pro-form, comprising an extracellular<br />

EGF-like domain, a transmembrane segment, and a short cytoplasmic tail. The EGF-like<br />

domain is sufficient to activate <strong>the</strong> EGF receptor, whereas <strong>the</strong> function <strong>of</strong> <strong>the</strong> cytoplasmic tail<br />

remains unknown. Previously we showed that in <strong>the</strong> case <strong>of</strong> one EGF family member,<br />

amphiregulin (AREG), shedding stimuli result in release <strong>of</strong> soluble EGFR ligand and migration<br />

<strong>of</strong> <strong>the</strong> pro-form (proAREG) to <strong>the</strong> nuclear envelope (NE) and induction <strong>of</strong> global histone<br />

H3K9methylation.<br />

Here we found that NE-localized proAREG activates cell migration. This activation was<br />

dependent on <strong>the</strong> cytoplasmic tail <strong>of</strong> proARG but independent <strong>of</strong> <strong>the</strong> extracellular growth factor<br />

domain. The activation <strong>of</strong> cell migration did not require EGFR function, but did occur<br />

concomitantly with a slight increase in a specific histone modification, histone H3 lysine9<br />

methylation (H3K9me3). Expression <strong>of</strong> <strong>the</strong> H3K9 methyltransferase SUV39H1 activated cell<br />

migration; inversely, knockdown <strong>of</strong> SUV39H1 or chemical inhibition by chaetocin reduced cell<br />

migration. Chaetocin also suppressed <strong>the</strong> cell migration activated by NE-localized proAREG.<br />

Moreover, impairment <strong>of</strong> H3K9me3 suppressed <strong>the</strong> proper formation <strong>of</strong> cell polarity and led to a<br />

reduction in directional cell movement. Thus, NE-localized proAREG regulates cell migration via<br />

histone modification, indicating that chromatin organization influences both cell polarity and<br />

motility.


<strong>SUNDAY</strong><br />

2017<br />

Samp1 is functionally associated with <strong>the</strong> LINC complex and A-type lamina networks.<br />

S. Gudise 1 , R. Figueroa 2 , V. Larsson 3 , E. Hallberg 3 ; 1 Department <strong>of</strong> Biosciences and Nutrition,<br />

Karolinska Institute, Sweden, 2 Department <strong>of</strong> Biochemistry and Biophysics, Stockholm<br />

University, 3 Department <strong>of</strong> Neurochemistry, Stockholm University, Stockholm, Sweden<br />

The transmembrane inner nuclear membrane (INM) protein Samp1 is required for anchoring<br />

centrosomes near <strong>the</strong> nuclei. Using highresolution<br />

fluorescence microscopy we show that Samp1 is distributed in a distinct and characteristic<br />

pattern in <strong>the</strong> nuclear envelope (NE), where it partially colocalizes with <strong>the</strong> LINC complex<br />

protein Sun1. By studying <strong>the</strong> localization <strong>of</strong> Samp1 deletion mutants and fusion proteins, we<br />

conclude that <strong>the</strong> cysteine-rich N-terminal half <strong>of</strong> Samp1 is nucleoplasmically exposed and is<br />

responsible for targeting to <strong>the</strong> INM. It contains four conserved CxxC motifs with <strong>the</strong> potential to<br />

form zinc fingers. The distribution <strong>of</strong> cysteine-toalanine substitution mutants, designed to<br />

prevent zinc finger formation, showed that NE localization <strong>of</strong> Samp1 depends on intact CxxC<br />

motifs. Overexpression <strong>of</strong> Samp1 zinc finger mutants produced an abnormal dominant<br />

phenotype characterized by disrupted organization<br />

<strong>of</strong> a selective subset NE proteins, including emerin, Sun1, endogenous Samp1 and, in some<br />

cases, lamin A/C, but not lamin B, Sun2<br />

or nucleoporins. Silencing <strong>of</strong> Samp1 expression showed that emerin depends on Samp1 for its<br />

correct localization in <strong>the</strong> NE. Our results demonstrate that Samp1 is functionally associated<br />

with <strong>the</strong> LINC complex protein Sun1 and proteins <strong>of</strong> <strong>the</strong> A-type lamina network.<br />

2018<br />

NE localization <strong>of</strong> <strong>the</strong> LINC complex requires multiple elements <strong>of</strong> SUN and KASH<br />

proteins.<br />

Y. G. Turgay 1 , R. Ungricht 2 , A. Rothballer 2 , A. Kiss 2 , G. Csucs 2 , P. Horvath 2 , U. Kutay 2 ; 1 Institute<br />

<strong>of</strong> Biochemistry, ETH Zurich, Zurich, Switzerland, 2 ETH Zurich, Zurich, Switzerland<br />

Inner nuclear membrane (INM) proteins are co-translationally inserted into <strong>the</strong> endoplasmic<br />

reticulum membrane (ER) and need to cross <strong>the</strong> nuclear pore complex (NPC) to reach <strong>the</strong>ir final<br />

destination. Originally, targeting <strong>of</strong> INM proteins to <strong>the</strong> INM was solely explained by a “diffusionretention”<br />

model. However, recent studies on yeast INM proteins revealed <strong>the</strong> existence <strong>of</strong><br />

classical nuclear localization signals (cNLS) that mediate <strong>the</strong> active transport <strong>of</strong> <strong>the</strong> proteins<br />

across <strong>the</strong> NPC. Whe<strong>the</strong>r active transport mechanisms also play a role in <strong>the</strong> targeting <strong>of</strong> INM<br />

proteins in mammalian cells is still unclear. We have used human SUN2 to study INM targeting<br />

and identified three different elements, which collectively mediate NE targeting. The N-terminal<br />

nucleoplasmic domain <strong>of</strong> SUN2 comprises a cNLS and a Golgi retrieval signal. The cNLS was<br />

shown to constitute a functional binding site for <strong>the</strong> heterodimeric transport receptor importin<br />

α/β. A nearby arginine cluster was found to serve as binding platform for <strong>the</strong> coatomer complex,<br />

which mediates retrieval <strong>of</strong> SUN2 from <strong>the</strong> Golgi to <strong>the</strong> ER and ensures efficient INM targeting.<br />

The conserved SUN domain at <strong>the</strong> C terminus represents <strong>the</strong> third targeting element we have<br />

identified in our study. Toge<strong>the</strong>r, our study shows that multiple elements in SUN2 contribute to<br />

NE targeting and that <strong>the</strong>se elements are not limited to cytoplasmic or transmembrane domains.


<strong>SUNDAY</strong><br />

2019<br />

LINC complexes mediate <strong>the</strong> positioning <strong>of</strong> cone photoreceptor nuclei in mouse retina.<br />

D. Razafsky 1 , S. Zang 1 , D. Hodzic 1 ; 1 Ophthalmology and Visual Sciences, Washington<br />

University School <strong>of</strong> Medicine, St Louis, MO<br />

LINC complexes form through <strong>the</strong> interaction between inner nuclear membrane Sun proteins<br />

(Sun 1 and Sun 2) and outer nuclear membrane Nesprins (Nesprin 1, 2, 3 and 4). The Suns and<br />

Nesprins interact within <strong>the</strong> perinuclear space <strong>the</strong>reby connecting <strong>the</strong> interior <strong>of</strong> <strong>the</strong> nucleus to<br />

<strong>the</strong> cytoplasm. In C.elegans and D. melanogaster, mutation <strong>of</strong> Sun and Nesprin orthologs<br />

prevent nuclear anchorage and/or migration within cells or syncitia. Mouse models deficient for<br />

both Sun1 and Sun2 expression display abnormal migration <strong>of</strong> cortical neurons and <strong>the</strong>reby<br />

phenocopies human lissencephaly phenotypes associated with mutations in Lis1 and Dcx. Our<br />

goal is to identify <strong>the</strong> role <strong>of</strong> <strong>the</strong> LINC complexes in mammalian tissue development and<br />

homeostasis. To this end, retinal development provides an excellent model for two reasons: 1)<br />

different types <strong>of</strong> nucleokinetic events such as interkinetic nuclear migration and postmitotic<br />

neuronal migration can be studied and 2) several waves <strong>of</strong> neuronal migration culminate in <strong>the</strong><br />

development <strong>of</strong> a stratified tissue whereby nuclei <strong>of</strong> various cell types are anchored specific<br />

spatial positions. Here, we characterized <strong>the</strong> expression <strong>of</strong> LINC complex components during<br />

mouse retinal development and present a new versatile transgenic mouse model allowing for<br />

<strong>the</strong> conditional expression <strong>of</strong> a dominant negative EGFP-KASH2 protein. Using this model, we<br />

disrupted LINC complexes within <strong>the</strong> photoreceptor layer <strong>of</strong> mouse retina and report <strong>the</strong> ectopic<br />

positioning <strong>of</strong> cone photoreceptor nuclei. These studies show, for <strong>the</strong> first time, <strong>the</strong> relevance <strong>of</strong><br />

LINC complexes in <strong>the</strong> positioning <strong>of</strong> cell nuclei in a mammalian CNS tissue.<br />

2020<br />

<strong>Molecular</strong> Basis <strong>of</strong> <strong>the</strong> SUN-KASH interaction.<br />

B. A. Sosa 1 , A. Rothballer 2 , U. Kutay 2 , T. U. Schwartz 1 ; 1 <strong>Biology</strong>, MIT, Cambridge, MA, 2 ETH<br />

Zurich, Switzerland<br />

The position <strong>of</strong> <strong>the</strong> nucleus within <strong>the</strong> eukaryotic cell is defined. To achieve specific anchorage<br />

and movement nucleoskeleton and cytoskeleton are mechanically connected across <strong>the</strong> doublelayered<br />

membrane <strong>of</strong> <strong>the</strong> nuclear envelope. These bridges, also called LINC-complexes, are<br />

formed by KASH-peptide containing proteins spanning <strong>the</strong> outer nuclear membrane tightly<br />

bound to SUN-domain proteins spanning <strong>the</strong> inner nuclear membrane. We present <strong>the</strong> crystal<br />

structure <strong>of</strong> <strong>the</strong> SUN domain <strong>of</strong> human Sun2 in complex with <strong>the</strong> KASH peptides <strong>of</strong> Nesprin-1<br />

and -2, respectively. The unexpected structure reveals several key features <strong>of</strong> this novel<br />

interaction that help explain how it can work as <strong>the</strong> mechanical te<strong>the</strong>r it is proposed to be.<br />

2021<br />

Building SUN-KASH Complexes at <strong>the</strong> Nuclear Envelope.<br />

A. Rothballer 1 , B. A. Sosa 2 , T. U. Schwartz 2 , U. Kutay 1 ; 1 Institute <strong>of</strong> Biochemistry, ETH Zurich,<br />

Zurich, Switzerland, 2 Department <strong>of</strong> <strong>Biology</strong>, Massachusetts Institute <strong>of</strong> Technology,<br />

Cambridge, MA<br />

Physical connections between <strong>the</strong> nuclear envelope and <strong>the</strong> cytoskeleton are an essential<br />

feature <strong>of</strong> eukaryotic cells. They are required for diverse biological processes such as nuclear<br />

anchorage and migration, or homologous chromosome pairing. Nucleo-cytoskeletal connections<br />

are built by SUN and KASH domain proteins, transmembrane proteins <strong>of</strong> <strong>the</strong> inner and <strong>the</strong> outer<br />

nuclear membrane, respectively. A key element lies in <strong>the</strong> perinuclear space where <strong>the</strong><br />

conserved SUN and KASH domains form a complex. Thereby, SUN-KASH protein assemblies<br />

bridge <strong>the</strong> entire nuclear envelope to connect nuclear structures with <strong>the</strong> cytoskeleton.


<strong>SUNDAY</strong><br />

We set out to identify <strong>the</strong> determinants <strong>of</strong> SUN-KASH interaction and to understand complex<br />

formation in molecular detail. Using pulldown experiments, we were able to reconstitute SUN-<br />

KASH binding in vitro. We analyzed luminal constructs <strong>of</strong> human SUN1 and SUN2 expressed<br />

ei<strong>the</strong>r in <strong>the</strong> ER <strong>of</strong> Hela cells or purified from E. coli. Both <strong>the</strong> conserved SUN domains and <strong>the</strong><br />

preceding coiled-coil regions were required for interaction. Replacing <strong>the</strong> endogenous coiledcoils<br />

<strong>of</strong> SUN1 or SUN2 by an unrelated coiled-coil domain allowed efficient KASH binding. This<br />

indicates that oligomerization <strong>of</strong> SUN domains by coiled-coils is a key determinant <strong>of</strong> SUN-<br />

KASH interaction.<br />

The crystal structure <strong>of</strong> <strong>the</strong> complex between <strong>the</strong> SUN domain <strong>of</strong> human SUN2 and <strong>the</strong><br />

Nesprin-2 KASH domain (see poster presented by Brian Sosa and Thomas Schwartz) revealed<br />

essential features <strong>of</strong> SUN-KASH interaction in molecular detail. Using mutational analyses <strong>of</strong><br />

both binding partners, we confirmed <strong>the</strong> key determinants in in vitro binding assays.<br />

Fur<strong>the</strong>rmore, we could show that <strong>the</strong> disruption <strong>of</strong> SUN-KASH interaction affects nuclear<br />

envelope targeting <strong>of</strong> both SUN and KASH proteins.<br />

2022<br />

NSrp70 is a novel nuclear speckle-related protein and modulates alternative pre-mRNA<br />

splicing in vivo.<br />

Y-D. Kim 1 , J-Y. Lee 1 , K-M. Oh 1 ; 1 Life Science, Gwangju Inst Science/Technol, Gwangju, Korea<br />

Nuclear speckle is known to serve as <strong>the</strong> storing place <strong>of</strong> mRNA splicing regulators. We report<br />

here <strong>the</strong> identification and characterization <strong>of</strong> a novel speckle protein, referred to as NSrp70,<br />

based on its sub-cellular location and apparent molecular weight. This protein was firstly<br />

identified as <strong>the</strong> NSrp70, as <strong>the</strong> product <strong>of</strong> <strong>the</strong> NIH Mammalian Gene Collection, while no<br />

function has been assigned yet. NSrp70 was co-localized and physically interacted with both<br />

SC35 and ASF/SF2 in speckles. NSrp66 has putative RNA recognition motif, RE/RD rich<br />

domain, and two coiled-coil domains, suggesting role in RNA processing. Accordingly, using<br />

CD44, Tra2b1 and Fas constructs as splicing reporter minigenes, we found that NSrp70<br />

modulated alternative splice site selection in vivo. The C-terminal ten amino acids (aa) (531-<br />

540) including 536 RD 537 were identified as <strong>the</strong> novel nuclear localization signal, and <strong>the</strong> region<br />

spanning 290 to 471 aa was critical for <strong>the</strong> speckle localization and <strong>the</strong> binding to SC35 and<br />

ASF/SF2. The N-terminal region (107-161) was essential for <strong>the</strong> pre-mRNA splicing activity.<br />

Finally, we found that knockout <strong>of</strong> NSrp70 gene in mice produced no progeny including fetal<br />

embryos. Collectively, we demonstrate that <strong>the</strong> NSrp70 is a novel splicing regulator and<br />

essentially required earlier during embryonic development.<br />

2023<br />

Quality control and stress response in mammalian ribosomal biogenesis.<br />

M. Wang 1,2 , D. Pestov 2 ; 1 UMDNJ-GSBS, Stratford, NJ, 2 Cell <strong>Biology</strong>, UMDNJ-SOM, Stratford,<br />

NJ<br />

Ribosome is <strong>the</strong> subcellular component that translates mRNA to proteins. This complex<br />

molecular machine is composed <strong>of</strong> two subunits, and contains four different ribosomal RNAs<br />

(rRNAs) and 79 ribosomal proteins (RPs). The assembly <strong>of</strong> eukaryotic ribosomes is a multi-step<br />

process involving numerous <strong>of</strong> proteins and RNA factors. Compromised ribosome integrity<br />

contributes to premature aging syndromes, cancer and o<strong>the</strong>r diseases. Surveillance<br />

mechanisms act throughout <strong>the</strong> entire ribosome syn<strong>the</strong>sis pathway to eliminate defective<br />

intermediates and <strong>the</strong>reby promote structural and functional integrity <strong>of</strong> <strong>the</strong> final ribosomal<br />

particles. However, ribosome biogenesis is very sensitive to various disturbances <strong>of</strong> cellular


<strong>SUNDAY</strong><br />

metabolism. Multiple studies have shown that “nucleolar stress” caused by disruption <strong>of</strong><br />

ribosome biogenesis activates <strong>the</strong> p53 pathway. However, <strong>the</strong> critical component(s) in ribosome<br />

syn<strong>the</strong>sis machinery that trigger <strong>the</strong> p53 response are unknown. We propose that<br />

nucleophosmin (NPM), a molecular chaperone with predominant nucleolar localization, is able<br />

to transmit stress signal via binding to ribosomal proteins or o<strong>the</strong>r factors involved in ribosome<br />

syn<strong>the</strong>sis. Although NPM is thought to be required for ribosome biogenesis, its down-regulation<br />

in NIH3T3 cells, unlike o<strong>the</strong>r ribosome syn<strong>the</strong>sis factors, does not trigger cell growth arrest. Our<br />

immuno-precipitation data show that certain amount <strong>of</strong> NPM could be pulled down by ribosomal<br />

proteins RPL5 and RPL11. This suggests that instead <strong>of</strong> participating in pre-ribosome<br />

processing directly, NPM may associate and stabilize free RPs in <strong>the</strong> nucleolus, <strong>the</strong>reby<br />

preventing binding <strong>of</strong> Mdm2 to RPs. We also found that expressing a mutant form <strong>of</strong> NPM with<br />

two amino acids substitutions in its oligomerization domain triggered cell growth arrest but did<br />

not affect pre-rRNA processing. Immunostaining also indicates that <strong>the</strong> mutant NPM becomes<br />

distributed over <strong>the</strong> whole nucleus. We are currently testing <strong>the</strong> ability <strong>of</strong> NPM (both wild-type<br />

and mutant forms) to bind RP in <strong>the</strong> in vitro system. Our data support <strong>the</strong> idea that under normal<br />

growth condition, free ribosomal proteins (RPL5 or RPL11) are sequestered by nucleolar NPM<br />

and thus do not interact with Mdm2, an inhibitor <strong>of</strong> p53. Upon stress, delocalization <strong>of</strong> NPM may<br />

liberate ribosomal protein and make <strong>the</strong>m available for binding to Mdm2 in <strong>the</strong> nucleoplasm<br />

which eventually leads to stabilization p53.<br />

2024<br />

Nucleologenesis in Giardia lamblia by light, electron and atomic force microscopy<br />

Nucleolog.<br />

L. F. Jiménez-García 1 , R. Lara-Martínez 2 , I. de la Mora-de la Mora 2 , H. Reyes-Vivas 3 , G. López-<br />

Velázquez 4 , M. Segura-Valdez 2 ; 1 Cell <strong>Biology</strong>, UNAM, México City, Mexico, 2 Cell <strong>Biology</strong>,<br />

UNAM, Mexico, 3 Instituto Nacional de Pediatría, 4 Instituto Nacional de Pediatría, Mexico<br />

The nucleolus is <strong>the</strong> site <strong>of</strong> most ribosome biogenesis in eukaryotes. During cell division, <strong>the</strong><br />

nucleolar elements are redistributed from prophase and <strong>the</strong>y reorganize during telophase by a<br />

process called nucleologenesis. There are several studies on <strong>the</strong> nucleologenesis in animal and<br />

plant cells Nucleologenesis has been studied in animal and plant cells, but less work is known in<br />

protest cells. The smallest nucleolus was described recently in Giardia lamblia. Here we show<br />

by light, electron and atomic force microscopy, that nucleolar elements are present during cell<br />

division in G. lamblia. Samples <strong>of</strong> trophozoites growing onto glass coverslips were processed<br />

for standard transmission electron microscopy and silver staining for NOR both for light and<br />

electron microscopy. Atomic force microscopy was conducted as described using semithin<br />

sections intended for electron microscopy. During cell division, silver staining was observed at<br />

<strong>the</strong> internal periphery <strong>of</strong> each cell nuclei. Atomic force microscopy revealed some nuclear<br />

peripheral material. We conclude that nucleolar elements are present throughout cell division <strong>of</strong><br />

G. lamblia (This work is supported by UNAM DGAPA PAPIIT IN227810).<br />

2025<br />

Mitotic Lamin disassembly is triggered by a lipid-mediated mitotic Lipin signaling<br />

pathway.<br />

M. Mall 1 , T. Walter 1 , M. Gorjánácz 1 , J. Ellenberg 1 , I. W. Mattaj 1 ; 1 European <strong>Molecular</strong> <strong>Biology</strong><br />

Laboratory (EMBL), Heidelberg, Germany<br />

Disassembly <strong>of</strong> <strong>the</strong> lamina, an intermediate filament meshwork that underlies <strong>the</strong> nuclear<br />

envelope, is a key step in early mitosis. While <strong>the</strong> activity <strong>of</strong> several kinases including cyclindependent<br />

kinase 1 (CDK1) and protein kinase C (PKC) have been reported to trigger this<br />

process, <strong>the</strong> relative contribution <strong>of</strong> <strong>the</strong>se kinases to lamina disassembly remains unclear.


<strong>SUNDAY</strong><br />

We have established an automated image-processing pipeline that allows us to quantitatively<br />

monitor lamina disassembly in live HeLa cells. Using kinase inhibitors, we could show that both<br />

CDK1 and PKCs significantly contribute to efficient Lamin B1 disassembly in HeLa cells.<br />

Similarly, overexpression <strong>of</strong> Lamin B1 reporters with mutated CDK1 or PKC consensus<br />

phosphorylation sites also impeded Lamin B1 disassembly. Since <strong>the</strong> role <strong>of</strong> PKC-dependent<br />

pathways during mitosis and <strong>the</strong>ir effects on lamina disassembly are poorly understood, we<br />

focused on elucidating this process in more detail. Using RNAi we found that specific PKC<br />

isoenzymes that require diacylglycerol (DAG) for <strong>the</strong>ir activation trigger rate-limiting steps during<br />

mitotic Lamin B1 disassembly in our in vivo assay.<br />

Interestingly, we recently showed that <strong>the</strong> enzyme Lipin, which produces DAG by<br />

dephosphorylation <strong>of</strong> phosphatidate, is essential for Lamin disassembly in <strong>the</strong> nematode C.<br />

elegans (Gorjánácz and Mattaj, 2009). This suggested that Lipin might act upstream <strong>of</strong> DAGdependent<br />

PKCs. To investigate this possibility we used RNAi to deplete <strong>the</strong> three redundant<br />

human Lipin enzymes and monitored Lamin B1 disassembly. Strikingly, Lipin depletion inhibited<br />

Lamin B1 disassembly to a similar extent as did PKC depletion. Fur<strong>the</strong>rmore, <strong>the</strong> effect <strong>of</strong> Lipin<br />

RNAi could be rescued by <strong>the</strong> addition <strong>of</strong> a DAG analogue.<br />

This supports a model where Lipins function in a lipid-mediated manner to activate a PKCdependent<br />

pathway <strong>of</strong> mitotic Lamin disassembly.<br />

Gorjánácz, M., and Mattaj, I.W. (2009). Lipin is required for efficient breakdown <strong>of</strong> <strong>the</strong> nuclear<br />

envelope in Caenorhabditis elegans. J Cell Sci 122, 1963-1969.<br />

Signal Transduction and Signaling Networks I<br />

2026<br />

Negative regulation <strong>of</strong> Notch1 signaling by Serum- and glucocorticoid-inducible kinase 1.<br />

J-H. Yoon 1 , E-J. Ann 1 , J-S. Ahn 1 , H-S. Park 1 ; 1 Hormone Research Center, School <strong>of</strong> Biological<br />

Sciences and Technology, Chonnam National University, Gwangju, Korea<br />

Notch is a transmembrane protein that acts as a transcriptional factor in <strong>the</strong> Notch signaling<br />

pathway for cell survival, cell death, and cell differentiation. However, little is known about<br />

mechanisms and regulators that are responsible for attenuating <strong>the</strong> Notch signaling pathway<br />

and protein stability. Here, we report that SGK1 remarkably reduced <strong>the</strong> protein stability <strong>of</strong><br />

Notch1 through Fbw7. The protein level and transcriptional activity <strong>of</strong> <strong>the</strong> Notch1 intracellular<br />

domain were higher in SGK1 null cells than in SGK1 wild-type cells. Notch1-IC was able to form<br />

a trimeric complex with Fbw7 and SGK1, <strong>the</strong>reby SGK1 enhanced <strong>the</strong> protein degradation <strong>of</strong><br />

Notch1-IC via a Fbw7-dependent proteasomal pathway. Fur<strong>the</strong>rmore, activated SGK1<br />

phosphorylated Fbw7 at Ser227, an effect inducing Notch1-IC protein degradation and<br />

ubiquitination. Moreover, accumulated dexamethasone-induced SGK1 facilitated <strong>the</strong><br />

degradation <strong>of</strong> Notch1-IC through phosphorylation <strong>of</strong> Fbw7. Altoge<strong>the</strong>r our results suggest that<br />

SGK1 inhibits <strong>the</strong> Notch1 signaling pathway via phosphorylation <strong>of</strong> Fbw7.


<strong>SUNDAY</strong><br />

2027<br />

Secretory Protein Expression Under Microgravity Conditions.<br />

M. Mednieks 1 , I. Khan 2 , A. Khatri 3 , J. Perrier 1 , A. Hand 4 ; 1 Oral Health and Diagnostic Sciences,<br />

University <strong>of</strong> Connecticut Health Center, Farmington, CT, 2 University <strong>of</strong> Connecticut School <strong>of</strong><br />

Dental Medicine, Farmington, CT, 3 Wesleyan University, Middletown, CT, 4 Departments <strong>of</strong><br />

Crani<strong>of</strong>acial Sciences and Cell <strong>Biology</strong>, University <strong>of</strong> Connecticut Health Center, Farmington,<br />

CT<br />

A major goal <strong>of</strong> <strong>the</strong> space shuttle biological experiments is to identify <strong>the</strong> physiological<br />

subsystems that require in-flight monitoring and post-landing testing to evaluate spaceflight<br />

crew status and to devise countermeasures for longer duration missions. Previous studies have<br />

shown that exposure <strong>of</strong> rodents to microgravity results in changes in <strong>the</strong> regulation <strong>of</strong> protein<br />

secretion. To test <strong>the</strong> hypo<strong>the</strong>sis that signaling via cyclic AMP-dependent pathways is affected<br />

by microgravity <strong>the</strong> expression <strong>of</strong> several salivary proteins (both regulated by cyclic AMP and<br />

not) was determined. Methods: Adult female C57Bl/6J mice housed in Animal Enclosure<br />

Modules (AEMs) were flown on <strong>the</strong> space shuttles Discovery and Atlantis. Findings from flight<br />

animals were compared with those from ground control animals housed in AEMs and from<br />

vivarium-housed controls. Tissue samples were collected within 5 hours <strong>of</strong> landing, and glands<br />

were fixed and processed for ultrastructure and frozen for biochemistry. Specific proteins,<br />

including <strong>the</strong> PKA type II regulatory subunit (RII), were identified by immunocytochemistry and<br />

by electrophoresis and Western Blotting (WB), digitized and quantified using NIH ImageJ.<br />

Results: The ultrastructure <strong>of</strong> serous and mucous secretory cells, as well as ducts, was similar<br />

in flight and control animals. Quantitative analysis <strong>of</strong> immunogold labeled thin sections <strong>of</strong> parotid<br />

acinar cells <strong>of</strong> flight animals showed a decrease (p < 0.01) in <strong>the</strong> expression <strong>of</strong> RII, amylase,<br />

and proline-rich proteins (PRPs), whereas parotid secretory protein (PSP) expression was<br />

unchanged. In <strong>the</strong> sublingual gland <strong>the</strong> expression<strong>of</strong> PSP in serous demilune cells and mucin<br />

(Muc-19) in mucous acinar cells was increased (p < 0.001), while RII was slightly increased in<br />

demilune cells (p = 0.23). Demilune cell and parotid protein (DCPP) was increased in<br />

intercalated ducts <strong>of</strong> <strong>the</strong> parotid (p < 0.001), whereas a slight decrease occurred in <strong>the</strong><br />

sublingual gland (p = 0.27). Electrophoretic banding patterns were not markedly altered in flight<br />

parotid or submandibular glands compared to controls. Analysis by WB, however, showed that<br />

in <strong>the</strong> submandibular gland expression <strong>of</strong> IgA (p < 0.05) was decreased while <strong>the</strong>re was an<br />

increase in RII (p < 0.02). Recent data from <strong>the</strong> last flight <strong>of</strong> <strong>the</strong> space shuttle Discovery show<br />

that protein expression in flight samples begins to return to normal by day 5 after landing and<br />

approaches control values by day 7. Conclusions: The expression <strong>of</strong> several parotid,<br />

submandibular and sublingual gland secretory proteins is altered by travel in space. These data<br />

also indicate that <strong>the</strong> responses <strong>of</strong> <strong>the</strong> different salivary glands to microgravity are cell, tissue,<br />

and organ specific. As <strong>the</strong>se secretory proteins are present in saliva, <strong>the</strong>se findings can be used<br />

to establish this easily collected bi<strong>of</strong>luid as a diagnostic tool to assess physiological responses<br />

<strong>of</strong> astronauts as well as for clinical applications on Earth. Support: NASA grant NNX09AP13G<br />

2028<br />

A-kinase Anchoring Proteins: Mediators in Neonatal Rat Schwann Cell Proliferation.<br />

A. L. Asirvatham 1 , R. Stahl 2 , C. M. Schworer 2 , E. Shoemaker 1 , D. J. Carey 2 ; 1 <strong>Biology</strong>,<br />

Misericordia University, Dallas, PA, 2 Weis Center for Research, Geisinger Clinic, Danville, PA<br />

A-kinase Anchoring Proteins: Mediators in Neonatal Rat Schwann Cell Proliferation<br />

Schwann cell proliferation in <strong>the</strong> peripheral nervous system is mediated by <strong>the</strong><br />

heregulin/neuregulin family <strong>of</strong> growth factors that are secreted by neurons. Previous studies<br />

have revealed that culture <strong>of</strong> neonatal rat Schwann cells exhibit a synergistic response in<br />

growth when treated with both heregulin and forskolin, in comparison to cultures incubated with


<strong>SUNDAY</strong><br />

ei<strong>the</strong>r heregulin or forskolin. The molecular mechanisms that mediate <strong>the</strong> proliferation <strong>of</strong><br />

Schwann cells in response to heregulin and forskolin are not well known. This study was<br />

undertaken to identify <strong>the</strong> role <strong>of</strong> <strong>the</strong> cyclic AMP protein kinase A signaling pathway in Schwann<br />

cells that are cultured with heregulin and forskolin. Studies conducted previously have shown<br />

that <strong>the</strong> A-Kinase anchoring proteins <strong>of</strong> <strong>the</strong> cAMP/PKA pathway play an important role in<br />

Schwann cell division. Incubation <strong>of</strong> neonatal rat Schwann cells with SiRNA oligos syn<strong>the</strong>sized<br />

against AKAP95 and AKAP150 resulted in a reduction in protein levels <strong>of</strong> both AKAPs<br />

accompanied by a decrease in cell proliferation. In addition to a decrease in AKAP protein<br />

levels, Schwann cells cultured with <strong>the</strong> SiRNA oligos against AKAP95 and AKAP150, exhibited<br />

a reduction in protein levels <strong>of</strong> cyclin D3 and PKB/Akt respectively. To investigate if <strong>the</strong> loss <strong>of</strong><br />

AKAPs along with cyclin D3 occurred at <strong>the</strong> level <strong>of</strong> syn<strong>the</strong>sis or degradation, neonatal rat<br />

Schwann cells were cultured with <strong>the</strong> protein syn<strong>the</strong>sis inhibitor cycloheximide and <strong>the</strong><br />

proteasome inhibitor lactacystin. Immunoblot analysis <strong>of</strong> Schwann cells incubated with<br />

cycloheximide and SiRNA oligos or cycloheximide alone showed a reduction in both AKAP95<br />

and cyclin D3 levels. Schwann cells cultured with lactacystin and SiRNA oligos showed a<br />

marked reduction in AKAP95 and cyclin D3 levels in comparison to cultures incubated with<br />

lactacystin only. These preliminary observations suggests that <strong>the</strong> loss <strong>of</strong> AKAP95 along with<br />

<strong>the</strong> cyclin D3 proteins does not occur at <strong>the</strong> syn<strong>the</strong>sis or degradation levels and instead may be<br />

mediated by a different mechanism. To examine if AKAP150 associates with PKB/Akt to<br />

stimulate Schwann cell proliferation, cells were incubated with modified N2 media or modified N2<br />

media with heregulin and forskolin. Using immun<strong>of</strong>luorescent staining techniques, preliminary<br />

results demonstrate that Schwann cells treated with heregulin and forskolin exhibit distinct<br />

colocalization patterns for AKAP150 and PKB/Akt. These results suggest that in <strong>the</strong> presence <strong>of</strong><br />

forskolin and heregulin, AKAP150 may associate with PKB/Akt to mediate cell proliferation.<br />

2029<br />

Turn motif phosphorylation negatively regulates activation-loop phosphorylation in Akt.<br />

D. Hiraoka 1 , E. Okumura 1 , T. Kishimoto 1 ; 1 Tokyo Institute <strong>of</strong> Technology, Yokohama,<br />

Kanagawa, Japan<br />

Akt, also known as PKB, plays a central role in various signaling pathways that regulate cellular<br />

processes such as metabolism, proliferation and survival. Under stimulation, phosphorylation <strong>of</strong><br />

<strong>the</strong> activation loop (A-loop) and hydrophobic motif (HM) <strong>of</strong> Akt by <strong>the</strong> kinase, PDK1, and <strong>the</strong><br />

mammalian target <strong>of</strong> rapamycin complex 2 (mTORC2), respectively, results in its activation. A<br />

well-conserved threonine in <strong>the</strong> turn motif (TM) is also constitutively phosphorylated by<br />

mTORC2 and contributes to <strong>the</strong> stability <strong>of</strong> Akt. However, <strong>the</strong> relationship between TM<br />

phosphorylation and <strong>the</strong> phosphorylation <strong>of</strong> Akt on <strong>the</strong> HM and A-loop has not been sufficiently<br />

evaluated. Using starfish oocytes as a model system, our study provides <strong>the</strong> first evidence that<br />

TM phosphorylation plays a negative role in A-loop phosphorylation. In this system, <strong>the</strong><br />

maturation-inducing hormone, 1-methyladenine, stimulates Akt to induce meiotic reinitiation<br />

through <strong>the</strong> activation <strong>of</strong> cyclin B-Cdc2. The phosphorylation status <strong>of</strong> Akt was monitored via<br />

introduction <strong>of</strong> exogenous human Akt in starfish oocytes. Injection <strong>of</strong> an anti-starfish TOR<br />

antibody inhibited TM and HM phosphorylation, suggesting that phosphorylation <strong>of</strong> <strong>the</strong>se sites<br />

depends on TOR, as reported in mammalian cells. Precise analyses <strong>of</strong> single or double alanine<br />

substitution mutants at each <strong>of</strong> three phosphorylation residues revealed that TM<br />

phosphorylation renders Akt susceptible to dephosphorylation on <strong>the</strong> A-loop. When A-loop<br />

phosphatase was inhibited by okadaic acid, TM phosphorylation still reduced A-loop<br />

phosphorylation, suggesting that <strong>the</strong> effect is caused at least partially through reduction <strong>of</strong><br />

sensitivity to PDK1. By contrast, HM phosphorylation enhanced A-loop phosphorylation and<br />

achieved full activation <strong>of</strong> Akt via a mechanism at least partially independent <strong>of</strong> TM


<strong>SUNDAY</strong><br />

phosphorylation. These observations provide new insight into <strong>the</strong> mechanism controlling Akt<br />

phosphorylation in <strong>the</strong> cell.<br />

2030<br />

MK-STYX Reduces eIF2α Phosphorylation.<br />

J. E. Barr 1 , S. D. Hinton 1 ; 1 <strong>Biology</strong>, College <strong>of</strong> William and Mary, Williamsburg, VA<br />

The pseudophosphatase MK-STYX [MAPK (mitogen-activated protein kinase)<br />

phosphoserine/threonine/tyrosine-binding protein] has been implicated in <strong>the</strong> stress response<br />

pathway. MK-STYX interacts with Ras-GTPase activating protein SH3 domain binding protein-1<br />

(G3BP-1), and inhibits stress granule assembly. Stress granules, cytoplasmic storage sites for<br />

mRNA, form as a protective mechanism against stress caused by UV irradiation, hypoxia, and<br />

heat shock. In addition, <strong>the</strong> overexpression <strong>of</strong> G3BP-1, when dephosphorylated induces stress<br />

granule assembly. Initially, we hypo<strong>the</strong>sized that MK-STYX inhibition <strong>of</strong> stress granules was<br />

G3BP-1 phosphorylation-dependent. However, data with G3BP-1 phosphomimetic and nonphosphorylatable<br />

mutants suggest differently so we turned our focus to <strong>the</strong> eukaryotic initiation<br />

factor 2 alpha (eIF2�). eIF2�� initiates translation by forming a ternary complex with methionine<br />

bound to tRNA and GTP. Phosphorylation <strong>of</strong> eIF2� arrests translation and results in stress<br />

granule formation, whereas dephosphorylation promotes polysomes. To determine if MK-STYX<br />

inhibits stress granule assembly via upstream interactions, we investigated its affects on eIF2�<br />

phosphorylation. HeLa cells were transfected with pMT2-FLAG-MK-STYX-FLAG or pMT2<br />

expression vectors, and heat shocked for an hour to induce stress. Cells were lysed and<br />

immunoblots for phosphorylation <strong>of</strong> eIF2� were performed. We show that <strong>the</strong> presence <strong>of</strong> MK-<br />

STYX reduced <strong>the</strong> phosphorylation <strong>of</strong> eIF2�. Fur<strong>the</strong>rmore, MK-STYX also reduced <strong>the</strong> eIF2�<br />

phosphorylation in heat shocked cells, which showed a drastic increase in phosphorlyation in<br />

<strong>the</strong> absence <strong>of</strong> MK-STYX. These data are significant because <strong>the</strong>y provide more insight into<br />

how MK-STYX inhibits stress granule assembly.<br />

2031<br />

VEGF modulates Col I expression in human adipose derived stem cells through Akt<br />

dependent mechanisms.<br />

C-J. Li 1 , V. Madhu 1 , A. Dighe 1 , G. Balian 1,2 , Q. Cui 1,3 ; 1 Orthopaedic Research Labs, University <strong>of</strong><br />

Virginia, Charlottesville, VA, 2 Biochemistry and <strong>Molecular</strong> Genetics, University <strong>of</strong> Virginia,<br />

Charlottesville, VA, 3 Orthopaedic Surgery, University <strong>of</strong> Virginia, Charlottesville, VA<br />

Objective:<br />

We earlier demonstrated that in vivo osteogenesis induced by mouse bone marrow derived<br />

osteoprogenitor cells expressing bone morphogenetic protein – 6 (BMP-6) or Lim Mineralization<br />

Protein - 1 (LMP-1) was enhanced significantly by co-expressing vascular endo<strong>the</strong>lial growth<br />

factor (VEGF) gene. The facts that increase in osteogenesis did not completely correlate with<br />

VEGF-induced angiogenesis and that LMP-1 is downstream activator <strong>of</strong> BMP-6 signaling<br />

pathway clearly indicated interaction between VEGF and o<strong>the</strong>r signaling pathways. In order to<br />

understand mechanisms <strong>of</strong> <strong>the</strong> interaction and to determine if similar interactions occurred in<br />

human mesenchymal stem cells we determined in vitro osteogenesis <strong>of</strong> human adipose derived<br />

stem cells (hADSCs) in presence <strong>of</strong> purified VEGF and BMP-6 proteins.<br />

Results:<br />

The results <strong>of</strong> calcium deposition showed no significant difference between control, BMP-6,<br />

VEGF or BMP-6+VEGF group in hADSC at day 14th. However, significant increase in<br />

expression <strong>of</strong> Col I α2 gene was observed in VEGF group. At day 14, a synergistic increase in<br />

Col I α2 expression was observed in VEGF+BMP-6 group. Addition <strong>of</strong> Akt1/2/3 inhibitor


<strong>SUNDAY</strong><br />

suppressed Akt and Smad1/5 phosphorylation, calcium deposition and Col I α2 mRNA<br />

expression in hADSCs. Most importantly, blockage <strong>of</strong> Akt activation significantly inhibited <strong>the</strong><br />

synergistic enhancement <strong>of</strong> Col I α2 mRNA expression, which was induced by BMP-6 and<br />

VEGF combined treatment.<br />

Conclusion and Discussion:<br />

Our data showed a striking observation that VEGF and BMP-6 synergistically enhanced Col I α2<br />

gene expression in hADSCs. Fur<strong>the</strong>rmore, biochemical blockage <strong>of</strong> Akt activation significantly<br />

inhibited <strong>the</strong> enhancing effects <strong>of</strong> Col I α2 mRNA expression by BMP-6 and VEGF combined<br />

treatment. It is reported that PI3-kinase catalytic is<strong>of</strong>orm is coupled to VEGFR2 and regulates<br />

<strong>the</strong> bulk <strong>of</strong> Akt activity. Moreover, dominant-negative Akt inhibited Smad5 phosphorylation. Our<br />

data and <strong>the</strong>se reported observations toge<strong>the</strong>r establish a novel axis <strong>of</strong> control <strong>of</strong> Col I<br />

expression as VEGF-VEGFR2-PI3K-Akt-Smad1/5-Col I in hADSCs. However, Akt-dependent<br />

BMP-6 modulation <strong>of</strong> this axis remains to be investigated in near future.<br />

2032<br />

A novel gene encoding a candidate CK2 substrate involves in cancer cell proliferation.<br />

H. Yang 1 , S. Liu 1 , H. Zheng 1 , H. Jiang 1 , Y. Qu 1 , Y. Li 1 ; 1 Harbin Institute <strong>of</strong> Technology, Harbin,<br />

China<br />

CK2, a ubiquitous protein serine/threonine kinase, is essential for <strong>the</strong> decision <strong>of</strong> cell survival<br />

and cell death. In <strong>the</strong> present study, we identified a novel gene, Homo Sapiens Chromosome 1<br />

ORF109 which protein product might be a candidate CK2 substrate. The C1ORF109 protein<br />

presented in <strong>the</strong> phosphorylated form in vivo, and could be phosphorylized by <strong>the</strong> protein kinase<br />

CK2 in vitro. Truncated constructs and site direct mutagenesis analysis revealed that Ser104,<br />

Ser 134, and Ser182 were phosphorylated sites. Both TATA box and CAAT box within <strong>the</strong><br />

crucial region <strong>of</strong> c1orf109 promoter are required for maximal transcription <strong>of</strong> <strong>the</strong> c1orf109 gene.<br />

The C1ORF109 protein was mainly located in <strong>the</strong> nucleus and cytoplasm. Upregulated<br />

expression <strong>of</strong> c1orf109 was detected in multiple breast cancer cells. Exogenous expression <strong>of</strong><br />

C1ORF109 in breast cancer Hs578T cells promoted colony formation and cell proliferation<br />

along with increased level <strong>of</strong> PCNA and cyclinD1. Meanwhile, knockdown <strong>of</strong> c1orf109 by siRNA<br />

in breast cancer MDA-MB-231 cells resulted in inhibition <strong>of</strong> cell proliferation. Taken toge<strong>the</strong>r, our<br />

findings suggest that C1ORF109 may be <strong>the</strong> down stream target <strong>of</strong> protein kinase CK2 and<br />

involved in <strong>the</strong> regulation <strong>of</strong> cancer cell proliferation.<br />

Key words: promoter, transcription, CK2 kinase, c1orf109, proliferation<br />

2033<br />

Quantitative detection <strong>of</strong> phosphorylation <strong>of</strong> STAT3 and p38 by Western blotting.<br />

K. Söderquist 1 , S. Grimsby 1 , M. Winkvist 1 ; 1 GE Healthcare, Uppsala, Sweden<br />

Introduction<br />

Western blotting is a well established technique used to study proteins from a wide variety <strong>of</strong><br />

sources. The technique is used throughout life sciences from basic research to medical<br />

diagnostic applications. Western blotting is at best considered as semi-quantitative and hence<br />

limited to studies involving large protein differences. However development <strong>of</strong> detection<br />

reagents as well as imagers has open up for quantitative applications when high sensitivity and<br />

broad dynamic range is needed.<br />

Here we demonstrate how <strong>the</strong> Western blotting technique can be used to determine<br />

phosphorylation levels <strong>of</strong> various proteins in whole cell lysates upon stimulation with various<br />

well known inducers.


<strong>SUNDAY</strong><br />

Methods<br />

HeLa cells were stimulated with INF-β. Phosphorylation level <strong>of</strong> STAT3 was determined by<br />

Western blotting performed according to standard procedure and protocol supplied with <strong>the</strong><br />

Amersham ECL Prime reagent using optimized blocking solution and antibodies<br />

concentrations.<br />

293T cells where stimulated with TGF-β. Phosphorylation level <strong>of</strong> p38 was determined by<br />

Western blotting performed according to standard procedure and protocol supplied with <strong>the</strong><br />

Amersham ECL Prime reagent using optimized blocking solution and antibodies concentrations.<br />

Detection reagent was added to <strong>the</strong> membrane and <strong>the</strong> chemiluminescent signal was captured<br />

using CCD cameras.<br />

Results<br />

The result demonstrates that phosphorylation <strong>of</strong> STAT3 upon stimulation with INF-β can be<br />

quantitatively determined by Western blotting. In addition, <strong>the</strong> result demonstrates that time<br />

dependent phosphorylation <strong>of</strong> p38 upon stimulation with TGF-β can be quantitatively<br />

determined by Western blotting.<br />

Conclusions<br />

The Western blotting technique can be used for accurate quantitative analysis <strong>of</strong><br />

phosphorylation level <strong>of</strong> various proteins, such as p38 and STAT3, by combining <strong>the</strong> use <strong>of</strong> a<br />

detection reagent, providing high sensitivity and broad dynamic range, with a CCD camera.<br />

2034<br />

A distinctive salt bridge arrangement in RSK P-loop enables discovery <strong>of</strong> is<strong>of</strong>ormselective<br />

RSK1/2 inhibitors that protect from myocardial infarction injury.<br />

V. O. Paavilainen 1 , I. Serafimova 2 , M. S. Cohen 3 , M. Spreafico 2 , J-I. Abe 4 , M. Jacobson 2 , J.<br />

Taunton 2 ; 1 Cellular and <strong>Molecular</strong> Pharmacology, UC-San Francisco, San Francisco, CA, 2 UC-<br />

San Francisco, 3 Oregon Health and Science University, Portland, OR, 4 University <strong>of</strong> Rochester,<br />

Rochester, NY<br />

p90 ribosomal protein S6 kinases (RSK) are serine/threonine kinases under <strong>the</strong> MAPK/ERK<br />

signaling pathway, which have been implicated in a variety <strong>of</strong> cellular functions such as cell<br />

proliferation and survival. Their expression is upregulated in primary breast and prostate<br />

cancers, and mutations in RSK2 gene lead to C<strong>of</strong>fin-Lowry syndrome, an X-linked form <strong>of</strong><br />

mental retardation. We sought to discover potent RSK1 inhibitors with high selectivity over<br />

RSK2. These inhibitors utilized diverse electrophilic warheads that covalently target a nonconserved<br />

cysteine in <strong>the</strong> RSK active site and led to <strong>the</strong> identification <strong>of</strong> an irreversible RSK<br />

inhibitor, fmk, with ~20-fold selectivity difference between <strong>the</strong> highly identical RSK1 and RSK2<br />

is<strong>of</strong>orms both in vitro and in vivo. Detailed biophysical and structural experiments showed that<br />

this selectivity arises from differences in <strong>the</strong> reversible binding affinity mediated by subtle<br />

differences in <strong>the</strong> structure and dynamics <strong>of</strong> <strong>the</strong> kinase P-loop region due to a specific salt<br />

bridge arrangement around <strong>the</strong> P-loop region. In a mouse model <strong>of</strong> myocardial infarction (MI),<br />

fmk treatment resulted in a dramatic improvement in survival and heart function, possibly by<br />

inhibition <strong>of</strong> NHE1 phosphorylation. Future studies will aim to dissect <strong>the</strong> roles <strong>of</strong> different RSK<br />

is<strong>of</strong>orms during MI injury.


<strong>SUNDAY</strong><br />

2035<br />

Inhibition <strong>of</strong> Notch1 signaling by APP intracellular domain.<br />

E-J. Ann 1 , J-H. Yoon 1 , J-S. Ahn 1 , H-S. Park 1 ; 1 Chonnam National University, Gwangju, Korea<br />

The Notch1 receptor is a crucial controller <strong>of</strong> cell fate decisions, and is also a key regulator <strong>of</strong><br />

cell growth and differentiation in a variety <strong>of</strong> contexts. In this study, we have demonstrated that<br />

<strong>the</strong> APP intracellular domain (AICD) attenuates Notch1 signaling by accelerated degradation <strong>of</strong><br />

<strong>the</strong> Notch1 intracellular domain (Notch1-IC) and RBP-Jk, through different degradation<br />

pathways. AICD suppresses Notch1 transcriptional activity by <strong>the</strong> dissociation <strong>of</strong> <strong>the</strong> Notch1-IC–<br />

RBP-Jk complex after processing by gamma-secretase. Notch1-IC is capable <strong>of</strong> forming a<br />

trimeric complex with Fbw7 and AICD, and AICD enhances <strong>the</strong> protein degradation <strong>of</strong> Notch1-<br />

IC through an Fbw7-dependent proteasomal pathway. AICD downregulates <strong>the</strong> levels <strong>of</strong> RBP-<br />

Jk protein through <strong>the</strong> lysosomal pathway. AICD-mediated degradation is involved in <strong>the</strong><br />

preferential degradation <strong>of</strong> non-phosphorylated RBP-Jk. Collectively, our results demonstrate<br />

that AICD functions as a negative regulator in Notch1 signaling through <strong>the</strong> promotion <strong>of</strong><br />

Notch1-IC and RBP-Jk protein degradation.<br />

2036<br />

Isoprenylcysteine methylation is required for growth and endocytosis in Dictyostelium<br />

discoideum.<br />

K. J. McQuade 1 , J. Bollan 1 , K. Bailey 1 , J. Fritz 1 ; 1 Colorado Mesa University, Grand Junction, CO<br />

Many members <strong>of</strong> <strong>the</strong> Ras superfamily <strong>of</strong> small monomeric GTPases and o<strong>the</strong>r membraneassociated<br />

proteins ending in carboxy-terminal –CAAX motifs are proteolytically processed and<br />

reversibly methylated on isoprenylated terminal cysteine residues. Methylation is thought to<br />

regulate localization and activity <strong>of</strong> <strong>the</strong>se proteins. Ras GTPases are <strong>of</strong>ten overactive in cancer<br />

cells and inhibitors <strong>of</strong> isoprenylcysteine methyltransferase are being tested as potential<br />

chemo<strong>the</strong>rapies, but <strong>the</strong> cellular effects <strong>of</strong> <strong>the</strong>se inhibitors are unclear. We are characterizing<br />

<strong>the</strong> effects <strong>of</strong> loss <strong>of</strong> isoprenylcysteine methyltransferase activity in <strong>the</strong> social amoeba,<br />

Dictyostelium discodeum. Amoebae lacking methyltransferase activity grow slowly on bacterial<br />

lawns and do not grow in shaking or adherent axenic culture. Methyltransferase-deficient cells<br />

also migrate slowly towards folate, have reduced rates <strong>of</strong> endocytosis and are sensitive to<br />

osmotic shock. The mechanisms underlying <strong>the</strong>se defects are being investigated.<br />

2037<br />

Understanding <strong>the</strong> cellular function <strong>of</strong> poly(ADP-ribose) and its importance in cancer.<br />

K. Krukenberg 1 , T. Mitchison 2 ; 1 Systems <strong>Biology</strong>, Harvard Medical School, Boston, MA,<br />

2 Systems <strong>Biology</strong>, Harvard Medical School<br />

Poly(ADP-ribose) is a unique but poorly understood post-translational modification. It has been<br />

implicated in multiple cellular processes including DNA damage, cell death, inflammation,<br />

protein stability and cell division. In <strong>the</strong> clinic, inhibition <strong>of</strong> poly(ADP-ribose) formation is showing<br />

promise as a cancer <strong>the</strong>rapeutic. Poly(ADP-ribose) polymerases (PARPs) catalyze <strong>the</strong> addition<br />

<strong>of</strong> ADP-ribose onto acceptor proteins using NAD+ as a substrate. Though <strong>the</strong> importance <strong>of</strong><br />

PARPs and poly(ADP-ribose) is clear, <strong>the</strong>ir biological functions are not well understood. We are<br />

working on developing tools for characterizing <strong>the</strong> function <strong>of</strong> PARPs and poly(ADP-ribose) and<br />

are using <strong>the</strong>se tools to fur<strong>the</strong>r identify and define <strong>the</strong> biological roles <strong>of</strong> <strong>the</strong>se intriguing and<br />

essential molecules. Here we describe an assay for quantifying <strong>the</strong> levels <strong>of</strong> poly(ADP-ribose) in<br />

cells that has increased sensitivity as compared to previous approaches. Using this assay, we<br />

quantified <strong>the</strong> extent to which poly(ADP-ribose) levels vary throughout <strong>the</strong> cell cycle. We have<br />

also found that different cancer cell types have different amounts <strong>of</strong> poly(ADP-ribose) and <strong>the</strong>se


<strong>SUNDAY</strong><br />

levels appear to be largely attributable to PARP1 activity. Current efforts are focused on better<br />

understanding <strong>the</strong> molecular basis for <strong>the</strong> differences in polymer levels. Not only will this provide<br />

insight into <strong>the</strong> biological function <strong>of</strong> poly(ADP-ribose) but poly(ADP-ribose) levels may be<br />

useful as an indicator <strong>of</strong> which patients will respond best to PARP inhibitors or o<strong>the</strong>r treatments.<br />

2038<br />

Monitoring Post-Translational Modifications Using Antibody-Based Real-Time PCR.<br />

R. Bruinsma 1 , K. G. Huwiler 1 , B. D. Marks 1 , M. Shannon 2 , D. Ruff 2 , B. Schweitzer 3 ; 1 Life<br />

Technologies, Madison, WI, 2 Life Technologies, Foster City, CA, 3 Life Technologies, Carlsbad,<br />

CA<br />

Protein post-translational modifications (PTM) are covalent modifications to specific amino acids<br />

within a protein. PTMs <strong>of</strong>ten occur via reversible enzymatically catalyzed reactions and<br />

numerous types <strong>of</strong> PTMs exist, including phosphorylation, acetylation, methylation, and<br />

ubiquitination. The importance <strong>of</strong> PTMs resides in <strong>the</strong>ir ability to influence <strong>the</strong> function, stability,<br />

and localization <strong>of</strong> <strong>the</strong> modified protein within cells. These changes in protein properties due to<br />

PTMs are critical to <strong>the</strong> regulation <strong>of</strong> cellular signaling networks and <strong>the</strong> deregulation <strong>of</strong> protein<br />

PTMs is associated with many diseases, including cancer. Two proteins that are recognized to<br />

be important in cancer biology and that undergo PTMs are p53 and AKT. p53 is central to<br />

maintaining <strong>the</strong> integrity <strong>of</strong> <strong>the</strong> genome and is found to be mutated in ~50% <strong>of</strong> all cancers.<br />

Phosphorylation <strong>of</strong> p53 at Ser15 has been shown to result in p53 protein stabilization, which can<br />

result in transcriptional activation. The PI3K/AKT pathway is critical for normal cell growth and<br />

survival, but persistent uncontrolled activation <strong>of</strong> this pathway is associated with cancer. Full<br />

kinase activity <strong>of</strong> AKT and downstream cellular signaling involves phosphorylation <strong>of</strong> Ser473<br />

and Thr308. We report <strong>the</strong> application <strong>of</strong> a technique that partners <strong>the</strong> specificity <strong>of</strong> antibody<br />

detection with <strong>the</strong> signal amplification <strong>of</strong> real-time PCR (RT-PCR) to monitor changes in PTMs.<br />

Specifically, we were able to quantitatively monitor changes in p53 Ser15 phosporylation and<br />

AKT Ser473 phosphorylation from very small cellular samples. To our knowledge, this is <strong>the</strong> first<br />

demonstration <strong>of</strong> monitoring PTMs for p53 and AKT via RT-PCR. This is a powerful method for<br />

monitoring PTMs due to <strong>the</strong> high specificity, high sensitivity, and small sample requirement.<br />

2039<br />

Association <strong>of</strong> Connexin 36 with synaptic scaffolding proteins in <strong>the</strong> retina.<br />

A. Vila 1 , C. Whitaker 1 , J. O'Brien 1 ; 1 Opthalmology and Vision, University <strong>of</strong> Texas Health<br />

Science Center, Houston, TX<br />

Our visual world contains an incredible range <strong>of</strong> light intensities. This necessitates adaption at<br />

many levels <strong>of</strong> <strong>the</strong> visual system to produce useful vision. In <strong>the</strong> retina, electrical synapses<br />

composed <strong>of</strong> gap junctions display extensive plasticity associated with light adaptation. Recent<br />

studies have found that NMDA-type glutamate receptors regulate coupling among AII amacrine<br />

cells and that NMDA receptors are localized near Cx36 gap junctions on AII amacrine cells.<br />

These studies indicate that a localized Ca 2+ signal derived from NMDA receptor activation drives<br />

Cam kinase II-dependent phosphorylation <strong>of</strong> Cx36 to produce activity-dependent enhancement<br />

<strong>of</strong> coupling. To maintain <strong>the</strong> integrity <strong>of</strong> this signaling mechanism, we hypo<strong>the</strong>sized that<br />

scaffolding proteins may assemble Cx36 gap junctions and NMDA receptors into a complex<br />

analogous to <strong>the</strong> post-synaptic density <strong>of</strong> conventional synapses. To explore this hypo<strong>the</strong>sis, we<br />

examined <strong>the</strong> association <strong>of</strong> Cx36 with traditional synaptic scaffold proteins Synapse-<br />

Associated Protein 97 (SAP97), SAP102, and Post-Synaptic Density 95 (PSD95) by<br />

immun<strong>of</strong>luorescence in rabbit retina and 3-D image reconstruction. We fur<strong>the</strong>r examined <strong>the</strong><br />

association <strong>of</strong> <strong>the</strong>se proteins by co-immunoprecipitation and western blot techniques.<br />

Antibodies to SAP97 and SAP102 gave punctate labeling throughout <strong>the</strong> inner synaptic layer <strong>of</strong>


<strong>SUNDAY</strong><br />

<strong>the</strong> retina, in which AII amacrine cells make <strong>the</strong>ir gap junctions. Antibodies to PSD95 did not<br />

label <strong>the</strong> inner synaptic layer, but did label <strong>the</strong> outer synaptic layer, where photoreceptor<br />

synapses are located. In <strong>the</strong> inner synaptic layer, approximately 50% <strong>of</strong> <strong>the</strong> Cx36 gap junctions<br />

were co-localized with ei<strong>the</strong>r SAP97 or SAP102, and 15% co-localized with both. SAP102 colocalized<br />

with about twice as many Cx36 gap junctions as did SAP97. Fur<strong>the</strong>rmore, <strong>of</strong> <strong>the</strong><br />

SAP102 puncta in <strong>the</strong> inner synaptic layer, fully one third were associated with Cx36 gap<br />

junctions. In <strong>the</strong> outer synaptic layer, some PSD95 immunoreactivity was closely associated<br />

with Cx36. Our data show that synaptic scaffolding proteins are associated not only with<br />

conventional synapses, but also with electrical synapses. These scaffolding proteins may<br />

facilitate assembly <strong>of</strong> non-synaptic neurotransmitter receptors, such as <strong>the</strong> NMDA receptors on<br />

AII amacrine cells, with Cx36 gap junctions. They may alternatively, or in addition, facilitate <strong>the</strong><br />

association <strong>of</strong> conventional synapses and gap junctions to form mixed synapses.<br />

2040<br />

A structural biology approach to predict proteolytic sites <strong>of</strong> accessory gland proteins in<br />

D. Melanogaster.<br />

C. A. Del Carpio 1 , M-T. Yamamoto 1 ; 1 Drosophila Genetic Resource Center (DGRC), Kyoto<br />

Institute <strong>of</strong> Technology, Kyoto, Japan<br />

Extra and intra-cellular protein processing and degradation accounts for one <strong>of</strong> <strong>the</strong> main<br />

biochemical processes involved in series <strong>of</strong> signal transduction and regulation pathways.<br />

Moreover, genome-wide studies <strong>of</strong> higher organisms have unveiled that a large proportion <strong>of</strong><br />

encoded proteins are enzymes oriented to <strong>the</strong> cleavage <strong>of</strong> o<strong>the</strong>r proteins. Thus predicting <strong>the</strong><br />

cleavage sites <strong>of</strong> substrate proteins and <strong>the</strong> enzyme involved is highly required in functional<br />

genome analyses including <strong>the</strong> elucidation <strong>of</strong> intra and extra-cellular mechanisms at <strong>the</strong> origin<br />

<strong>of</strong> cellular malfunction and diseases. Sequence based inference <strong>of</strong> proteolytic cleavage sites in<br />

polypeptides frequently leads to several sites that are located in <strong>the</strong> core <strong>of</strong> <strong>the</strong> molecule or are<br />

sterically hindered and hardly accessible to <strong>the</strong> cleaving enzyme. Catering to <strong>the</strong> need <strong>of</strong><br />

including 3D structural factors in <strong>the</strong> assessment <strong>of</strong> cleavage sites in polypeptides and reduce<br />

<strong>the</strong> number <strong>of</strong> candidates output at <strong>the</strong> primary structure level, we propose a bio-computational<br />

strategy to firstly evaluate substrate-enzyme affinity and subsequently infer putative scissible<br />

bonds. Affinity is evaluated by docking <strong>the</strong> 3D structures <strong>of</strong> <strong>the</strong> substrate and <strong>the</strong> enzyme, and<br />

molecular dynamics (MD) simulation is performed to assess <strong>the</strong> formation <strong>of</strong> <strong>the</strong> transition state<br />

(TS) complex triggering <strong>the</strong> proteolytic reaction. The cleavage sites are <strong>the</strong>n inferred by tracing<br />

<strong>the</strong> main atomic interactions throughout <strong>the</strong> MD simulation leading to formation <strong>of</strong> <strong>the</strong> TS<br />

species. The methodology involves protein structure prediction, as well as protein docking to<br />

assess substrate-enzyme interaction. As a case study we applied <strong>the</strong> methodology to <strong>the</strong><br />

prediction <strong>of</strong> cleavage sites for Acp36DE, a D. Melanogaster seminal fluid protein that plays an<br />

important role in sperm storage and competition in multiply mated females. The results show<br />

that among several candidate regions amenable to protease attack, <strong>the</strong> region LEU514-<br />

ASP517, located at a disordered region between two alpha helices shows geometrical<br />

complementarity and energetic affinity for an astacin like protease. The TS species derived<br />

through <strong>the</strong> MD simulation shows <strong>the</strong> metal (Zn) in <strong>the</strong> astacin pulling <strong>the</strong> LYS515.O, and <strong>the</strong><br />

contraction <strong>of</strong> <strong>the</strong> distance between <strong>the</strong> SER516.N and GLU101.OE2 in astacin suggests <strong>the</strong><br />

scissible peptide bond. Repeating <strong>the</strong> calculations with serine proteases reported to be<br />

expressed at copulation shows a higher affinity region for <strong>the</strong> hydrophobic region that includes<br />

amino acids SER21-GLU22-SER23-PHE24 <strong>the</strong> cleavage bond being GLU22-SER23. Cleavage<br />

<strong>of</strong> Acp36DE at LYS515-SER516 generates fragments <strong>of</strong> Acp36DE <strong>of</strong> molecular weights that<br />

compare well with reported experimental findings


<strong>SUNDAY</strong><br />

2041<br />

Evolutionary analysis <strong>of</strong> <strong>the</strong> phosphorylation signaling reveals functionally conserved<br />

motifs.<br />

H. Yoshizaki 1 , S. Okuda 2 ; 1 Department <strong>of</strong> Biomedical Sciences, College <strong>of</strong> Life Sciences,<br />

Ritsumeikan University, Kusatsu, Shiga, Japan, 2 Department <strong>of</strong> Bioinformatics, College <strong>of</strong> Life<br />

Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan<br />

(Background) Protein kinases regulate various cellular processes by phosphorylating specific<br />

substrates. Phosphoproteomic studies recently have shown that tens <strong>of</strong> thousands <strong>of</strong> sites are<br />

phosphorylated. In addition, in silico systemes biological analyses have revealed protein<br />

phosphorylations dynamically regulate cellular networks. These studies provide powerful tools<br />

for studying intra-cellular signal transduction pathways. High throughput proteomic techniques<br />

provide a massive amount <strong>of</strong> protein information but we can not directly retrieve protein-protein<br />

interactions organizing cellular processes. Toward understanding <strong>the</strong> complicate intracellular<br />

signaling networks, we performed a comparative analysis about evolutionally conserved<br />

phosphorylation motifs. (Methods and Results) First, we retrieved <strong>the</strong> known phosphorylated<br />

protein sequences from <strong>the</strong> PhoshoSitePlus database (http://www.phosphosite.org/) and<br />

determined phosphorylation motifs by clustering <strong>the</strong>m. As a result, we acquired 186 original<br />

phosphorylation motifs from 434 clusters obtained by <strong>the</strong> MCL clustering method. Subsequently,<br />

we assigned kinase information to a motif based on <strong>the</strong> known kinase-subustrate combinations<br />

stored in <strong>the</strong> PhosphoSitePlus database and predicted a probable kinase phosphorylating <strong>the</strong><br />

motif. Fur<strong>the</strong>rmore, we evaluated <strong>the</strong> evolutionary conservation <strong>of</strong> <strong>the</strong> phosphorylation motifs by<br />

a comparative sequence analysis from yeast to human. We found that most <strong>of</strong> <strong>the</strong> motifs<br />

conserved along <strong>the</strong> general evolutionary scenario, but some motifs showed <strong>the</strong> evolutionary<br />

patterns different from <strong>the</strong>m. Additional protein functional analysis showed that <strong>the</strong>se<br />

phosphorylation sites were involved in a specific protein function. Our comparative evolutionary<br />

analysis <strong>of</strong> phosphorylation motifs is useful methods to highlight protein groups with specific<br />

functions from complicated signal transduction pathways.<br />

2042<br />

Network topologies evolve during neutrophil polarization.<br />

C-J. Ku* 1 , Y. Wang* 1 , O. Weiner 2 , S. J. Altschuler 1 , L. F. Wu 1 ; 1 Dept <strong>of</strong> Pharmacology, Green<br />

Ctr for Systems <strong>Biology</strong>, Simmons Cancer Ctr, University <strong>of</strong> Texas Southwestern Med Ctr,<br />

Dallas, TX, 2 Cardiovascular Research Institute and Department <strong>of</strong> Biochemistry, University <strong>of</strong><br />

California, San Francisco, San Francisco, CA<br />

*These authors contributed equally to <strong>the</strong> work.<br />

Much attention has focused on how complex cellular behaviors can emerge from static<br />

networks. Here, we found that <strong>the</strong> rapid response <strong>of</strong> neutrophils to stimulus arises from a timevarying<br />

series <strong>of</strong> distinct signaling network topologies. Not only do <strong>the</strong> signaling topologies<br />

change over time, but <strong>the</strong> topology also depends on <strong>the</strong> phenotype. Surprisingly, <strong>the</strong> set <strong>of</strong><br />

persistent links revealed an underlying simplicity <strong>of</strong> <strong>the</strong> network structure: a linear cascade<br />

controlling signaling activities, and a feed-forward network in <strong>the</strong> opposite direction controlling<br />

signaling polarity. Our work provides a clear example that simple circuits orchestrating cellular<br />

behaviors may lay hidden beneath <strong>the</strong> complexity <strong>of</strong> comprehensive “everything-connects-toeverything”<br />

networks obtained by combining links from different times and phenotypes.


2043<br />

The Drosophila Transcription Factor Protein Interactome.<br />

D. Y. Rhee 1 , B. Zhai 1 , C. Wong 1 , C. Beekman 1 , S. Gygi 1 , R. Obar 1 , S. Artavanis-Tsakonas 1 ;<br />

1 Department <strong>of</strong> Cell <strong>Biology</strong>, Harvard Medical School, Boston, MA<br />

<strong>SUNDAY</strong><br />

From <strong>the</strong> earliest embryo to <strong>the</strong> adult, <strong>the</strong> spatiotemporal expression <strong>of</strong> genes is essential for<br />

normal development and physiology. At <strong>the</strong> basis <strong>of</strong> this is <strong>the</strong> regulation <strong>of</strong> transcription via<br />

transcription factors (TFs), proteins that physically bind DNA to activate or suppress gene<br />

expression. As <strong>the</strong> target <strong>of</strong> signaling pathways, TFs represent a crucial point <strong>of</strong> regulation<br />

relating to <strong>the</strong> vast majority <strong>of</strong> cellular processes and as a rule function through interactions with<br />

o<strong>the</strong>r proteins. Consequently, <strong>the</strong> characterization <strong>of</strong> protein-protein interactions involving TFs is<br />

essential for understanding how <strong>the</strong>y function to regulate gene expression and, in turn, <strong>the</strong><br />

biology <strong>of</strong> <strong>the</strong> cell. Towards this end, we have generated a co-AP/MS-based interaction map<br />

encompassing more than 500 from <strong>the</strong> total <strong>of</strong> 750 TFs estimated, by various criteria, to<br />

represent <strong>the</strong> entire gamut <strong>of</strong> TFs in Drosophila melanogaster.<br />

Cell Migration and Motility<br />

2044<br />

Macrophage Strength: Quantifying Phagocytic Forces and Kinetics.<br />

N. Sosale 1 , T. Rouhi 2 , P. Rodriguez 1 , R. Lipowsky 2 , D. Discher 1 ; 1 Biophysical Engineering Lab,<br />

University <strong>of</strong> Pennsylvania, Philadelphia, PA, 2 Max Planck Institute <strong>of</strong> Colloids and Interfaces,<br />

Potsdam, Germany<br />

Macrophages act as immunological gatekeepers at <strong>the</strong> interface <strong>of</strong> tissue, blood, and lymph as<br />

<strong>the</strong>se cells take up antigens from <strong>the</strong> extracellular environment and <strong>the</strong>n present <strong>the</strong>m to <strong>the</strong><br />

immune system stimulating lymphocytes. We have studied <strong>the</strong> force and kinetics <strong>of</strong><br />

phagocytosis by human macrophages. To this aim, we studied phagocytosis <strong>of</strong> opsonized red<br />

blood cells. The force calculation is based on blood cell's shape deformation and elastic<br />

properties. Our analysis shows that <strong>the</strong> range <strong>of</strong> <strong>the</strong> resultant force imposed by Macrophages is<br />

up to 100 pN and is imparted within 10 minutes after initial contact.<br />

2045<br />

Integrin Linked Kinase Modulates Phagocytosis through Rac1 Activation in Epidermal<br />

Keratinocytes.<br />

S. Sayedyahossein 1,2 , L. Nini 1 , L. Dagnino 1,2 ; 1 Physiology and Pharmacology, The University <strong>of</strong><br />

Western Ontario, London, ON, Canada, 2 Children’s Health Research Institute, London, ON,<br />

Canada<br />

Integrin linked kinase (ILK) is an adaptor protein that regulates numerous biological processes,<br />

such as cell survival and migration. ILK is also essential for keratinocyte interactions with <strong>the</strong><br />

extracellular matrix and development <strong>of</strong> polarity, but <strong>the</strong> exact role <strong>of</strong> ILK in o<strong>the</strong>r epidermal<br />

functions, such as melanosome phagocytosis, is not clear. We examined alterations in<br />

phagocytic capacity in ILK-deficient epidermal keratinocytes, using 0.5-µm latex microspheres.<br />

Inactivation <strong>of</strong> <strong>the</strong> Ilk gene resulted in severe impairment <strong>of</strong> microsphere uptake. Fur<strong>the</strong>r,<br />

stimulation <strong>of</strong> <strong>the</strong> keratinocyte growth factor (KGF) receptor or PAR-2 failed to increase<br />

phagocytosis in <strong>the</strong>se cells. Activation <strong>of</strong> ERK in response to KGF stimulation is unaltered in<br />

ILK-deficient keratinocytes, indicating that <strong>the</strong> KGF receptor likely signals normally in <strong>the</strong><br />

absence <strong>of</strong> ILK. In contrast, ILK deficiency was associated with impaired Rac1 activation in<br />

response to KGF. The latter was accompanied by reduced formation <strong>of</strong> membrane ruffles and


<strong>SUNDAY</strong><br />

abnormal F-actin dynamics. Thus, ILK is essential for normal keratinocyte phagocytosis in<br />

response to KGF.<br />

Supported by <strong>the</strong> Canadian Institutes <strong>of</strong> Health Research (CIHR).<br />

2046<br />

Myristolated Alanine-Rich C-Kinase Substrate (MARCKS) is an Important Regulator <strong>of</strong><br />

Equine Neutrophil Migration and Adhesion.<br />

M. K. Sheats 1 , K. L. Carren 2 , E. M. Hefner 3 , E. J. Sung 2 , K. B. Adler 4 , S. L. Jones 2 ; 1 <strong>Molecular</strong><br />

Biological Sciences, North Carolina State University, Holly Springs, NC, 2 Clinical Sciences,<br />

North Carolina State University, 3 Animal Science, North Carolina State University, 4 <strong>Molecular</strong><br />

Biological Sciences, North Carolina State University<br />

Neutrophil infiltration is a prominent feature in a number <strong>of</strong> pathologic conditions including<br />

recurrent airway obstruction and ischemic reperfusion injury. Cell signaling components<br />

involved in neutrophil migration represent targets for novel anti-inflammatory <strong>the</strong>rapies. This<br />

study was conducted to test <strong>the</strong> hypo<strong>the</strong>sis that <strong>the</strong> 32 kD protein, Myristolated Alanine-Rich C-<br />

Kinase Substrate (MARCKS) is involved in equine neutrophil migration and adhesion.<br />

In o<strong>the</strong>r species (i.e. human) MARCKS phosphorylation and dephosphorylation causes <strong>the</strong><br />

protein to cycle between <strong>the</strong> cell membrane and cytosol, respectively. To investigate MARCKS<br />

phosphorylation in horses, neutrophils were isolated from whole blood using Ficoll gradient<br />

centrifugation and stimulated with platelet activating factor (PAF), leukotriene B4 (LTB4) or<br />

phorbol myristate acetate (PMA). Western blot was performed using specific phospho-MARCKS<br />

and total MARCKS primary antibodies. These results determined that MARCKS phosphorylation<br />

in equine neutrophils is maximal 30 seconds following stimulation with 100 nM PAF or LTB4 and<br />

that dephosphorylation occurs within 3 minutes.<br />

MARCKS interacts with <strong>the</strong> resting neutrophil cell membrane through <strong>the</strong> combined efforts <strong>of</strong> its<br />

myristolated N-terminus and basic amino acid effector domain; potentially regulating interactions<br />

between PIP2 and actin binding proteins. To investigate <strong>the</strong> requirement for MARCKS in equine<br />

neutrophil chemotaxis, isolated neutrophils were pre-treated with MANS (a cell permeant<br />

peptide identical to <strong>the</strong> N-terminal 24 amino acids <strong>of</strong> MARCKS), RNS (a scrambled sequence<br />

control peptide) or vehicle control prior to migration toward known neutrophil chemoattractants<br />

(LTB4 or PAF). Pre-treatment <strong>of</strong> equine neutrophils with <strong>the</strong> MANS peptide significantly inhibited<br />

LTB4 and PAF stimulated migration while <strong>the</strong> control RNS peptide had no effect.<br />

Beta-integrin upregulation and adhesion is an essential process to neutrophil chemotaxis. To<br />

investigate <strong>the</strong> requirement for MARCKS in equine neutrophil adhesion, isolated neutrophils<br />

were pretreated with MANS, RNS or vehicle control and stimulated to adhere to Immulon 2<br />

plates coated with 5% FBS (a β-integrin dependent substrate) in PBS with 10 ng/ml PMA or<br />

vehicle control. Pre-treatment <strong>of</strong> equine neutrophils with <strong>the</strong> MANS peptide significantly inhibited<br />

PMA induced adhesion; while <strong>the</strong> control RNS peptide had no effect.<br />

Pre-treatment <strong>of</strong> equine neutrophils with a cell permeant peptide identical to <strong>the</strong> N-terminus <strong>of</strong><br />

MARCKS significantly inhibited equine neutrophil PMA induced adhesion and LTB4 and PAF<br />

induced migration. These results indicate that MARCKS is an important regulator <strong>of</strong> equine<br />

neutrophil migration and adhesion and MARCKS regulation <strong>of</strong> neutrophil chemotaxis has been<br />

conserved across evolutionary lines.


<strong>SUNDAY</strong><br />

2047<br />

The Scar/WAVE complex is necessary for proper regulation <strong>of</strong> traction stresses during<br />

amoeboid motility.<br />

E. E. Bastounis 1 , R. Meili 2 , B. Alonso Latorre 2 , J-C. del Alamo 2 , J. Lasheras 2 , R. Firtel 2 ;<br />

1 Bioengineering, University <strong>of</strong> California, San Diego, San Diego, CA, 2 University <strong>of</strong> California,<br />

San Diego<br />

Chemotaxis, or directed cell migration, is involved in a broad spectrum <strong>of</strong> biological phenomena,<br />

ranging from <strong>the</strong> metastatic spreading <strong>of</strong> cancer to <strong>the</strong> active migration <strong>of</strong> neutrophils during<br />

wound healing or in response to bacterial infection. Chemotaxis requires a tightly regulated,<br />

spatiotemporal coordination <strong>of</strong> underlying biochemical processes. SCAR/WAVE-mediated<br />

dendritic F-actin polymerization at <strong>the</strong> cell’s leading edge plays a key role in cell migration. Our<br />

study identifies a mechanical and biochemical role for <strong>the</strong> SCAR/WAVE complex in modulating<br />

<strong>the</strong> traction stresses that drive cell movement. We demonstrate that <strong>the</strong> traction stresses <strong>of</strong><br />

wild-type Dictyostelium cells or cells lacking <strong>the</strong> SCAR/WAVE complex protein PIR121 (pirA-) or<br />

SCAR (scrA-) exert stresses <strong>of</strong> different strength that correlate with <strong>the</strong>ir levels <strong>of</strong> F-actin. By<br />

processing <strong>the</strong> time records <strong>of</strong> <strong>the</strong> cell length and <strong>the</strong> strain energy exerted by <strong>the</strong> cells on <strong>the</strong>ir<br />

substrate, we show that wild-type cells migrate by repeating a specific set <strong>of</strong> mechanical steps<br />

(motility cycle), whereby <strong>the</strong> cell length (L) and <strong>the</strong> strain energy exerted by <strong>the</strong> cells on <strong>the</strong>ir<br />

substrate (Us) vary periodically. Our analysis also revealed that scrA- cells exhibit an altered<br />

motility cycle with a longer period (T=160s) and a lower migration velocity (V =6μm/min)<br />

compared to those <strong>of</strong> wild-type cells (T=80s, V =13μm/min). In marked contrast to <strong>the</strong>se strains,<br />

pirA- cells, although <strong>the</strong>y have a higher F-actin content, migrate as slowly as scrA- cells but <strong>the</strong>ir<br />

migration occurs in a seemingly random manner in that <strong>the</strong>y lack <strong>the</strong> periodic changes in<br />

traction stresses that are observed for <strong>the</strong> o<strong>the</strong>r two strains. Finally, by quantifying <strong>the</strong> level <strong>of</strong><br />

F-actin in <strong>the</strong> leading edge in combination with <strong>the</strong> traction stresses, we demonstrated that <strong>the</strong><br />

level <strong>of</strong> leading edge, SCAR/WAVE complex-mediated F-actin polymerizations is critical for <strong>the</strong><br />

level and spatiotemporal control <strong>of</strong> <strong>the</strong> traction stresses, cell-substrate interactions, and <strong>the</strong><br />

motility cycle.<br />

2048<br />

Self-Generated EGF Gradients Guide Epi<strong>the</strong>lial Cell Migration.<br />

D. Irimia 1 , A. J. Aranyosi 1 , B. Kulemann 1 , S. Thayer 1 , M. Toner 1 , O. Illiopoulos 1 , C. Scherber 1 ;<br />

1 BioMEMS Resource Center, Massachusetts General Hospital/Harvard Medical School, Boston,<br />

MA<br />

Migrating epi<strong>the</strong>lial cells can contribute to wound healing processes, or can trigger lethal<br />

complications during cancer through invasion and distant metastasis. During <strong>the</strong>ir migration,<br />

cells must be able to orient accurately and it is generally assumed that gradients <strong>of</strong> chemokines<br />

or growth factors are required to guide epi<strong>the</strong>lial cells towards <strong>the</strong>ir target location.<br />

Unexpectedly, we uncovered a novel strategy for epi<strong>the</strong>lial cell orientation during migration that<br />

does not require pre-existent chemical gradients. Using microscale engineering techniques, we<br />

demonstrate that <strong>the</strong> strategy is dependent on <strong>the</strong> competition between epidermal growth factor<br />

(EGF) uptake by <strong>the</strong> cells and <strong>the</strong> restricted diffusion <strong>of</strong> EGF from surrounding<br />

microenvironment. Both normal and malignant epi<strong>the</strong>lial cells are capable <strong>of</strong> EGF uptake and<br />

can use this strategy when placed in confined environment. The strategy has some surprising<br />

consequences for epi<strong>the</strong>lial cells, like <strong>the</strong> ability to efficiently disperse through channels, or to<br />

navigate along <strong>the</strong> shortest path to exit from various microscopic mazes. Inhibition <strong>of</strong> EGFreceptor<br />

but not <strong>the</strong> inhibition <strong>of</strong> chemokine receptors, reduces <strong>the</strong> ability <strong>of</strong> <strong>the</strong> cells to orient,<br />

without altering <strong>the</strong>ir velocity <strong>of</strong> migration. Better understanding <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lial cells guidance<br />

strategy by EGF uptake-dependent self-generated gradients could lead to approaches for


<strong>SUNDAY</strong><br />

restricting <strong>the</strong> migration <strong>of</strong> malignant cells to delay local invasion and distant metastases, or<br />

enhance <strong>the</strong> migration <strong>of</strong> normal cells to promote wound healing.<br />

2049<br />

Three dimensional traction forces exerted by migrating amoeboid cells.*<br />

R. Meili 1 , B. Alvarez-Gonzalez 2 , J. C. del Alamo 2 , R. A. Firtel 1 , J. C. Lasheras 2 ; 1 Division <strong>of</strong> Cell<br />

and Developmental <strong>Biology</strong>, University <strong>of</strong> California, San Diego, La Jolla, CA, 2 MAE<br />

Department, University <strong>of</strong> California, San Diego<br />

Cell migration is essential for many physiological processes in multicellular organisms including<br />

embryonic development, wound healing and cancer metastasis. Amoeboid motility requires <strong>the</strong><br />

spatiotemporal coordination <strong>of</strong> forces generated by biochemical processes. This results in<br />

directional motility by repeated protrusion <strong>of</strong> <strong>the</strong> front and retraction <strong>of</strong> <strong>the</strong> rear driven by actin<br />

polymerization and actomyosin contraction.<br />

Mechanically, movement requires <strong>the</strong> modulation <strong>of</strong> <strong>the</strong> cell’s interactions with <strong>the</strong> environment,<br />

ei<strong>the</strong>r with surrounding cells or with extracellular matrix. For quantitative analysis, <strong>the</strong>se<br />

interactions have so far typically been characterized as tangential traction stresses imposed by<br />

<strong>the</strong> cell on a flat substrate.<br />

We have expanded <strong>the</strong> method for measuring traction stresses to determine <strong>the</strong> stresses<br />

exerted perpendicular to <strong>the</strong> substrate in addition to <strong>the</strong> in-plane stresses.<br />

We find that <strong>the</strong> deformation perpendicular to <strong>the</strong> substrate is significant and that calculating <strong>the</strong><br />

stresses using one <strong>of</strong> <strong>the</strong> existing two-dimensional traction cytometry methods misses an<br />

important aspect <strong>of</strong> cellular mechanics.<br />

We used our new method to better understand <strong>the</strong> mechanical role <strong>of</strong> individual cytoskeletal<br />

components by comparing traction stress maps and dynamics <strong>of</strong> wild type cells moving over flat<br />

substrates with measurements <strong>of</strong> Dictyostelium cells with mutations in cytoskeletal proteins. We<br />

focused our study on mutant cell lines with crosslinking defects, such as myosin II-null cells and<br />

cortexilin-null cells.<br />

We find that cells from all cell lines studied push downward on <strong>the</strong> substrate near <strong>the</strong> center <strong>of</strong><br />

<strong>the</strong> cell and pull up near <strong>the</strong> periphery. The magnitude <strong>of</strong> <strong>the</strong> perpendicular forces is<br />

comparable to <strong>the</strong> magnitude <strong>of</strong> <strong>the</strong> tangential forces exerted on <strong>the</strong> substrate; <strong>the</strong>refore <strong>the</strong><br />

perpendicular component is expected to play a significant role in <strong>the</strong> cell behavior and cannot<br />

be neglected. Our initial findings show that <strong>the</strong> effects <strong>of</strong> <strong>the</strong> crosslinking mutations on <strong>the</strong><br />

parallel forces do not track with <strong>the</strong> effects on <strong>the</strong> perpendicular forces. For example, myosin IInull<br />

cells show a significant reduction <strong>of</strong> <strong>the</strong> front to back organization <strong>of</strong> <strong>the</strong> parallel traction<br />

forces while <strong>the</strong> push pull distribution <strong>of</strong> forces remains unaffected. Our observations suggest<br />

that <strong>the</strong> generation <strong>of</strong> <strong>the</strong> stresses perpendicular and tangential to <strong>the</strong> substrate is possibly<br />

controlled independently.<br />

*Work supported by NIH grant 1RO1 GM084227.<br />

2050<br />

Pr<strong>of</strong>ilin-1: a biomarker for breast cancer malignancy?<br />

Z. Ding 1 , R. Bhargava 2 , N. Lakshman 3 , M. Petroll 4 , P. Roy 5 ; 1 Bioengineering, University <strong>of</strong><br />

Pittsburgh, Pittsburgh, PA, 2 University <strong>of</strong> Pittsburgh, Pittsburgh, PA, 3 University <strong>of</strong> Texas<br />

Southwestern Medical Center Dallas, Dallas, TX, 4 Ophthalmology, University <strong>of</strong> Texas<br />

Southwestern Medical Center Dallas, Dallas, TX, 5 Bioengineering and Pathology, University <strong>of</strong><br />

Pittsburgh, Pittsburgh, PA<br />

This study unexpectedly reveals that stable loss <strong>of</strong> expression <strong>of</strong> pr<strong>of</strong>ilin1 (Pfn1), an actinbinding<br />

protein that has been traditionally shown to be required for cell proliferation and<br />

migration, in fact promotes tumor growth and overall distant metastasis <strong>of</strong> breast cancer


<strong>SUNDAY</strong><br />

xenografts in mice. Comparative studies in spontaneous vs experimental metastasis models<br />

suggest that loss <strong>of</strong> Pfn1 expression likely promotes metastasis-promoting events at <strong>the</strong> primary<br />

tumor site. ECM invasion (an important step for tumor metastasis) <strong>of</strong> breast cancer cells is<br />

increased upon loss <strong>of</strong> Pfn1 expression. Experiments involving selective perturbation <strong>of</strong><br />

signaling pathway downstream <strong>of</strong> PI3-Kinase (PI3K) fur<strong>the</strong>r revealed that hyper-invasiveness <strong>of</strong><br />

Pfn1-depleted cells is mediated by a PI3K-PI(3,4)P2-lamellipodin signaling axis. Finally, as a<br />

clinical correlate <strong>of</strong> <strong>the</strong>se in vitro and in vivo findings, analyses <strong>of</strong> Pfn1 expression in primary<br />

tumors revealed that Pfn1 downregulation occurs in all molecular subtypes <strong>of</strong> breast cancer,<br />

somewhat more prominently in estrogen receptor-negative tumors. Fur<strong>the</strong>rmore, tumors<br />

involving significant lymph node infiltration and/or distant metastasis present <strong>the</strong> most dramatic<br />

downregulation in Pfn1 expression. These findings imply that Pfn1 could serve as a new<br />

molecular marker for breast cancer malignancy.<br />

2051<br />

Reestablishing <strong>the</strong> polarity <strong>of</strong> Dictyostelium cells to understand <strong>the</strong> organization <strong>of</strong> <strong>the</strong><br />

cytoskeleton and signaling complexes during chemotaxis.<br />

D. Jowhar 1 , G. Wright 1 , C. Janetopoulos 1,2 ;<br />

1 Biological Sciences, Vanderbilt University, Nashville, TN, 2 Cell and Developmental <strong>Biology</strong>,<br />

Vanderbilt University, Nashville, TN<br />

Cells migrating in a chemical gradient typically have a polarized morphology, where <strong>the</strong>y display<br />

a distinct front and back. Cells can also have different localized sensitivities to chemoattractants<br />

depending on <strong>the</strong>ir degree and type <strong>of</strong> polarity. To better understand <strong>the</strong>se phenomena, we<br />

used <strong>the</strong> social amoeba Dictyostelium discoideum that expressed GFP-tagged proteins known<br />

to localize to distinct regions <strong>of</strong> <strong>the</strong> cell during cell migration. Dictyostelium cells were lured into<br />

a specialized micr<strong>of</strong>luidic device developed in our laboratory which confined <strong>the</strong> cells in narrow<br />

3-dimensional Polydimethylsiloxane (PDMS) channels. The chemoattractant gradient in this<br />

experimental platform could be reversed so<br />

that <strong>the</strong> rear <strong>of</strong> <strong>the</strong> cell was exposed to a higher concentration <strong>of</strong> chemoattractant than <strong>the</strong> front<br />

<strong>of</strong> <strong>the</strong> cell. Cells in <strong>the</strong> channels would freeze, become unpolarized, and were <strong>the</strong>n capable <strong>of</strong><br />

forming a new front at <strong>the</strong> former rear <strong>of</strong> <strong>the</strong> cell. We have observed <strong>the</strong> temporal and spatial<br />

loss and gain <strong>of</strong> localization <strong>of</strong> various signaling and cytoskeletal molecules at both <strong>the</strong> rear and<br />

front <strong>of</strong> <strong>the</strong> cell as polarity is broken down and re-established. We monitored <strong>the</strong> localization <strong>of</strong><br />

PHcrac (a biosensor for PI(3,4)P2 and PI(3,4,5)P3), RBD (a biosensor for Ras activity), PTEN-<br />

GFP (a phosphatase which elevates levels <strong>of</strong> PI(4,5)P2), LimE (a marker for F-actin), and<br />

TACC-GFP (a microtubule centrosome and plus-end<br />

marker.) The signaling markers showed a re-distribution to opposite sides <strong>of</strong> <strong>the</strong> cell during <strong>the</strong><br />

reversals, as did LimE. We also monitored <strong>the</strong> microtubules and centrosomes and tracked <strong>the</strong>ir<br />

redistribution during polarity re-establishment using TACC. Fur<strong>the</strong>r experiments were performed<br />

in <strong>the</strong> absence <strong>of</strong> an actin cytoskeleton and in a variety <strong>of</strong> signaling mutants. These experiments<br />

have helped elucidate <strong>the</strong> molecular mechanisms that contribute to <strong>the</strong> establishment and<br />

maintenance <strong>of</strong> cell polarity.<br />

2052<br />

Alteration <strong>of</strong> α6 integrin expression and chemotactic motility <strong>of</strong> microglia upon ADP<br />

stimulation.<br />

S-H. Lee 1 , C. Y. Chung 2 ; 1 Pharmacology, Vanderbilt University Medical Center, Nashville, TN,<br />

2 Vanderbilt University Medical Center, Nashville, TN<br />

Microglia are <strong>the</strong> immune effector cells that are rapidly activated in response to even minor<br />

pathological changes in <strong>the</strong> central nervous system. It has been demonstrated that microglia


<strong>SUNDAY</strong><br />

attach well to fibronectin and vitronectin, but only weakly to laminin, and that laminin exerts a<br />

dominant anti-adhesive effect on microglial adhesion. Microglial activation is reported to be<br />

accompanied by <strong>the</strong> alteration <strong>of</strong> integrin expression. Inflammatory cytokines increased<br />

expression <strong>of</strong> α4β1, α5β1, and Mac-1 integrins on microglia but changes <strong>of</strong> integrin expression<br />

upon ADP (a chemoattractant for microglia) stimulation remain unknown. We, in this study,<br />

investigated if ADP can induce <strong>the</strong> alteration <strong>of</strong> integrin species on cell surface, leading to<br />

changes in chemotactic ability on different ECM. FACS scans and surface biotinylation assays<br />

showed that ADP stimulation induced a significant increase in <strong>the</strong> expression <strong>of</strong> α6 integrin, but<br />

not α5 on <strong>the</strong> surface <strong>of</strong> microglia cells. Examination <strong>of</strong> microglia motility on different ECM<br />

revealed that cells have greater motility on laminin than fibronectin upon ADP stimulation,<br />

presumably due to <strong>the</strong> increased expression <strong>of</strong> α6 integrin. The alteration <strong>of</strong> integrin-mediated<br />

adhesion may regulate <strong>the</strong> extent <strong>of</strong> microglial infiltration into <strong>the</strong> site <strong>of</strong> damage by controlling<br />

<strong>the</strong>ir chemotactic ability.<br />

2053<br />

Directing cell migration with nano-ridges/grooves.<br />

X. Sun 1 , M. Driscoll 2 , J. Fourkas 1 , W. Losert 2 ; 1 Chemistry and Biochemistry, University <strong>of</strong><br />

Maryland, College Park, MD, 2 Physics, University <strong>of</strong> Maryland, College Park, MD<br />

We evaluated <strong>the</strong> contact guidance <strong>of</strong> nano-ridges/grooves on <strong>the</strong> amoeba Dictyostelium<br />

discoideum, a model system for studying cell migration. Nano-ridges were fabricated with<br />

Multiphoton Absorption Polymerization (MAP). These nano-topographies, as well as<br />

complementary nano-grooves, were replicated with high fidelity from an improved composite<br />

polydimethylsiloxane (PDMS) mold. By analyzing <strong>the</strong> cellular velocity, orientation, boundary<br />

curvature and shape dynamics, we found that <strong>the</strong> ridges and grooves exert bidirectional<br />

guidance on migrating cells. More cells migrate in <strong>the</strong> direction <strong>of</strong> <strong>the</strong> nano-ridges/grooves than<br />

perpendicular to <strong>the</strong>m. We also found that cells aligning in <strong>the</strong> direction <strong>of</strong> <strong>the</strong> nanoridges/grooves<br />

are more elongated and that cells migrating in <strong>the</strong> direction <strong>of</strong> <strong>the</strong> nanoridges/grooves<br />

migrate faster. Contact guidance is only achieved with topotraphies <strong>of</strong> proper<br />

pitch. Nano-ridges/grooves with a pitch smaller than 0.2 μm or greater than 10 μm do not exhibit<br />

effective guidance. This study may <strong>of</strong>fer insights into fabricating complex 3D scaffolds for tissue<br />

engineering with MAP.<br />

2054<br />

The N-formylpeptide Receptor FPR1 Is Functionally Expressed in Lens Epi<strong>the</strong>lial Cells<br />

and is Critical for Lens Maintenance in Mice.<br />

J-L. Gao 1 , J. Tuo 1 , E. H. Schneider 1 , D. Despres 1 , M. Lizak 1 , A. Maminishkis 1 , C. C. Chan 1 , P.<br />

M. Murphy 1 ; 1 National Institutes <strong>of</strong> Health, Be<strong>the</strong>sda, MD<br />

Most mouse knockouts for inflammatory chemoattractants and <strong>the</strong>ir receptors lack spontaneous<br />

phenotypes. We considered whe<strong>the</strong>r an aging study might identify slowly developing<br />

phenotypes in such mice. Here we show that mice lacking Fpr1 gene (Fpr1-/- mice) have severe<br />

cataracts at an early age. FPR1 is known to mediate pro-inflammatory chemotactic responses<br />

by phagocytic leukocytes to N-formylpeptides produced by bacteria or mitochondria and plays a<br />

role in host defense against certain bacteria. Fpr1-/- mice developed small eyes starting at 3<br />

month <strong>of</strong> age because <strong>of</strong> lens degeneration and rupture. Histopathological analysis did not<br />

show any infection or inflammation in <strong>the</strong>se eyes, but showed abnormal lens cell migration.<br />

Consistent with this, we detected FPR1 mRNA in both mouse and human lens epi<strong>the</strong>lial cells,<br />

and FPR1 proteins on primary human fetal lens cells with <strong>the</strong> level compatible to that detected<br />

on human neutrophils. Stimulation <strong>of</strong> lens epi<strong>the</strong>lial cells with FPR1 ligands induced intracellular


<strong>SUNDAY</strong><br />

calcium release and actin polymerization. Taken toge<strong>the</strong>r, <strong>the</strong> data implicate FPR1 in mediating<br />

lens cell migration, a critical event for lens development and maintenance.<br />

2055<br />

PRL-3 promotes migration and invasion by up-regulating MMP-7 in human colorectal<br />

cancer cells.<br />

Y-M. Han 1 , S-K. Lee 2 , D-S. Sin 2 , Y-R. Ha 2 , J. Kim 2 , C-W. Lee 3 ; 1 University <strong>of</strong> Science and<br />

Technology, Daejeon, Korea, 2 Laboratory <strong>of</strong> Chemical <strong>Biology</strong> and Genomics, Korea Research<br />

Institute <strong>of</strong> Bioscience and Biotechnology, Daejeon, Korea, 3 Bio-Evaluation Center, Korea<br />

Research Institute <strong>of</strong> Bioscience and Biotechnology, Chungbuk, Korea<br />

PRL-3, a member <strong>of</strong> a subgroup <strong>of</strong> protein tyrosine phosphatases (PTPs) that can stimulate <strong>the</strong><br />

degradation <strong>of</strong> <strong>the</strong> extracellular matrix, is over-expressed in metastatic colorectal cancer relative<br />

to primary tumors. To determine whe<strong>the</strong>r PRL-3-induced enhancement <strong>of</strong> migration and<br />

invasion is dependent on <strong>the</strong> expression <strong>of</strong> matrix metalloproteases (MMPs), PRL-3 was<br />

expressed in DLD-1 human colorectal cancer cells. The motility, migration and invasion<br />

characteristics <strong>of</strong> <strong>the</strong> cells were examined and metastasis to <strong>the</strong> lung was confirmed in a nude<br />

mouse using PRL-3-overexpressing DLD-1 cells [DLD-1 (PRL-3)]. Migration and invasion <strong>of</strong> <strong>the</strong><br />

cells were inhibited by phosphatase and farnesyl transferase inhibitors. Expression <strong>of</strong> MMPs<br />

was enhanced 3- to 10-fold in comparison to control cells, and migration and invasion were<br />

partially inhibited by siRNA knockdown <strong>of</strong> MMP-2, -13, or -14. Importantly, siRNA knockdown <strong>of</strong><br />

MMP-7 completely inhibited <strong>the</strong> migration and invasion <strong>of</strong> DLD-1 (PRL-3) cells, while<br />

overexpression <strong>of</strong> MMP-7 increased migration. The expression <strong>of</strong> MMP-7 was also downregulated<br />

by phosphatase and farnesyl transferase inhibitors. It was found that PRL-3 induced<br />

MMP-7 through oncogenic pathways including PI3K/AKT and ERK, and that <strong>the</strong>re is a<br />

relationship between <strong>the</strong> expression <strong>of</strong> PRL-3 and MMP-7 in human tumor cell lines. The<br />

expression <strong>of</strong> MMP-13 and MMP-14 was very sensitive to <strong>the</strong> inhibition <strong>of</strong> farnesyl transferase;<br />

however, <strong>the</strong> migration and invasion <strong>of</strong> DLD-1 (PRL-3) cells did not strongly depend on <strong>the</strong><br />

expression <strong>of</strong> MMP-13 or -14. These results suggest that <strong>the</strong> migration and invasion <strong>of</strong> PRL-3expressing<br />

colorectal cancer cells depends primarily on <strong>the</strong> expression <strong>of</strong> MMP-7.<br />

2056<br />

Serine phosphorylation in paxillin is important for focal adhesion formation during cell<br />

adhesion onto collagen type I.<br />

T. Kwak 1 , J. W. Lee 1 ; 1 Department <strong>of</strong> Pharmacy, Research Institute <strong>of</strong> Pharmaceutical<br />

Sciences, College <strong>of</strong> Pharmacy, Seoul National University, Seoul, Korea<br />

Integrin-mediated adhesion to extracellular matrix proteins is dynamically regulated during<br />

morphological change and cell migration. Upon cell adhesion, protein-protein interaction at focal<br />

adhesions consisting <strong>of</strong> FAK, talin, paxillin, and o<strong>the</strong>rs, plays major roles in regulation <strong>of</strong> cell<br />

morphogenesis and migration. Although phosphorylation at tyrosines <strong>of</strong> paxillin has been well<br />

known to be critically involved in adhesion-mediated signaling, little is known about <strong>the</strong><br />

significance <strong>of</strong> paxillin phosphorylation at serine 85 in cell migration. Here in this study, we<br />

provide evidences that paxillin phosphorylation at serine 85 occurred during HeLa cell adhesion<br />

onto collagen I, being concomitant with FAK and talin phosphorylations. However,<br />

unphosphorylatable mutant S85A paxillin impaired cell spreading, focal adhesion turnover, and<br />

migration toward collagen I but not to serum components. Fur<strong>the</strong>r, <strong>the</strong> physical interaction<br />

between paxillin and talin was through <strong>the</strong> C-terminal tail <strong>of</strong> talin leading to focal adhesions at<br />

<strong>the</strong> cell boundary, but S85A paxillin did not bind talin and caused random focal adhesion<br />

formation. Toge<strong>the</strong>r, <strong>the</strong>se observations suggest that paxillin phosphorylation at Serine 85<br />

depending on cell adhesion is important for interaction with talin and for cell morphological and


<strong>SUNDAY</strong><br />

migratory ability. [This work was supported by NRF by senior researchers program (Leap<br />

research, 2011-0001160) and Global Frontier Project grant (NRF-M1AXA002-2010-0029778),<br />

and a grant <strong>of</strong> <strong>the</strong> Korean Health Technology R&D Project (A100727), MHWFA, Korea to JW<br />

Lee].<br />

Key words: focal adhesion, paxillin, talin, morphology, migration<br />

2057<br />

Nck Is Required for Focal Adhesion Maturation and Polarized Cell Motility.<br />

S. P. Chaki 1 , R. Barhoumi 2 , R. Yog 1 , M. E. Berginski 3 , S. M. Gomez 3 , G. M. Rivera 1 ; 1 Veterinary<br />

Pathobiology, Texas A&M University, College Station, TX, 2 Veterinary Integrative Biosciences,<br />

Texas A&M University, College Station, TX, 3 University <strong>of</strong> North Carolina at Chapel Hill, NC<br />

The establishment <strong>of</strong> cell polarity is essential for directional migration <strong>of</strong> endo<strong>the</strong>lial cells and<br />

<strong>the</strong> organization and function <strong>of</strong> endo<strong>the</strong>lial layers. The SH2/SH3 domain-containing adaptors<br />

Nck (Nck1 and Nck2) link signaling by tyrosine phosphorylation with effectors that mediate<br />

remodeling <strong>of</strong> <strong>the</strong> actin cytoskeleton. We previously determined an important role for Nck<br />

adaptors in endo<strong>the</strong>lial cell migration, invasion, and in vitro morphogenesis. To understand <strong>the</strong><br />

mechanisms by which Nck promotes <strong>the</strong>se processes, we used differential interference contrast<br />

(DIC) and total internal reflection fluorescence (TIRF) microscopy to compare membrane and<br />

adhesion dynamics <strong>of</strong> human umbilical vein endo<strong>the</strong>lial cells (HUVEC) with or without<br />

perturbation <strong>of</strong> Nck signaling. Expression <strong>of</strong> specific short hairpin RNAs induced > 90% and<br />

50% reduction in Nck1 and Nck2 levels, respectively. Double knockdown cells (Nck1 and Nck2)<br />

were rescued by expression <strong>of</strong> a siRNA-resistant Nck2 cDNA. Kymographs derived from timelapse<br />

DIC images showed a significant (p


<strong>SUNDAY</strong><br />

2058<br />

The F-BAR domain protein PACSIN2 associates to Rac1 and regulates cell spreading and<br />

migration.<br />

B-J. de Kreuk 1 , M. Ne<strong>the</strong> 1,2 , M. Fernandez-Borja 1 , E. Anthony 1 , P. Hensbergen 3 , A. Deelder 3 , M.<br />

Plomann 4 , P. Hordijk 1 ; 1 <strong>Molecular</strong> Cell <strong>Biology</strong>, Sanquin Research, University <strong>of</strong> Amsterdam,<br />

Amsterdam, Ne<strong>the</strong>rlands, 2 University <strong>of</strong> Virginia, Charlottesville, VA, 3 Leiden University Medical<br />

Center, Leiden, Ne<strong>the</strong>rlands,, 4 Institute for Biochemistry 2, University <strong>of</strong> Cologne, Germany<br />

The Rac1 GTPase controls cytoskeletal dynamics and is a key regulator <strong>of</strong> cell spreading and<br />

migration via signaling through effector proteins such as <strong>the</strong> PAK kinases and <strong>the</strong> Scar/WAVE<br />

proteins. We previously identified a series <strong>of</strong> regulatory proteins that associate with Rac1<br />

through its hypervariable C-terminal domain. These include <strong>the</strong> Rac1 activator β-Pix, which<br />

recruits Rac1 to focal adhesions, and <strong>the</strong> membrane adapter Caveolin1, which regulates <strong>the</strong><br />

poly-ubiquitylation and degradation <strong>of</strong> activated Rac1. Here, we show that Rac1 associates with<br />

<strong>the</strong> F-BAR domain protein PACSIN2, an inducer <strong>of</strong> membrane tubulation and a regulator <strong>of</strong><br />

endocytosis. We show that loss <strong>of</strong> PACSIN2 increases Rac1-GTP loading and promotes cell<br />

spreading and –migration. Conversely, expression <strong>of</strong> PACSIN2 reduces Rac1-GTP levels in a<br />

fashion which is dependent on <strong>the</strong> PACSIN2-Rac1 interaction, on <strong>the</strong> membrane-tubulating<br />

capacity <strong>of</strong> PACSIN2, and on Dynamin. In addition, we show that GEF-mediated Rac1-GTP<br />

loading is unaffected when PACSIN2 is expressed. These results demonstrate that PACSIN2 is<br />

an important regulator <strong>of</strong> <strong>the</strong> small GTPase Rac1 and that PACSIN2 limits Rac1-GTP signaling<br />

by promoting Rac1 inactivation.<br />

2059<br />

Control over acquisition <strong>of</strong> cell motility and cell migration by <strong>the</strong> RNA binding protein<br />

Dead end.<br />

M. Goudarzi 1 , T. U. Banisch 1 , E. Raz 1 ; 1 Cell <strong>Biology</strong>, ZMBE, Muenster, Germany<br />

Specification <strong>of</strong> zebrafish primordial germ cells (PGCs) depends on maternally inherited<br />

determinants. These factors, collectively termed <strong>the</strong> germ-plasm direct cells to enter <strong>the</strong><br />

germline fate and to acquire proper cell behavior. An important such maternally-provided germplasm<br />

component in zebrafish is encoded by <strong>the</strong> dead end (dnd) gene. The Dnd protein<br />

contains an RNA recognition motif and is specifically expressed in PGCs, where it is localized to<br />

<strong>the</strong> perinuclear granules. Knockdown <strong>of</strong> dead end results in severe defects in cell behavior (e.g.<br />

<strong>the</strong> acquisition <strong>of</strong> motility), cell fate maintenance and survival.<br />

We could previously show that Dnd functions by protecting specific mRNAs (e.g. tdrd7, nanos1<br />

and hub) from micro RNA–mediated translational inhibition and degradation in <strong>the</strong> germ cells, a<br />

process by which <strong>the</strong> same RNAs are repressed and degraded in somatic cells. To determine<br />

<strong>the</strong> molecular basis <strong>of</strong> <strong>the</strong> dramatic changes in PGC properties upon loss <strong>of</strong> Dnd, we identified<br />

RNAs regulated by miRNAs and <strong>the</strong> Dnd protein. The functional role <strong>the</strong> proteins encoded by<br />

<strong>the</strong>se mRNAs play in controlling cell shape and behavior will be discussed.<br />

2060<br />

Nonmuscle Myosin II Is<strong>of</strong>orms regulate microtubule dynamics in three-dimensional cell<br />

migration through <strong>the</strong> distinct MLC phosphorylations.<br />

S. Komatsu 1 , M. Ikebe 2 ; 1 Microbiology and Physiological Systems, UMass Medical School,<br />

Worcester, MA, 2 UMass Medical School, Worcester, MA<br />

Cell migration is an extremely complex process which is controlled by <strong>the</strong> rearrangement <strong>of</strong><br />

cytoskeletal systems. Most studies <strong>of</strong> cell migration have been focused on controlling <strong>the</strong><br />

actomyosin and microtubule networks, and studied a migrating cell on two-dimensional (2D)


<strong>SUNDAY</strong><br />

surfaces such as plastic or glass. However, cells under physiological environment interact with<br />

extracellular matrices on all surfaces, not just on <strong>the</strong> basal surface. This raises an idea that cells<br />

showing rounded spindle shape in 3D matrices migrate with <strong>the</strong> different mechanism from flatshaped<br />

cells in 2D surface. To address this question, we use a 3D migration in collagen gel as<br />

an in vitro model system for identifying <strong>the</strong> mechanisms that regulate cell migration under<br />

physiological conditions.<br />

First, we observed distribution <strong>of</strong> nonmuscle myosin IIA (NMIIA) and IIB (NMIIB) is<strong>of</strong>orms in 3D<br />

migrating cells by using NMII is<strong>of</strong>orms specific antibodies and fluorescent protein-fusion NMII<br />

is<strong>of</strong>orms, mCherry-NMIIA and GFP-NMIIB. Although fluorescent signals <strong>of</strong> <strong>the</strong> NMIIA and <strong>the</strong><br />

NMIIB were observed at both anterior and posterior regions in <strong>the</strong> 3D migrating cells, <strong>the</strong><br />

detailed distribution <strong>of</strong> both is<strong>of</strong>orms were different from each o<strong>the</strong>r. The strong signals <strong>of</strong> NMIIA<br />

were particularly detected at <strong>the</strong> tips <strong>of</strong> cell extensions, while <strong>the</strong> signals <strong>of</strong> NMIIB were<br />

accumulated at <strong>the</strong> cortex around <strong>the</strong> spindle-shaped cell body. Inhibition <strong>of</strong> ei<strong>the</strong>r actomyosin<br />

or microtubule networks by blebbistatin or nocodazole resulted in a decrease in 3D cell<br />

migration. Is<strong>of</strong>orm-specific knockdowns by recombinant adenovirus siRNA for NMIIA and NMIIB<br />

reveal that NMII is<strong>of</strong>orms are respectively involved in <strong>the</strong> regulation <strong>of</strong> <strong>the</strong> microtubule dynamics<br />

in <strong>the</strong> 3D migrating cells. We also studied <strong>the</strong> localization <strong>of</strong> myosin II is<strong>of</strong>orms phosphorylated<br />

at functionally distinct sites <strong>of</strong> myosin light chain (MLC) in <strong>the</strong> 3D migrating cells. Interestingly,<br />

our results showed that NMIIA phosphorylated at Ser19 localized at <strong>the</strong> anterior tip <strong>of</strong> cell<br />

protrusion, while NMIIB phosphorylated at Ser1 localized at posterior region <strong>of</strong> <strong>the</strong> protrusion.<br />

On <strong>the</strong> o<strong>the</strong>r hand, di-phosphorylated myosin II at Thr18 and Ser19 colocalized with cortical<br />

actin structure.<br />

Our results suggest that <strong>the</strong> functionally distinct MLC phosphorylation plays an important role in<br />

<strong>the</strong> function <strong>of</strong> NMII is<strong>of</strong>orms in 3D cell migration. Fur<strong>the</strong>r studies are in progress to elucidate<br />

<strong>the</strong> role <strong>of</strong> <strong>the</strong> functionally distinct MLC phosphorylation in <strong>the</strong> 3D cell migration.<br />

2061<br />

Wound healing: The combination use <strong>of</strong> regenerative agents and bioavailable antioxidant<br />

constituents <strong>of</strong> vitamin E and vitamin D in a model in vitro system.<br />

A. Bettica 1 , N. Christensen 2 , T. Ragasha 2 ; 1 <strong>Biology</strong>, Manhattanville College, Purchase, NY,<br />

2 Manhattanville College, Purchase, NY<br />

Wound healing models have fostered a greater understanding <strong>of</strong> <strong>the</strong> cell types involved and <strong>the</strong><br />

many complex series <strong>of</strong> events from <strong>the</strong> site <strong>of</strong> trauma through <strong>the</strong> phases <strong>of</strong> inflammation,<br />

proliferation, and remodeling. Many in vitro studies have concentrated on <strong>the</strong> proliferation<br />

stages to elucidate agents that may aid migrating fibroblasts in producing growth factors, laying<br />

down fibers and ground substance, and attracting epi<strong>the</strong>lial cells to <strong>the</strong> site. Fat-soluble vitamins<br />

such as vitamin D3 and vitamin E are potent antioxidant and anti-inflammatory agents and have<br />

been shown to be effective in ameliorating dental and surgical wounds, burns, and scars. To<br />

determine <strong>the</strong> optimal effectiveness <strong>of</strong> <strong>the</strong>se properties, appropriate formulations <strong>of</strong> <strong>the</strong> isomeric<br />

constituents must be tested. The combination <strong>of</strong> <strong>the</strong>se vitamin species may show additive<br />

effects and enhanced efficacy under in vitro wound healing conditions. In dental wound healing,<br />

liners and sealers containing varying amounts <strong>of</strong> calcium hydroxide can trigger a cascade <strong>of</strong><br />

events which include increased cell proliferation, growth factor syn<strong>the</strong>sis, and cell differentiation.<br />

Skin cells, keratinocytes, and fibroblasts are susceptible to a range <strong>of</strong> UVA and UVB<br />

wavelengths that cause cellular and DNA damage, including <strong>the</strong> formation <strong>of</strong> thymine dimers.<br />

Avobenzone and zinc oxide (ZnO) provide true broad-spectrum protection against UVA<br />

wavelengths above 360 nm and are <strong>the</strong> only two sunscreen active ingredients approved in <strong>the</strong><br />

US. Studies suggest that proper formulation strategies are necessary for superior attenuation <strong>of</strong><br />

UVA wavelengths and may be achievable at 3% avobenzone or 5% ZnO. An in vitro wound<br />

healing model was standardized to test <strong>the</strong> combination <strong>of</strong> vitamin D3 and vitamin E isomers for


<strong>SUNDAY</strong><br />

additive effectiveness with proven or suggested regenerative agents. WS1 human skin<br />

fibroblasts are seeded at a density <strong>of</strong> 105 cells/well on a 6-well culture plate. Using a 4mm<br />

sterile biopsy punch, three wound areas are created in each well at confluency. WS1 cell<br />

wounds are exposed to 10-fold serial dilutions <strong>of</strong> calcium hydroxide, avobenzene, or ZnO in<br />

appropriate solvents for 24 hours in control wells, or before cells are exposed to UVA. Effective<br />

concentrations are selected from standard dose response curves for <strong>the</strong> various regenerative<br />

reagents and are used in combination with <strong>the</strong> vitamin D3 and E isomers. During <strong>the</strong> test<br />

periods, wound areas are digitally recorded beginning at day 0, for every 12 hours over a four<br />

day period. Measurements <strong>of</strong> <strong>the</strong> radius (rw - radius <strong>of</strong> <strong>the</strong> minimal diameter <strong>of</strong> <strong>the</strong> wound), <strong>the</strong><br />

wound area (Aw) are taken using Motic 2.0 s<strong>of</strong>tware and compared to day 0 (r0 and A0).<br />

Triplicate data sets are statistically analyzed using a one-way ANOVA with Tukey’s post hoc<br />

tests. Effective concentrations <strong>of</strong> calcium/ D3 and cholcalciferol alone are seen at 10-4 mg/ml.<br />

Vitamin E isomers did improve cell proliferation and overall wound healing. The most effective<br />

doses were 10-7 mg/ml for alpha-tocopherol and 10-8 mg/ml for delta-tocotrienol, showing<br />

complete wound healing between 48 and 60 hours. It is suggested that agents with greater<br />

antioxidant activities may shorten healing times, reduce <strong>the</strong> incidence <strong>of</strong> wound infection and, in<br />

<strong>the</strong> most serious wound cases, lower <strong>the</strong> mortality rate in burn patients.<br />

2062<br />

Arp 2/3 Plays a Crucial Role in Focal Adhesion Initiation at <strong>the</strong> Leading Edge.<br />

R. J. Vasquez 1 , K. Sayegh 2 , J. Stricker 3 , Y. Beckham 3 , M. Gardel 3 ; 1 Department <strong>of</strong> Pediatrics,<br />

Section <strong>of</strong> Hematology, Oncology and Stem Cell Transplantation, University <strong>of</strong> Chicago,<br />

Chicago, IL, 2 University <strong>of</strong> Chicago, Chicago, IL, 3 Department <strong>of</strong> Physics, University <strong>of</strong> Chicago,<br />

Chicago, IL<br />

The lamellipodia is a dense, Arp-2/3 mediated actin network at <strong>the</strong> leading edge <strong>of</strong> migrating<br />

cells that generates forces sufficient to generate sheet-like protrusions <strong>of</strong> <strong>the</strong> cell edge. In<br />

addition to this force generating role, <strong>the</strong> initiation <strong>of</strong> focal adhesions also occurs within <strong>the</strong><br />

lamellipodia. However, <strong>the</strong> role <strong>of</strong> lamellipodial actin in focal adhesion initiation is not well<br />

understood. We used a recently identified inhibitor <strong>of</strong> <strong>the</strong> ARP 2/3 complex, CK 869, to<br />

investigate <strong>the</strong> role <strong>of</strong> <strong>the</strong> lamellipodia in leading edge protrusion and focal adhesion formation<br />

in cell motility and adhesion in two cell types- MCF10A cells (a human breast epi<strong>the</strong>lial cell line)<br />

and U20S cells (a human osteosarcoma cell line). We find that treatment with CK 869 disrupts<br />

or inhibits <strong>the</strong> formation <strong>of</strong> an organized lamellipod, as demonstrated by phalloidin staining and<br />

immun<strong>of</strong>luoresce with cortactin, a protein enriched in <strong>the</strong> lamellipod. Motility assays <strong>of</strong> single<br />

cells and scratch assays with MCF-10A cells demonstrate that treatment with CK-869 reduced<br />

<strong>the</strong> number <strong>of</strong> motile cells and reduced <strong>the</strong> motility rate, and in <strong>the</strong> case <strong>of</strong> <strong>the</strong> scratch assay,<br />

greatly increased <strong>the</strong> time to closure <strong>of</strong> <strong>the</strong> cell sheet. Despite <strong>the</strong>se effects on cell migration,<br />

protrusions <strong>of</strong> <strong>the</strong> cell edge persist, suggesting that ARP 2/3 mediated actin assembly is not <strong>the</strong><br />

only mechanism through which to generate sheet-like membrane protrusions. We hypo<strong>the</strong>sized<br />

that <strong>the</strong>se membrane protrusions might differ from lamellipodial protrusions in <strong>the</strong>ir ability to<br />

form new focal adhesions. Immuno-staining <strong>of</strong> <strong>the</strong> focal adhesion proteins paxillin and vinculin<br />

demonstrated that <strong>the</strong> ARP 2/3 inhibited cells had fewer but larger peripheral adhesions and<br />

fewer central focal adhesions. Live cell imaging with GFP-paxillin in U2OS cells revealed that<br />

<strong>the</strong> protrusions formed in <strong>the</strong> presence <strong>of</strong> <strong>the</strong> ARP-inhibitor revealed reduced focal adhesion<br />

assembly. Collectively, <strong>the</strong>se results indicate that ARP 2/3 is not necessary for sheet-like<br />

protrusions <strong>of</strong> <strong>the</strong> cell edge but plays a crucial role in focal adhesion formation.


2063<br />

Quantitative Analysis <strong>of</strong> Protein Binding Dynamics in Live Cells with Photoswitchable<br />

Fluorescent Tags.<br />

M. Rubashkin 1 , M. J. Paszek 1 , C. C. Dufort 1 , P. Kumar 2 , T. Wittmann 2 , V. M. Weaver 1,3 ;<br />

1 Surgery and Center for Bioengineering and Tissue Regeneration, University <strong>of</strong> California at<br />

San Francisco, San Francisco, CA, 2 Cell and Tissue <strong>Biology</strong>, University <strong>of</strong> California at San<br />

Francisco, San Francisco, CA, 3 Anatomy, Bioengineering and Therapeutic Sciences, Eli and<br />

Edy<strong>the</strong> Broad Center <strong>of</strong> Regeneration Med., University <strong>of</strong> California at San Francisco, San<br />

Francisco, CA<br />

<strong>SUNDAY</strong><br />

The kinetics <strong>of</strong> individual proteins can dictate focal adhesion dynamics and cellular behaviors<br />

such as polarity, motility and migration. We developed a new quantitative fluorescence<br />

microscopy technique, RAPS (Regression After Photo-Switching), to study <strong>the</strong> binding state and<br />

kinetics <strong>of</strong> proteins in live cells. In brief, RAPS involves tagging a protein with a photoswitchable<br />

fluorescent protein, such as mEOS2. Initially, an image is taken <strong>of</strong> <strong>the</strong> protein in <strong>the</strong> unswitched<br />

green state, 488nm, to identify focal adhesions <strong>of</strong> interest. Then, a digital micro-mirror device<br />

concurrently illuminates all adhesions <strong>of</strong> interest at <strong>the</strong> activation wavelength, 405nm,<br />

converting a subpopulation <strong>of</strong> <strong>the</strong> protein to a stable red fluorescent state. The cell is imaged in<br />

<strong>the</strong> converted state, 568nm, to measure <strong>the</strong> dynamics <strong>of</strong> <strong>the</strong> protein. Lastly, kinetic modeling<br />

analysis is implemented to determine <strong>the</strong> K<strong>of</strong>f and mobile fraction <strong>of</strong> <strong>the</strong> protein. In comparison<br />

to existing numerical fluorescence photobleaching techniques, RAPS reduces <strong>the</strong> time and<br />

intensity <strong>of</strong> light needed for activation, enables imaging in a secondary wavelength to increase<br />

contrast and minimize noise, permits diffusion kinetics to be ignored in analysis, and facilitates<br />

simultaneous investigation <strong>of</strong> multiple cell structures within a single cell. In order to verify that<br />

RAPS can be used to quantify changes in protein binding kinetics, we studied focal adhesion<br />

proteins in MCF10a breast cancer epi<strong>the</strong>lial cells. In <strong>the</strong> study <strong>of</strong> talin, we observed a K<strong>of</strong>f rate <strong>of</strong><br />

0.024 ± 0.007 (1/s) and a bound protein fraction <strong>of</strong> 63 ± 12%. Fur<strong>the</strong>rmore, we detected that <strong>the</strong><br />

mobile fraction <strong>of</strong> talin significantly increased after inducing expression <strong>of</strong> <strong>the</strong> glycoprotein<br />

Mucin1, a cancer oncogene associated with changes in cell motility.<br />

Development and Morphogenesis I<br />

2064<br />

Expression <strong>of</strong> new growth factor <strong>of</strong> <strong>the</strong> VEGF family in sea urchin development.<br />

Y. O. Kipryushina 1,2 , K. V. Yakovlev 1 , N. A. Odintsova 1,2 ; 1 A.V. Zhirmunsky Institute <strong>of</strong> Marine<br />

<strong>Biology</strong>, Far Eastern Branch <strong>of</strong> <strong>the</strong> Russian Academy <strong>of</strong> Sciences, Vladivostok, Russia, 2 Far<br />

Eastern Federal University, Vladivostok, Russia<br />

Growth factors regulate many processes, including cell proliferation, differentiation, migration<br />

and apoptosis, and play an important role in embryonic development. Among <strong>the</strong>m vascular<br />

endo<strong>the</strong>lial growth factors (VEGF), <strong>of</strong> <strong>the</strong> VEGF/PDGF superfamily, are important signaling<br />

proteins involved in induction <strong>of</strong> angiogenesis and playing a central role in <strong>the</strong> regulation <strong>of</strong><br />

vasculogenesis in mammals. In invertebrates, such as insects and nematodes, VEGF/PDGF<br />

superfamily members guide cell migration and cell differentiation during development. The aim<br />

<strong>of</strong> our study is to investigate <strong>the</strong> role <strong>of</strong> VEGFs during development <strong>of</strong> <strong>the</strong> sea urchin<br />

Strongylocentrotus intermedius. Three Sp-Vegf genes have been predicted in <strong>the</strong> genome <strong>of</strong> a<br />

closely related sea urchin S. purpuratus. One <strong>of</strong> <strong>the</strong>m, Sp-Vegf3, has been reported to play an<br />

essential role in sea urchin development (Duloquin et al., 2007). In this study, we investigated<br />

<strong>the</strong> expression <strong>of</strong> a new gene <strong>of</strong> <strong>the</strong> VEGF family, Si-Vegf2, and expression <strong>of</strong> <strong>the</strong> Si-<br />

Pdgfr/vegfrL gene in sea urchin development by RT-PCR. We demonstrated a high similarity <strong>of</strong>


<strong>SUNDAY</strong><br />

<strong>the</strong> central part <strong>of</strong> <strong>the</strong> new gene, which we found in <strong>the</strong> sea urchin S. intermedius, with that <strong>of</strong><br />

<strong>the</strong> predicted gene Sp-Vegf2 in S. purpuratus. The Si-Vegf2 was found to be expressed from<br />

<strong>the</strong> zygote stage, whereas Si-Pdgfr/vegfrL expression began later at <strong>the</strong> blastula stage. Our<br />

data have shown that a nonspecific inhibitor <strong>of</strong> heparine-binding GF receptors, suramin, results<br />

in significant disturbances in embryonic development: archenteron formation is suppressed, and<br />

spicule growth is inhibited. An addition <strong>of</strong> human PDGF-AB to embryonic culture leads to partial<br />

recovery <strong>of</strong> archenteron growth. These results match <strong>the</strong> findings on ano<strong>the</strong>r sea urchin species<br />

(Katow and Aizu, 2002). In summary, we have discovered <strong>the</strong> expression <strong>of</strong> a new VEGF family<br />

member in sea urchin development and shown that temporal expression pr<strong>of</strong>iles <strong>of</strong> <strong>the</strong> Si-Vegf2<br />

and Si-Pdgfr/vegfrL are not similar in sea urchin development, but signaling through<br />

VEGF/PDGF pathway is essential for embryonic development. (This work was supported by<br />

Presidium <strong>of</strong> <strong>the</strong> FEB RAS (grants 09-I-P22-04, 09-II-SB-06-001) and Program at <strong>the</strong> Far<br />

Eastern Federal University (grant 11 G34.31.0010)).<br />

2065<br />

How “acellular” embryos gastrulate: Investigation <strong>of</strong> cytoplasm movements during<br />

gastrulation in developing Drosophila melanogaster embryos.<br />

B. He 1 , O. Polyakov 2 , E. Wieschaus 1,3 ; 1 Department <strong>of</strong> <strong>Molecular</strong> <strong>Biology</strong>, Princeton University,<br />

Princeton, NJ, 2 Department <strong>of</strong> Physics, Princeton University, Princeton, NJ, 3 Howard Hughes<br />

Medical Institute<br />

During Drosophila gastrulation, <strong>the</strong> mesoderm precursor cells are internalized from <strong>the</strong> ventral<br />

surface <strong>of</strong> <strong>the</strong> embryo and form a ventral furrow. The invagination <strong>of</strong> <strong>the</strong>se cells is accompanied<br />

by apical constriction, a process driven by cycles <strong>of</strong> pulsed contraction and stabilization <strong>of</strong> <strong>the</strong><br />

actin-myosin network underneath <strong>the</strong> apical membrane. Previous studies have shown that in<br />

response to individual constriction pulses, <strong>the</strong> ventral cells elongate along <strong>the</strong>ir apical-basal axis<br />

and <strong>the</strong>ir nuclei move basally in an incremental manner. However, it is unclear how contractile<br />

forces constrained to <strong>the</strong> vicinity <strong>of</strong> <strong>the</strong> apical surface drives cell shape change along <strong>the</strong> entire<br />

cell length. We recently identified a cellularization mutant in which <strong>the</strong> periplasm <strong>of</strong> <strong>the</strong> syncytial<br />

blastoderm is never partitioned into individual cells. Intriguingly, <strong>the</strong>se “acellular” embryos still<br />

gastrulate, and exhibit initial gastrulation movements largely reminiscent <strong>of</strong> those in normal<br />

cellular embryos. In particular, a contractile actin-myosin network forms on <strong>the</strong> ventral cortex<br />

and drives apical constriction, which leads to formation <strong>of</strong> a pseudo-ventral-furrow. The<br />

observation suggests that <strong>the</strong> cytoplasm can respond to cellular forces and undergo<br />

morphogenetic movements even in <strong>the</strong> absence <strong>of</strong> cell membrane. To test this, we investigated<br />

<strong>the</strong> micromechanical property <strong>of</strong> <strong>the</strong> cytoplasm and its response to constriction pulses by<br />

tracking injected fluorescent polystyrene microspheres. Constriction pulses were identified by<br />

simultaneously imaging a GFP-tagged membrane marker or myosin subunit. We found that <strong>the</strong><br />

periplasm in both cellular and acellular embryos was highly viscous (~3 orders magnitude<br />

higher than water) but on <strong>the</strong> o<strong>the</strong>r hand did not show measurable elasticity over<br />

developmentally relevant timescales. In <strong>the</strong> cellular embryos, upon gastrulation, beads within<br />

<strong>the</strong> ventral cells moved basally in an incremental manner that largely correlated with pulsed<br />

constriction. Interestingly, beads initially localized to <strong>the</strong> apical region above <strong>the</strong> nucleus were<br />

never observed to flow past <strong>the</strong> nucleus upon apical constriction. Fur<strong>the</strong>rmore, <strong>the</strong> distance<br />

between beads localized in <strong>the</strong> same cell remained correlated during cell leng<strong>the</strong>ning and did<br />

not change abruptly over time, as if <strong>the</strong> cytoplasm was a gel that underwent gradual<br />

deformation under force. Strikingly, this behavior does not appear to rely on <strong>the</strong> presence <strong>of</strong> cell<br />

membrane, as in <strong>the</strong> acellular embryos <strong>the</strong> beads within <strong>the</strong> ventral periplasm display similar<br />

global movements as in <strong>the</strong> cellular embryos. Based on <strong>the</strong>se observations we propose that<br />

during Drosophila ventral furrow formation <strong>the</strong> cytoplasm responds to cellular forces in a more<br />

“gel-like”, ra<strong>the</strong>r than a “liquid-like”, manner. This “gel-like” property would allow for an


<strong>SUNDAY</strong><br />

instantaneous transmittance <strong>of</strong> apically generated force throughout <strong>the</strong> entire tissue to drive cell<br />

shape change and <strong>the</strong>ir corresponding invagination. Our study also implicates that cellular<br />

components that determine <strong>the</strong> viscoelastic property <strong>of</strong> <strong>the</strong> cytoplasm are potential targets <strong>of</strong><br />

regulation during tissue morphogenesis.<br />

2066<br />

Probing tension and dynamics in actomyosin mediated cell shape change.<br />

C. D. Higgins 1 , U. S. Tulu 2 , L. Gao 3 , E. Betzig 3 , D. P. Kiehart 2 , B. Goldstein 1 ; 1 <strong>Biology</strong>, UNC<br />

Chapel Hill, Chapel Hill, NC, 2 <strong>Biology</strong>, Duke University, Durham, NC, 3 Janelia Farm Research<br />

Campus, Howard Hughes Medical Institute, Ashburn, VA<br />

Apical constriction is a cell shape change critical for morphogenesis in diverse organisms.<br />

Apical constriction requires <strong>the</strong> activity <strong>of</strong> a dynamic, contractile actomyosin meshwork that is<br />

enriched at <strong>the</strong> apical cortex <strong>of</strong> <strong>the</strong> cell. In <strong>the</strong> 26-cell C. elegans embryo, two cells apically<br />

constrict and internalize. We have found previously that contractility in <strong>the</strong> apical actomyosin<br />

meshwork <strong>of</strong> <strong>the</strong>se cells is initially uncoupled from apical constriction, and embryos deficient for<br />

RacGEF ced-5 and classical cadherin hmr-1 display normal myosin dynamics that fail to drive<br />

apical constriction. These findings suggest a model where <strong>the</strong> contractile actomyosin meshwork<br />

must become coupled to apical cell-cell contacts to drive cell shape change. Here, we use<br />

particle image velocimetry (PIV) to map <strong>the</strong> movements <strong>of</strong> <strong>the</strong> actomyosin meshwork and cellcell<br />

contacts over time. An unbiased PIV algorithm confirms our manual measurements<br />

indicating that actomyosin contractility precedes cell shape change. Fur<strong>the</strong>rmore, we estimate<br />

<strong>the</strong> tensile properties <strong>of</strong> <strong>the</strong> meshwork by cortical laser ablation and show that contractile forces<br />

are established prior to cell shape change. We also show that tension in <strong>the</strong> apical meshwork is<br />

higher in apically constricting cells than neighboring cells. To examine actomyosin dynamics<br />

and cell shape change with improved axial resolution, we have also taken advantage <strong>of</strong> Bessel<br />

beam structured plane illumination microscopy. Toge<strong>the</strong>r, our results suggest that both apical<br />

actomyosin contractility and coupling to cell-cell contacts are required to facilitate cell shape<br />

change.<br />

2067<br />

Imaging Assays for High-throughput Quantification <strong>of</strong> Synaptic Formation and Neurite<br />

Dynamics in Human iPSC-derived Neurons.<br />

J. A. Hesley 1 , L. Chase 2 , M. Strathman 2 , O. Sirenko 1 , P. Tavormina 1 ; 1 <strong>Molecular</strong> Devices, LLC,<br />

Sunnyvale, CA, 2 Cellular Dynamics International, Inc., Madison, WI<br />

Central nervous system (CNS) disorders affect millions <strong>of</strong> people worldwide. Considerable effort<br />

has been made in an attempt to develop relevant in vitro human model systems for studying<br />

degenerative (i.e. Alzheimer’s and Parkinson’s disease) and genetic (i.e. Huntington's disease<br />

and muscular dystrophy) disorders as well as neurotoxicity. Induced pluripotent stem cells<br />

(iPSCs) <strong>of</strong> human origin provide an ideal model for scientists both within <strong>the</strong> academic and<br />

pharmaceutical settings. By pairing <strong>the</strong>se cells with an automated high content imaging system,<br />

endpoint and live-cell assays can be used to determine whe<strong>the</strong>r pharmaceutical drug<br />

candidates or environmental contaminants exert neurotrophic, neuroprotective, or neurotoxic<br />

effects. High content imaging <strong>of</strong> neurons allows scientists to both characterize and measure<br />

changes in neuronal networks such as neurite number, length, and branching, as well as to<br />

quantitate synaptogenesis.<br />

iPSC-derived iCell® Neurons were treated with neurite modulating compounds at varying doses<br />

and images were acquired over time using <strong>the</strong> ImageXpress® Micro High Content System.<br />

These experiments were done using time-lapse imaging <strong>of</strong> live cells with transmitted light or<br />

fluorescence as well as using immun<strong>of</strong>luorescence staining <strong>of</strong> fixed cells with <strong>the</strong> neuronal


<strong>SUNDAY</strong><br />

marker class III beta tubulin (TuJ1). Images <strong>of</strong> <strong>the</strong> neural networks were analyzed with<br />

MetaXpress® S<strong>of</strong>tware to identify numerous aspects <strong>of</strong> <strong>the</strong> cells and <strong>the</strong>ir outgrowths. The data<br />

shows over a doubling <strong>of</strong> neurite outgrowths over a 12 hour period without stimulation and a<br />

clear response to drug doses as well. In addition, emergence <strong>of</strong> GABAergic and glutamatergic<br />

synaptic phenotypes was seen by tracking <strong>the</strong> number <strong>of</strong> vGAT+ and vGLUT2+ puncta within a<br />

1-3 week post-thaw timecourse.<br />

These results demonstrate that human iPSC-derived neurons paired with high content imaging<br />

techniques provide valuable information for assessing <strong>the</strong> effects <strong>of</strong> compounds on neuronal<br />

networks and can be applied to various academic and pharmaceutical applications.<br />

2068<br />

Growth Hormone Regulates Survival <strong>of</strong> Primary Cultures <strong>of</strong> Embryonic Neural Precursor<br />

Cells.<br />

C. Regalado Santiago 1 , M. L. Lopez Meraz 2 , E. Juarez Aguilar 3 ; 1 Doctorado en Ciencias<br />

Biomedicas, Instituto de Ciencias de la Salud Universidad Veracruzana, Xalapa, Mexico,<br />

2 Centro de Investigaciones Cerebrales Universidad Veracruzana, Xalapa, Mexico,<br />

3 Departamendo de Biomedicina, Instituto de Ciencias de la Salud Universidad Veracruzana,<br />

Xalapa, Mexico<br />

During central nervous system development, cellular proliferation and cellular survival play a<br />

critical role. It has been suggested that <strong>the</strong>se biological processes are regulated in an<br />

orchestrated way by numerous neurotrophic factors. In addition to <strong>the</strong> well known effects <strong>of</strong><br />

growth hormone (GH) on growth and metabolism regulation, <strong>the</strong>re is a growing body <strong>of</strong><br />

evidence that suggest a central role <strong>of</strong> this hormone in <strong>the</strong> development and function <strong>of</strong> <strong>the</strong><br />

brain. The presence <strong>of</strong> <strong>the</strong> GH and its receptor has been widely reported in different regions <strong>of</strong><br />

<strong>the</strong> brain. In spite <strong>of</strong> <strong>the</strong>se findings, <strong>the</strong> biological role <strong>of</strong> GH in <strong>the</strong> CNS is not completely<br />

understood. In order to analyze <strong>the</strong> effect <strong>of</strong> GH during CNS development, we performed<br />

studies on primary cultures <strong>of</strong> embryonic (~E14) neural precursor cells from striatum. In<br />

presence <strong>of</strong> <strong>the</strong> epidermal growth factor (EGF), this population gives rise to neurospheres <strong>of</strong><br />

undifferentiated cells that can generate neurons, oligodendrocytes and glia. In a first approach,<br />

we showed that GH was unable to stimulate <strong>the</strong> formation <strong>of</strong> neurospheres in absence <strong>of</strong> EGF.<br />

Secondly, we incubated primary cultures <strong>of</strong> striatum with different concentrations <strong>of</strong> GH (1, 10,<br />

50, 100, 500 and 1000 ng/ml) in presence <strong>of</strong> EGF and determined <strong>the</strong> total number <strong>of</strong><br />

neurospheres produced. The results showed a typical “bell shaped” curve with an increase<br />

since 1 ng/ml <strong>of</strong> GH and a maximal response at 10 ng/ml <strong>of</strong> hormone. In order to discriminate<br />

whe<strong>the</strong>r <strong>the</strong> increase in <strong>the</strong> number <strong>of</strong> neurospheres stimulated by GH was due to a mitogenic<br />

effect, primary cultures <strong>of</strong> striatum were incubated with different doses <strong>of</strong> GH and <strong>the</strong> analogue<br />

<strong>of</strong> thymidine BrdU (10 µM), during 24 hours. Counting <strong>of</strong> BrdU positive cells showed no<br />

difference between control and cultures treated with different doses <strong>of</strong> GH (10 ng/ml - 100<br />

ng/ml), while this hormone inhibited BrdU incorporation at doses <strong>of</strong> 500 ng/ml and 1000 ng/ml.<br />

Interestingly, GH stimulated a proliferative effect on primary cultures <strong>of</strong> striatum in absence <strong>of</strong><br />

EGF. Next, we evaluated if GH has a neuroprotective action on <strong>the</strong> primary cultures using an<br />

apoptosis assay (TUNEL). Results showed a decrease in <strong>the</strong> apoptosis rate on cultures treated<br />

with GH at 10 ng/ml. Altoge<strong>the</strong>r, <strong>the</strong>se results suggest that GH could be considered a survival<br />

factor ra<strong>the</strong>r than a mitogenic factor during embryonic development <strong>of</strong> <strong>the</strong> CNS at <strong>the</strong> E14<br />

stage.


<strong>SUNDAY</strong><br />

2069<br />

Endo<strong>the</strong>lium effects on <strong>the</strong> differentiation <strong>of</strong> iPSCs to insulin-producing cells.<br />

D. Talavera 1 , S. Kurtovic 1 , D. C. Dafoe 1 ; 1 Regenerative Medicine Institute, Cedars-Sinai Medical<br />

Center, Los Angeles, CA<br />

Formation <strong>of</strong> fully differentiated insulin-producing beta cells depends <strong>of</strong> <strong>the</strong> interaction between<br />

endo<strong>the</strong>lial cells (ECs) and pre-patterned endodermal cells in vivo. It has been described that<br />

endo<strong>the</strong>lial-derived bone morphogenetic proteins (BMPs) play an important role in this<br />

differentiation process. However, such EC signals are not fully characterized. Therefore, our<br />

objective is to study <strong>the</strong> effects <strong>of</strong> ECs in <strong>the</strong> differentiation <strong>of</strong> induced pluripotent stem cells<br />

(iPSCs) to insulin-producing cells and <strong>the</strong> role <strong>of</strong> EC-BMPs in this process. To accomplish this<br />

objective, we generated human embryoid bodies (EBs) from iPSCs. Different groups <strong>of</strong> EBs<br />

were cultured in a three-dimensional system <strong>of</strong> collagen-laminin gels that allowed EB-EC<br />

interaction for longer periods. After four weeks, <strong>the</strong> differentiated cells from EBs were analyzed<br />

by immunocytochemistry (ICC), immunohistochemistry (IHC), qRT-PCR, and Western blot (WB)<br />

looking for <strong>the</strong> expression <strong>of</strong> specific beta-cell, progenitor-cell, and islet-cell markers. To study<br />

<strong>the</strong> role <strong>of</strong> BMP-2/-4 and BMPR1A in <strong>the</strong> differentiation process, expression <strong>of</strong> BMP-2/-4, and<br />

BMPR1A as well as co-expression <strong>of</strong> C-peptide and phosphorylated Smad1/5/8 (pSmad1/5/8)<br />

were analyzed in all EB groups. Our results showed a significant increase in <strong>the</strong> expression <strong>of</strong><br />

specific beta-cell markers (insulin, PDX-1, GLUT2, Nkx6.1, Kir6.2, SUR1, GKS, PC1/3, and<br />

PC2), progenitor-cell markers (Isl-1, Ngn3, Pax4, Pax6, Nkx2.2, Nkx6.1, and Ptf1a), and isletcell<br />

markers (GCG and STT) in those EBs co-cultured with ECs in comparison to EBs cultured<br />

alone. Islet-like structures composed <strong>of</strong> cells that co-expressed C-peptide and PDX-1 or Cpeptide<br />

and pSmad1/5/8 were identified exclusively in co-cultured EBs. In addition, higher<br />

expression <strong>of</strong> BMP-2, BMP-4, and BMPR1A was found in <strong>the</strong>se EBs in contrast to EBs cultured<br />

alone. Our results indicate that ECs promote <strong>the</strong> differentiation <strong>of</strong> iPSCs to insulin-producing<br />

cells and that <strong>the</strong> BMP pathway activation plays a key role in this process.<br />

2070<br />

Myogenic progenitors regulating skeletal muscle homeostasis through Bmpr1a<br />

signaling.<br />

P. Huang 1 , T. J. Schulz 2 , A. Beauvais 1 , Y-H. Tseng 2 , E. Gussoni 1 ; 1 Children's Hospital Boston,<br />

Harvard Medical School, Boston, MA, 2 Joslin Diabetes Center, Harvard Medical School, Boston,<br />

MA<br />

Skeletal muscle maintenance and regeneration require finely coordinated interaction <strong>of</strong> multiple<br />

cell types. The molecular mechanism regulating this coordination is not clear. Bmp-signaling is<br />

suggested to balance proliferation and differentiation <strong>of</strong> myogenic cells in vitro. Here, we ablated<br />

expression <strong>of</strong> Bmpr1a in Myf5+ and MyoD+ progenitors in vivo (Myf5+/cre:Bmpr1af/f and<br />

MyoD+/cre:Bmpr1af/f ). Mutant Myf5+/cre:Bmpr1af/f mice were born runted and remained<br />

significantly smaller in size throughout life. Ablation <strong>of</strong> Bmpr1a resulted in increased fat<br />

infiltration and smaller regenerated my<strong>of</strong>ibers in acutely injured Myf5+/cre:Bmpr1af/f and<br />

MyoD+/cre:Bmpr1af/f muscles. Interstitial myogenic cells (IMCs, Sca-1posCD45neg<br />

CD31posPDGFRaneg) isolated from mutant mice proliferated more slowly as compared to wild<br />

type cells. Following culture and differentiation in vitro, Sca-1posCD45neg cells from wildtype<br />

mice gave rise to myogenic and adipogenic progenitors, whereas Sca-1posCD45neg cells from<br />

mutant mice generated, nearly exclusively, adipocytes. Ablation <strong>of</strong> Bmpr1a expression in<br />

Bmpr1a f/f Sca-1posCD45neg progenitors also resulted in adipocyte differentiation, as opposed<br />

to myogenic cells and adipocyte when Bmpr1a is expressed. Altoge<strong>the</strong>r, <strong>the</strong>se results show that<br />

Bmpr1a signaling is necessary for <strong>the</strong> commitment and proliferation <strong>of</strong> interstitial progenitors<br />

towards <strong>the</strong> myogenic lineage, and its presence might regulate intramuscular adipogenesis.


<strong>SUNDAY</strong><br />

2071<br />

Human mesenchymal stem cell differentiation to <strong>the</strong> osteogenic or adipogenic lineage is<br />

regulated by AMP-activated protein kinase.<br />

E-K. Kim 1 , S. Lim 1 , J-M. Park 1 , J. Seo 1 , J. Kim 2 , K. Kim 3 , S. Ryu 3 , P-G. Suh 1 ; 1 School <strong>of</strong> Nano-<br />

Bioscience & Chemical Engineering, UNIST, Ulsan, Korea, 2 Physiology, Pusan National<br />

University, Busan, Korea, 3 Division <strong>of</strong> <strong>Molecular</strong> and Life Sciences, POSTECH, Pohang, Korea<br />

AMP-activated protein kinase (AMPK) is an energy-sensing kinase that has recently been<br />

shown to regulate <strong>the</strong> differentiation <strong>of</strong> preadipocytes and osteoblasts. However, <strong>the</strong> role <strong>of</strong><br />

AMPK in stem cell differentiation is largely unknown. Using in vitro culture models, <strong>the</strong> present<br />

study demonstrates that AMPK is a critical regulatory factor for osteogenic differentiation. We<br />

observed that expression and phosphorylation <strong>of</strong> AMPK were increased during osteogenesis in<br />

human adipose tissue–derived mesenchymal stem cells (hAMSC). To elucidate <strong>the</strong> role <strong>of</strong><br />

AMPK in osteogenic differentiation, we investigated <strong>the</strong> effect <strong>of</strong> AMPK inhibition or knockdown<br />

on mineralization <strong>of</strong> hAMSC. Compound C, an AMPK inhibitor, reduced mineralized matrix<br />

deposition and suppressed <strong>the</strong> expression <strong>of</strong> osteoblast-specific genes, including alkaline<br />

phosphatase (ALP), runt-related transcription factor 2 (RUNX2), and osteocalcin (OCN).<br />

Knockdown <strong>of</strong> AMPK by shRNA-lentivirus infection also reduced osteogenesis. In addition,<br />

inhibition or knockdown <strong>of</strong> AMPK during osteogenesis inhibited ERK phosphorylation, which is<br />

required for osteogenesis. Interestingly, inhibition <strong>of</strong> AMPK induced adipogenic differentiation <strong>of</strong><br />

hAMSC, even in osteogenic induction medium (OIM). These results provide a potential<br />

mechanism involving AMPK activation in osteogenic differentiation <strong>of</strong> hAMSC and suggest that<br />

commitment <strong>of</strong> hAMSC to osteogenic or adipogenic lineage is governed by activation or<br />

inhibition <strong>of</strong> AMPK, respectively.<br />

2072<br />

Thymic nurse cells express signature markers consistent with epi<strong>the</strong>lial progenitor<br />

phenotype.<br />

R. V. Chilukuri 1 , V. K. Patel 1 , M. D. Samms 2 , J. C. Guyden 1 ; 1 <strong>Biology</strong>, The City College <strong>of</strong> New<br />

York, New York, NY, 2 The City College <strong>of</strong> New York, The City College <strong>of</strong> New York, New York,<br />

NY<br />

Thymic nurse cells (TNCs) represent a subset <strong>of</strong> thymic cortical epi<strong>the</strong>lial cell with <strong>the</strong> unique<br />

ability to entose o<strong>the</strong>r cell types. Our earlier studies demonstrated that TNCs bind and<br />

internalize immature thymocytes bearing <strong>the</strong> αβ TCR lo CD4 + CD8 + phenotype. Subsequent<br />

studies showed that <strong>the</strong> binding and internalization <strong>of</strong> thymocytes by TNCs is mediated by a<br />

TNC-specific antigen that is recognized by <strong>the</strong> monoclonal antibody, pH91. Internalized<br />

thymocyte subset received life or death signals during <strong>the</strong> process <strong>of</strong> MHC restriction, resulting<br />

in survival and release <strong>of</strong> some thymocytes or <strong>the</strong> induction <strong>of</strong> apoposis in <strong>the</strong> thymocyte subset<br />

that remained trapped within specialized TNC compartments. In this study, we examined <strong>the</strong><br />

ontogenesis <strong>of</strong> <strong>the</strong> TNC-specific antigen. Using confocal microscopic analyses, <strong>the</strong> onset<br />

expression <strong>of</strong> <strong>the</strong> pH91-specific antigen occurs as early as E11.5 stage <strong>of</strong> development and<br />

persists throughout embryological development as well as postnatally in mice. Fur<strong>the</strong>r analyses<br />

<strong>of</strong> PH91 + cells show <strong>the</strong> co-expression <strong>of</strong> K5, K8, Foxn1 and P63. At E17.5 stage <strong>of</strong><br />

development, we observe <strong>the</strong> formation <strong>of</strong> <strong>the</strong> multicellular, lympho-epi<strong>the</strong>lial complexes that<br />

exhibit <strong>the</strong> pH91 + K8 + K5Foxn1 + P63 + phenotype. These markers have been implicated in <strong>the</strong><br />

identification <strong>of</strong> thymic epi<strong>the</strong>lial progenitors elsewhere. Interestingly, in a previous report, we<br />

showed pH91 + K5 + K8 TNCs to be strategically localized within <strong>the</strong> cortico-medullary junction <strong>of</strong><br />

<strong>the</strong> thymus, thus possibly allowing for differentiation <strong>of</strong> K8 + cells to proceed toward cortex and<br />

K5 + cells to <strong>the</strong> medulla. By E18.5 stage <strong>of</strong> development, <strong>the</strong> pH91 + K8 + K5 + Foxn1 + P63 + TNCs<br />

express elevated levels <strong>of</strong> MHC-class II antigens on <strong>the</strong>ir cell surfaces. In addition, confocal


<strong>SUNDAY</strong><br />

analyses show that in post-natal mice 26% <strong>of</strong> pH91 + TNCs also express K8 and K5, whereas<br />

76% <strong>of</strong> <strong>the</strong> K8 + K5 + cells in <strong>the</strong> thymus are TNCs. Taken toge<strong>the</strong>r our data suggest that<br />

pH91 + K8 + K5 + Foxn1 + P63 + TNCs have thymic epi<strong>the</strong>lial progenitor potential.<br />

2073<br />

The Role <strong>of</strong> Endogenous FGF-2 in heterogeneity and pluripotency <strong>of</strong> human embryonic<br />

stem cells.<br />

K. Matulka 1 , L. Eiselleova 1 , M. Kunova 1 , A. Hampl 2,3 , Y-M. Sun 1 , P. Dvorak 1,3 ; 1 Department <strong>of</strong><br />

<strong>Biology</strong>, Masaryk University, Brno, Czech Republic, 2 Department <strong>of</strong> Histology, Masaryk<br />

University, Brno, Czech Republic, 3 st. Anne´s University Hospital, International Clinical<br />

Research Center, Brno, Czech Republic<br />

Exogenous fibroblast growth factor-2 (FGF-2) is one <strong>of</strong> crucial supplements in media for human<br />

embryonic stem (ES) cells. In our systematic work, we investigate its roles in plating efficiency<br />

and survival rate under stressed conditions. We show that exogenous FGF-2 enhances <strong>the</strong><br />

expression <strong>of</strong> high molecular mass (HMM) is<strong>of</strong>orms <strong>of</strong> FGF-2, which have been implicated in<br />

promoting proliferation and survival in most cell types. Stably over expression <strong>of</strong> HMM FGF-2<br />

is<strong>of</strong>orms has been shown to link to malignant phenotypes.<br />

We found that all is<strong>of</strong>orms <strong>of</strong> FGF-2 are expressed in undifferentiated human ES cells, whereas<br />

<strong>the</strong> low molecular mass FGF-2 retains its expression when cells undergo differentiation. The<br />

withdrawal <strong>of</strong> exogenous FGF-2 results in cell differentiation depending upon <strong>the</strong> size <strong>of</strong><br />

colonies. Our data suggest that exogenous FGF-2 is involved in <strong>the</strong> early establishment <strong>of</strong><br />

heterogeneous population. Fur<strong>the</strong>rmore, FGF-2 knockdown human ES cells using shRNA are<br />

predisposed to differentiate, suggesting that FGF2 plays a pivotal role in pluripotency.<br />

We hypo<strong>the</strong>sise that FGF-2 signalling maintains <strong>the</strong> undifferentiated growth <strong>of</strong> hESCs. This<br />

action is enhanced by exogenous FGF-2. Both, extrinsic and intrinsic, activities may synergize<br />

to regulate transition between self-renewal and differentiation <strong>of</strong> hESCs.<br />

2074<br />

Mutant frequency increases with <strong>the</strong> length <strong>of</strong> cultivation in human embryonic stem cells.<br />

M. Kruta 1 , L. Balek 1 , R. Hejnova 1 , M. Seneklova 1 , A. Hampl 2 , P. Dvorak 1 , V. Rotrekl 1 ;<br />

1 Department <strong>of</strong> <strong>Biology</strong>, Faculty <strong>of</strong> Medicine, Masaryk University, Brno, Czech Republic,<br />

2 Department <strong>of</strong> Histology and Embryology, Faculty <strong>of</strong> Medicine, Masaryk University, Brno,<br />

Czech Republic<br />

Human embryonic stem cells (hESCs) derived from blastocyst <strong>of</strong> early embryo are capable <strong>of</strong><br />

differentiation into all three germ layers and can produce all cell types <strong>of</strong> human body. Although<br />

hESCs immortality and <strong>the</strong>ir differenciation potential make <strong>the</strong>m promising candidates to<br />

become powerful tool <strong>of</strong> regenerative medicine, <strong>the</strong>ir long-term propagation in vitro induces<br />

accumulation <strong>of</strong> genetic changes as demonstrated by karyotyping, SNP analysis or analysis <strong>of</strong><br />

copy number variation. These changes might render <strong>the</strong> cells useless in regenerative medicine.<br />

We have shown one <strong>of</strong> <strong>the</strong> mechanisms contributing to <strong>the</strong>se changes, Our data demonstrate<br />

that decreased efficiency <strong>of</strong> base excision repair (BER) in late passage hESCs contributes to<br />

<strong>the</strong> genome instability. Decrease <strong>of</strong> apurinic/apyrimidinic endonuclease APE1, a key enzyme <strong>of</strong><br />

BER, with increasing time in culture leads to changes in DNA repair dynamics as well as to<br />

changes in DNA damage signaling. To dissect <strong>the</strong> role <strong>of</strong> such changes in DNA repair and<br />

damage signaling during prolonged hESC cultivation we have designed specific mutation assay.<br />

It is a method based on mutagenesis <strong>of</strong> hypoxanthine-phosphoribosyltransferase (HPRT) gene.<br />

Despite <strong>the</strong> current thinking based on computer models, that time in culture does not increase<br />

<strong>the</strong> mutagenic pressure, we have shown significantly increased both spontaneous and IRinduced<br />

frequency <strong>of</strong> mutants in late passage hESCs in comparison with early passage.


<strong>SUNDAY</strong><br />

However <strong>the</strong> higher doses <strong>of</strong> IR (above 1Gy) induce similar effect in early and late passage,<br />

suggesting that <strong>the</strong> most dramatic effect happens under non stressing conditions. Taken<br />

toge<strong>the</strong>r, our results suggest that <strong>the</strong> changes in <strong>the</strong> APE1 level during long-term cultivation <strong>of</strong><br />

hESCs play an important role in <strong>the</strong>ir response to acquired DNA damage and that <strong>the</strong> long-term<br />

propagation increases <strong>the</strong> mutagenic burden and that <strong>the</strong>se events present a chain <strong>of</strong> events<br />

from DNA repair failure all <strong>the</strong> way to genome instability, presented as increased mutation<br />

burden.<br />

Supported by Ministry <strong>of</strong> Education, Youth and Sports <strong>the</strong> Czech Republic (LC06077) and by<br />

<strong>the</strong> MZ CR (grant NS10237-3)<br />

2075<br />

Primary Cilia <strong>of</strong> Proliferating Cells in Rodent Dorsal Root Ganglia.<br />

M. V. Coronel 1 , D. Hao 1 , W. Huang 1 , H. D. Schwark 1 ; 1 <strong>Biology</strong>, University <strong>of</strong> North Texas,<br />

Denton, TX<br />

Primary cilia are non-motile, microtubule based organelles that arise from <strong>the</strong> membrane <strong>of</strong><br />

most vertebrate cells. Mutations in primary cilia genes can lead to severe developmental<br />

abnormalities. In <strong>the</strong> nervous system <strong>the</strong>se include hydrocephaly, neural tube patterning defects<br />

and sensory deficits that may include anosmia, hearing loss, retinal degeneration and impaired<br />

somatic sensory inputs. Many <strong>of</strong> <strong>the</strong> developmental defects seen in primary cilia mutants have<br />

been attributed to impaired sonic hedgehog signaling. The mitogen sonic hedgehog is essential<br />

in development <strong>of</strong> neural crest-derived structures such as <strong>the</strong> dorsal root ganglia (DRG):<br />

disruption <strong>of</strong> sonic hedgehog signaling results in failure <strong>of</strong> primary sensory neurons to develop.<br />

This essential developmental role played by primary cilia may continue into adulthood.<br />

Each sensory neuron in <strong>the</strong> DRG is tightly enshea<strong>the</strong>d by surrounding satellite glial cells. These<br />

cells arise around E9.5 (neurons) and E11 (glia) from multipotent progenitors derived from <strong>the</strong><br />

neural crest. Neurons, and perhaps satellite glial cells, continue to be added to <strong>the</strong> postnatal<br />

DRG, but <strong>the</strong> origin <strong>of</strong> <strong>the</strong>se new cells is unknown. It seems likely that primary cilia play a role in<br />

postnatal proliferation and maintenance <strong>of</strong> DRG homeostasis for <strong>the</strong> following reasons: primary<br />

cilia play an active role during <strong>the</strong> cell cycle; proliferation <strong>of</strong> stem cell niches is aberrant in<br />

primary cilia mutants; peripheral nerve injury induces sonic hedgehog expression in <strong>the</strong> DRG;<br />

and patients with abnormal primary cilia have sensory deficits.<br />

We have begun our investigation into <strong>the</strong> role <strong>of</strong> primary cilia in <strong>the</strong> developing and adult rodent<br />

DRG by determining <strong>the</strong> incidence and lengths <strong>of</strong> <strong>the</strong> cilia. In <strong>the</strong> rat DRG, primary cilia<br />

decrease in length from P3 (2.6 um) to P14 (2.1 um) to adult (1.9 um). A similar trend <strong>of</strong><br />

decreasing cilium length with development is seen in proliferating satellite glial cells labeled by<br />

antibodies to <strong>the</strong> cell cycle marker Ki67. At P1 <strong>the</strong> average cilium length is 1.4 um, and at P7<br />

<strong>the</strong> average length is 1.2 um. The percentage <strong>of</strong> ciliated proliferating cells also decreased<br />

across <strong>the</strong>se ages, from 35.5% to 28.5%. Finally, we also identified primary cilia in cells<br />

expressing <strong>the</strong> transcription factor Sox2, which is thought to maintain certain cell types in an<br />

undifferentiated state. Cilia characteristics in this small sample <strong>of</strong> cells were not quantified. The<br />

decreasing lengths and incidence <strong>of</strong> primary cilia with development may underlie a decreased<br />

sensitivity to sonic hedgehog signaling.


2076<br />

HNF-4α control <strong>of</strong> pancreatic endocrine differentiation from pluripotent stem cells.<br />

E. C. Moorefield 1 , X. Guan 1 , M. E. Furth 1 , C. E. Bishop 1 ; 1 Wake Forest University, Winston-<br />

Salem, NC<br />

<strong>SUNDAY</strong><br />

The HNF4A gene codes for hepatocyte nuclear factor (HNF)-4α, which has an important role in<br />

pancreatic development and maintenance <strong>of</strong> β-cell function. Haplo-insufficiency <strong>of</strong> this<br />

transcription factor causes a form <strong>of</strong> monogenic type 2 diabetes (T2D), maturity-onset diabetes<br />

<strong>of</strong> <strong>the</strong> young 1 (MODY1). Recently, <strong>the</strong> advancement <strong>of</strong> embryonic stem (ES) cell and induced<br />

pluripotent stem (iPS) cell technology makes study <strong>of</strong> <strong>the</strong> genetic basis <strong>of</strong> <strong>the</strong> disease<br />

achievable. Pluripotent stem cells can be differentiated into insulin producing β-cells using stepwise<br />

protocols that recapitulate signaling pathways necessary for endocrine pancreas<br />

development in vivo. Growth factors and small molecules are applied in a step-wise fashion to<br />

replicate <strong>the</strong> stages in β-cell development which include endoderm formation, pancreas<br />

specification, endocrine specification and β-cell maturation. By applying this protocol to normal<br />

human ES cells we have shown that HNF-4α is expressed at high levels during pancreas<br />

specification and continues to be expressed at lower levels in throughout <strong>the</strong> differentiation<br />

process. To investigate <strong>the</strong> effect <strong>of</strong> HNF-4α deficiency on pancreatic β-cell development and<br />

function we have examined two systems. First, we have generated iPS cells from MODY1<br />

patient fibroblasts using a polycistronic human lentivirus carrying Oct4, Sox2, Klf4 and c-myc. In<br />

addition, we have created dox-inducible HNF-4α knockdown hES cells. The stemness <strong>of</strong> both<br />

systems has been confirmed by analysis <strong>of</strong> a panel <strong>of</strong> pluripotency markers. However, <strong>the</strong><br />

MODY1 iPS showed a large clone to clone variation in differentiation capability toward insulinproducing<br />

cells. On <strong>the</strong> contrary, we have confirmed that HNF-4α expression can be efficiently<br />

knocked down throughout in vitro differentiation to endocrine pancreas by shRNA delivery.<br />

Thus, <strong>the</strong> HNF-4α KD hES system will allow us to elucidate <strong>the</strong> impact <strong>of</strong> genetic variation on<br />

<strong>the</strong> development and function <strong>of</strong> pancreatic β-cells which may lead to identification <strong>of</strong> novel<br />

<strong>the</strong>rapeutic targets for treatment <strong>of</strong> T2D.<br />

2077<br />

RNAi screen <strong>of</strong> predicted E3 ubiquitin ligases that may mark MEX-3 for degradation in<br />

<strong>the</strong> Caenorhabditis elegans embryo.<br />

A. Lin 1 , K. Reichard 1 , A. Desai 2 , P. Heng 2 , K. Schwartz 2 , N. Huang 1 ; 1 <strong>Biology</strong>, Colorado College,<br />

Colorado Springs, CO, 2 Colorado College, Colorado Springs, CO<br />

Early embryonic development in C. elegans depends on precise timing <strong>of</strong> division and<br />

differentiation, which requires precise regulation <strong>of</strong> maternally supplied cell fate determinants.<br />

MEX-3, a maternally supplied RNA binding protein, is present throughout <strong>the</strong> oocyte, 1-cell, and<br />

2-cell embryo. By <strong>the</strong> 4-cell stage, MEX-3 becomes depleted in <strong>the</strong> posterior blastomeres. By<br />

binding to <strong>the</strong> 3’ UTR <strong>of</strong> pal-1 mRNA, MEX-3 prevents translation <strong>of</strong> PAL-1 in anterior<br />

blastomeres. PAL-1 is a homeodomain transcription factor that promotes posterior<br />

development, including muscle development. In mex-3 mutants, PAL-1 is expressed throughout<br />

<strong>the</strong> early embryo, leading to muscle excess and embryonic lethality. After <strong>the</strong> 4-cell stage <strong>of</strong><br />

embryogenesis, MEX-3 is rapidly broken down. The E3 ubiquitin ligase component <strong>of</strong> <strong>the</strong><br />

ubiquitin-proteosome system determines protein specificity <strong>of</strong> ubiquitination, which leads to<br />

degradation. There are numerous predicted E3 ubiquitin ligases in <strong>the</strong> C. elegans genome.<br />

The goal <strong>of</strong> this project is to use RNA inhibition (RNAi) to knock out predicted E3 ubiquitin<br />

ligases, and determine which one(s) mark MEX-3 for degradation in <strong>the</strong> C. elegans embryo.<br />

Genes encoding predicted E3 ubiquitin ligases that were previously shown to be maternally<br />

expressed and/or that produced embryonic lethality in previous RNAi experiments were chosen


<strong>SUNDAY</strong><br />

as candidate genes to knock out. We first determined if knockout <strong>of</strong> <strong>the</strong> candidate genes<br />

resulted in embryonic lethality higher than 20%. Then, fluorescence microscopy was used to<br />

determine whe<strong>the</strong>r RNAi effected <strong>the</strong> degradation <strong>of</strong> a GFP::MEX-3 fusion protein. If an E3<br />

ubiquitin ligase contributing to MEX-3 degradation was successfully knocked out, GFP::MEX-3<br />

would remain present throughout <strong>the</strong> embryo. RNAi was induced by feeding young adults with<br />

bacteria expressing dsRNA, or by directly injecting <strong>the</strong> dsRNA. To date, numerous predicted<br />

HECT-domain, U-box, and monomeric RING finger E3 ubiquitin ligases have been knocked out<br />

by RNAi. Depletion <strong>of</strong> five <strong>of</strong> <strong>the</strong>se genes resulted in high levels <strong>of</strong> embryonic lethality, with two<br />

resulting in abnormal patterns <strong>of</strong> GFP::MEX-3 expression. However, mis-expression <strong>of</strong> MEX-3<br />

does not appear to be <strong>the</strong> primary defect in <strong>the</strong>se RNAi embryos. The screen is being continued<br />

with additional predicted E3 ubiquitin ligases.<br />

2078<br />

Osteoblast response to a nanostructured titanium surface.<br />

M. M. Beloti 1 , R. B. Kato 1 , A. Nanci 2 , P. T. de Oliveira 1 , A. L. Rosa 1 ; 1 School <strong>of</strong> Dentistry <strong>of</strong><br />

Ribeirao Preto, University <strong>of</strong> Sao Paulo, Ribeirao Preto, Brazil, 2 Faculté de Médecine Dentaire,<br />

Université de Montréal, Canada<br />

Titanium (Ti) surface nanotopography obtained by chemical deoxidation and controlled<br />

reoxidation affects early events <strong>of</strong> osteoblast phenotype development in cultures derived from<br />

rat calvaria. The aim <strong>of</strong> our study was to evaluate <strong>the</strong> effect <strong>of</strong> <strong>the</strong> nanostructured Ti surface on<br />

<strong>the</strong> responses <strong>of</strong> human osteoblastic cells. Machined Ti discs, 12 mm in diameter, were<br />

chemically treated with H2SO4/H2O2 for 2 hours. Osteoblastic cells were obtained by enzymatic<br />

digestion <strong>of</strong> human alveolar bone and cultured under osteogenic condition until subconfluence.<br />

First passage cells were cultured (2�10 4 cells/disc) on nanostructured and machined Ti discs for<br />

periods <strong>of</strong> up to 21 days. For cell attachment assay, adherent cells were counted at 24 hours<br />

and data were expressed as a percentage <strong>of</strong> <strong>the</strong> initial cell number. For proliferation, cells were<br />

counted at 1 and 7 days and data were expressed as doubling time in hours. At day 7, cell<br />

viability was evaluated using trypan blue and expressed as a percentage <strong>of</strong> viable cells. At days<br />

1, 3, and 7, cell morphology was evaluated with phalloidin and DAPI; alkaline phosphatase<br />

(ALP) labeling was performed using a primary mouse anti-human ALP antibody. Extracellular<br />

matrix mineralization was detected by Alizarin red staining at 21 days and data were expressed<br />

as a percentage <strong>of</strong> <strong>the</strong> total disc area. Quantitative data were compared by Mann-Whitney test<br />

(n=3). There were no statistically significant differences between nanostructured and machined<br />

Ti in terms <strong>of</strong> cell attachment (34.7±6.4 and 40.3±10.5; p=0.70), proliferation (50.4±2.7 and<br />

48.1±2.1; p=0.28), and viability (92.1±2.8 and 93.1±1; p=0.83). At day 1, cells were spread and<br />

exhibited an elongated polygonal shape on machined Ti whereas on nanostructured Ti <strong>the</strong>y<br />

were mostly stellate-shaped. At day 7, both Ti surfaces exhibited areas <strong>of</strong> cell multilayering and<br />

cells were predominantly elongated. ALP positive cells were observed at all time points with a<br />

similar labeling pattern on both Ti surfaces. It was noticed five times more extracellular matrix<br />

mineralization (2.7±0.2 and 15.2±4.6; p=0.01) on nanostructured compared with machined Ti. In<br />

conclusion, our results indicate that nanotopography induces a significant increase in <strong>the</strong> late<br />

key event <strong>of</strong> in vitro osteogenesis, i.e.: extracellular matrix mineralization and this surface<br />

modification may, <strong>the</strong>refore, be considered a useful strategy to accelerate/enhance <strong>the</strong> process<br />

<strong>of</strong> Ti implant osseointegration.<br />

Acknowledgements: FAPESP (Grant # 2010/18395-3, 2010/19280-5)


2079<br />

Effects <strong>of</strong> hedgehog signaling on human differentiated osteoblasts.<br />

F. S. Oliveira 1 , G. B. Junqueira 1 , M. M. Beloti 1 , A. L. Rosa 1 ; 1 School <strong>of</strong> Dentistry <strong>of</strong> Ribeirao<br />

Preto, University <strong>of</strong> Sao Paulo, Ribeirao Preto, Brazil<br />

<strong>SUNDAY</strong><br />

Hedgehog (Hh) signaling is involved in many developmental processes including osteoblast<br />

differentiation <strong>of</strong> mesenchymal stem cells. Here, we aimed at determining <strong>the</strong> effect <strong>of</strong> Hh<br />

signaling modulation on phenotype expression <strong>of</strong> differentiated osteoblasts. Cells harvested<br />

from human dental alveolar bone were cultured in osteogenic media under four conditions: nonsupplemented<br />

(control), supplemented with <strong>the</strong> Hh agonist purmorphamine (2 �M),<br />

supplemented with <strong>the</strong> Hh antagonist KAAD-cyclopamine (100 nM), and supplemented with<br />

both purmorphamine and KAAD-cyclopamine. Hh signaling was evaluated by gene expression<br />

<strong>of</strong> Gli1 at day 7. Osteoblast phenotype expression was evaluated by activity and gene<br />

expression <strong>of</strong> alkaline phosphatase (ALP) at day 7, and extracellular matrix mineralization at<br />

day 21. Data (n=3) were compared by Kruskall-Wallis test followed by Fisher’s test when<br />

appropriated. Hh pathway was stimulated by purmorphamine as revealed by an increase <strong>of</strong><br />

almost four times in <strong>the</strong> expression <strong>of</strong> Gli1 compared to control (p=0.001). This increase was<br />

only partially reversed by KAAD-cyclopamine (p=0.05) that by itself did not present any effect<br />

when compared to control (p>0.05). Despite this, ALP activity (p>0.05), ALP gene expression<br />

(p>0.05) and matrix mineralization (p>0.05) were not modified by any culture condition. These<br />

results suggest that <strong>the</strong> modulation <strong>of</strong> Hh signaling does not affect <strong>the</strong> phenotypic expression <strong>of</strong><br />

human differentiated osteoblasts.<br />

Acknowledgments: FAPESP and CNPq<br />

2080<br />

Bavachinin A modulates <strong>the</strong> differentiation <strong>of</strong> hADMSCs on adipogenesis and<br />

osteogenesis.<br />

H-J. Jang 1,2 , S. Lim 1 , J-M. Kim 1,2 , S. Ryu 2 , P-G. Suh 1 ; 1 School <strong>of</strong> Nano-Bioscience and Chemical<br />

Engineering, UNIST, Ulsan Metropolitan, Korea, 2 Division <strong>of</strong> <strong>Molecular</strong> and Life Science,<br />

POSTECH, POHANG, Korea<br />

Human mesenchymal stem cells are an attractive model not only to investigate molecular<br />

mechanisms <strong>of</strong> cell growth and differentiation under normal and pathological conditions but also<br />

to use cell-based <strong>the</strong>rapies for regenerative medicine. Therefore, it is important to understand<br />

how <strong>the</strong>ir differentiation is regulated. We screened <strong>the</strong> molecules which could regulate<br />

adipogenesis using natural compound library. As a screening result, we found that bavachinin A<br />

enhanced adipogenesis <strong>of</strong> human adipose-derived mesenchymal stem cells(hADMSCs).<br />

Bavachinin A also increased <strong>the</strong> mRNA level <strong>of</strong> adipocyte markers, such as CCAAT-enhancerbinding<br />

proteins α, peroxisome proliferator-activated receptor γ(PPAR-γ) and adipocyte fatty<br />

acid–binding protein during adipocyte differentiation. Fur<strong>the</strong>rmore, PPARγ activity was<br />

increased by treatment <strong>of</strong> bavachinin A. To evaluate <strong>the</strong> effect <strong>of</strong> bavachinin A in vivo, we used<br />

implanted de novo adipose tissue formation system. It showed that intraperitoneal<br />

administration <strong>of</strong> bavachinin A in mice, implanted with mass <strong>of</strong> 3t3-f442A cells, increased de<br />

novo adipogenesis and weight <strong>of</strong> inborn adipose tissues. Whereas bavachinin A inhibited<br />

osteogenesis <strong>of</strong> hADMSCs. Our results suggest that bavachinin A is a potent activator <strong>of</strong><br />

adipogenesis and a inhibitor <strong>of</strong> osteogenesis via activation <strong>of</strong> PPAR-γ.


2081<br />

Directed Stem Cell Differentiation Using PEG/α-CD-derived Biomaterials.<br />

A. Singh 1 , J. Zhan 1 , J. Elisseeff 1,2 ; 1 BME, Johns Hopkins University, Baltimore, MD,<br />

2 Ophthalmology, Wilmer Eye Institute, Baltimore, MD<br />

<strong>SUNDAY</strong><br />

Adult stem cells (ASCs), e.g., mesenchymal and adipose-derived (MSCs and ADSCs) hold<br />

great promise in tissue engineering because <strong>of</strong> <strong>the</strong>ir proliferative capacity and ability to<br />

differentiate into several phenotypes, and ultimately built new tissues. However, to enable<br />

translation <strong>of</strong> stem cell <strong>the</strong>rapies, <strong>the</strong>re is a significant need to generate materials that can be<br />

used to study fundamentals in biology and modulate or enhance differentiation. Poly(ethylene<br />

glycol) (PEG)-based hydrogels are frequently utilized to encapsulate cells in a 3D environment.<br />

Unfortunately, PEG based biomaterials lack functionality to incorporate multiple chemical and<br />

biological moieties as signals for stem cells to probe behavior and guide development.<br />

Conventional methods to modify PEG hydrogels include copolymerization and chain extension,<br />

which leads to changes in mechanical properties <strong>of</strong> <strong>the</strong> material. Therefore, we developed an<br />

advanced multifunctional hydrogel by combining PEG- and α-Cyclodextrin (α-CD). α-CD is a six<br />

membered oligosaccharide ring, which threads onto <strong>the</strong> PEG chains. [1] The hydroxyl groups <strong>of</strong><br />

α-CD provide necessary functional sites to introduce various moieties, such as hydrophobic<br />

groups (CH3), hydrophilic groups (PO4 - ) and integrin-binding peptide RGD by chemical<br />

modifications. This simple strategy provides us a unique ability to independently control<br />

chemical functionality, mechanical and bioadhesive properties <strong>of</strong> <strong>the</strong> hydrogels to probe stem<br />

cell behavior.<br />

ASCs were ei<strong>the</strong>r seeded onto or encapsulated in UV-photopolymerizable PEG-α-CD hydrogels<br />

with varying stiffness (0.5 kPa to 30 kPa), adhesive peptide concentrations (0% to 10% w/v) and<br />

different functional groups. In an initial experiment, <strong>the</strong> enhanced chondrogenic differentiation <strong>of</strong><br />

gMSCs in 10% PEGDA/1% α-CD hydrogel was evident from increased gene expression for Col<br />

II and Aggrecan, and Safranin O staining for GAG deposition. At <strong>the</strong> similar mechanical<br />

properties <strong>of</strong> <strong>the</strong> hydrogel, introducing a hydrophobic group (CH3) in α-CD enhanced adipogenic<br />

differentiation, while a hydrophilic group (PO4 - ) increased osteogenic differentiation <strong>of</strong> hADSCs.<br />

We also found that cell spreading and morphology are dependent on both stiffness and<br />

concentrations <strong>of</strong> integrin-binding peptide. On <strong>the</strong> stiffer substrate, hMSCs adopted elongated<br />

morphology at all concentrations <strong>of</strong> RGD. Interestingly, on <strong>the</strong> s<strong>of</strong>ter substrate hMSCs adopted<br />

elongated morphology at higher concentrations <strong>of</strong> RGD. RT-PCR experiments showed that<br />

s<strong>of</strong>ter matrix upregulates gene-expression markers for adipogenesis at both lower and higher<br />

cell-binding peptide concentrations, while moderately stiffer substrate with lower cell-binding<br />

peptide concentration upregulates markers for myogenesis. The stiffer substrate enhances both<br />

chondrogenesis and osteogenesis at higher concentrations <strong>of</strong> integrin-binding peptide. This<br />

indicates that both substrate-stiffness[2] and concentration <strong>of</strong> cell-binding peptide sites dictate<br />

stem cell differentiation. In summary, this biomaterial enables us to probe stem cell behavior,<br />

such as proliferation and lineage specific differentiation by independently controlling <strong>the</strong><br />

functionality, mechanical and bioadhesive properties <strong>of</strong> <strong>the</strong> material.<br />

References:<br />

1. A. Harada, J. Li, M. Kamachi, Nature, 1992, 356, 325-327.<br />

2. A.J. Engler, S. Sen, H.L. Sweeny, D.E. Discher, Cell, 2006, 126(4), 677-689.<br />

Acknowledgements: Maryland Stem Cell (TEDCO) fellowship, Dr. O. Manuel Uy, and Cindy<br />

Berlinicke, and JHE group.


Cell <strong>Biology</strong> <strong>of</strong> <strong>the</strong> Neuron<br />

2082<br />

PINK1 and Parkin Target Miro for Phosphorylation and Degradation to Arrest<br />

Mitochondrial Motility.<br />

X. Wang 1 , D. Winter 2 , G. Ashrafi 2 , J. Schlehe 3 , Y. Wong 4 , D. Selkoe 3 , S. Rice 4 , J. Steen 2 , M.<br />

LaVoie 3 , T. Schwarz 3 ; 1 Stanford University/Harvard Medical School, Boston, MA, 2 Children's<br />

Hospital Boston, MA, 3 Harvard Medical School, Boston, MA, 4 Northwestern University,<br />

Evanston, IL<br />

<strong>SUNDAY</strong><br />

Cells keep <strong>the</strong>ir energy balance and avoid oxidative stress by regulating mitochondrial<br />

movement, distribution, and clearance. We report here that two Parkinson's disease proteins,<br />

<strong>the</strong> Ser/Thr-kinase PINK1 and ubiquitin-ligase Parkin, participate in this regulation by arresting<br />

mitochondrial movement. PINK1 phosphorylates Miro, a component <strong>of</strong> <strong>the</strong> primary<br />

motor/adaptor complex that anchors kinesin to <strong>the</strong> mitochondrial surface. The phosphorylation<br />

<strong>of</strong> Miro activates proteasomal degradation <strong>of</strong> Miro in a Parkin-dependent manner. Removal <strong>of</strong><br />

Miro from <strong>the</strong> mitochondrion also detaches kinesin from its surface. By preventing mitochondrial<br />

movement, <strong>the</strong> PINK1/Parkin pathway may quarantine damaged mitochondria prior to <strong>the</strong>ir<br />

clearance. PINK1 has been shown to act upstream <strong>of</strong> Parkin but <strong>the</strong> mechanism corresponding<br />

to this relationship has not been known. We propose that PINK1 phosphorylation <strong>of</strong> substrates<br />

is necessary for <strong>the</strong> subsequent action <strong>of</strong> Parkin and <strong>the</strong> proteasome.<br />

2083<br />

Essential Role <strong>of</strong> a Conserved Homophilic Interaction Domain <strong>of</strong> SYD-2/Liprin-alpha<br />

Protein in Presynaptic Assembly in C.elegans.<br />

H. Taru 1 ; 1 Creative Research Institution, Hokkaido University, Sapporo, Japan<br />

Background<br />

Synapses are asymmetric structures that are specialized for neuronal signal transduction. A<br />

unique set <strong>of</strong> proteins is present at <strong>the</strong> presynaptic active zone, which is a core structure<br />

essential for neurotransmitter release. C. elegans SYD-2, a Liprin-α family protein, is localized<br />

at <strong>the</strong> center <strong>of</strong> presynaptic active zones and acts toge<strong>the</strong>r with a GAP protein SYD-1 to<br />

promote active zone organization in various types <strong>of</strong> neurons. Liprin-α proteins are scaffolding<br />

proteins and composed <strong>of</strong> coiled-coil rich regions in <strong>the</strong> N-terminal halves, which mediate<br />

interactions with adapter proteins at <strong>the</strong> presynaptic active zone, and three SAM domains in <strong>the</strong><br />

C-termini, which bind proteins such as LAR receptor tyrosine phosphatase.<br />

Objective<br />

To address <strong>the</strong> molecular mechanism by which SYD-2 activity in presynaptic assembly is<br />

regulated, core functional domains <strong>of</strong> SYD-2 were dissected out by transgenic structure-function<br />

analyses and characterized biochemically.<br />

Methods<br />

A series <strong>of</strong> SYD-2 deletion constructs were transgenically expressed in C.elegans, and were<br />

analyzed for <strong>the</strong>ir abilities to rescue presynaptic assembly defects in HSN neuron in loss-<strong>of</strong>function<br />

mutants <strong>of</strong> syd-2 or syd-1. Recovery <strong>of</strong> presynaptic assembly was evaluated by<br />

restored egg-laying behavior and localization <strong>of</strong> fluorescent tagged presynaptic protein reporters<br />

such as synaptobrevin::YFP. Biochemical property <strong>of</strong> recombinant SYD-2 coiled-coil region was<br />

analyzed by pull-down, crosslinking and gel-filtration assay. Clustering <strong>of</strong> human Liprin-α was<br />

analyzed by Blue Native-PAGE and live-cell imaging <strong>of</strong> GFP in cultured cells.<br />

Results<br />

Transgenic structure-function studies <strong>of</strong> SYD-2 revealed that <strong>the</strong> N-terminal half <strong>of</strong> SYD-2 is<br />

minimally required for rescuing presynaptic defects in syd-2 mutants. A highly conserved short


<strong>SUNDAY</strong><br />

coiled-coil segment named Liprin Homology 1 (LH1) domain is both necessary and sufficient to<br />

suppress presynaptic defects in syd-1 mutants. Biochemical analyses revealed that <strong>the</strong> LH1<br />

domains <strong>of</strong> SYD-2 and Liprin-α form dimer in vitro. The role <strong>of</strong> SYD-2 LH1 domain in<br />

presynaptic assembly can be partially complemented by artificial dimerization. In human Liprin-α<br />

deletion <strong>of</strong> <strong>the</strong> LH1 domain impaired proper clustering and/or complex formation in cells.<br />

Conclusions<br />

These findings suggest a model by which <strong>the</strong> self-assembly <strong>of</strong> SYD-2/Liprin-α proteins mediated<br />

by <strong>the</strong> evolutionally conserved coiled-coil LH1 domain is one <strong>of</strong> <strong>the</strong> key steps to <strong>the</strong><br />

accumulation <strong>of</strong> presynaptic components at nascent synaptic junctions.<br />

2084<br />

Quantum-dot based presynaptic probe reveals dynamic modulation <strong>of</strong> synaptic vesicle<br />

trafficking behaviors by synaptic activity.<br />

S. Lee 1 , S. Chang 1 ; 1 Biomedical science, SNU, Seoul, Korea<br />

Presynaptic contribution during synaptic plasticity is a matter <strong>of</strong> debate. A small nature <strong>of</strong><br />

presynaptic bouton and a lack <strong>of</strong> suitable technique, however, hamper detailed analysis <strong>of</strong><br />

synaptic vesicle (SV) behaviors at synapse. Here, we created a quantum dot-based presynaptic<br />

probe and characterized dynamic behaviors <strong>of</strong> individual SVs in CA3-CA1 synapse <strong>of</strong> cultured<br />

neurons. As previously reported, SVs show multiple exchanges between neighboring boutons.<br />

Actin disruption induces a dramatic decrease in diffusive behaviors <strong>of</strong> SVs at synapse while<br />

microtubule disruption only reduce extrasynaptic mobility. Chemical LTP induces NMDA<br />

receptor-dependent increase in synaptic mobility <strong>of</strong> SVs and in inter-boutonal traffickings <strong>of</strong> SVs<br />

between neighboring boutons, causing a bias to incorporation <strong>of</strong> SVs into synapses. NMDA<br />

induced-LTD decreased diffusion coefficient <strong>of</strong> SVs at synapse. Toge<strong>the</strong>r, our results suggest<br />

that a dynamic modulation <strong>of</strong> SV diffusive behaviors could be <strong>the</strong> one <strong>of</strong> key mechanisms <strong>of</strong><br />

regulating presynaptic efficacy during presynaptically expressed long-term synaptic plasticity.<br />

2085<br />

Characterization <strong>of</strong> <strong>the</strong> DOPA transporter in an in vitro cellular model <strong>of</strong> dopaminergic<br />

neurons.<br />

E. Moura 1,2 , E. Magalhães Ribeiro 1 , E. Silva 3,4 , M. P. Serrão 1 , J. Guimarães 5 , M. A. Vieira<br />

Coelho 1,2 ; 1 Pharmacology and Therapeutics, Faculty <strong>of</strong> Medicine University <strong>of</strong> Porto, Porto,<br />

Portugal, 2 Neuropharmacology, Institute for <strong>Molecular</strong> and Cell <strong>Biology</strong>, Porto, Portugal,<br />

3 Laboratório de Biologia Celular e <strong>Molecular</strong>, Faculty <strong>of</strong> Medicine University <strong>of</strong> Porto, Porto,<br />

Portugal, 4 Departamento de Stress in Animals, Instituto de Biologia Celular e <strong>Molecular</strong>, Porto,<br />

Portugal, Portugal, 5 Neurology Department, Faculty <strong>of</strong> Medicine University <strong>of</strong> Porto & Hospital<br />

de São João<br />

L-3,4-dihydroxyphenylalanine (DOPA) is <strong>the</strong> immediate product <strong>of</strong> <strong>the</strong> rate-limiting step in<br />

catecholamine biosyn<strong>the</strong>sis and <strong>the</strong> precursor <strong>of</strong> all <strong>the</strong> endogenous catecholamines Treatment<br />

with DOPA remains to date <strong>the</strong> most effective treatment <strong>of</strong> <strong>the</strong> slowness <strong>of</strong> movement,<br />

increased muscle tone, and tremor that are typical <strong>of</strong> Parkinson’s disease (PD). The human<br />

neuroblastoma cell line SH-SY5Y has been widely used as a cellular model <strong>of</strong> dopaminergic<br />

neurons for PD research. In this study we investigated <strong>the</strong> transporter involved in DOPA uptake<br />

in SH-SY5Y cells.<br />

DOPA levels in cells were evaluated by high performance liquid chromatography with<br />

electrochemical detection. Results are presented as arithmetic mean ± standard error mean.<br />

SH-SY5Y cells take up DOPA in a time dependent (linear until 6 minutes) and concentration<br />

dependent (2.5-2500 µM) manner. Non-linear analysis <strong>of</strong> <strong>the</strong> saturation curves revealed for<br />

DOPA a KM (in µM) <strong>of</strong> 570±97 and a Vmax (in nmol/mg protein/6 min) <strong>of</strong> 611±34. The uptake <strong>of</strong>


<strong>SUNDAY</strong><br />

DOPA (2.5 µM) was reduced by <strong>the</strong> inhibitor <strong>of</strong> <strong>the</strong> L-type amino acid transporters 2aminobicyclo-(2,2,1)-heptane-2-carboxylic<br />

acid (BCH, 0.1-1000 µM) (IC50 = 47 ± 2 nM; Emax =<br />

24 ± 10 % control uptake) and neutral amino acids (1mM), but no by <strong>the</strong> inhibitor <strong>of</strong> <strong>the</strong> A-type<br />

amino acid transporters N-(methylamino)-isobutyric acid (MeAIB, 0.1-1000 µM) nor by <strong>the</strong> acidic<br />

and basic amino acids (1mM). DOPA uptake (2.5 µM) was unaltered by lowering <strong>the</strong> pH from<br />

7.4 to 6.2. In <strong>the</strong> absence <strong>of</strong> Na + <strong>the</strong>re was a 20% reduction in <strong>the</strong> Vmax values for DOPA<br />

uptake. Accumulation <strong>of</strong> DOPA in SH-SY5Y cells was largely inhibited by <strong>the</strong> L-isomers <strong>of</strong> <strong>the</strong><br />

small and large neutral amino acids (alanine, serine, threonine, cysteine, leucine, isoleucine,<br />

phenylalanine, methionine, and tyrosine), histidine, tryptophan, valine, asparagine and<br />

glutamine. Whereas <strong>the</strong> amino acids glycine, proline and <strong>the</strong> basic amino acid arginine also<br />

produced an inhibition <strong>of</strong> DOPA uptake, albeit minor, <strong>the</strong> basic amino acids lysine and cystine,<br />

and acidic amino acids aspartate and glutamate did not inhibit <strong>the</strong> uptake <strong>of</strong> L-DOPA.<br />

In conclusion, DOPA uptake in SH-SY5Y cells was sensitive to inhibition by BCH, but not to<br />

MeAIB, and was more sensitive to inhibition by neutral than to basic and acidic amino acids.<br />

Although most <strong>of</strong> DOPA was entering <strong>the</strong> cells in a Na + -independent manner, a minor<br />

component <strong>of</strong> DOPA uptake (25%) was found to require extracellular Na + . In general, <strong>the</strong>se<br />

findings support <strong>the</strong> view that DOPA may be transported by systems B 0 (Na + -dependent) and L<br />

(Na + -independent). The fraction <strong>of</strong> DOPA that is handled through system L is through a highaffinity<br />

(KM values in <strong>the</strong> micromolar range) and pH-insensitive transport, which are<br />

characteristic <strong>of</strong> <strong>the</strong> LAT1.<br />

2086<br />

Deciphering <strong>the</strong> molecular interaction between sodium channel Nav1.8 and ankyrin G.<br />

F. Castets 1,2 , A. Montersino 1,2 , M-P. Fache 1,2 , B. Dargent 1,2 ; 1 INSERM U641, Marseille, France,<br />

2 Aix-Marseille University, Marseille, France<br />

In <strong>the</strong> central nervous system (CNS), <strong>the</strong> accumulation <strong>of</strong> <strong>the</strong> voltage-gated sodium channels,<br />

Nav1 (Nav1-2 and Nav1.6) at <strong>the</strong> axon initial segment (AIS) results from a direct interaction<br />

between <strong>the</strong> ankyrin-binding motif <strong>of</strong> Nav1 and ankyrin G. This interaction is regulated in a<br />

phospho-dependent fashion by protein caseine kinase 2 (CK2). The peripheral channel Nav1.8<br />

is predominantly expressed in dorsal root ganglion neurons. However, Nav1.8 is ectopically<br />

expressed in Purkinje cells in EAE animal model and in human multiple sclerosis. Interestingly,<br />

this channel possesses a motif close to <strong>the</strong> conserved ankyrin-binding motif found in <strong>the</strong> Nav1<br />

expressed in CNS.<br />

This study was aimed at deciphering <strong>the</strong> molecular interaction between Nav1.8 and ankyrin G.<br />

We observed by GST pull-downs that Nav1.8 associates with endogenous ankyrin G from rat<br />

brain membranes. Using surface plasmon resonance, we found that Nav1.8 constitutively<br />

interacts with ankyrin G 190 kDa in contrast to Nav1.2, which requires <strong>the</strong> phosphorylation <strong>of</strong><br />

CK2 to bind to ankyrin G 190 kDa. Fur<strong>the</strong>rmore when <strong>the</strong> ankyrin-binding motif <strong>of</strong> Nav1.8 was<br />

fused to <strong>the</strong> somatodendritic channel Kv2.1, <strong>the</strong> resulting chimeric protein was not only<br />

clustered at <strong>the</strong> AIS <strong>of</strong> transfected primary hippocampal neurons but also acted as a dominant<br />

negative displacing <strong>the</strong> endogenous Nav1 from AIS <strong>of</strong> hippocampal neurons.<br />

Altoge<strong>the</strong>r, <strong>the</strong>se results indicate that Nav1.8 carries all <strong>the</strong> information required for clustering at<br />

<strong>the</strong> AIS and that its accumulation is not regulated by protein kinase CK2, unlike CNS Nav1. Our<br />

work shows a new kind <strong>of</strong> interaction between sodium channel and <strong>the</strong> scaffold protein ankyrin<br />

G, which is likely to contribute to <strong>the</strong> pathological aspects <strong>of</strong> multiple sclerosis.


<strong>SUNDAY</strong><br />

2087<br />

N-cadherin induces synapse formation in heterologous cultures <strong>of</strong> brainstem cholinergic<br />

neurons and CHO cells.<br />

R. Flannery 1 , J. L. Brusés 2 ; 1 Department <strong>of</strong> Anatomy and Cell <strong>Biology</strong>, University <strong>of</strong> Kansas<br />

School <strong>of</strong> Medicine, Kansas City, KS, 2 Department <strong>of</strong> Anatomy and Cell <strong>Biology</strong> & Department<br />

<strong>of</strong> Psychiatry and Behavioral Sciences, University <strong>of</strong> Kansas School <strong>of</strong> Medicine, Kansas City,<br />

KS<br />

N-cadherin is a cell adhesion molecule that contributes to synaptic differentiation, synaptic<br />

vesicle recycling, and synaptic plasticity. It promotes <strong>the</strong> contact between pre and postsynaptic<br />

membranes and acts co-operatively with synaptogenic proteins to induce synapse development.<br />

Much <strong>of</strong> what is known about <strong>the</strong> regulation <strong>of</strong> synapse formation and function by N-cadherin<br />

has been elucidated from experiments with glutamatergic synapses. Here we have used<br />

heterologous cell cultures with primary cholinergic neurons and transfected Chinese Hamster<br />

Ovary (CHO) cells to determine whe<strong>the</strong>r N- cadherin is sufficient to initiate formation <strong>of</strong><br />

functional cholinergic synapses. Central cholinergic neurons were cultured from brainstem<br />

nuclei explants <strong>of</strong> transgenic mice expressing EGFP under <strong>the</strong> control <strong>of</strong> choline<br />

acetyltransferase (ChAT) transcriptional regulatory elements, and cultured with CHO cells<br />

(which do not express endogenous N-cadherin) transfected with N-cadherin or a fluorescent<br />

protein (FP) as control. CHO cells were co-transfected with nicotinic acetylcholine receptor<br />

(nAChR) subunits alpha 3 and beta 4 forming functional AChRs that are activated by<br />

acetylcholine. Immunostaining for synapsin I and SV2 demonstrated that synaptic vesicle<br />

proteins accumulate at contact sites between cholinergic axons and N-cadherin expressing<br />

CHO cells. There was a ~3-fold increase in <strong>the</strong> total area <strong>of</strong> SV2 immuno- labeling and a ~2-fold<br />

increase in <strong>the</strong> amount <strong>of</strong> synapsin I immuno-puncta accumulation per axon length on Ncadherin<br />

expressing CHO cells as compared to FP expressing CHO cells. N-cadherin<br />

expression in CHO cells in contact with axons extending from cholinergic neurons resulted in<br />

<strong>the</strong> appearance <strong>of</strong> excitatory postsynaptic potentials (EPSPs), as measured by whole cell<br />

recordings <strong>of</strong> <strong>the</strong> CHO transfected cells. While 67% <strong>of</strong> N-cadherin transfected CHO cells<br />

displayed synaptic events (6 out <strong>of</strong> 9 cells) only 11% <strong>of</strong> control cells (1 out <strong>of</strong> 9 cells) showed<br />

activity. The mean EPSP amplitude for heterologous synapses was 66.7 +/- 13.0 pA (227.7 +/-<br />

36.7 pA after normalizing current density). While <strong>the</strong> mean decay time constant for heterologous<br />

cholinergic synapses was 10.1 +/- 1.0 msec, events in one cell had a biphasic decay with time<br />

constants tau1 = 42.4 +/- 10.4 msec and tau2 = 5.2 +/- 0.6 msec, suggesting <strong>the</strong> presence <strong>of</strong><br />

immature synaptic release sites. Miniature EPSPs were not detected in control (n=7) or Ncadherin<br />

transfected CHO cells (n=12), which we attribute to a low release probability intrinsic to<br />

brainstem cholinergic neurons. These results indicate that expression <strong>of</strong> N-cadherin in nonneuronal<br />

cells is sufficient to initiate formation <strong>of</strong> functional synapses with brainstem cholinergic<br />

neurons; however, complete development or maturation <strong>of</strong> release sites may require expression<br />

<strong>of</strong> additional synaptogenic proteins.<br />

2088<br />

Fast-scanning AFM analysis <strong>of</strong> agonist-induced height transition <strong>of</strong> <strong>the</strong> NMDA receptor.<br />

Y. Suzuki 1 , T. A. Goetze 2 , D. Stroebel 3 , S. H. Yoshimura 1 , P. Paoletti 3 , R. M. Henderson 2 , K.<br />

Takeyasu 1 , J. M. Edwardson 2 ; 1 Laboratory <strong>of</strong> Plasma Membrane and Nuclear Signaling,<br />

Graduate School <strong>of</strong> Biostudies, Kyoto University, Kyoto, Japan, 2 Department <strong>of</strong> Pharmacology,<br />

University <strong>of</strong> Cambridge, Cambridge, United Kingdom, 3 Institut de Biologie de l'Ecole Normale<br />

Supérieure (IBENS), Ecole Normale Supérieure, Paris, France<br />

NMDA receptors (NMDARs) are widely expressed in <strong>the</strong> central nervous system, and play a<br />

major role in excitatory synaptic transmission. Activation <strong>of</strong> NMDARs requires <strong>the</strong> binding <strong>of</strong> two


<strong>SUNDAY</strong><br />

agonists, glutamate and glycine (or D-serine). Here, we used fast-scanning atomic force<br />

microscopy (AFM) to analyze agonist-induced structural changes in <strong>the</strong> extracellular domain <strong>of</strong><br />

<strong>the</strong> heterotetrameric GluN1/GluN2A receptor integrated into lipid bilayers. In <strong>the</strong> absence <strong>of</strong><br />

agonist, <strong>the</strong> frequency distribution <strong>of</strong> heights <strong>of</strong> <strong>the</strong> receptors above <strong>the</strong> bilayer surface had two<br />

peaks, at 3.5 nm and 8.5 nm. The taller particles were specifically recognized by an anti-GluN1<br />

antibody that recognizes <strong>the</strong> S2 segment <strong>of</strong> <strong>the</strong> agonist-binding domain, suggesting that <strong>the</strong>se<br />

particles represent <strong>the</strong> extracellular region <strong>of</strong> <strong>the</strong> receptor. Receptor activation was triggered by<br />

UV photolysis <strong>of</strong> caged glutamate. Release <strong>of</strong> glutamate into a glycine-containing buffer<br />

resulted in a significant structural change, with <strong>the</strong> height <strong>of</strong> <strong>the</strong> receptor decreasing from 8.5<br />

nm to 7.2 nm. This height shift did not occur in <strong>the</strong> absence <strong>of</strong> glycine or in <strong>the</strong> presence <strong>of</strong> <strong>the</strong><br />

selective NMDAR antagonist, D-(-)-2-amino-5-phosphonopentanoic acid (D-AP5). Hence, <strong>the</strong><br />

height <strong>of</strong> <strong>the</strong> extracellular domain <strong>of</strong> <strong>the</strong> receptor is reduced upon agonist activation. Such a<br />

conformational change could affect interactions that NMDARs engage with synaptic partners.<br />

2089<br />

Potential <strong>of</strong> neuronal progenitor stem cell <strong>the</strong>rapy for Alzheimer's disease.<br />

D. Raman 1 , M. Choudhary 1 , J. P. Cleary 1 , R. R. Punzalan 1 , C. V. Sharma 1 , M. V. Sharma 1 , R.<br />

Balmiki 2 , N. Chopra 2 , D. K. Lahiri 2 , J. P. Sharma 3 ; 1 Stem Cell <strong>Biology</strong>, Celprogen Inc, San<br />

Pedro, CA, 2 Institute <strong>of</strong> Psychiatric Research, Indiana University School <strong>of</strong> Medicine,<br />

Indianapolis, IN, 3 Celprogen Inc, San Pedro, CA<br />

Alzheimer's disease (AD) is a progressive neurodegenerative disorder which impairs <strong>the</strong><br />

memory and intellectual abilities <strong>of</strong> <strong>the</strong> affected individuals.The basal forebrain cholinergic<br />

system is <strong>the</strong> population <strong>of</strong> neurons most affected by <strong>the</strong> neurodegenerative process.<br />

Depositions <strong>of</strong> beta-amyloid or Abeta (Abeta) protein, neur<strong>of</strong>ibrillary tangles and neuronal loss<br />

are <strong>the</strong> neuropathological hallmarks <strong>of</strong> AD. The goal <strong>of</strong> our labs is to unveil <strong>the</strong> mechanism <strong>of</strong><br />

neurodegeneration and devise ways to prevent /reverse neurodegeneration by various<br />

approaches in primary brain cultures and AD animal models. For example, we have recently<br />

shown that rivastigmine can preserve or enhance neuronal and synaptic terminal<br />

markers in degenerating primary embryonic cerebrocortical cultures (Bailey et al, PLOS One,<br />

2011). Fur<strong>the</strong>r, we demonstrated neuroprotective and neurorescue effects <strong>of</strong> a novel polymeric<br />

nanoparticle in neuronal cultures and AD animal model (Ray et al- JAD-2011). Similarly <strong>the</strong> role<br />

<strong>of</strong> specific micro-RNA species is being studied (Long & Lahiri-CMC-2011). From <strong>the</strong><br />

pharmacological -chemical perspective to si RNA approach, here we report <strong>of</strong> testing a novel<br />

cell replacement <strong>the</strong>rapy based on <strong>the</strong> hypo<strong>the</strong>sis that cell replacement <strong>the</strong>rapy would provide<br />

cure by compensating <strong>the</strong> lost neuronal systems. Neuronal progenitor stem cells (NPSCs)<br />

obtained from adult human post mortem brains from transplant rejects were grafted into <strong>the</strong><br />

intact brain <strong>of</strong> mice (10) with matching controls (10), which was <strong>the</strong>n followed by <strong>the</strong>ir<br />

incorporation into <strong>the</strong> host parenchyma and differentiation into functional neurons. In <strong>the</strong><br />

lesioned mouse brains (SCID), NPSCs exhibited a significant targeted migration towards <strong>the</strong><br />

damaged regions <strong>of</strong> <strong>the</strong> brain, where <strong>the</strong>y engrafted, proliferated and matured into functional<br />

neurons. These NPSCs were intravenously administered and migrated into brain damaged<br />

areas and induce functional recovery. These results in animal models <strong>of</strong> AD suggest that<br />

transplanted NPSCs survive, migrate, and differentiate into cholinergic neurons, astrocytes, and<br />

oligodendrocytes with possible amelioration <strong>of</strong> <strong>the</strong> learning/memory deficits. In addition to<br />

replenish lost or damaged cells, NPSCs most likely stimulate endogenous neural precursors,<br />

enhance structural neuroplasticity, and down regulate proinflammatorycytokines and neuronal<br />

apoptotic death. Fur<strong>the</strong>r, <strong>the</strong> NPSCs could be genetically modified to express growth factors<br />

into <strong>the</strong> brain. An attempt at mobilizing this endogenous pool <strong>of</strong> resident neuronal progenitor<br />

stem cell-like cells would provide ano<strong>the</strong>r attractive approach for <strong>the</strong> treatment <strong>of</strong> AD. This<br />

paved <strong>the</strong> way towards exploring <strong>the</strong> possible cell based <strong>the</strong>rapy <strong>of</strong> neurogenesis


which would <strong>of</strong>fer an alternative approach for <strong>the</strong> treatment <strong>of</strong> AD.<br />

<strong>SUNDAY</strong><br />

2090<br />

DSCAM Associates with UNC5C in Netrin-1-Mediated Growth Cone Collapse.<br />

A. Purohit 1 , C. Qu 1 , W. Li 2 , K-L. Guan 3 , G. Liu 1 ; 1 Department <strong>of</strong> Biological Sciences, University<br />

<strong>of</strong> Toledo, Toledo, OH, 2 University <strong>of</strong> Michigan Health Systems, University <strong>of</strong> Michigan, Ann<br />

Arbor, MI, 3 Department <strong>of</strong> Pharmacology, University <strong>of</strong> California San Diego, La Jolla, CA<br />

In <strong>the</strong> developing nervous system, neuronal growth cones explore <strong>the</strong> extracellular environment<br />

for guidance cues, which can attract or repel axons and guide <strong>the</strong>m along specific trajectories<br />

towards <strong>the</strong>ir targets. Netrin-1, a bifunctional guidance cue, binds to receptors deleted in<br />

colorectal cancer (DCC) and Down syndrome cell adhesion molecule (DSCAM) mediating axon<br />

attraction, and uncoordinated-5 (UNC5) mediating axon repulsion. Here, we show that DSCAM<br />

interacts with UNC5C and netrin-1 stimulation increases this interaction in primary cortical<br />

neurons and postnatal cerebellar granule cells. Netrin-1 induces mouse cerebellum external<br />

granule layer (EGL) cell growth cone collapse and knocking-down DSCAM or UNC5C, by<br />

specific shRNAs or blocking <strong>the</strong>ir signaling by over-expressing mutants lacking <strong>the</strong> intracellular<br />

domains, eliminates netrin-1-induced growth cone collapse. Simultaneous knockdown <strong>of</strong><br />

DSCAM and UNC5C blocks netrin-1 induced growth cone collapse in EGL cells as well. Netrin-<br />

1 increases <strong>the</strong> tyrosine phosphorylation <strong>of</strong> endogenous DSCAM, UNC5C, FAK, Fyn, and<br />

PAK1, and promotes <strong>the</strong> formation <strong>of</strong> a protein-protein interaction complex <strong>of</strong> DSCAM with<br />

<strong>the</strong>se signaling molecules in primary postnatal cerebellar neurons. PP2, a Src family kinase<br />

inhibitor, efficiently inhibits <strong>the</strong> tyrosine phosphorylation <strong>of</strong> <strong>the</strong>se molecules and netrin-1-induced<br />

mouse EGL cell growth cone collapse. These data present <strong>the</strong> first evidence that DSCAM<br />

coordinates with UNC5C in netrin-1 repulsion.<br />

2091<br />

p75 neurotrophin receptor mediates ephrin-B3 signaling for axonal growth inhibition.<br />

N. Uesugi 1 , T. Yamashita 1 ; 1 Osaka University, Suita, Japan<br />

p75 neurotrophin receptor, a member <strong>of</strong> <strong>the</strong> TNF (tumor necrosis factor) receptor family, is<br />

known as a key molecule for axonal growth inhibition in <strong>the</strong> central nervous system (CNS). Axon<br />

elongation in <strong>the</strong> CNS is inhibited by molecules expressed in oligodendrocytes, e.g. MAG,<br />

Nogo, OMgp. These inhibitory molecules bind to p75 receptor indirectly, activate RhoA, known<br />

as a cytoskeletal regulator, thus suppressing axonal elongation. It has been reported that<br />

ephrin-B3, a member <strong>of</strong> <strong>the</strong> ephrin family, is also expressed in oligodendrocytes in <strong>the</strong> CNS and<br />

inhibits axonal elongation in vitro. Here we report that p75 neurotrophin receptor mediates <strong>the</strong><br />

signal <strong>of</strong> ephrin-B3 for axonal growth inhibition. The axonal growth <strong>of</strong> cortical neurons was<br />

inhibited on ephrin-B3 coated dishes. This inhibitory effect <strong>of</strong> ephrin-B3 was reversed by<br />

treatment with Pep5, an inhibitory peptide <strong>of</strong> <strong>the</strong> signal transduction <strong>of</strong> p75 receptor.<br />

Fur<strong>the</strong>rmore, knockdown <strong>of</strong> p75 receptor by siRNA also silenced <strong>the</strong> inhibitory effect <strong>of</strong> ephrin-<br />

B3. Our results reveal a novel signaling cascade <strong>of</strong> ephrin-B3-induced axonal growth inhibition<br />

via p75 receptor. Moreover, in this study, we assessed if regeneration <strong>of</strong> optic nerve was<br />

induced by Pep5 application after crush injury.


2092<br />

Maintenance <strong>of</strong> Dendritic Spine Morphology by PAR1b through Regulation <strong>of</strong><br />

Microtubule Growth.<br />

K. Hayashi 1 , A. Suzuki 1 , C. C. Hoogenraad 2 , S. Ohno 1 ; 1 <strong>Molecular</strong> and Cellullar <strong>Biology</strong>,<br />

Yokohama City University Graduate School <strong>of</strong> Medical Science, Yokohama, Japan, 2 Cell<br />

<strong>Biology</strong>, Faculty <strong>of</strong> Science, Utrecht University, The Ne<strong>the</strong>rlands<br />

<strong>SUNDAY</strong><br />

Dendritic spines are postsynaptic structures that receive excitatory synaptic input from<br />

presynaptic terminals. Actin and its regulatory proteins play a central role in morphogenesis <strong>of</strong><br />

dendritic spines. In addition, recent studies have revealed that microtubules are indispensable<br />

for <strong>the</strong> maintenance <strong>of</strong> mature dendritic spine morphology by stochastically invading dendritic<br />

spines and regulating dendritic localization <strong>of</strong> p140Cap, which is required for actin<br />

reorganization. However, <strong>the</strong> regulatory mechanisms <strong>of</strong> microtubule dynamics remains poorly<br />

understood.<br />

PAR1b, a cell polarity-regulating serine/threonine protein kinase, is thought to regulate<br />

microtubule dynamics by inhibiting microtubule binding <strong>of</strong> microtubule-associated proteins.<br />

Results from <strong>the</strong> present study demonstrated that PAR1b participates in <strong>the</strong> maintenance <strong>of</strong><br />

mature dendritic spine morphology. Immun<strong>of</strong>luorescent analysis revealed PAR1b localization in<br />

<strong>the</strong> dendrites, which was concentrated in dendritic spines <strong>of</strong> mature neurons. PAR1b<br />

knockdown cells exhibited decreased mushroom-like dendritic spines, as well as increased<br />

filopodia-like dendritic protrusions, with no effect on <strong>the</strong> number <strong>of</strong> protrusions. Live imaging <strong>of</strong><br />

microtubule plus-end tracking proteins directly revealed decreases in distance and duration <strong>of</strong><br />

microtubule growth following PAR1b knockdown in a neuroblastoma cell line and in dendrites <strong>of</strong><br />

hippocampal neurons. In addition, reduced accumulation <strong>of</strong> GFP-p140Cap in dendritic<br />

protrusions was confirmed in PAR1b knockdown neurons. In conclusion, <strong>the</strong> present results<br />

suggested a novel function for PAR1b in <strong>the</strong> maintenance <strong>of</strong> mature dendritic spine morphology<br />

by regulating microtubule growth and <strong>the</strong> accumulation <strong>of</strong> p140Cap in dendritic spines.<br />

2093<br />

PDZ-RhoGEF ubiquitination by Cullin3-KLHL20 controls neurotrophin-induced neurite<br />

outgrowth.<br />

M. Y. Lin 1,2 , Y. M. Lin 1,3 , T. C. Kao 1,3 , H. H. Chuang 1 , R. H. Chen 1,3 ; 1 Institute <strong>of</strong> Biological<br />

Chemistry, Academia Sinica, Taipei, Taiwan, 2 Institute <strong>of</strong> <strong>Molecular</strong> Medicine, College <strong>of</strong><br />

Medicien, National Taiwan University, Taipei, Taiwan, 3 Institute <strong>of</strong> Biochemical Science,<br />

National Taiwan University, Taipei, Taiwan<br />

The induction <strong>of</strong> neurite outgrowth and arborization is critical for developmental and<br />

regenerative processes. Here we report that <strong>the</strong> BTB-kelch protein KLHL20 promoted neurite<br />

outgrowth and arborization in hippocampal and cortical neurons through its interaction with<br />

Cullin3 to form a ubiquitin ligase complex. This complex targeted PDZ-RhoGEF, a protein<br />

abundantly expressed in brain, for ubiquitin-dependent proteolysis, <strong>the</strong>reby restricting RhoA<br />

activity and facilitating growth cone spreading and neurite outgrowth. Importantly, targeting<br />

PDZ-RhoGEF to KLHL20 required PDZ-RhoGEF phosphorylation by p38 MAPK. In response to<br />

p38-activating neurotrophins, such as brain-derived neurotrophic factor and neurotrophin-3,<br />

KLHL20-mediated PDZ-RhoGEF destruction was potentiated, leading to neurotrophin-induced<br />

neurite outgrowth. Our study identified a ubiquitin-dependent pathway that targets PDZ-<br />

RhoGEF destruction to facilitate neurite outgrowth, and indicates a key role <strong>of</strong> this pathway in<br />

neurotrophin-induced neuronal morphogenesis.


<strong>SUNDAY</strong><br />

2094<br />

c-Abl inhibition decreases <strong>the</strong> levels <strong>of</strong> Amyloid beta peptide in plasma <strong>of</strong> Alzheimer<br />

transgenic mice.<br />

L. D. Estrada 1 , D. chamorro 1 , N. C. inestrosa 2 , A. R. alvarez 1 ; 1 Lab. Señalizacion celular, Depto<br />

Biología Celular y <strong>Molecular</strong>, P. University <strong>of</strong> Catolica De Chile, Santiago, Chile, 2 Centro de<br />

Envejecimiento y Regeneración, P. Universidad Catolica de Chile, Santiago, Chile<br />

Introduction: AD is a progressive degenerative disorder characterized by memory loss,<br />

confusion and a variety <strong>of</strong> cognitive disabilities. The major neuropathological features <strong>of</strong> AD are<br />

<strong>the</strong> presence <strong>of</strong> amyloid plaques and neur<strong>of</strong>ibrillary tangles. The main component <strong>of</strong> amyloid<br />

plaques is <strong>the</strong> amyloid-beta peptide (Aβ), which is a 39-42 amino acid peptide part <strong>of</strong> a much<br />

larger transmembrane protein; <strong>the</strong> amyloid precursor protein (APP). Accumulation <strong>of</strong> toxic Aβoligomers<br />

in <strong>the</strong> brain <strong>of</strong> <strong>the</strong> AD patients appears to be central in <strong>the</strong> pathogenesis <strong>of</strong> AD.<br />

Our group previously report a dramatic clearance <strong>of</strong> amyloid plaques in old AD transgenic mice<br />

treated with <strong>the</strong> c-Abl kinase inhibitor; STI571.<br />

Objective:<br />

The aim <strong>of</strong> this work was to investigated <strong>the</strong> effect <strong>of</strong> STI571 on peripheral blood and evaluate<br />

<strong>the</strong> cellular mechanisms involved in this clearance.<br />

Material and Methods:<br />

APPsw/PSEN1ΔE9 mice (6-months) were treated with imatinib (STI571, Novartis) or vehicle<br />

every o<strong>the</strong>r day for 14-days by intraperitoneal injection. Plasma fractions were isolated using a<br />

Ficoll gradient and amyloid beta was measured using an anti-A� ELISA. Brains were removed<br />

and fixed in paraformaldehide for histological analysis. In addition, primary hippocampal<br />

neurons and HT22-hippocampal cells lines expressing APP-GFP were treated with STI571 and<br />

APP localization and processing was evaluated.<br />

Results: The inhibition <strong>of</strong> c-abl by peripheral administration <strong>of</strong> <strong>the</strong> inhibitor STI571 decreased<br />

both, cerebral and plasmatic levels <strong>of</strong> A�-oligomers in APPswe/PSEN1ΔE9 transgenic mice. In<br />

addition, hippocampal neurons and HT22-APP(swe) cells treated with STI571 show differences<br />

in <strong>the</strong> distribution <strong>of</strong> APP and decreased levels <strong>of</strong> A� in comparison with control cells.<br />

Conclusion: It has been shown that c-Abl participates in endosomal regulation and since APP<br />

amyloidogenic proteolytic cleavage occurs in endosomal compartments, we believe that this<br />

processing may be regulated by inhibiting c-abl by STI571, thus decreasing A� production.<br />

Besides, <strong>the</strong> fact that <strong>the</strong> administration <strong>of</strong> STI571, a drug that poorly cross <strong>the</strong> blood–brain<br />

barrier, reduced accumulation <strong>of</strong> Aβ in both peripherical blood and <strong>the</strong> brain, suggest a<br />

peripheral clearance <strong>of</strong> A�. The findings <strong>of</strong> <strong>the</strong>se experiments support a <strong>the</strong>rapeutic value for<br />

STI571 and related compounds in Alzheimer's disease.<br />

Fondecyt-Postoctorado-3110052/Fondecyt-1080221<br />

2095<br />

Septin-dependent microtubule control during mammalian neurite outgrowth.<br />

N. Ageta-Ishihara 1 , M. Kinoshita 1 ; 1 Graduate School <strong>of</strong> Science, Nagoya University, Nagoya,<br />

Japan<br />

The outgrowth, arborization, connection and functional organization <strong>of</strong> neurites (axons and<br />

dendrites) are prerequisites for <strong>the</strong> development <strong>of</strong> <strong>the</strong> nervous system. Previous studies<br />

revealed <strong>the</strong> implications <strong>of</strong> mammalian septins in multiple steps in neural development<br />

including neuritogenesis and synaptogenesis. However, <strong>the</strong> mechanism <strong>of</strong> action remains<br />

unclear due to <strong>the</strong> functional redundancy and pleiotropic roles <strong>of</strong> this cytoskeletal/scaffold<br />

protein family. Here we focus on <strong>the</strong> role <strong>of</strong> septins in neurite outgrowth <strong>of</strong> mouse cerebral<br />

cortical neurons. Quantitative morphometric analysis demonstrated that shRNA-mediated<br />

depletion <strong>of</strong> <strong>the</strong> pivotal subunit SEPT7 inhibited <strong>the</strong> outgrowth <strong>of</strong> both axons and dendrites in


<strong>SUNDAY</strong><br />

vitro, which was recapitulated in vivo. Septin-depleted neurons contained more acetylated αtubulin,<br />

indicating hyperstabilized microtubules are responsible for <strong>the</strong> phenotype at least in<br />

part. Tubacin, a small molecule inhibitor <strong>of</strong> <strong>the</strong> major tubulin deacetylase HDAC6 gave a<br />

phenocopy <strong>of</strong> septin depletion, i.e., neurite outgrowth and hyperacetylated α-tubulin.<br />

Biochemical analyses indicated that septins, HDAC6 and acetylated α-tubulin can physically<br />

interact and that septin depletion can reduce <strong>the</strong> interaction between HDAC6 and acetylated αtubulin.<br />

These and o<strong>the</strong>r data indicate that septins serve as a scaffold that facilitates <strong>the</strong><br />

interaction between HDAC6 and acetylated α/β-tubulin heterodimer, which ensures microtubule<br />

remodeling for neurite outgrowth by optimizing <strong>the</strong> acetylation level <strong>of</strong> α-tubulin.<br />

2096<br />

Neuroproteomic Analysis <strong>of</strong> Homer2 Knockout and Wildtype Mice Reveals Novel Gene<br />

Products Contributing to Alcohol Aversion Behavior.<br />

S. P. Goulding 1,2 , N. Shulman 3 , N. W. Bateman 2 , M. J. MacCoss 3 , K. K. Szumlinski 4 , C. C. Wu 2 ;<br />

1 Neuroscience Program, University <strong>of</strong> Colorado Anschutz Medical Campus, Aurora, CO,<br />

2 Department <strong>of</strong> Cell <strong>Biology</strong> and Physiology, University <strong>of</strong> Pittsburgh School <strong>of</strong> Medicine,<br />

Pittsburgh, PA, 3 Department <strong>of</strong> Genome Sciences, University <strong>of</strong> Washington, Seattle, WA,<br />

4 Department <strong>of</strong> Psychology and <strong>the</strong> Neuroscience Research Institute, University <strong>of</strong> California,<br />

Santa Barbara, Santa Barbara, CA<br />

In excitatory synapses <strong>of</strong> <strong>the</strong> mammalian central nervous system, <strong>the</strong> Homer family <strong>of</strong> proteins<br />

mediates excitatory signal transduction and receptor plasticity. The product <strong>of</strong> three genes,<br />

Homer1/2/3, this family shares domain features that include a conserved amino-terminal<br />

Ena/Vasp Homology 1 (EVH1) domain and two leucine zippers that form <strong>the</strong> hydrophobic core<br />

<strong>of</strong> a coiled-coil motif unique to <strong>the</strong> carboxyl-terminal <strong>of</strong> long, but not short, Homer is<strong>of</strong>orms.<br />

EVH1 containing Homers bind proline-rich sequences found in numerous postsynaptic<br />

glutamatergic signaling proteins such as group 1 metabotropic glutamate receptors (mGluRs)<br />

and <strong>the</strong> N-methyl D-aspartate (NMDA) receptor adaptor protein Shank. Coiled-coil, or long<br />

Homer is<strong>of</strong>orms, oligomerize into scaffolds and regulate signaling through interactions with<br />

cytoskeletal constituents like Drebrin, an actin-binding protein. Consistent with structural<br />

observations, Homer2 knockout mice present with numerous molecular, neurochemical and<br />

behavioral phenotypes in response to acute and chronic alcohol administration. Since ethanol<br />

alters glutamate neurotransmission in parts <strong>of</strong> <strong>the</strong> brain implicated in alcohol addiction, such as<br />

<strong>the</strong> hippocampus, and Homers regulate excitatory neural signaling through interactions with<br />

binding partners in <strong>the</strong> postsynaptic density (PSD), we set out to investigate alcohol avoiding<br />

behavior <strong>of</strong> <strong>the</strong> Homer2 knockout mouse at <strong>the</strong> neuroproteomic level. To better understand<br />

molecular mechanisms contributing to alcohol aversion, we studied differentially expressed<br />

peptides in <strong>the</strong> hippocampus <strong>of</strong> wildtype and Homer2 knockout mice using label-free mass<br />

spectrometry. By combining liquid chromatography-tandem mass spectrometry (LC-MS/MS)<br />

and a suite <strong>of</strong> s<strong>of</strong>tware tools capable <strong>of</strong> detecting chromatographic features <strong>of</strong> differential<br />

peptide peak areas between samples, we demonstrate that Homer2 deletion alters global gene<br />

products between wildtype and knockout mice. While Homer2 deletion is necessary and<br />

sufficient for alcohol aversion in mice, <strong>the</strong>se data suggest that differentially abundant gene<br />

products also contribute to <strong>the</strong> alcohol aversive phenotype and provide a readout <strong>of</strong> functional<br />

protein effectors <strong>of</strong> <strong>the</strong> Homer2 protein.


<strong>SUNDAY</strong><br />

2097<br />

Seizure susceptibility in mice lacking somatostatin type 3 receptors.<br />

S. K. Evans 1 , S. Lewis 1 , J. L. Fuchs 1 ; 1 Biological Sciences, University <strong>of</strong> North Texas, Denton,<br />

TX<br />

The role <strong>of</strong> primary cilia as essential mediators <strong>of</strong> cell proliferation in <strong>the</strong> developing brain is well<br />

established, but <strong>the</strong> function <strong>of</strong> cilia in mature, postmitotic neurons is just beginning to be<br />

investigated. The somatostatin type 3 receptor (Sst3R) is concentrated mainly on neuronal<br />

primary cilia in most brain regions. We found that this receptor was typically absent in cilia <strong>of</strong><br />

neural progenitors and immature neurons, but was present in cilia <strong>of</strong> more mature neurons in<br />

postnatal mice. Given <strong>the</strong> generally homeostatic, inhibitory, anti-mitotic effects <strong>of</strong> <strong>the</strong> hormone<br />

somatostatin, we hypo<strong>the</strong>sized that Sst3R on neuronal cilia serves to protect neurons against<br />

excitotoxicity, thus enhancing neuron survival in <strong>the</strong> face <strong>of</strong> stressful conditions. We compared<br />

seizure-related behaviors in C57BL/6 wildtype versus SstR3 knockout mice, in response to<br />

intranasal kainic acid (KA) administered under is<strong>of</strong>lurane anes<strong>the</strong>sia. Mice were observed<br />

periodically for 24 hours. The mutant mice showed behavioral symptoms <strong>of</strong> seizures at level 4-5<br />

on a standard seizure scale, where 0 is no symptoms and 5 is <strong>the</strong> most severe. In contrast,<br />

wildtypes scored only 0-1, even with up to two additional doses <strong>of</strong> KA (0.3 mg total per mouse).<br />

After a 3-day post-KA survival period, <strong>the</strong> brains <strong>of</strong> control and KA-treated mice were<br />

immunostained for GFAP to visualize fibrous astrocytes and adenylate cyclase type 3 to<br />

visualize primary cilia. KA-treated mutants showed evident astrogliosis, a hallmark <strong>of</strong> epilepsy,<br />

whereas KA-treated wildtypes showed only mild astrogliosis. We examined cilia in <strong>the</strong> hilus <strong>of</strong><br />

<strong>the</strong> dentate gyrus in all four groups <strong>of</strong> mice and found that KA administration was associated<br />

with reductions in <strong>the</strong> numerical density <strong>of</strong> immunostained cilia in <strong>the</strong> mutants. These<br />

observations support <strong>the</strong> hypo<strong>the</strong>sis that ciliary Sst3R has a neuroprotective role in mature,<br />

postmitotic neurons.<br />

2098<br />

SNX26, a GAP for Cdc42, Interacts with PSD-95 and Regulates Dendritic Spine Formation<br />

in Neurons.<br />

Y. Kim 1 , S. Chang 1 ; 1 Department <strong>of</strong> Physiology and Biomedical Sciences, Seoul National<br />

University College <strong>of</strong> Medicine, Seoul, Korea<br />

Dendritic spines are tiny protrusions composed <strong>of</strong> highly specialized actin micr<strong>of</strong>ilaments that<br />

receive <strong>the</strong> majority <strong>of</strong> excitatory glutamatergic synaptic inputs in <strong>the</strong> CNS. SNX26 (Sorting<br />

nexin 26) is a brain-enriched Rho GTPase-activating protein known to be localized in spines,<br />

though little is known about its neuronal function. Here, we report that SNX26 interacts with<br />

PSD-95, a well-known glutamatergic postsynaptic scaffolding protein and regulates spine<br />

formation in cultured hippocampal neurons. SNX26/TCGAP is present in dendritic spines where<br />

it colocalizes with PSD-95. We found that C-terminal domain <strong>of</strong> SNX26 interacts with SH3<br />

domain <strong>of</strong> PSD-95. Overexpression <strong>of</strong> SNX26 decreased <strong>the</strong> number <strong>of</strong> dendritic spines while it<br />

increased <strong>the</strong> number <strong>of</strong> filopodial form <strong>of</strong> protrusions. RhoGAP domain is responsible for its<br />

effect on spines to filopodia transition. Accordingly R350I, a GAP defective mutant form <strong>of</strong><br />

SNX26/TCGAP, has no effect on spines to filopodia transition. We fur<strong>the</strong>r found that expression<br />

<strong>of</strong> SNX26 largely decreases <strong>the</strong> F-actin contents in COS-7 cells as well as in neurons,<br />

suggesting that SNX26 as a RhoGAP destabilizes F-actin, which may mediate spines to<br />

filopodia transition in neurons. Evidently, knocking-down endogenous expression <strong>of</strong> SNX26 by<br />

shRNA affects <strong>the</strong> number and morphology <strong>of</strong> dendritic spines. Taken toge<strong>the</strong>r, our results<br />

indicate that SNX26 acts as a GAP for Cdc42, interacts with PSD-95 in dendritic spines, and<br />

regulates spine formation. Our results also raise <strong>the</strong> possibility that coordinated interaction <strong>of</strong>


<strong>SUNDAY</strong><br />

Cdc42 with SNX26 and o<strong>the</strong>r GEF proteins during dendritic spinogenesis in developing neurons<br />

may also play a pivotal role during synaptic plasticity in mature neurons.


MONDAY, <strong>DECEMBER</strong> 5- <strong>Late</strong> <strong>Abstracts</strong> 2<br />

Science Education<br />

MONDAY<br />

2099<br />

Industry Engagement in STEM Education.<br />

S. P. Denker 1 , L. Lindburg 2 , G. Maderis 3 ; 1 BayBio Institute, South San Francisco, CA, 2 BayBio<br />

Institute, 3 BayBio<br />

Bio-Community: Building <strong>the</strong> Next Generation <strong>of</strong> Life Science Pr<strong>of</strong>essionals is <strong>the</strong> BayBio<br />

Institute′s Science Education program aimed at bringing quality science, technology,<br />

engineering and math (STEM) educational experiences to students <strong>of</strong> diverse socio-economic<br />

and ethnic backgrounds. It is a hands-on program designed to connect students and educators<br />

to life science pr<strong>of</strong>essionals, and to bring <strong>the</strong>se pr<strong>of</strong>essionals as volunteers into middle/high<br />

school and community college classrooms. Members <strong>of</strong> <strong>the</strong> Bio-Community are Nor<strong>the</strong>rn<br />

California life science companies and educators committed to improving STEM education. The<br />

program and network provide a mechanism to establish <strong>the</strong> critical partnerships between<br />

industry, community based organizations and schools necessary to improve STEM education,<br />

teacher impact and student motivation. The objectives <strong>of</strong> Bio-Community are to 1) enhance<br />

STEM education in <strong>the</strong> region′s middle, high schools, and community colleges, 2) engage life<br />

science employees and companies in STEM-related activities and 3) increase student interest in<br />

STEM subjects, STEM-related majors, and awareness <strong>of</strong> <strong>the</strong> variety <strong>of</strong> careers available within<br />

<strong>the</strong> life science industry. The program was developed and launched in Fall, 2010 and has<br />

reached nearly 900 students thus far.<br />

2100<br />

Case-based learning to understand Cell Communication.<br />

S. W. Gorsich 1 ; 1 <strong>Biology</strong>, Central Michigan University, Mount Pleasant, MI<br />

Cell Communication is an advanced biology course. The goal <strong>of</strong> this course is for students to<br />

learn various cell communication mechanisms, become familiar with critically evaluating <strong>the</strong><br />

primary literature, and to improve critical thinking skills. Thirteen students were enrolled in <strong>the</strong><br />

class and <strong>the</strong>y were divided into five groups with each group having a lap top computer. During<br />

<strong>the</strong> first week <strong>of</strong> <strong>the</strong> course an introduction to how cells communicate was given in a more<br />

traditional lecture style. In addition during this first week I introduced <strong>the</strong> Osternsky family to <strong>the</strong><br />

class. Beginning in <strong>the</strong> second week we began a 3-4 day cycle <strong>of</strong> 1) case presentation, 2)<br />

discussion <strong>of</strong> cell communication topic, and 3) discussion/presentation <strong>of</strong> primary literature.<br />

During <strong>the</strong> case presentation a member <strong>of</strong> <strong>the</strong> Osternsky family is presented, which <strong>of</strong>ten<br />

centers on a medical concern. As <strong>the</strong> case unfolds students are provided leading information for<br />

<strong>the</strong>m to think about and make predictions at each stage. At <strong>the</strong> end <strong>of</strong> <strong>the</strong> case students will<br />

diagnose what’s wrong with <strong>the</strong> family member and <strong>the</strong>n link it to a cell communication problem.<br />

For instance in week one <strong>the</strong> class diagnosed E. Osternsky as having diabetes. They <strong>the</strong>n<br />

found a primary paper connecting G-protein coupled receptors to diabetes. During <strong>the</strong> next<br />

class I provide an interactive lecture introducing <strong>the</strong> specific cell communication pathway <strong>the</strong><br />

students previously identified (e.g. G-protein coupled receptors). On <strong>the</strong> final day <strong>of</strong> <strong>the</strong> cycle<br />

we critically discuss <strong>the</strong> paper. Everyone in <strong>the</strong> class is expected to read <strong>the</strong> entire paper, but<br />

each group has one figure <strong>the</strong>y present. To assure students are reading <strong>the</strong> paper before class,<br />

individual and group quizzes are given over <strong>the</strong> main take home points <strong>of</strong> <strong>the</strong> paper and <strong>the</strong><br />

communication pathway being discussed. The three-part cycle is <strong>the</strong>n repeated with a different<br />

Osternsky family member. Some outcomes that have resulted include: 1) students came more<br />

prepared to class, 2) students became pr<strong>of</strong>icient in critically evaluating and presenting <strong>the</strong>


MONDAY<br />

primary literature, 3) improved attendance, 4) retention <strong>of</strong> material improved, and 5) at a<br />

qualitative level student engagement, critical thinking, curiosity, and enthusiasm were high, 6)<br />

improved confidence, and 7) student’s became invested in <strong>the</strong> individual family members.<br />

Within this course additional learning strategies were used. They include: multiple quizzing,<br />

research ethic case studies, and oral midterm and final exams. With <strong>the</strong> success <strong>of</strong> this course I<br />

plan to implement a similar strategy in my larger classes.<br />

2101<br />

Go Figure: Comparing Graphics in <strong>Biology</strong> Textbooks.<br />

K. Chiem 1 , N. K. Dave 1 , R. Ghasemian 1 , E. J. Ortiz 1 , D. R. Perusse 1 , J. T. Sandu 1 , I. M. Tabel 1 ,<br />

B. A. Velasco 1 , T. Vu 1 , M. L. Casem 1 , S. E. Walker 1 ; 1 Biological Science, California State<br />

University, Fullerton, Fullerton, CA<br />

The quality <strong>of</strong> a biology textbook can immensely affect a student’s ability to learn, comprehend<br />

and integrate concepts. Figures, drawings or photographs, are <strong>of</strong>ten used in textbooks to<br />

convey large and complex concepts that would be o<strong>the</strong>rwise difficult to follow. We explored <strong>the</strong><br />

quality and quantity <strong>of</strong> figures in majors, non-majors, and mixed biology textbooks. A rubric was<br />

developed to measure <strong>the</strong> quality <strong>of</strong> figures. Figures were evaluated for three different topics<br />

typically taught in a general biology course: meiosis, alternation <strong>of</strong> generations, and<br />

endosymbiosis. We hypo<strong>the</strong>sized that major’s textbooks would provide greater quality and<br />

quantity <strong>of</strong> figures than non-majors/mixed textbooks. On average <strong>the</strong> majors textbooks scored<br />

higher in all quality categories, except for figure location and number <strong>of</strong> figures when compared<br />

to non-majors. Major’s textbooks tend to include entire complex processes in one large figure<br />

versus a non-majors text that might break down a complicated, lengthy concept (e.g. meiosis)<br />

into smaller more easily understandable pieces.<br />

2102<br />

An interdisciplinary team approach in undergraduate biological instruction and research.<br />

V. C. Ware 1 , N. G. Simon 1 ; 1 Biological Sciences, Lehigh University, Bethlehem, PA<br />

Lehigh University has an undergraduate science education award from <strong>the</strong> Howard Hughes<br />

Medical Institute (HHMI) that prepares students to integrate perspectives from multiple<br />

disciplines to address complex questions in <strong>the</strong> biosciences. Lehigh is also a member <strong>of</strong> <strong>the</strong><br />

HHMI-sponsored Science Education Alliance (SEA), where students and faculty nationwide use<br />

multidisciplinary approaches in mycobacteriophage research. A curricular model around <strong>the</strong><br />

<strong>the</strong>me <strong>of</strong> “biosystems dynamics” begins in <strong>the</strong> first semester and provides continuing access to<br />

courses and research opportunities that emphasize multidisciplinary approaches. Key<br />

programmatic elements are a “Bioscience in <strong>the</strong> 21st Century” survey course and <strong>the</strong><br />

“Biosystems Dynamics Summer Institute (BDSI),” a 10-week, research-intensive experience for<br />

interdisciplinary teams <strong>of</strong> undergraduates, graduate students, and faculty. The survey course<br />

introduces students to diverse perspectives on bioscience issues through lectures from faculty<br />

spanning science, engineering, and o<strong>the</strong>r disciplines to imbue a broad, trans-disciplinary<br />

perspective as a core scientific philosophy. The BDSI has supported 23 research teams,<br />

spanning projects in cell and molecular biology, bioengineering, and neuroscience with faculty<br />

from ma<strong>the</strong>matics, electrical engineering, chemistry, mechanical engineering, chemical<br />

engineering, computer science and biology. Student team members (which have included some<br />

SEA alumni) are drawn from different disciplines to reinforce a multidisciplinary<br />

conceptualization to problem-solving. Participation in BDSI has had several positive effects:<br />

publications and presentations that include student authors; grant proposals to support<br />

continued collaboration; undergraduate participation in research laboratories by most students<br />

after <strong>the</strong> BDSI experience; and several graduates <strong>of</strong> <strong>the</strong> BDSI program altered <strong>the</strong>ir post-


MONDAY<br />

graduate plans to include graduate programs that <strong>of</strong>fered interdisciplinary opportunities or job<br />

opportunities in a discipline outside <strong>of</strong> <strong>the</strong>ir major. BDSI support has generated a “ripple effect”<br />

that significantly expanded collaborative, multidisciplinary research efforts on campus. As <strong>the</strong><br />

HHMI-supported program has evolved over <strong>the</strong> project period, we see changes in institutional<br />

culture in teaching and research strategies designed to address complex problems in <strong>the</strong> life<br />

sciences more broadly.<br />

2103<br />

Undergraduate research experiences based on genomic, proteomic and gene expression<br />

libraries from Tetrahymena: launching students into cellular research on ciliated protists.<br />

L. A. Hufnagel 1 ; 1 Cell Mol <strong>Biology</strong>, University <strong>of</strong> Rhode Island, Kingston, RI<br />

To encourage undergraduate students to consider research on <strong>the</strong> cell and molecular biology <strong>of</strong><br />

ciliated protists in <strong>the</strong>ir future career plans, I have established in my laboratory an informal<br />

educational program that combines training in 1) laboratory methods for Tetrahymena<br />

<strong>the</strong>rmophila, 2) in silico gene sequence analysis and 3) immunocytochemistry. Each student<br />

who joins my laboratory is encouraged to choose a human disease <strong>of</strong> interest, do background<br />

reading to establish <strong>the</strong> current understanding <strong>of</strong> molecular and cellular events underlying <strong>the</strong><br />

disease, and identify a protein <strong>of</strong> particular importance in <strong>the</strong> disease process. The student next<br />

learns to use BLAST-searching to identify possible T. <strong>the</strong>rmophila orthologues <strong>of</strong> <strong>the</strong> relevant<br />

protein, as well as orthologues in o<strong>the</strong>r organisms, from ciliates to humans, using portals such<br />

as TGD-Wiki and Pubmed. The orthologues identified in this way have <strong>of</strong>ten not yet been<br />

functionally characterized, so a variety <strong>of</strong> o<strong>the</strong>r on-line-accessible tools (available at NCBI, EBI<br />

and elsewhere) are <strong>the</strong>n used, to fur<strong>the</strong>r analyze <strong>the</strong> functional properties <strong>of</strong> <strong>the</strong> Tetrahymena<br />

orthologues, and <strong>the</strong>ir evolutionary relationships with orthologues in o<strong>the</strong>r protists, invertebrates<br />

and chordates. In preparation for immunocytochemical studies to characterize <strong>the</strong> spatial and<br />

temporal expression <strong>of</strong> <strong>the</strong> identified proteins, sequence alignment tools are used to compare<br />

sequences <strong>of</strong> immunogens used in preparation <strong>of</strong> commercially available antibodies with <strong>the</strong><br />

sequences <strong>of</strong> <strong>the</strong> tetrahymena orthologues. To determine whe<strong>the</strong>r <strong>the</strong> predicted protein may<br />

actually be expressed, <strong>the</strong> student learns to use <strong>the</strong> expression data available at TGED, which<br />

provides temporal information that also helps to predict when during <strong>the</strong> cell cycle, starvation<br />

and mating <strong>the</strong> cells will most likely immunolabel. Using this approach, my undergraduate<br />

students are 1) characterizing <strong>the</strong> cell surface distribution <strong>of</strong> a Tetrahymena ribosomal protein,<br />

phosphoprotein P0, identified as a possible target for vaccine development in apicomplexan<br />

parasites; 2) investigating <strong>the</strong> Tetrahymena orthologue <strong>of</strong> JAK2, a protein mutated in individuals<br />

with polycy<strong>the</strong>mia vera (a rare blood disorder), and 3) analyzing <strong>the</strong> Tetrahymena orthologue <strong>of</strong><br />

human FANCM, a protein implicated in Fanconi anemia and cancer.<br />

2104<br />

Converting a senior-level cell biology course to a fully active learning format.<br />

D. J. Mat<strong>the</strong>s 1 ; 1 Genetics, Cell <strong>Biology</strong> & Development, University <strong>of</strong> Minnesota, Minneapolis,<br />

MN<br />

While <strong>the</strong>re have been efforts nationwide to convert traditional introductory college biology<br />

courses to an active learning format, <strong>the</strong>re have been few reports <strong>of</strong> shifting high-enrollment<br />

senior-level cell biology courses to incorporate such innovative instructional strategies. Here I<br />

report such a course conversion, where a modified Team Based Learning approach has been<br />

adopted for cell biology courses with approximately 100 students meeting in a SCALE-UP style<br />

active learning classroom at a large public university. Particular strategies (cell biology concept<br />

inventories, learning readiness quizzes, team challenges, and a team project to design<br />

nanoparticle-based cell-targeting <strong>the</strong>rapies) will be described. Learning retention as assessed


MONDAY<br />

by pre- and post-test concept inventories has improved significantly with this course format<br />

compared to <strong>the</strong> same material presented by <strong>the</strong> same instructor in similar lecture format<br />

courses.<br />

Microtubule Cytoskeleton<br />

2105<br />

Analysis <strong>of</strong> <strong>the</strong> expression <strong>of</strong> microtubule plus-end tracking proteins (+TIPs) during<br />

Xenopus development.<br />

E. C. Park 1,2 , H. Lee 3 , M. Kim 3 , J-K. Han 3 , G-H. Kim 1,2 ; 1 Division <strong>of</strong> Life Science, Korea Basic<br />

Science Institute, Daejeon, Korea, 2 Pioneer Research Center for Protein Network Exploration,<br />

Korea Basic Science Institute, Daejeon, Korea, 3 Division <strong>of</strong> <strong>Molecular</strong> and Life Sciences,<br />

Pohang University <strong>of</strong> Science and Technology, Pohang, Korea<br />

Microtubules are a component <strong>of</strong> <strong>the</strong> cytoskeleton and are important for maintaining cell<br />

structure and providing platforms for intracelluar transport, as well as o<strong>the</strong>r cellular processes.<br />

Microtubule plus-end tracking proteins (+TIPs) are a structurally and functionally diverse group<br />

<strong>of</strong> proteins. +TIPs are specifically accumulated in microtubule plus end and regulate dynamic<br />

microtubules behavior. We characterized <strong>the</strong> +TIPs, XClip-170, Xp150glued, XClasp1b, XLis1,<br />

XStim1 and XStim2, in Xenopus laevis and report <strong>the</strong> expression patterns during<br />

embryogenesis. All six Xenopus +TIP genes are maternally expressed and show similar<br />

expression patterns during Xenopus embryo development. The expressions <strong>of</strong> Xenopus +TIPs<br />

are localized in <strong>the</strong> animal hemisphere and ectoderm region at early stages. As embryos<br />

develop to later stages, <strong>the</strong> ectodermal expression <strong>of</strong> Xenopus +TIPs persists in head and<br />

neural tube structures. XClasp1b and Xp150glued,especially, shows ectopic expressions in <strong>the</strong><br />

cranial ganglia. Remarkably, Xenopus +TIPs are also expressed in <strong>the</strong> involuting mesoderm<br />

during gastrulation. This is <strong>the</strong> first study <strong>of</strong> developmental expression patterns <strong>of</strong> +TIPs and our<br />

analysis highlights <strong>the</strong> future insight <strong>of</strong> research fields <strong>of</strong> microtubules in vertebrate<br />

development, gastrulation cell movements and neurogenesis.<br />

2106<br />

Nuclear gamma-tubulin associates with nucleoli and interacts with tumor suppressor<br />

protein C53.<br />

B. Horejší 1 , S. Vinopal 1 , E. Dráberová 1 , V. Sulimenko 1 , T. Sulimenko 1 , A. Philimonenko 2 , P.<br />

Hozák 2 , P. Dráber 1 ; 1 Dept. <strong>Biology</strong>. Cytoskeleton, Inst. Mol. Genet AS CR, Prague, Czech<br />

Republic, 2 Dept. Cell Nucleus, Inst. Mol. Genet AS CR, Prague<br />

γ-Tubulin is assumed to be a typical cytosolic protein necessary for nucleation <strong>of</strong> microtubules<br />

from microtubule organizing centers. Using immunolocalization and cell fractionation techniques<br />

in combination with siRNAi and expression <strong>of</strong> FLAG-tagged constructs, we have obtained<br />

evidence that γ-tubulin is also present in nucleoli <strong>of</strong> mammalian interphase cells <strong>of</strong> diverse<br />

cellular origins. Immunoelectron microscopy has revealed γ-tubulin localization outside fibrillar<br />

centers where transcription <strong>of</strong> ribosomal DNA takes place. γ-Tubulin was associated with<br />

nucleolar remnants after nuclear envelope breakdown and could be translocated to nucleoli<br />

during mitosis. Pretreatment <strong>of</strong> cells with leptomycin B did not affect <strong>the</strong> distribution <strong>of</strong> nuclear γtubulin,<br />

making it unlikely that rapid active transport via nuclear pores participates in <strong>the</strong><br />

transport <strong>of</strong> γ-tubulin into <strong>the</strong> nucleus. This finding was confirmed by heterokaryon assay and<br />

time-lapse imaging <strong>of</strong> photoconvertible protein Dendra2 tagged to γ-tubulin.<br />

Immunoprecipitation from nuclear extracts combined with mass spectrometry revealed an<br />

association <strong>of</strong> γ-tubulin with tumor suppressor protein C53 located at multiple subcellular


MONDAY<br />

compartments including nucleoli. The notion <strong>of</strong> an interaction between γ-tubulin and C53 was<br />

corroborated by pull-down and co-immunoprecipitation experiments. Overexpression <strong>of</strong> γtubulin<br />

antagonized <strong>the</strong> inhibitory effect <strong>of</strong> C53 on DNA damage G2/M checkpoint activation.<br />

The combined results indicate that aside from its known role in microtubule nucleation, γ-tubulin<br />

may also have nuclear-specific function(s).<br />

2107<br />

Cap-gly proteins at microtule plus ends: is EB1 detyrosination involved?<br />

A. Bosson 1 , J-M. Soleilhac 1 , O. Valiron 1 , D. Job 1 , A. Andrieux 1 , M-J. Moutin 1 ; Grenoble Institut<br />

Des Neurosciences, Grenoble, France<br />

Localization <strong>of</strong> CAP-Gly proteins such as CLIP170 at microtubule + ends results from <strong>the</strong>ir dual<br />

interaction with α-tubulin and EB1 through <strong>the</strong>ir C-terminal amino acids -EEY. Detyrosination <strong>of</strong><br />

α-tubulin by tubulin-carboxypeptidase abolishes CLIP170 binding. Can detyrosination affect<br />

EB1 as well? We developed specific antibodies to discriminate tyrosinated vs detyrosinated<br />

forms <strong>of</strong> EB1 and detected only tyrosinated EB1 in fibroblasts, astrocytes, and total brain tissue.<br />

Over-expressed EB1 was not detyrosinated in cells and chimeric EB1 with <strong>the</strong> eight C-terminal<br />

amino acids <strong>of</strong> α-tubulin was only barely detyrosinated. Our results indicate that detyrosination<br />

regulates CLIPs interaction with tubulin, but not with EB1. They highlight <strong>the</strong> specificity <strong>of</strong><br />

carboxypeptidase toward tubulin.<br />

2108<br />

Tubulin Acetylation Is Required for <strong>the</strong> Maintenance <strong>of</strong> Stable Microtubules Arrays<br />

During Cell Migration.<br />

Z. Xu 1,2 , M. Nachury 2 ; 1 Department <strong>of</strong> Dermotology, Stanford University School <strong>of</strong> Medicine,<br />

Palo Alto, CA, 2 Department <strong>of</strong> <strong>Molecular</strong> and Cellular Physiology, Stanford University School <strong>of</strong><br />

Medicine, Palo Alto, CA<br />

Migrating cells possess a subset <strong>of</strong> polarized stable microtubules oriented towards <strong>the</strong> leading<br />

edge. These long lived microtubules that are highly acetylated on lysine 40 site are generally<br />

considered as tracks for delivery <strong>of</strong> signaling molecular for cell migration. However, <strong>the</strong><br />

functional significance <strong>of</strong> tubulin acetylation is still elusive in large part because <strong>the</strong> tubulin<br />

acetyltransferase remains unidentified.<br />

Recently, in <strong>the</strong> course <strong>of</strong> proteomic study, we identified a protein with distant structural<br />

similarity to <strong>the</strong> GNAT clade <strong>of</strong> acetylatransferases that is exclusively and universally conserved<br />

in ciliated organisms. Strikingly, this putative acetyltransferase specifically acetylates a-tubulin<br />

at K40 in vitro and was <strong>the</strong>refore named aTAT1.<br />

In a variety <strong>of</strong> cell lines, overexpression <strong>of</strong> aTAT1 is sufficient to induce massive a-tubulin K40<br />

acetylation. Fur<strong>the</strong>rmore, a catalytically inactive mutant <strong>of</strong> aTAT1 fails to acetylate tubulin in<br />

vivo. Thus, <strong>the</strong> enzymatic activity <strong>of</strong> aTAT1 is necessary for acetylation a-tubulin at K40 in cells.<br />

Meanwhile, Depleting aTAT1 from hTERT-RPE cells abolishes MT acetylation and affects <strong>the</strong><br />

kinetics <strong>of</strong> primary cilium assembly. Additionally, aTAT1 is required for <strong>the</strong> maintenance <strong>of</strong><br />

stable microtubule arrays and for directional cell migration. Interestingly, we found that <strong>the</strong><br />

median length <strong>of</strong> nocodazole-resistant MTs falls from 4 μm in control-treated cells to less than 1<br />

μm in αTAT1-depleted cells. This result suggests that in <strong>the</strong> absence <strong>of</strong> acetylation, stable MTs<br />

become highly fragile and fragmented. We conclude that aTAT1 is <strong>the</strong> major a-tubulin K40<br />

acetylatransferase in mammalian cells and that a-tubulin K40 aceytlation enable <strong>the</strong> assembly<br />

<strong>of</strong> stable microtubule tracks utilized in various cellular processes.


MONDAY<br />

2109<br />

HURP regulates chromosome congression by modulating kinesin Kif18A function.<br />

F. Ye 1 , L. Tan 1 , Q. Yang 1 , Y. Xia 1 , L-W. Deng 2 , M. Murata-Hori 3 , Y-C. Liou 1 ; Department <strong>of</strong><br />

Biological Sciences, National University <strong>of</strong> Singapore, Singapore, 2 Department <strong>of</strong> Biochemistry,<br />

National University <strong>of</strong> Singapore, Singapore, 3 Temasek Life Sciences Laboratory, Singapore<br />

Chromosome biorientation and congression during mitosis require precise control <strong>of</strong> microtubule<br />

dynamics. The dynamics <strong>of</strong> kinetochore microtubules (K-MTs) are regulated by a variety <strong>of</strong><br />

microtubule associated proteins (MAPs). Recently, a MAP known as HURP (hepatoma upregulated<br />

protein) was identified. During mitosis, RanGTP releases HURP from <strong>the</strong> Importin â<br />

inhibitory complex and allows it to localize to <strong>the</strong> kinetochore fiber (K-fiber). HURP bundles and<br />

stabilizes K-fibers and promotes chromosome congression. However, <strong>the</strong> molecular mechanism<br />

underlying <strong>the</strong> role <strong>of</strong> HURP in regulating chromosome congression remains elusive. Here, we<br />

show that overexpression <strong>of</strong> <strong>the</strong> N-terminal microtubule binding domain (1-278 aa, HURP278)<br />

<strong>of</strong> HURP induces a series <strong>of</strong> mitotic defects including lagging chromosomes during <strong>the</strong> late<br />

prometaphase to metaphase stage, prolonged mitosis and increased kinetochore oscillation<br />

amplitude, mimicking <strong>the</strong> effects <strong>of</strong> Kif18A depletion. In addition, co-immunoprecipitation and<br />

BiFC assays identify Kif18A as a novel interaction partner <strong>of</strong> HURP. Fur<strong>the</strong>rmore, quantitative<br />

results from live cell imaging analyses illustrate that HURP regulates Kif18A localization and<br />

dynamics at <strong>the</strong> plus-end <strong>of</strong> K-MTs. Lastly, misaligned chromosomes in HURP278overexpressing<br />

cells can be partially rescued by <strong>the</strong> overexpression <strong>of</strong> Kif18A, suggesting <strong>the</strong><br />

specificity <strong>of</strong> Kif18A regulation by HURP. Our results demonstrate in part <strong>the</strong> regulatory<br />

mechanism for kinesin Kif18A at <strong>the</strong> K-MTs during chromosome congression and provide new<br />

insights into <strong>the</strong> mechanism <strong>of</strong> chromosome movement at <strong>the</strong> metaphase plate.<br />

2110<br />

MTWD40 is a Novel Microtubule Regulatory Protein Expressed in Neurons.<br />

W. Wang 1 , V. Lundin 1 , I. Millan 1 , M. Maloney 1 , M. Kapur 1 , N. Chen 1 ; 1 Neurology, Stanford<br />

School <strong>of</strong> Medicine, Stanford, CA<br />

Neurons are uniquely dependent on a highly regulated microtubule (MT) cytoskeleton. Several<br />

MT-associated proteins (MAPs) are involved in maintaining MT stability and dynamics, which<br />

support essential neuronal functions such as axonal transport. Given <strong>the</strong> critical role <strong>of</strong> MAPs in<br />

regulating MTs in both healthy and diseased neurons, we sought to understand how this<br />

regulation is achieved. Here, we identify a novel protein containing a WD40 repeat domain,<br />

tentatively named microtubule-related WD40 (MTWD40), as a potential regulator <strong>of</strong> MAPdependent<br />

MT dynamics. mRNA and protein expression analyses in mice indicate that<br />

MTWD40 is enriched in <strong>the</strong> nervous system. Protein expression is detected as early as E11 and<br />

continues throughout adulthood. At <strong>the</strong> cellular level, MTWD40 localizes as puncta along <strong>the</strong><br />

MT network in both COS7 cells and primary cortical neurons. Interestingly, co-overexpression<br />

with MAP8 causes MTWD40 to lose its punctate pattern and instead colocalize with MAP8<br />

uniformly along MTs. These results suggest that MTWD40 may serve an important role in<br />

regulating <strong>the</strong> neuronal cytoskeleton through an interaction with MAP8.


MONDAY<br />

2111<br />

Role <strong>the</strong> Microtubule (+)-End Binding protein EB1 in differentiating osteoblasts.<br />

S. Pustylnik 1 , N. Nabavi 2 , R. Harrison 3 ; 1 Department <strong>of</strong> Biological Sciences, University <strong>of</strong><br />

Toronto, Toronto, ON, Canada, 2 Department <strong>of</strong> Cell and Systems <strong>Biology</strong>, University <strong>of</strong> Toronto,<br />

3 University <strong>of</strong> Toronto, Toronto, ON, Canada<br />

Osteogenesis is a strictly controlled developmental process in which exogenous factors, such<br />

as hormones and growth factors activate osteoblast-specific signaling proteins and transcription<br />

factors required for osteoblast differentiation. Our microarray analysis identified <strong>the</strong> microtubule<br />

(+)-End-Binding protein (EB1) as an upregulated gene in 5-day ascorbate-induced<br />

differentiation <strong>of</strong> mouse MC3T3 mouse cells compared to undifferentiated control cells. The<br />

main functions <strong>of</strong> EB1 are regulation <strong>of</strong> microtubule dynamics, cell polarity, chromosome<br />

stability and cell migration. EB1 is known to bind to <strong>the</strong> tumor suppressor APC (Adenomatosis<br />

Polyposis Coli). This interaction has led to <strong>the</strong> speculation that EB1 is a component <strong>of</strong> <strong>the</strong><br />

canonical APC tumor suppressor pathway, which regulates <strong>the</strong> levels <strong>of</strong> β-catenin, a key Wnt<br />

signaling effector.<br />

In this study, we examined <strong>the</strong> role <strong>of</strong> EB1 in osteoblasts during differentiation to elucidate <strong>the</strong><br />

possible involvement <strong>of</strong> EB1 in Wnt signaling. Immunoblotting <strong>of</strong> extracts from undifferentiated<br />

and differentiated osteoblasts showed that EB1 could be detected in freshly plated cells and its<br />

level increases during <strong>the</strong> first 5 days <strong>of</strong> differentiation. Osteoblasts in which EB1 was knocked<br />

down by RNA interference showed changes in cell morphology and osteoblasts specific gene<br />

expression. EB1 and β-catenin co-immunoprecipitate in osteoblasts and we see an induction in<br />

β-catenin protein levels during differentiation using immunoblotting. Interestingly, EB1 protein<br />

knockdown led to a decrease <strong>of</strong> nuclear accumulation <strong>of</strong> β-catenin by immun<strong>of</strong>luorescence.<br />

These results suggest that EB1 is important for β-catenin degradation events that lead to its<br />

nuclear accumulation and potential activation <strong>of</strong> Wnt signaling genes. Variations in cell density<br />

significantly influenced <strong>the</strong> whole differentiation process while block <strong>of</strong> adherent junctions with<br />

E-cadherin antibody resulted in decreased cell-cell adhesions and altered bone matrix protein<br />

expression. We are now using live imaging to discern <strong>the</strong> cellular location <strong>of</strong> interactions<br />

between <strong>the</strong>se 2 proteins to show a definitive role for EB1 in osteoblast differentiation.<br />

2112<br />

Characterization <strong>of</strong> HIV Rev and Tubulin Interactions.<br />

J. L. Chang 1 , B. E. Dukes 1 , M. W. Miller 1 ; 1 Biological Sciences, Wright State University, Dayton,<br />

OH<br />

The HIV Rev protein has <strong>the</strong> ability to bind tubulin heterodimers and depolymerize microtubules<br />

(MTs) in vitro producing bilayered rings called Rev-tubulin toroids (RTTs) (Watts et al. 2000. J.<br />

Cell Biol. 150: 349-360). These interactions may account for MT defects observed in HIV<br />

infected cells or cells that over-express Rev. Watts et al. hypo<strong>the</strong>sized Rev interacts with MTs<br />

by a mechanism shared with Kinesin-13 (Kin13) proteins owing to <strong>the</strong> presence <strong>of</strong> a shared<br />

amino acid sequence. Kin13 proteins are potent MT depolymerizing agents affecting MT<br />

behavior during mitosis.<br />

To test this hypo<strong>the</strong>sis, point mutations were introduced into Rev substituting amino acids<br />

shared with Kin13. In one set <strong>of</strong> experiments, <strong>the</strong> abilities <strong>of</strong> four mutant Rev proteins (A37D,<br />

R42A, E47A, and E57A) to form RTTs when mixed with colchicine-treated tubulin were<br />

compared to wild-type Rev. With <strong>the</strong> exception <strong>of</strong> RevR42A, wild-type and mutant Rev formed<br />

comparably sized RTTs at a high frequency. In contrast, mutating arginine at position 42 to<br />

alanine completely abrogated formation <strong>of</strong> RTTs. This observation suggests this amino acid is<br />

critical for Rev-tubulin interactions, consistent with our hypo<strong>the</strong>sis.


MONDAY<br />

In a second set <strong>of</strong> experiments, wild-type and seven mutant Rev proteins (T34A, A37D, R39A,<br />

R42A, E47A, R50A, E57A, and <strong>the</strong> E47A/E57A double mutant E2A) fused to YFP were overexpressed<br />

in HeLa cells and <strong>the</strong> ability <strong>of</strong> <strong>the</strong>se cells to grow in culture and elaborate MT<br />

cytoskeletons was measured. Previous results show over-expression <strong>of</strong> wild-type Rev slows<br />

growth in culture and alters cell cycle progression. If <strong>the</strong>se defects are due to Rev-MT<br />

interactions, mutation <strong>of</strong> residues critical for <strong>the</strong>se interactions should mitigate <strong>the</strong>se defects.<br />

Several Rev mutants, notably A37D, E47A and <strong>the</strong> double mutant E47A/E57A (E2A), alter cell<br />

morphology and growth in culture suggesting <strong>the</strong> affected amino acids are dispensable.<br />

Curiously, over-expression <strong>of</strong> RevR42A also alters doubling times and increases <strong>the</strong> frequency<br />

<strong>of</strong> multinucleated cells. Taken toge<strong>the</strong>r, Rev over-expression can lead to defects not directly<br />

attributable to Rev-MT interactions.<br />

2113<br />

Differential Interaction <strong>of</strong> Dystrophin and Utrophin with Microtubules.<br />

J. J. Belanto 1,2 , D. M. Henderson 1 , M. A. Jaeger 1 , J. M. Ervasti 1 ; 1 Biochemisty, <strong>Molecular</strong><br />

<strong>Biology</strong>, and Biophysics, University <strong>of</strong> Minnesota, Minnapolis, MN, 2 <strong>Molecular</strong>, Cellular,<br />

Developmental <strong>Biology</strong>, and Genetics, University <strong>of</strong> Minnesota, Minneapolis, MN<br />

The dystrophin gene resides on <strong>the</strong> X chromosome and encodes a 427 kD cytoplasmic protein<br />

expressed in striated muscle which links <strong>the</strong> costameric actin cytoskeleton to <strong>the</strong> extracellular<br />

matrix via a sarcolemmal-associated glycoprotein complex. Mutations in dystrophin that abolish<br />

or reduce its expression lead to Duchenne muscular dystrophy (DMD) or Becker muscular<br />

dystrophy (BMD), respectively. Patients with DMD succumb to fatal cardiac and/or respiratory<br />

failure in <strong>the</strong>ir late teens to early twenties while BMD patients are more mildly affected.<br />

Currently, <strong>the</strong>re is no effective treatment available for DMD or BMD.<br />

Our lab previously demonstrated that dystrophin directly binds and organizes microtubules into<br />

a rectilinear lattice beneath <strong>the</strong> sarcolemma (Prins et al., J. Cell Biol. 2009). Using an in vitro<br />

cosedimentation assay, we show that dystrophin binds to microtubules with high affinity<br />

(KD=0.20µM). We also show that transgenic expression <strong>of</strong> nearly full-length dystrophin on <strong>the</strong><br />

dystrophin-deficient mdx background restores microtubule lattice organization whereas mdx<br />

mice present with a disorganized microtubule lattice. Based on cosedimentation assays with an<br />

array <strong>of</strong> short dystrophin is<strong>of</strong>orms and truncation constructs, we conclude/suggest that<br />

microtubule binding activity maps to a region located between spectrin-like repeats 9 and 19 <strong>of</strong><br />

<strong>the</strong> large middle rod domain.<br />

Utrophin is <strong>the</strong> fetal homolog <strong>of</strong> dystrophin that is down-regulated in skeletal muscle at birth but<br />

ubiquitously expressed at low levels in all tissues, including skeletal muscle, in adults. Because<br />

utrophin binds to nearly all <strong>of</strong> dystrophin’s previously identified molecular partners, several<br />

<strong>the</strong>rapies under development for DMD aim to upregulate utrophin in skeletal muscle. We show<br />

that utrophin binds microtubules in vitro with 10-fold lower affinity (KD=2.34µM) than dystrophin<br />

and that transgenic over-expression <strong>of</strong> utrophin is not sufficient to rescue <strong>the</strong> disorganized<br />

microtubule network in mdx muscle. Our results suggest that any deficiency in microtubule<br />

trafficking or function caused by loss <strong>of</strong> dystrophin may not be restored by upregulation <strong>of</strong><br />

utrophin. Therefore, it remains important to determine whe<strong>the</strong>r any specific pathologies can be<br />

attributed to <strong>the</strong> microtubule disorganization associated with loss <strong>of</strong> dystrophin protein<br />

expression.<br />

This work was supported by <strong>the</strong> NIH Training Program in Muscle Research AR007612 and NIH<br />

RO1 AR042423.


2114<br />

XMAP215 uses avidity to promote microtubule growth.<br />

P. O. Widlund 1 , J. Howard 1 , A. A. Hyman 1 ; 1 Max Planck Inst Moleculcar and Cell<br />

Biolology/Genetics, Dresden, Germany<br />

MONDAY<br />

Microtubule-associated proteins adjust <strong>the</strong> dynamic properties <strong>of</strong> microtubules so that <strong>the</strong>y can<br />

be used to carry out cellular functions such as chromosome segregation, vesicle transport, and<br />

cell motility. The XMAP215/Dis1 family <strong>of</strong> proteins has been shown to dramatically promote<br />

microtubule growth and <strong>the</strong>ir activities significantly contribute to <strong>the</strong> rapid growth rates <strong>of</strong><br />

microtubules seen in cells. Repeats at <strong>the</strong>ir N-termini, called TOG domains, are important for<br />

this function. While TOG domains directly bind tubulin dimers, it is unclear how this interaction<br />

translates to polymerase activity. Understanding <strong>the</strong> functional roles <strong>of</strong> TOG domains is fur<strong>the</strong>r<br />

complicated by <strong>the</strong> fact that <strong>the</strong> number <strong>of</strong> <strong>the</strong>se domains present in <strong>the</strong> proteins <strong>of</strong> different<br />

species varies. We determined <strong>the</strong> contributions <strong>of</strong> <strong>the</strong> individual TOG domains to microtubule<br />

growth and looked at <strong>the</strong> affinities <strong>of</strong> <strong>the</strong> TOG domains for tubulin. Based on <strong>the</strong>se experiments,<br />

we have developed a model where TOG domains each have low affinity for tubulin dimers, but<br />

<strong>the</strong>ir combination allows for high overall avidity. This makes it possible for TOG proteins to<br />

efficiently recruit tubulin dimers, but also be able to release <strong>the</strong>m into <strong>the</strong> growing microtubule<br />

end.<br />

2115<br />

A High-Throughput and High-Content Microscopy Approach to Identify Microtubule<br />

Regulators.<br />

X. E. Studera 1,2 , V. Graml 1,2 , M. Bortfeld-Miller 2 , R. E. Carazo Salas 1,2 ; 1 The Wellcome<br />

Trust/Cancer Research UK Gurdon Institute, Cambridge, United Kingdom, 2 Institute <strong>of</strong><br />

Biochemistry/ETH Zurich, Zurich, Switzerland<br />

Microtubules (MT) are key determinants <strong>of</strong> countless eukaryotic cellular features, such as cell<br />

shape, intracellular transport, organelle positioning and cell division, yet to this day a<br />

comprehensive (genomic) catalogue <strong>of</strong> microtubule regulators is lacking even for <strong>the</strong> simplest<br />

cells.<br />

We have carried out a screen for novel microtubule regulators, using a commercial genomewide<br />

gene-knockout (‘deletion’) collection <strong>of</strong> haploid mutant strains <strong>of</strong> <strong>the</strong> fission yeast<br />

(Schizosaccharomyces pombe) and high-throughput/high-content microscopy. So far, only 26<br />

genes are known from previous studies to be bona fide microtubule regulators in fission yeast,<br />

although <strong>the</strong> Gene Ontology database suggests that <strong>the</strong>re could be a few hundred more.<br />

Using a high-throughput spinning disk confocal microscope, we imaged approximately 3 000<br />

non-essential gene knockout strains expressing GFP-labelled tubulin, allowing us to inspect<br />

<strong>the</strong>ir intact microtubules in live cells. The images were screened visually to look for strains<br />

displaying microtubule defects. Over 500 000 images were also analyzed computationally using<br />

custom-made, automated microtubule detection algorithms to look for knockouts with<br />

statistically significantly aberrant microtubule structures.<br />

After preliminary quality control, we have identified approximately 100 gene knockouts<br />

displaying a phenotypic defect in microtubule organization. About 40 % <strong>of</strong> <strong>the</strong> corresponding<br />

genes have homologues in o<strong>the</strong>r organisms and 50 % <strong>of</strong> <strong>the</strong>m have been experimentally<br />

characterized but not reported to regulate microtubules, and are hence potentially novel<br />

regulators. We are in <strong>the</strong> course <strong>of</strong> carrying out fur<strong>the</strong>r quality control and detailed strain<br />

analysis to confirm which <strong>of</strong> those preliminary hits are conserved, new microtubule regulators


MONDAY<br />

and to clarify <strong>the</strong>ir mode <strong>of</strong> action. Here we describe <strong>the</strong> strategy <strong>of</strong> our screen and our<br />

preliminary results.<br />

2116<br />

Calcium-Dependent Reorganization <strong>of</strong> Microtubules in Activated Mast Cells.<br />

V. Sulimenko 1 , Z. Hájková 1 , S. Vinopal 1 , E. Dráberová 1 , P. Dráber 1 ; 1 Institute <strong>of</strong> <strong>Molecular</strong><br />

Genetics, Prague, Czech Republic<br />

Mast cells play a pivotal role in innate immunity, allergy and inflammation. They express plasma<br />

membrane-associated high-affinity IgE receptors (FcεRIs), <strong>the</strong> aggregation <strong>of</strong> which by<br />

multivalent antigen-IgE complexes triggers mast cell activation resulting in degranulation and<br />

release <strong>of</strong> inflammatory mediators such as histamine, proteases, lipid mediators, and cytokines.<br />

Tyrosine kinases <strong>of</strong> <strong>the</strong> Src family are crucial for triggered signaling pathways. Although FcεRI<br />

aggregation leads to reorganization <strong>of</strong> microtubules and <strong>the</strong>ir accumulation in <strong>the</strong> cell periphery<br />

(J. Cell Biol. 170:115,2005; J. Immunol. 176:7243,2006), <strong>the</strong> molecular mechanisms that control<br />

microtubule rearrangement after cell activation are poorly understood. Here we show that<br />

changes in cytosolic Ca 2+ concentration, controlled by store-operated Ca 2+ entry (SOCE),<br />

affected microtubule plus-end dynamics detected by plus-end tracking protein EB1 and led to<br />

generation <strong>of</strong> protrusions containing microtubules. Experiments with knock-down or<br />

reexpression <strong>of</strong> STIM1, <strong>the</strong> essential regulator <strong>of</strong> SOCE, confirmed <strong>the</strong> important role <strong>of</strong> STIM1<br />

in this process. Changes in Ca 2+ concentration also modulated interactions <strong>of</strong> γ-tubulin, <strong>the</strong> key<br />

player in microtubule nucleation, with γ-tubulin complex proteins (GCPs) as well as with o<strong>the</strong>r<br />

proteins that are substrates for tyrosine kinases <strong>of</strong> <strong>the</strong> Src family. Combined data suggest that<br />

rearrangement <strong>of</strong> microtubules in activated mast cells depends on STIM1-induced SOCE, and<br />

that Ca 2+ regulates <strong>the</strong> formation <strong>of</strong> microtubule protrusions in activated mast cells.<br />

2117<br />

Assembly <strong>of</strong> a millimeter-sized microtubule aster in a model cytoplasm.<br />

K. Ishihara 1 , C. M. Field 1 , A. C. Groen 1 , P. A. Nguyen 1 , T. J. Mitchison 1 ; 1 Department <strong>of</strong><br />

Systems <strong>Biology</strong>, Harvard Medical School, Boston, MA<br />

The cytoplasms <strong>of</strong> large egg and embryonic cells are organized primarily by radial arrays <strong>of</strong><br />

microtubules called asters. Prior to cell division, asters expand to cover <strong>the</strong> entire millimetersized<br />

cytoplasm in 20 minutes. Such rapid exploration <strong>of</strong> cell shape and size is thought to be<br />

crucial for positioning <strong>of</strong> centrosomes and cleavage plane determination (Wuhr et al., 2010, Curr<br />

Biol 20, 2040-45). It is unknown how microtubules (MTs) assemble at such a large distance<br />

from <strong>the</strong>ir presumable nucleation centers or centrosomes. We hypo<strong>the</strong>size three distinct<br />

processes that contribute to addition <strong>of</strong> MT at <strong>the</strong> aster periphery: (1) MT polymerization at plusends,<br />

(2) MT transport by minus-end directed motor proteins and (3) MT-stimulated MT<br />

nucleation. We used interphase Xenopus egg extract with ei<strong>the</strong>r centrosomes or beads coated<br />

with Aurora A antibody (Tsai & Zheng, 2005, Curr Biol 15, 2156-63) to recapitulate aster growth<br />

and assembly. To quantify <strong>the</strong> MT polymerization dynamics in <strong>the</strong> interphase extract, we<br />

tracked single MT plus-ends using time lapse microscopy. MT transport was assayed within a<br />

growing aster by fluorescent speckle microscopy. We explored <strong>the</strong> consequences <strong>of</strong> noncentrosomal<br />

MT nucleation on aster growth by constructing a biophysical model <strong>of</strong> plus-end<br />

distribution. Our model suggests that MT-mediated MT nucleation, when coupled with plus-end<br />

dynamic instability, contribute to <strong>the</strong> aster expansion velocity depending on parameter values. In<br />

<strong>the</strong> depleting limits <strong>of</strong> a hypo<strong>the</strong>tical MT nucleator, <strong>the</strong> model <strong>of</strong>fers an explanation to how MT<br />

density is maintained at <strong>the</strong> aster periphery. Our work begins to explore <strong>the</strong> biophysical<br />

mechanisms underlying <strong>the</strong> assembly <strong>of</strong> a millimeter-sized cytoskeletal structure.


MONDAY<br />

2118<br />

Evaluating models <strong>of</strong> self-organisation <strong>of</strong> linear microtubule arrays by computer<br />

simulation.<br />

Y-C. Oei 1,2 , X. E. Studera 1 , R. E. Carazo Salas 1 , T. Duke 3 ; 1 The Gurdon Institute, University <strong>of</strong><br />

Cambridge, Cambridge, United Kingdom, 2 Cavendish Laboratory, University <strong>of</strong> Cambridge,<br />

Cambridge, United Kingdom, 3 London Centre for Nanotechnology, University College London,<br />

London, United Kingdom<br />

In a wild-type fission yeast cell in interphase, microtubules (MTs) are organized in 3-4 bundles<br />

<strong>of</strong> 2-7 dynamic, anti-parallel MTs (interphase microtubule arrays, IMAs) whose correct number<br />

and organisation are essential for longitudinal cell growth and precise nuclear centring prior to<br />

mitosis (Sawin and Tran, Yeast 2006). Disruption <strong>of</strong> MT organisation, in cells mutant for certain<br />

genes or in cells treated with MT poisons, correlates with defects in cellular growth, shape<br />

and/or function.<br />

Within IMAs, MTs are thought to self-organise in a process that has been shown dependent on<br />

<strong>the</strong> bundling protein Ase1 and <strong>the</strong> motor protein Klp2, resulting in a configuration in which MT<br />

plus-tips point towards <strong>the</strong> cell periphery, with a central region where MTs overlap which is<br />

traditionally referred to as <strong>the</strong> microtubule organising centre (MTOC) (Janson et al, Cell 2007).<br />

Given <strong>the</strong> small number <strong>of</strong> constituent MTs, <strong>the</strong>ir continuous turnover, and <strong>the</strong> implied dynamic<br />

regeneration <strong>of</strong> IMAs, it is intriguing that <strong>the</strong> bundle number remains so stable at 3-4. Also,<br />

strikingly, cells that have had <strong>the</strong>ir nucleus and all IMAs removed eventually manage to reestablish<br />

IMAs in a configuration that resembles <strong>the</strong> wild-type (Carazo-Salas and Nurse, NCB<br />

2006).<br />

Aiming to understand how IMA number is regulated, we developed a computational Langevin<br />

simulation <strong>of</strong> interphase MT organisation, and use it to explore <strong>the</strong> influence <strong>of</strong> cell shape, <strong>of</strong> <strong>the</strong><br />

concentrations <strong>of</strong> bundle constituents, and <strong>of</strong> <strong>the</strong> properties commonly attributed to <strong>the</strong>m.<br />

2119<br />

Communication between Flagellar Outer and Inner Dynein Arms.<br />

T. Oda 1 , T. Yagi 1 , M. Kikkawa 1 ; 1 Department <strong>of</strong> Cell <strong>Biology</strong>, University <strong>of</strong> Tokyo Graduate<br />

School <strong>of</strong> Medicine, Tokyo, Japan<br />

The beating motion <strong>of</strong> cilia and flagella is driven by two rows <strong>of</strong> dynein arms: <strong>the</strong> outer dynein<br />

arms (ODA) and <strong>the</strong> inner dynein arms (IDA). The ODAs are required to generate <strong>the</strong> normal<br />

beat frequency and <strong>the</strong> IDAs are responsible for <strong>the</strong> amplitude <strong>of</strong> <strong>the</strong> waveform. Although<br />

structural connections between <strong>the</strong> ODA and <strong>the</strong> IDA have been observed by cryo-electron<br />

tomography <strong>of</strong> Chlamydomonas axoneme, <strong>the</strong> functional communication between <strong>the</strong> two<br />

species <strong>of</strong> <strong>the</strong> dynein arms has not been elucidated. In this study, we investigated <strong>the</strong> roles <strong>of</strong><br />

<strong>the</strong> two intermediate chains (IC1 and IC2) <strong>of</strong> Chlamydomonas ODA. We constructed <strong>the</strong><br />

expression vectors <strong>of</strong> IC1 and IC2 with biotinylation-tags added to <strong>the</strong> N- and <strong>the</strong> C-termini and<br />

introduced <strong>the</strong>m into <strong>the</strong> Chlamydomonas strains deficient in <strong>the</strong> respective ICs. The<br />

transformants were selected with drug resistance and <strong>the</strong> recovery <strong>of</strong> <strong>the</strong> beat frequency. The<br />

ODAs containing <strong>the</strong> biotinylated ICs were extracted from <strong>the</strong> axoneme <strong>of</strong> <strong>the</strong> transformants.<br />

We solved <strong>the</strong> three dimensional structures <strong>of</strong> <strong>the</strong> biotin-streptavidin labeled ODA-microtubule<br />

complexes using cryo-electron microscopy. We located <strong>the</strong> positions <strong>of</strong> <strong>the</strong> termini <strong>of</strong> <strong>the</strong> IC1<br />

and IC2 near <strong>the</strong> β and <strong>the</strong> γ heavy chains, respectively. It is noteworthy that <strong>the</strong> location <strong>of</strong> <strong>the</strong><br />

N-terminus <strong>of</strong> IC2 is close to <strong>the</strong> previously observed ODA-IDA linker. The biotin-tags at <strong>the</strong> Nterminus<br />

<strong>of</strong> IC1 and <strong>the</strong> C-terminus <strong>of</strong> IC2 did not alter <strong>the</strong> swimming phenotypes <strong>of</strong> <strong>the</strong>


MONDAY<br />

transformed Chlamydomonas cells, but <strong>the</strong> biotin-tag added to <strong>the</strong> N-terminus <strong>of</strong> IC2 reduced<br />

<strong>the</strong> amplitude <strong>of</strong> <strong>the</strong> beating by half while <strong>the</strong> beat frequency was not decreased. These results<br />

suggest <strong>the</strong> N-terminus <strong>of</strong> IC2 mediates <strong>the</strong> communication between <strong>the</strong> ODA and <strong>the</strong> IDA.<br />

2120<br />

A Gas8 Knockout Mouse Model Reveals Defects in Cilia Motility.<br />

W. R. Lewis 1 , E. Malarkey 1 , R. Pasek 1 , N. Berbari 1 , B. Yoder 1 ; 1 Cell <strong>Biology</strong>, University <strong>of</strong><br />

Alabama-Birmingham, Birmingham, AL<br />

Mouse Growth Arrest Specific 8 (Gas8) has been associated with microtubules and more<br />

specifically <strong>the</strong> Dynein Regulatory Complex (DRC) where it is thought to play a role in cilia and<br />

flagella motility. While previous work on Gas8 has been done utilizing in vitro knockdown<br />

approaches and in model organisms such as Trypanosoma brucei and Chlamydomonas<br />

reinhardtii, this gene has not been studied in an in vivo mammalian genetic model. We have<br />

generated a Gas8 mutant mouse and found that mutants survive gestation; however <strong>the</strong>y do not<br />

survive to sexual maturity. Consistent with a role in <strong>the</strong> DRC, it appears that <strong>the</strong> Gas8 mutant<br />

mice die as a result <strong>of</strong> severe hydrocephalus. Fur<strong>the</strong>rmore, analysis <strong>of</strong> ependymal cilia <strong>of</strong> <strong>the</strong><br />

lateral ventricles <strong>of</strong> <strong>the</strong> brain indicated a severe defect in motility based on fluorescent bead<br />

tracking. Intriguingly, Gas8 is also expressed in cells that lack motile cilia, suggesting it has an<br />

additional role outside <strong>of</strong> <strong>the</strong> DRC. Recently, Gas8 has been implicated in Sonic Hedgehog<br />

signaling pathway using knockdown approaches where it has been proposed to be a positive<br />

regulator <strong>of</strong> <strong>the</strong> pathway activator Smoo<strong>the</strong>ned. However, our analysis <strong>of</strong> <strong>the</strong> Sonic Hedgehog<br />

pathway in <strong>the</strong> Gas8 genetic mutants using neural tube and limb bud patterning as readouts for<br />

pathway activity has yet to reveal any abnormalities consistent with a hedgehog defect. Future<br />

studies <strong>of</strong> this mouse model will focus on determining <strong>the</strong> precise nature <strong>of</strong> <strong>the</strong> motile cilia<br />

defect using high speed video waveform analysis and analyzing potential novel roles for Gas8 in<br />

cells with primary cilia.<br />

2121<br />

Cryo-Electron Tomography <strong>of</strong> Chlamydomonas Flagella Reveals Radial Spoke<br />

Heterogeneity and Connectivity.<br />

C. F. Barber 1 , T. Heuser 1 , B. I. Carbajal-Gonzalez 1 , V. V. Botchkarev Jr. 1 , D. Nicastro 1 ;<br />

1 <strong>Biology</strong> Department, Rosenstiel Center, Brandeis University, Waltham, MA<br />

Cilia and flagella are highly conserved cellular organelles critical to many cellular, tissue and<br />

developmental processes. The core structure <strong>of</strong> cilia and flagella, called <strong>the</strong> axoneme, consists<br />

<strong>of</strong> 96-nm long units that repeat along <strong>the</strong> axoneme length. Within each repeat are two or three<br />

(depending on species) radial spokes (RSs) that transiently interact with <strong>the</strong> central pair<br />

complex (Warner and Satir, 1974). Studies <strong>of</strong> <strong>the</strong> unicellular alga Chlamydomonas reinhardtii<br />

have revealed that <strong>the</strong> RSs play a critical role in transmitting regulatory signals from <strong>the</strong> central<br />

pair complex to <strong>the</strong> axonemal dyneins, ultimately controlling <strong>the</strong> activity <strong>of</strong> dynein proteins<br />

(reviewed Porter and Sale, 2000; Smith and Yang, 2004). In both Chlamydomonas and<br />

humans, failure to assemble RSs results in flagellar and ciliary paralysis (Sturgess et al., 1979;<br />

Witman et al., 1978). While several molecular components <strong>of</strong> <strong>the</strong> regulatory pathway have been<br />

identified, little is known about <strong>the</strong>ir specific locations or physical interactions. To shed fur<strong>the</strong>r<br />

light on <strong>the</strong> mechanism <strong>of</strong> axonemal dynein regulation we used cryo-electron tomography and<br />

sub-tomogram averaging <strong>of</strong> Chlamydomonas axonemes to visualize <strong>the</strong> three-dimensional<br />

structure <strong>of</strong> <strong>the</strong> radial spokes in unprecedented detail.<br />

Unlike many o<strong>the</strong>r species, Chlamydomonas has only two spokes per 96-nm repeat, RS1 and<br />

RS2, which were till recently believed to be structurally and functionally homogenous. Our data


MONDAY<br />

reveal that while <strong>the</strong> overall structure <strong>of</strong> RS1 and RS2 is similar - including a two-pronged spoke<br />

base and direct connections to inner arm dynein tails - unique densities are seen in each spoke.<br />

The structural comparison <strong>of</strong> Chlamydomonas wild type and <strong>the</strong> mutant pf17 with head-less<br />

spokes, also revealed <strong>the</strong> morphology and boundaries <strong>of</strong> <strong>the</strong> RS head, including a direct RS1 to<br />

RS2 interaction. Finally, in place <strong>of</strong> a third radial spoke we found an uncharacterized, shorter<br />

density, <strong>the</strong> “Radial Spoke 3 Stand-in” (RS3S), which bears structurally no resemblance to RS1<br />

and RS2, and is unaltered in <strong>the</strong> pf17 mutant. These findings demonstrate that radial spokes<br />

are heterogeneous in structure and may play functionally distinct roles in regulating <strong>the</strong><br />

movement <strong>of</strong> cilia and flagella.<br />

References:<br />

Porter, M.E., and Sale, W.S. (2000). The 9 + 2 axoneme anchors multiple inner arm dyneins<br />

and a network <strong>of</strong> kinases and phosphatases that control motility. J Cell Biol 151, F37-42.<br />

Smith, E.F., and Yang, P. (2004). The radial spokes and central apparatus: mechano-chemical<br />

transducers that regulate flagellar motility. Cell Motil Cytoskeleton 57, 8-17.<br />

Sturgess, J.M., Chao, J., Wong, J., Aspin, N., and Turner, J.A. (1979). Cilia with defective radial<br />

spokes: a cause <strong>of</strong> human respiratory disease. N Engl J Med 300, 53-56.<br />

Warner, F.D., and Satir, P. (1974). The structural basis <strong>of</strong> ciliary bend formation. Radial spoke<br />

positional changes accompanying microtubule sliding. J Cell Biol 63, 35-63.<br />

Witman, G.B., Plummer, J., and Sander, G. (1978). Chlamydomonas flagellar mutants lacking<br />

radial spokes and central tubules. Structure, composition, and function <strong>of</strong> specific axonemal<br />

components. J Cell Biol 76, 729-747.<br />

2122<br />

How flagellum controls cell length.<br />

Y. Sun 1 , C. Y. He 1 ; 1 Department <strong>of</strong> Biological Sciences, National University <strong>of</strong> Singapore,<br />

Singapore<br />

Trypanosoma brucei is an extracellular parasite causing African sleeping sickness in humans<br />

and Nagana in cattle. This single-celled pathogen possesses a single flagellum attached to <strong>the</strong><br />

cell body via flagellum adhesion proteins that link plasma membrane to flagellar membrane, and<br />

a complex flagellar attachment zone (FAZ) mostly located in <strong>the</strong> cell body. In addition to being a<br />

motility and sensory organelle, <strong>the</strong> flagellum also acts as a molecular ruler that controls cell<br />

morphogenesis. However, it remains to be understood how <strong>the</strong> length information contained in<br />

<strong>the</strong> flagellum is transmitted to <strong>the</strong> FAZ, which directly controls cellular microtubule biogenesis<br />

and organization. Here we focused on TbFla1, a surface glycoprotein required for adhesion<br />

between flagellar and cell membranes. Using immunoprecipitation <strong>of</strong> YFP tagged Fla1 and<br />

mass spectrometry, a Fla1 binding protein (Fla1BP) was identified and was localized to<br />

flagellum membrane. Procyclic T. brucei lacking Fla1BP showed no apparent growth defects,<br />

produced flagellum <strong>of</strong> normal length but shorter FAZ <strong>of</strong> relatively fixed length, uncoupling <strong>the</strong><br />

regulation <strong>of</strong> cell length by flagellum. Our results provided a molecular basis for flagellum<br />

membrane adhesion to cell membrane and may explain how flagellum controls cell<br />

morphogenesis.<br />

2123<br />

A Neuronal Calcium Sensor Protein, Calaxin, is Essential for Modulation <strong>of</strong> Ciliary Beat<br />

Direction in Sea Urchin Embryos.<br />

K. Mizuno 1 , J. Yaguchi 1 , K. Shiba 1 , S. Yaguchi 1 , K. Inaba 1 ; 1 Shimoda Marine Research Center,<br />

University <strong>of</strong> Tsukuba, Shimoda, Shizuoka, Japan


MONDAY<br />

Calaxin is a neuronal calcium sensor (NCS)-family Ca 2+ -binding protein localized in cilia and<br />

flagella. Calaxin has three EF-hand Ca 2+ -binding motifs, and its orthologues are present in<br />

metazoan species, but not in yeast, green algae or plant. Previous research has shown that<br />

calaxin binds to outer arm dynein or β-tubulin in <strong>the</strong> presence or absence <strong>of</strong> Ca 2+ , respectively<br />

[Mizuno et al., Biol. Cell. 2009]. In addition, our recent studies showed that inhibiting calaxin<br />

function greatly reduced <strong>the</strong> duration <strong>of</strong> asymmetrical waveform, resulting in <strong>the</strong> suppression <strong>of</strong><br />

turning movement during sperm chemotaxis. These results indicate that calaxin has an<br />

essential role in <strong>the</strong> control <strong>of</strong> flagellar waveform through <strong>the</strong> control <strong>of</strong> outer arm dynein activity<br />

in Ca 2+ -dependent manner. However, we still do not understand <strong>the</strong> whole mechanisms <strong>of</strong><br />

calaxin function throughout <strong>the</strong> animal kingdom, especially in <strong>the</strong>ir cilia. Here, we tried <strong>the</strong><br />

functional analysis <strong>of</strong> calaxin in somatic ciliary movement in <strong>the</strong> embryo <strong>of</strong> <strong>the</strong> sea urchin,<br />

Hemicentrotus pulcherrimus, because <strong>the</strong>ir cilia are suitable for motility analysis and it is easy to<br />

perform knockdown experiments using morpholino antisense oligo (MO). Immunostaining<br />

showed calaxin was localized to <strong>the</strong> entire region <strong>of</strong> sea urchin embryonic cilia. To study <strong>the</strong><br />

calaxin function, we analyzed <strong>the</strong> motility <strong>of</strong> normal and calaxin-MO embryos using a system <strong>of</strong><br />

digital-camera equipped dark field illumination. calaxin morphants swam much slower than<br />

normal embryos and showed abnormal swimming trajectories. Detailed analysis using a highspeed<br />

camera, however, revealed that cilia <strong>of</strong> calaxin morphants beat with normal frequency.<br />

On <strong>the</strong> o<strong>the</strong>r hand, beating direction <strong>of</strong> cilia in calaxin morphants was greatly disorganized as<br />

compared with cilia <strong>of</strong> normal embryos that show highly organized beat direction from <strong>the</strong><br />

animal pole toward <strong>the</strong> vegetal pole. These results suggest that calaxin is required to coordinate<br />

ciliary beat direction by controlling outer arm dyneins. Fur<strong>the</strong>rmore, we observed that<br />

gastrulation was greatly inhibited in calaxin morphants. Although <strong>the</strong> mechanism how <strong>the</strong><br />

gastrulation was inhibited in calaxin morphants is not clear, it is possible that calaxin is involved<br />

in <strong>the</strong> establishment <strong>of</strong> planer cell polarity and contributes to <strong>the</strong> convergent extension <strong>of</strong><br />

embryonic cells.<br />

2124<br />

Loss <strong>of</strong> basal feet on Odf2 deletion perturbs polarization <strong>of</strong> basal bodies.<br />

K. Kunimoto 1 , Y. Yamazaki 1 , T. Nishida 2 , K. Shinohara 3 , H. Ishikawa 4 , T. Hasegawa 2 , T.<br />

Okanoue 5 , H. Hamada 3 , T. Noda 6 , A. Tamura 1 , S. Tsukita 7 , S. Tsukita 1 ; 1 Laboratory <strong>of</strong> Biological<br />

Science, Department <strong>of</strong> Biological Science, Osaka University, Suita, Japan, 2 Research Center<br />

for Ultra-high Voltage Electron Microscopy, Osaka University, Ibaraki, Japan, 3 Developmental<br />

Genetics Group, Department <strong>of</strong> Biological Science, Osaka University, Suita, Japan,<br />

4 Department <strong>of</strong> Biochemistry and Biophysics, UCSF, San Francisco, 5 Department <strong>of</strong><br />

Hepatology, Saiseikai Suita Hospital, 6 Department <strong>of</strong> Cell <strong>Biology</strong>, Cancer Institute <strong>of</strong> Japanese<br />

Foundation for Cancer Research, Tokyo, Japan, 7 Department <strong>of</strong> Cell <strong>Biology</strong>, Kyoto University,<br />

Kyoto, Japan<br />

Synergic multiciliary beating relies on cilia generated from basal bodies, with which basal feet<br />

are regularly associated through molecular mechanisms that remain unknown. Here we show<br />

that <strong>the</strong> coordinated multiciliary action is disturbed in Odf2 mutant mice, resulting in primary<br />

ciliary dyskinesia and a characteristic coughing/sneezing-like phenotype. Odf2 mutant depleted<br />

basal feet from basal bodies to perturb <strong>the</strong> planar cell polarity (PCP) <strong>of</strong> basal bodies, as shown<br />

by ultra-high voltage electron microscopic tomography <strong>of</strong> wild and Odf2 mutant tracheas. The<br />

apical microtubular lattice, which is organized by <strong>the</strong> keystone positioning <strong>of</strong> basal feet/basal<br />

bodies, was lost in Odf2-mutant animals, irrespective <strong>of</strong> normal localization <strong>of</strong> Vangl1, <strong>the</strong> PCP<br />

core protein. These findings demonstrate that Odf2 is required for <strong>the</strong> formation <strong>of</strong> basal feet.<br />

Odf2-based basal feet play a critical role in <strong>the</strong> PCP-based arrangement <strong>of</strong> <strong>the</strong> microtubular<br />

lattice and basal bodies, <strong>the</strong>reby enabling coordinated multiciliary beating.


MONDAY<br />

2125<br />

Functional Analysis <strong>of</strong> a Gliding-Associated Ciliary Protein with Putative Lipase Activity.<br />

E. Betleja 1 , T. Birch 1 , R. A. Bloodgood 2 , J. L. Watts 3 , D. G. Cole 1 ; 1 Dept <strong>of</strong> Biological Sciences,<br />

University <strong>of</strong> Idaho, Moscow, ID, 2 Dept <strong>of</strong> Cell <strong>Biology</strong>, University <strong>of</strong> Virginia School <strong>of</strong> Medicine,<br />

Charlottesville, VA, 3 School <strong>of</strong> <strong>Molecular</strong> Biosciences, Washington State University, Pullman,<br />

WA<br />

The unicellular green algae Chlamydomonas reinhardtii exhibits two independent forms <strong>of</strong><br />

whole cell locomotion, both utilizing <strong>the</strong> flagellum: swimming through a liquid medium and<br />

gliding across a solid substrate. Essential for <strong>the</strong> assembly and function <strong>of</strong> <strong>the</strong>se ciliary<br />

organelles is <strong>the</strong> bidirectional transport <strong>of</strong> protein particles known as intraflagellar transport<br />

(IFT). In a search for IFT-associated cargo, pull down assays <strong>of</strong> Chlamydomonas flagellar<br />

extract with <strong>the</strong> retrograde IFT motor, cytoplasmic dynein 1b/2, yielded a set <strong>of</strong> proteins, among<br />

which <strong>the</strong> most prominent were <strong>the</strong> major flagellar surface glycoprotein, FMG-1B and a flagellarassociated<br />

protein <strong>of</strong> unknown function, which we have named GAP12 (gliding-associated<br />

protein 12). This finding suggests that <strong>the</strong> retrograde IFT motor may play a role in ciliarydependent<br />

gliding motility. In contrast to a well established role for FMG-1B which acts as a<br />

transmembrane gliding receptor that contacts substrate during gliding motility, <strong>the</strong> biological<br />

function <strong>of</strong> GAP12 is unknown. To address <strong>the</strong> functional roles <strong>of</strong> GAP12, an artificial micro<br />

RNA interference was used to reduce expression <strong>of</strong> <strong>the</strong> GAP12 gene. Western blot analysis <strong>of</strong><br />

whole cellular extracts from RNAi transformants showed that in 14 out <strong>of</strong> 28 knockdown lines<br />

GAP12 protein level was significantly reduced. Selected GAP12-RNAi lines were assayed for<br />

gliding phenotype. Some <strong>of</strong> <strong>the</strong> knockdown strains had difficulty attaching to <strong>the</strong> glass slides<br />

and when <strong>the</strong>y did attach, <strong>the</strong>y would glide for only short distances <strong>of</strong> a few microns before<br />

detaching. The observation that GAP12-deficient cells are blocked at <strong>the</strong> stage <strong>of</strong> adherence<br />

suggests that GAP12 is involved in an early stage <strong>of</strong> gliding motility. Because GAP12 contains a<br />

putative lipase domain, we also looked for effects on lipid metabolism. By shifting cells into a<br />

nitrogen-deficient medium, Chlamydomonas is forced to accumulate greater levels <strong>of</strong><br />

triglycerides in specialized storage cytosolic organelles known as lipid bodies. When GAP12deficient<br />

strains were examined after 72 hrs <strong>of</strong> nitrogen starvation, <strong>the</strong> lipid-soluble fluorescent<br />

Bodipy dye revealed enlarged lipid bodies, which would be consistent with a possible role for<br />

GAP12 lipase activity in <strong>the</strong> degradation or hydrolysis <strong>of</strong> lipid body triglycerides. We are<br />

currently testing, at <strong>the</strong> biochemical level, if GAP12-RNAi knockdown strains accumulate<br />

specific fatty acids when starved in N-free medium. Any fur<strong>the</strong>r analysis <strong>of</strong> GAP12 will need to<br />

consider this potential role in lipid metabolism, which may or may not be independent <strong>of</strong> gliding<br />

motility.<br />

2126<br />

Monte Carlo simulation <strong>of</strong> centrosomal self-centering due to pushing by microtubules in<br />

large cells.<br />

S. K. Tang 1,2 , B. T. Castle 3,2 , D. J. Odde 2,3 ; 1 Department <strong>of</strong> Mechanical Engineering, Stanford<br />

University, Stanford, CA, 2 Physiology Course 2011, Woods Hole Marine Biological Laboratory,<br />

Woods Hole, MA, 3 Department <strong>of</strong> Biomedical Engineering, University <strong>of</strong> Minnesota,<br />

Minneapolis, MN<br />

The overarching questions <strong>of</strong> our work are: what drives self-centering in cells, and if cells with<br />

different sizes use different mechanisms for centering? Finding <strong>the</strong> center in a cell is critical for<br />

many processes such as cell division. Various models for self-centering exist. Here we<br />

implement a 1D Monte Carlo model to simulate <strong>the</strong> centering <strong>of</strong> centrosome (or o<strong>the</strong>r<br />

microtubule-nucleating site) due to <strong>the</strong> pushing forces generated when microtubules reach <strong>the</strong><br />

boundary <strong>of</strong> <strong>the</strong> cell. Our model consists <strong>of</strong> one centrosome that is originally positioned close to


MONDAY<br />

<strong>the</strong> cell boundary. Microtubules nucleate at this site, and grow/shrink in length governed by<br />

experimentally measured dynamic instability parameters. Our model assumes that <strong>the</strong> cell<br />

boundary is rigid, and that microtubules push <strong>the</strong> whole length <strong>of</strong> <strong>the</strong> microtubule backwards,<br />

causing <strong>the</strong> nucleating site to move.<br />

Our model shows that <strong>the</strong> growth and pushing <strong>of</strong> multiple microtubules (up to 1000 in number)<br />

in opposing directions can center <strong>the</strong> centrosome in cells smaller than 500-600 microns in<br />

diameter. However, with <strong>the</strong> experimental dynamic instability parameters, microtubules do not<br />

grow longer than 250-300 microns within ~90 min, a typical time scale for first division in<br />

Xenopus eggs (diameter ~ 1.2 mm). As microtubules do not reach <strong>the</strong> cell boundary, <strong>the</strong><br />

pushing model alone cannot explain centering <strong>of</strong> centrosomes for cells with diameters larger<br />

than 600 microns. In addition, <strong>the</strong> centering time scale is highly sensitive to small variations in<br />

microtubule dynamics. We conclude that <strong>the</strong> pushing model is not a robust centering<br />

mechanism in large cells and that o<strong>the</strong>r mechanisms must play a role in driving centering in<br />

<strong>the</strong>se cells.<br />

2127<br />

A Sas-4/tubulin module regulates <strong>the</strong> formation <strong>of</strong> centrosomal complexes and<br />

centrosome biogenesis.<br />

J. Gopalakrishnan 1 , Y-C. F. Chim 1 , A. Ha 1 , M. Basiri 1 , T. Avidor-Reiss 1 ; 1 Harvard Medical<br />

School, Boston, MA<br />

Centrosomes are conserved organelles that are essential for cilium formation and accurate cell<br />

division, and centrosome abnormality can result in genomic instability. A centrosome consists <strong>of</strong><br />

a pair <strong>of</strong> centrioles surrounded by a protein network <strong>of</strong> pericentriolar material that is essential for<br />

centrosome function. Pericentriolar material assembly is a tightly regulated, critical step in<br />

centrosome biogenesis that determines a centrosome’s size and capability. Here we show that<br />

a Sas-4/tubulin module regulates <strong>the</strong> formation <strong>of</strong> centrosomal complexes and centrosome<br />

biogenesis. Using a combination <strong>of</strong> genetic and biochemical approaches in Drosophila, we<br />

found that Sas 4 and tubulin regulate <strong>the</strong> formation <strong>of</strong> several cytoplasmic complexes, which<br />

include CNN, Asl and D-PLP (named S-CAP complexes) and γ-tubulin ring proteins (named S-γ<br />

complexes). The proteins <strong>of</strong> <strong>the</strong>se complexes are components <strong>of</strong> <strong>the</strong> pericentriolar material and<br />

are essential for pericentriolar material assembly and for <strong>the</strong> centrosome’s capacity to nucleate<br />

microtubule asters. Interestingly, <strong>the</strong> S-CAP complex contains several microcephaly-linked<br />

proteins and disturbing this complex causes abnormal centrosome formation. The finding that<br />

microcephaly-linked proteins form a common complex may explain why mutations in any <strong>of</strong><br />

<strong>the</strong>se lead to <strong>the</strong> same disorder. Tubulin (α-and β-tubulin heterodimer) is known to be a building<br />

block <strong>of</strong> microtubules. Our data points to a new and novel function for tubulin as a regulator <strong>of</strong><br />

centrosome complex formation, binding to Sas-4 and regulating <strong>the</strong> interaction <strong>of</strong> Sas-4 with<br />

o<strong>the</strong>r centrosomal proteins. By regulating centrosome complex formation, <strong>the</strong> Sas-4/tubulin<br />

module regulates centrosome biogenesis.<br />

2128<br />

Mechanisms <strong>of</strong> Centrosome Separation.<br />

B. R. Mardin 1 , F. G. Agircan 1 , C. Lange 1 , A. M. Fry 2 , E. Schiebel 1 ; 1 ZMBH Uni Heidelberg,<br />

Heidelberg, Germany, 2 University <strong>of</strong> Leicester, United Kingdom<br />

In human cells, duplicated centrosomes are first held toge<strong>the</strong>r by a proteinaceous linker that<br />

extends between <strong>the</strong> proximal ends <strong>of</strong> <strong>the</strong> two mo<strong>the</strong>r centrioles. To facilitate centrosome<br />

separation and bipolar spindle formation at mitotic entry, this linker is disassembled in G2/M in a<br />

process known as centrosome disjunction. Centrosome disjunction is driven by <strong>the</strong> Nek2A


MONDAY<br />

kinase, which phosphorylates <strong>the</strong> linker proteins, C-Nap1 and rootletin. Previously we<br />

demonstrated that <strong>the</strong> ability <strong>of</strong> Nek2A to initiate centrosome disjunction is controlled by two<br />

components <strong>of</strong> <strong>the</strong> Hippo pathway, <strong>the</strong> mammalian sterile 20-like kinase 2 (Mst2) and <strong>the</strong><br />

scaffold protein Salvador (hSav1). Conventionally, Hippo pathway proteins are tumor<br />

suppressors that are well known for <strong>the</strong>ir function in tissue growth and apoptosis. By forming a<br />

separate module, hSav1 and Mst2 associate with and regulate <strong>the</strong> localized activity <strong>of</strong> Nek2A by<br />

phosphorylation. Our recent data revealed that <strong>the</strong> Mst2-Nek2A kinase module is regulated by<br />

<strong>the</strong> Polo-like kinase 1 (Plk1). Plk1 selectively binds to and phosphorylates Mst2, which controls<br />

<strong>the</strong> binding <strong>of</strong> Nek2A to <strong>the</strong> phosphatase PP1γ providing a balance between <strong>the</strong> counteracting<br />

Nek2A and PP1γ activities on <strong>the</strong> centrosome linker.<br />

Importantly, we provided <strong>the</strong> first evidence that <strong>the</strong> Mst2-Nek2A module has overlapping<br />

functions with <strong>the</strong> kinesin motor protein Eg5 in centrosome separation and bipolar spindle<br />

formation. In fact, in cells with reduced Eg5 motor activity, <strong>the</strong> hSav1-Mst1/2-Nek2A pathway<br />

becomes essential for bipolar spindle formation. Plk1 is crucial for <strong>the</strong> targeting <strong>of</strong> Eg5 to <strong>the</strong><br />

centrosomes and this process is supported by <strong>the</strong> microtubule cytoskeleton. Taken toge<strong>the</strong>r,<br />

our data significantly contribute to <strong>the</strong> understanding <strong>of</strong> how centrosome disjunction and spindle<br />

formation are regulated on <strong>the</strong> molecular level.<br />

2129<br />

Differentiating Naegleria assemble two basal bodies de novo by counting from 0 to 1 to 2.<br />

L. Fritz-Laylin 1 , Y. Levy 2 , E. Levitan, S. Chen, Z. Cande 3 , E. Lai 4 , C. Fulton 4 ; 1 University <strong>of</strong><br />

California, San Francsico, San Francisco, CA, 2 CLARIFY Scientific and Medical<br />

Communications, 3 Department <strong>of</strong> <strong>Molecular</strong> and Cell <strong>Biology</strong>, University <strong>of</strong> California, Berkeley,<br />

4 Department <strong>of</strong> <strong>Biology</strong>, Brandeis University, Waltham, MA<br />

Naegleria gruberi differentiate from amoebae to flagellates in about an hour. This phenotypic<br />

transformation requires <strong>the</strong> de novo assembly <strong>of</strong> an entire microtubule cytoskeleton, including<br />

two basal bodies (centrioles) and flagella. The formation <strong>of</strong> new basal bodies occurs about 10<br />

minutes before <strong>the</strong> appearance <strong>of</strong> visible flagella. Here we use antibodies against tubulin,<br />

centrin, and SAS-6 to reveal that <strong>the</strong> sequence <strong>of</strong> basal body formation proceeds in two distinct<br />

steps: first one basal body forms de novo, after which a second basal body assembles adjacent<br />

to <strong>the</strong> first. This second round <strong>of</strong> basal body assembly is similar to <strong>the</strong> centriole assembly<br />

typically seen in human cells. Interestingly, when microtubule assembly is temporarily blocked<br />

using a pulse treatment <strong>of</strong> oryzalin, followed by inhibition <strong>of</strong> protein syn<strong>the</strong>sis with<br />

cycloheximide, cells form a single flagellum. Because <strong>the</strong>se cells successfully assemble two<br />

basal bodies but only a single flagellum, this demonstrates that <strong>the</strong>re is a moment in<br />

differentiation when one basal body is competent to template flagellar assembly while <strong>the</strong> o<strong>the</strong>r<br />

is not. Naegleria utilizes both commonly studied modes <strong>of</strong> centriole assembly, de novo and<br />

template-associated, and forms two basal bodies in a 0 to 1 to 2 sequence ra<strong>the</strong>r than<br />

simultaneously.<br />

2130<br />

Microtubule anchoring is facilitated by <strong>the</strong> shoulder/sidearm subunits <strong>of</strong> <strong>the</strong> dynactin<br />

complex.<br />

A. G. Le 1 , R. L. Schneider 1 , L. M. McCullough 2 , N. J. Quintyne 2 ; 1 Florida Atlantic University,<br />

Jupiter, FL, 2 Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL<br />

Dynactin is a multifunctional protein complex identified as a required c<strong>of</strong>actor for cytoplasmic<br />

dynein. Dynactin also plays a dynein-independent role in anchoring microtubules to <strong>the</strong><br />

centrosome during interphase. Dynactin is <strong>the</strong> only component <strong>of</strong> <strong>the</strong> microtubule anchoring<br />

complex that has been shown to directly bind to microtubules. Previous studies have shown that


MONDAY<br />

<strong>the</strong> shoulder/sidearm region <strong>of</strong> dynactin, consisting <strong>of</strong> p150Glued, dynamitin and p24 are<br />

integral to this anchoring process. The p150Glued subunit <strong>of</strong> dynactin contains two microtubule<br />

binding domains <strong>of</strong> different affinity. The CAP-Gly domain binds microtubules tightly, whereas<br />

<strong>the</strong> Basic domain has a much weaker affinity for <strong>the</strong> microtubule. The function <strong>of</strong> p24 remains<br />

largely uncharacterized. Using overexpression and siRNA-mediated knockdown, we have<br />

examined <strong>the</strong> contribution <strong>of</strong> two <strong>of</strong> <strong>the</strong> three shoulder/sidearm subunits to microtubule<br />

anchoring in more detail. We find that anchoring is abrogated when ei<strong>the</strong>r <strong>of</strong> <strong>the</strong>se subunits<br />

expression levels are changed. We observe both microtubule organization and regrowth<br />

patterns to vary when comparing knockdown and overexpression. Our data suggest a model<br />

where <strong>the</strong> different microtubule-binding domains <strong>of</strong> p150Glued contribute unequally to<br />

microtubule anchoring and p24 serves as reinforcement for maintaining shoulder/sidearm<br />

rigidity at <strong>the</strong> centrosome.<br />

2131<br />

Centrosome inactivation in Drosophila oogenesis.<br />

I. Bento 1 , M. Bettencourt-Dias 1 ; 1 Instituto Gulbenkian de Ciência, Oeiras, Portugal<br />

Centrosome is <strong>the</strong> primary microtubule-organizing center (MTOC) in animal cells, whose<br />

number is highly controlled. Duplication <strong>of</strong> this structure occurs in parallel with DNA replication.<br />

During development <strong>the</strong>re are tissues where centrosome number is differently regulated. The<br />

zygote formation is one <strong>of</strong> <strong>the</strong>m. During oogenesis, centrioles degenerate and female meiosis<br />

relies on an acentrosomal spindle. On <strong>the</strong> o<strong>the</strong>r hand, during spermatogenesis, <strong>the</strong> centriole<br />

that nucleates <strong>the</strong> axoneme looses pericentriolar (PCM) proteins. After fertilization, centrioles<br />

are paternally inherited and <strong>the</strong> PCM proteins are deposited by <strong>the</strong> mo<strong>the</strong>r. The absence <strong>of</strong><br />

centrioles in <strong>the</strong> egg has been hypo<strong>the</strong>sized as a strategy to avoid par<strong>the</strong>nogenesis. This is<br />

reinforced by <strong>the</strong> observations that naturally occurring par<strong>the</strong>nogenic species are able to form<br />

de novo centrosomes. Using Drosophila melanogaster oogenesis, we aim to understand how<br />

centrioles disappear during oogenesis. We are characterizing Drosophila melanogaster<br />

oogenesis by immunostaining. We observed that proteins such as Bld10, Asl, SAS6 or SAS4<br />

are present in <strong>the</strong> oocyte´s MTOC until stage 10-11 although <strong>the</strong>y are absent from <strong>the</strong> meiotic<br />

spindle. We noticed that <strong>the</strong> isolated overexpression <strong>of</strong> proteins involved in centriole biogenesis<br />

does not inhibit centriole disappearance in later stages <strong>of</strong> oocyte development, suggesting<br />

those are not limiting factors. Interestingly, Spd2, a PCM protein, is <strong>the</strong> first protein that<br />

disappears from <strong>the</strong> oocyte´s MTOC. Currently we are testing if PCM maintenance is crucial to<br />

keep centriole stability. Oogenesis characterization leads to better understanding <strong>of</strong> centriole<br />

inactivation and disappearance.<br />

2132<br />

Regulation <strong>of</strong> <strong>the</strong> Formation <strong>of</strong> Microtubule Organizing Centers.<br />

F. Leal 1,2 , S. Kandel-Lewis 2 , E. Karsenti 2 , M. Bettencourt-Dias 1 ; 1 Instituto Gulbenkian De<br />

Ciência, Oeiras, Portugal, 2 European <strong>Molecular</strong> <strong>Biology</strong> Laboratory, Heidelberg, Germany<br />

Centrosomes were originally discovered in <strong>the</strong> late 19 th century [1] , being named after <strong>the</strong>ir<br />

central position in <strong>the</strong> cell, and characterized by Theodor Boveri [2] . They are composed by a pair<br />

<strong>of</strong> orthogonally oriented centrioles, microtubule barrel shaped structures organized in a nine-fold<br />

symmetry, and a surrounding electron-dense material composed <strong>of</strong> many proteins, named<br />

pericentriolar material (PCM). The centrosome is <strong>the</strong> primary microtubule-organizing centre<br />

(MTOC) in animal cells regulating cell motility and polarity in Interphase and facilitating <strong>the</strong><br />

organization <strong>of</strong> <strong>the</strong> spindle poles during Mitosis. Also, in animals, centrioles retain <strong>the</strong> ability to<br />

migrate to <strong>the</strong> cell membrane and form basal bodies, thus templating <strong>the</strong> assembly <strong>of</strong> cilia and<br />

flagella, important structures in motility and signalling and sensory perception.


MONDAY<br />

Centrosomes undergo duplication once every cell cycle so that, like <strong>the</strong> genetic material, <strong>the</strong>ir<br />

number remains stable. Abnormalities in centrosome number and structure occur in many<br />

cancers and are associated with genomic instability. In 2005, two separate studies [3-4] showed<br />

<strong>the</strong> importance <strong>of</strong> SAK/Plk4, a kinase form <strong>the</strong> serine/threonine family <strong>of</strong> polo-like kinases, in<br />

centriole duplication. Depletion or overexpression <strong>of</strong> this kinase led to <strong>the</strong> depletion or<br />

overduplication <strong>of</strong> centrioles in both Drosophila and human cells. Although SAK/Plk4 has been<br />

shown to be essential for centrosome duplication and biogenesis, very little is known about its<br />

regulation, due to <strong>the</strong> lack <strong>of</strong> knowledge <strong>of</strong> interacting partners. In order to understand <strong>the</strong><br />

regulation and activity <strong>of</strong> this important kinase we are taking advantage <strong>of</strong> <strong>the</strong> biochemical<br />

system per excellence, <strong>the</strong> Xenopus laevis cell free extract to approach <strong>the</strong> following questions:<br />

1. How is SAK/Plk4 activity regulated?<br />

2. How does SAK/Plk4 activity regulate centriole formation?<br />

3. How do centrioles regulate <strong>the</strong>ir own biogenesis?<br />

Up to now we have seen that <strong>the</strong> predicted Xenopus laevis ortholog (XPLK4) is able to<br />

induce <strong>the</strong> formation <strong>of</strong> MTOCs which contain centriolar markers. The number and size is<br />

directly proportional to <strong>the</strong> concentration <strong>of</strong> XPLK4 used, suggesting that this kinase specifically<br />

controls centriole biogenesis. We have also seen that addition <strong>of</strong> this kinase in different stages<br />

<strong>of</strong> <strong>the</strong> cell cycle leads to different outcomes in terms <strong>of</strong> centriole biogenesis. Strikingly, meiotic<br />

extracts are not permissive for centriole biogenesis suggesting differential regulation <strong>of</strong> <strong>the</strong><br />

kinase activity or <strong>of</strong> o<strong>the</strong>r participants in this process.<br />

[1] Flemming, W. (1875). Studien uber die Entwicklungsgeschichte der Najaden. Sitzungber<br />

Akad Wissensch Wien; 71: 81-147;<br />

[2] Boveri, T. (1900). Zellen-Studien: Uber die Natur der Centrosomen. Fischer: Jena;<br />

[3] Bettencourt-Dias, M. et al. Curr Biol 15, 2199-207 (2005);<br />

[4] Habedanck, R. et al. Nat Cell Biol 7, 1140-6 (2005);<br />

2133<br />

The temperature-sensitive ipl1-2 mutation in yeast is suppressed by mutations altering<br />

kinetochore components Duo1 and Ndc80.<br />

K. Tatchell 1 , L. Brou 1 , J. Phillips 1 , E. P. Boswell 1 , L. C. Robinson 1 ; 1 Biochemistry and <strong>Molecular</strong><br />

<strong>Biology</strong>, Louisiana State University Health Sci Ctr, Shreveport, LA<br />

Ipl1/Aurora B is <strong>the</strong> catalytic subunit <strong>of</strong> a complex that is required for chromosome segregation<br />

and nuclear division. Prior to anaphase, Ipl1 localizes to kinetochores, where it is required to<br />

establish proper kinetochore-microtubule associations and regulate <strong>the</strong> spindle assembly<br />

checkpoint (SAC). At anaphase, <strong>the</strong> complex migrates to <strong>the</strong> spindle, where it stabilizes <strong>the</strong><br />

spindle and regulates cytokinesis. We have taken a genetic approach in budding yeast to<br />

investigate Ipl1/Aurora B activity by isolating and characterizing mutations that suppress <strong>the</strong><br />

inviability <strong>of</strong> <strong>the</strong> temperature-sensitive ipl1-2 mutant at non-permissive temperatures. These<br />

suppressors include three intragenic, second site revertants in IPL1, 19 mutations in<br />

components <strong>of</strong> <strong>the</strong> Glc7 phosphatase previously reported to suppress ipl1 (GLC7, SDS22,<br />

YPI1, and SHP1), and a null mutation in TCO89, which encodes a subunit <strong>of</strong> <strong>the</strong> TORC1<br />

complex. Two revertants contain missense mutations in kinetochore components. rev76<br />

contains a missense mutation in DUO1, an essential component <strong>of</strong> <strong>the</strong> microtubule binding<br />

DASH/DAM1 complex. The mutant is cold sensitive and arrests in G2-M due to activation <strong>of</strong> <strong>the</strong><br />

SAC. rev8 contains <strong>the</strong> missense mutation ndc80-K204E, which corresponds to K116 in <strong>the</strong> CH<br />

domain <strong>of</strong> human Ndc80/Hec1. In vitro biochemical studies on human Ndc80 indicate that<br />

Ndc80-K166E binds weakly to microtubules (Ciferri et al Cell 133, 427-439, 2008). In a wild-type<br />

IPL1 background, ndc80-K204E cells grow slowly and <strong>the</strong> SAC is activated, consistent with <strong>the</strong><br />

possibility that kinetochores bind more weakly to microtubules in <strong>the</strong> ndc80-K204E mutant. The<br />

slow growth and cell cycle delay <strong>of</strong> ndc80-K204E cells are partially alleviated by <strong>the</strong> ipl1-2


MONDAY<br />

mutation. These data are consistent with <strong>the</strong> published model that <strong>the</strong> NDC80 complex makes<br />

electrostatic interactions between negatively charged regions <strong>of</strong> Ndc80 and a positively charged<br />

C-terminal domain <strong>of</strong> tubulin. Reduction in <strong>the</strong> negative charge on Ndc80 can compensate for<br />

reduced Ipl1 activity.<br />

Cell-Matrix and Cell-Cell Interactions<br />

2134<br />

A discoidin domain <strong>of</strong> <strong>the</strong> Del1 protein is essential and sufficient for its deposition in <strong>the</strong><br />

extracellular matrix.<br />

C. Hidai 1 , H. Kitano 2 , A. Mamiya 1 , S. Kokubun 1 ; 1 Biomedical Sciences, Nihon University School<br />

<strong>of</strong> Medicine, Itabashi-ku, Japan, 2 Dental surgery, Nihon University School <strong>of</strong> Medicine, Itabashiku,<br />

Japan<br />

Objective: Because <strong>the</strong> organization <strong>of</strong> <strong>the</strong> ECM directly influences tissue structure and cell<br />

behavior, determination <strong>of</strong> how ECM organization is regulated can help clarify <strong>the</strong> mechanisms<br />

<strong>of</strong> development and diseases. In <strong>the</strong> present study, we analyzed how Del1, an ECM protein, is<br />

deposited in <strong>the</strong> ECM. Del1 consists <strong>of</strong> three epidermal growth factor repeats (E1–E3) at its Nterminus<br />

and two discoidin domains (C1 and C2) at its C-terminus. The aim <strong>of</strong> this study was to<br />

investigate which domains <strong>of</strong> Del1 mediate its deposition in <strong>the</strong> ECM.<br />

Materials & Methods: We prepared cDNAs corresponding to fusion proteins <strong>of</strong> heat-stable<br />

alkaline phosphatase (AP) and Del1 deletion mutants <strong>of</strong> various lengths. For in vitro<br />

experiments, Cos7 cells were transfected with <strong>the</strong>se cDNAs and <strong>the</strong> ratio <strong>of</strong> AP activity in <strong>the</strong><br />

ECM and <strong>the</strong> medium was measured. To evaluate deposition <strong>of</strong> Dell and its fragments into <strong>the</strong><br />

ECM in vivo, <strong>the</strong> cDNAs encoding <strong>the</strong> AP fusion proteins were cloned into a non-viral vector and<br />

injected intravenously into mice. Exogenous AP activity in serum or in liver was measured and<br />

AP staining <strong>of</strong> tissue sections was performed.<br />

Results: In vitro experiments using AP-fusion constructs <strong>of</strong> various deletion mutants <strong>of</strong> Del1<br />

showed that fragments containing <strong>the</strong> C-terminus <strong>of</strong> C1, which has a lectin-like structure, are<br />

deposited in <strong>the</strong> ECM. Digestion <strong>of</strong> <strong>the</strong>se ECM with hyaluronidase from bovine testis released<br />

Del1 from <strong>the</strong> ECM, suggesting that glycosaminoglycans are involved in <strong>the</strong> deposition <strong>of</strong> Del1.<br />

The efficiency <strong>of</strong> Del1 C1-dependent deposition varied according to <strong>the</strong> presence <strong>of</strong> o<strong>the</strong>r Del1<br />

domains. A Del1 fragment containing E3 and C1 showed <strong>the</strong> highest deposition. An<br />

immunoprecipitation assay suggested that C2, which is highly homologous to C1, inhibits C1<br />

deposition by competitive binding to E3. In vivo gene transfer experiments showed that fusion <strong>of</strong><br />

exogenous proteins with <strong>the</strong> C1 deposition domain <strong>of</strong> Del1 dramatically altered <strong>the</strong> distribution<br />

<strong>of</strong> <strong>the</strong>se proteins in mice.<br />

Conclusions: This study suggests that C1 is essential and sufficient for deposition <strong>of</strong> Del1 into<br />

<strong>the</strong> ECM in vitro and in vivo. The deposition <strong>of</strong> Del1 could be regulated intramolecularly by its<br />

E3 and C2 domains.<br />

2135<br />

Tissue tension controls spatially patterned invasion <strong>of</strong> mammary epi<strong>the</strong>lial tissue by<br />

regulating EMT.<br />

K. Lee 1,2 , N. Gjorevski 1 , C. M. Nelson 1,2 ; 1 Chemical & Biological Engineering, Princeton<br />

University, Princeton, NJ, 2 <strong>Molecular</strong> <strong>Biology</strong>, Princeton University, Princeton, NJ<br />

Cells within tissues constantly experience physical forces and dynamically adapt to <strong>the</strong>se<br />

mechanical cues by modifying <strong>the</strong>ir behavior and remodeling <strong>the</strong>ir environment. Here we<br />

characterized <strong>the</strong> mechanical stress pr<strong>of</strong>ile <strong>of</strong> mammary epi<strong>the</strong>lium and determined how tissue


MONDAY<br />

mechanics regulated tissue behavior. We found that <strong>the</strong> mechanical rigidity <strong>of</strong> <strong>the</strong><br />

microenvironment was distributed non-uniformly in three-dimensional tissues and this patterned<br />

mechanical tone played a critical role in determining sites <strong>of</strong> mammary invasion. Cells within<br />

regions <strong>of</strong> high mechanical stress were surrounded by a stiff extracellular matrix (ECM), and<br />

were also more prone to invade. Moreover, <strong>the</strong> spatial distribution <strong>of</strong> mechanical stress was<br />

significantly correlated with patterned expression <strong>of</strong> genes associated with epi<strong>the</strong>lialmesenchymal<br />

transition (EMT), which promotes <strong>the</strong> switch to a motile phenotype. Changes in<br />

tissue rigidity altered cellular behaviors including cell morphology and scattering, and <strong>the</strong><br />

expression <strong>of</strong> EMT markers. These findings indicate that tissue mechanics plays a pivotal role in<br />

regulating invasion <strong>of</strong> mammary epi<strong>the</strong>lial cells by promoting EMT.<br />

2136<br />

An EGF motif <strong>of</strong> Del1 induces apoptosis in vitro.<br />

H. Kitano 1 , A. Mamiya 1 , M. Komiya 1 , S. Kokubun 2 , C. Hidai 2 ; 1 Division <strong>of</strong> Oral Surgery, Nihon<br />

University School <strong>of</strong> Medicine, Tokyo, Japan, 2 Department <strong>of</strong> Biomedical Sciences, Nihon<br />

University School <strong>of</strong> Medicine<br />

Introduction: Most cells cannot survive without integrin-mediated adherence to <strong>the</strong> extracellular<br />

matrix (ECM). However, some ECM proteins induce cell death, such as CCN1, which induces<br />

apoptosis in fibroblasts via binding with integrin ?6?1 and syndecan-4. The ECM controls cell<br />

behavior by providing ei<strong>the</strong>r pro-survival or pro-death signals to cells. Dell, an ECM protein, is<br />

known to show pro-angiogenic or anti-angiogenic activities depending on <strong>the</strong> experimental<br />

conditions. It consists <strong>of</strong> two discoidin domains (C1, C2), and three epidermal growth factor<br />

(EGF) motifs (E1, E2, E3), <strong>of</strong> which E2 is reported to contain an RGD sequence that binds to<br />

integrin receptors and supports cell survival. In <strong>the</strong> present study, we provide evidence that E3<br />

induces apoptosis in vitro.<br />

Materials & Methods: Several cell lines were treated with recombinant Del1 or its mutants.<br />

Apoptosis <strong>of</strong> <strong>the</strong> cells was evaluated with DNA laddering, chromatin condensation, annexin V<br />

staining and TUNEL staining. The signal transduction <strong>of</strong> apoptosis was analyzed using an<br />

inhibitor <strong>of</strong> caspase.<br />

Results: Gene transfer <strong>of</strong> Del1 induced cell death in cultured cells. DNA laddering, and <strong>the</strong><br />

results <strong>of</strong> annexin V staining and TUNEL staining were consistent with apoptosis. The<br />

experiments with Del1 mutant proteins with deletions or point mutations revealed that apoptotic<br />

activity was localized in E3 (34 amino acids). Because an inhibitor <strong>of</strong> caspase suppressed E3<br />

and induced apoptosis, it appears that signaling by E3 is caspase dependent.<br />

Discussion: In <strong>the</strong> Del1 protein, <strong>the</strong>re is a cell death-inducing domain, E3, adjacent to <strong>the</strong> RGD<br />

sequence, which supports cell viability. The presence <strong>of</strong> domains with opposing activities next to<br />

one ano<strong>the</strong>r in <strong>the</strong> same protein could account for <strong>the</strong> seemingly contradictory characteristics <strong>of</strong><br />

Del1 activity in various cells. Thus, it is likely that Del1functions as a balancing factor between<br />

life and death. In <strong>the</strong> present study, we also showed that <strong>the</strong> efficiency <strong>of</strong> cancer gene <strong>the</strong>rapy<br />

with <strong>the</strong> cytotoxic FasL gene can be increased by <strong>the</strong> addition <strong>of</strong> <strong>the</strong> E3 domain.<br />

2137<br />

Exploring <strong>the</strong> syndecan-4 adhesome.<br />

M. Montesion 1 , C. Flynn 1 , J. Ye 1 , M. Bonnet 1 , M. Frigault 1 , R. Bellin 1 ; 1 <strong>Biology</strong>, Holy Cross<br />

College, Worcester, MA<br />

Syndecan-4 is a transmembrane heparan sulfate proteoglycan that has been found in almost all<br />

mammalian cell types. Specifically, syndecan-4 is localized most commonly to focal adhesion<br />

complexes, and is thought to play a role in linking cells to matrix adhesion sites. In our recent<br />

work, we have demonstrated that <strong>the</strong> attachment <strong>of</strong> cells to surfaces that are engineered to


MONDAY<br />

solely support syndecan-4-based connections caused <strong>the</strong> recruitment <strong>of</strong> <strong>the</strong> focal adhesion<br />

complex proteins vinculin and talin to adhesion sites. In addition, we have shown that cells<br />

adhered solely through syndecan-4 molecules are able to transmit external mechanical strain<br />

resulting in activation <strong>of</strong> <strong>the</strong> MAPK signaling pathway within <strong>the</strong> cell. Currently, we are working<br />

to build on <strong>the</strong>se findings by conducting experiments to identify <strong>the</strong> full collection <strong>of</strong> proteins that<br />

are recruited to sites <strong>of</strong> cell adhesion when cells are subjected to syndecan-4 specific adhesion<br />

conditions--i.e., <strong>the</strong> syndecan-4 adhesome. To conduct <strong>the</strong>se studies, fibroblast cells are grown<br />

on non-specific growth surfaces, and <strong>the</strong>n treated with anti-syndecan-4 core protein antibodies<br />

coated on magnetic core microbeads to cause <strong>the</strong> generation <strong>of</strong> new adhesion complexes at<br />

sites <strong>of</strong> cell attachment to <strong>the</strong> microbeads. After subjecting cells to a reversible, membranepermeable<br />

chemical crosslinker to stabilize protein interactions, <strong>the</strong> adhesion complexes are<br />

collected on a magnet after cell lysis, <strong>the</strong> cross-linker based connections are broken and<br />

proteomic techniques are used to probe for specific proteins <strong>of</strong> interest, or for <strong>the</strong> contents <strong>of</strong><br />

<strong>the</strong> entire complex. Efforts such as <strong>the</strong>se have been conducted with a focus on specific integrinbased<br />

adhesion conditions, and we are very interested to compare our syndecan-4 based<br />

findings to <strong>the</strong> integrin projects' results, especially in light <strong>of</strong> long term questions in <strong>the</strong> literature<br />

<strong>of</strong> <strong>the</strong> need for both syndecan-4 and integrin molecules within focal adhesion complexes.<br />

2138<br />

Inverse Regulation between Integrin α2β1 and Laminin Receptors.<br />

L. Dao 1 , C. M. Franz 1 ; 1 Center for Functional Nanostructures, Karlsruhe Institute <strong>of</strong> Technology,<br />

Karlsruhe, Germany<br />

Integrin α2β1, a well-characterized collagen receptor, has repeatedly been reported to function<br />

as a laminin receptor, but its relative contribution to collagen and laminin binding has not been<br />

determined. In this work, we compared α2β1-mediated adhesion to bifunctional adhesion<br />

substrates consisting <strong>of</strong> alternating collagen type I and laminin stripes. CHO cells which do not<br />

express collagen–binding integrins, showed a highly polarized morphology and adhere only on<br />

collagen stripes, whereas CHO cells stably expressing α2β1 (CHO-A2) polarized strongly on<br />

collagen, indicating a preference <strong>of</strong> α2β1 for collagen over laminin. To directly compare α2β1mediated<br />

adhesion strength to collagen and laminin, we quantitated adhesion forces using<br />

single-cell force spectroscopy (SCFS). As expected, CHO-A2 cells adhered more strongly to<br />

collagen than wildtype cells. Comparable results were obtained for α2β1-expressing (SAOS-A2)<br />

and α2β1-deficient wildtype (SAOS-WT) human osteosarcoma cells. In turn, CHO-WT and<br />

SAOS-WT cells showed significantly stronger adhesion to laminin than <strong>the</strong> corresponding α2β1expressing<br />

cells, pointing towards a suppressing effect <strong>of</strong> α2β1 expression on laminin-binding.<br />

In agreement, RT-qPCR and western blot analysis <strong>of</strong> α2β1-expressing cells revealed a<br />

downregulation <strong>of</strong> integrin subunits α6 and β4, both components <strong>of</strong> <strong>the</strong> major laminin-binding<br />

integrin receptors α6β1 and α6β4. In conclusion, we have shown that α2β1 is an efficient<br />

laminin but not collagen receptor. Instead, α2β1 expression decreased laminin binding and α6<br />

and β4 expression, suggesting an inverse regulation <strong>of</strong> α2β1 and laminin receptors.<br />

2139<br />

Prolines in βA-sheet <strong>of</strong> neural cadherin act as a switch to control <strong>the</strong> dynamics <strong>of</strong> <strong>the</strong><br />

equilibrium between monomer and dimer.<br />

N. Vunnam 1 , S. Pedigo 1 ; 1 Chemistry and Biochemistry, University <strong>of</strong> Mississippi, University, MS<br />

Neural cadherins dimerize through <strong>the</strong> formation <strong>of</strong> calcium-dependent strand-crossover<br />

structures. Dimerization <strong>of</strong> cadherins leads to cell-cell adhesion in multicellular organisms.<br />

Strand-crossover dimer forms exclusively between <strong>the</strong> first N-terminal extracellular modules<br />

(EC1) <strong>of</strong> <strong>the</strong> adhesive partners via swapping <strong>of</strong> <strong>the</strong>ir βA-sheets and docking <strong>of</strong> tryptophan-2 in


MONDAY<br />

<strong>the</strong> hydrophobic pocket. In <strong>the</strong> apo-state wild-type cadherin is predominantly monomer, which<br />

indicates that <strong>the</strong> dimerization is energetically unfavorable in <strong>the</strong> absence <strong>of</strong> calcium. Addition <strong>of</strong><br />

calcium favors dimer formation by creating strain in <strong>the</strong> monomer and lowering <strong>the</strong> energetic<br />

barrier between monomer and dimer. Dynamics <strong>of</strong> <strong>the</strong> monomer-dimer equilibrium is vital for<br />

plasticity <strong>of</strong> synapses. Prolines recurrently occur in proteins that form strand-crossover dimer<br />

and are believed to be <strong>the</strong> source <strong>of</strong> <strong>the</strong> strain in <strong>the</strong> monomer. N-cadherins have two proline<br />

residues in <strong>the</strong> βA-sheet. We focused our studies on <strong>the</strong> role <strong>of</strong> <strong>the</strong>se two prolines in calciumdependent<br />

dimerization. Spectroscopic, electrophoretic, and chromatopgraphic studies showed<br />

that mutations <strong>of</strong> both prolines to alanines increased <strong>the</strong> dimerization affinity by ~20-fold and<br />

relieved <strong>the</strong> requirement <strong>of</strong> calcium in dimerization. The P5A and P6A mutant formed very<br />

stable dimers that required denaturation <strong>of</strong> protein to disassemble in <strong>the</strong> apo conditions. In<br />

summary, <strong>the</strong> proline residues act as a switch to control <strong>the</strong> dynamics <strong>of</strong> <strong>the</strong> equilibrium<br />

between monomer and dimer which is crucial for <strong>the</strong> plasticity <strong>of</strong> synapses.<br />

2140<br />

Calmodulin-dependent CEACAM1 monomer-dimer equilibrium <strong>of</strong> in epi<strong>the</strong>lial cells.<br />

P. C. Patel 1 , H. S. Lee 1 , S. D. Gray-owen 1 ; 1 Department <strong>of</strong> <strong>Molecular</strong> Genetics, University <strong>of</strong><br />

Toronto, Toronto, ON, Canada<br />

Carcinoembryonic antigen-related cellular adhesion molecules (CEACAMs) can associate with<br />

<strong>the</strong>mself and/or o<strong>the</strong>r CEACAM members in a manner that controls cellular growth and<br />

differentiation. CEACAM1 has previously been shown to exist in both a cis- (parallel) and a<br />

trans- (antiparallel) homophilic dimer. In this study, we used epi<strong>the</strong>lial cells to determine what<br />

controls <strong>the</strong> homophilic dimerization <strong>of</strong> CEACAM1 and describe <strong>the</strong> relative effect <strong>of</strong> <strong>the</strong><br />

monomeric versus dimeric conformation on downstream effector recruitment and cellular<br />

responses that are mediated by CEACAM1. To establish a causal relationship between <strong>the</strong><br />

oligomeric state <strong>of</strong> CEACAM1 and its biological activities, we employed mutagenic analysis to<br />

identify specific residues that are important for CEACAM1 dimerization. In addition, we provide<br />

evidence that CEACAM1 dimerization is controlled by calmodulin activation, with increasing<br />

intracellular free calcium levels promoting dissociation <strong>of</strong> <strong>the</strong> oligomeric state in a process that<br />

requires <strong>the</strong> CEACAM1 cytoplasmic domain. This stabilization <strong>of</strong> <strong>the</strong> monomeric form has<br />

implications on tyrosine phosphatase SHP-1 recruitment, CEACAM1-dependent bacterial<br />

engulfment by epi<strong>the</strong>lial cells, and cellular aggregation.<br />

2141<br />

Cell-to-Cell Fusion and Links to Cancer Cell Migration<br />

J. E. Boutilier 1,2 , R. Duncan 1,2 ; 1 Microbiology and Immunology, Dalhousie University, Halifax,<br />

NS, Canada, 2 Biochemistry and <strong>Molecular</strong> <strong>Biology</strong>, Dalhousie University, Halifax, NS, Canada<br />

Growing evidence suggests that cell-cell fusion may play a role in <strong>the</strong> initiation and/or<br />

progression <strong>of</strong> cancer. Our lab studies a unique group <strong>of</strong> reovirus-encoded cellular fusogens<br />

termed FAST (fusion-associated small transmembrane) proteins, which serve as ideal models<br />

to study <strong>the</strong> links between cancer and cellular fusion. A genetic protein-protein interaction<br />

screen identified <strong>the</strong> receptor protein tyrosine phosphatase mu (PTPµ) as a binding partner <strong>of</strong><br />

<strong>the</strong> p14 FAST protein. PTPµ is a member <strong>of</strong> <strong>the</strong> Ig superfamily and is inhibitory to cellular<br />

migration via its involvement in regulating adherens junction formation and stability. In<br />

glioblastoma cells, migration and dispersal is regulated by <strong>the</strong> proteolytic cleavage and downregulation<br />

<strong>of</strong> PTPµ. Moreover, while normal prostate cancer cells express PTPµ, invasive<br />

prostate cancer cells do not. Co-immunoprecipitation and truncation studies indicated that p14<br />

interacts with <strong>the</strong> membrane-proximal portion <strong>of</strong> <strong>the</strong> cytoplasmic tail <strong>of</strong> PTPµ, which includes <strong>the</strong><br />

active phosphatase domain. Expression <strong>of</strong> a catalytically null PTPµ protein in HT1080


MONDAY<br />

fibrosarcoma cells, which lack endogenous PTPµ, increased p14-induced cell-cell fusion 1.7<br />

fold. Similarly, siRNA knockdown <strong>of</strong> endogenous PTPµ in A549 lung adenocarcinoma cells<br />

increased syncytiogenesis 2.5 fold. These results indicated that PTPµ, in addition to being<br />

inhibitory to cellular migration, is also inhibitory to cellular fusion. Interestingly, <strong>the</strong>re was<br />

temporal regulation <strong>of</strong> p14-PTPµ interactions. Using several fusion-dead p14 constructs and a<br />

kinetic analysis <strong>of</strong> co-immunoprecipitation, we determined that p14 only interacts with PTPµ<br />

after cell-cell fusion occurs. Syncytium formation induced by p14 requires dynamic regulation <strong>of</strong><br />

adherens junctions, with cadherin engagement being required to promote initial cell-cell<br />

association and membrane fusion, however, adherens junctions need to be disrupted to allow<br />

micr<strong>of</strong>usion pores to expand to <strong>the</strong> macropores needed for syncytiogenesis. We <strong>the</strong>refore<br />

hypo<strong>the</strong>size that <strong>the</strong> stabilization <strong>of</strong> adherens junctions by PTPµ is inhibitory to pore expansion<br />

during cellular fusion, and that cell-cell fusion triggers interactions between p14 and PTPµ that<br />

may allow p14 to downregulate PTPµ activity to promote syncytium formation. These studies<br />

reveal an interesting convergence <strong>of</strong> <strong>the</strong> processes that regulate cell migration during cancer<br />

progression and cell-cell fusion on cellular pathways that regulate cell-cell adhesion and<br />

membrane-cytoskeletal dynamics.<br />

2142<br />

Establishment <strong>of</strong> a cell-based assay system to screen compounds modulating tight<br />

junction barrier.<br />

A. Watari 1 , M. Kondoh 1 , K. Yagi 1 ; 1 Graduate School <strong>of</strong> Pharmaceutical Sciences, Osaka<br />

University, Suita, Japan<br />

Objective<br />

The intestinal epi<strong>the</strong>lial cells are a first physical barrier to prevent entry <strong>of</strong> foreign antigens,<br />

bacteria, toxins and allergens from <strong>the</strong> external environment. Tight junction (TJ) in <strong>the</strong> epi<strong>the</strong>lial<br />

cells mainly maintains <strong>the</strong> barrier function. Disruption <strong>of</strong> <strong>the</strong> intestinal epi<strong>the</strong>lial TJ barrier results<br />

in increase in intestinal paracellular permeability, and induces <strong>the</strong> paracellular permeation <strong>of</strong><br />

toxic luminal substances, which leads to inflammatory bowel disease. In this study, we tried to<br />

find compounds modulating permeability in intestinal epi<strong>the</strong>lial cell sheets from various food<br />

additives, which are recognized as safe, by using claudin expression monitoring system which<br />

based on cell stably expressing claudin promoter reporter gene.<br />

Methods<br />

The cell stably expressing luciferase gene under control <strong>of</strong> 500 bp claudin-4 promoter region<br />

has been prepared. To screen compound modulating reporter activity, validated 88 food<br />

additives were tested using <strong>the</strong> claudin-4 reporter system. After adding food additives to<br />

claudin-4 reporter cell for 48 hours, luciferase activities were measured. Effect <strong>of</strong> food additives<br />

on claudin-4 expression was verified by RT-PCR and western blotting analysis. Effect on barrier<br />

function <strong>of</strong> Caco-2 cells, human colorectal adenocarcinoma cells, was measured by transepi<strong>the</strong>lial<br />

electrical resistance assay.<br />

Results<br />

Potassium carbonate decreased <strong>the</strong> claudin-4 reporter activity, whereas thiabendazol, carotene<br />

and curcumin induced it. In accordance with <strong>the</strong> alteration <strong>of</strong> reporter activity, potassium<br />

carbonate decreased claudin-4 mRNA and protein expression. Thiabendazol, carotene and<br />

curcumin increased <strong>the</strong> expression. Fur<strong>the</strong>rmore, <strong>the</strong>se compounds modulated barrier functions<br />

<strong>of</strong> Caco-2 monolayer correlating with <strong>the</strong> reporter activity. These results show that we<br />

successfully identified novel intestinal epi<strong>the</strong>lial barrier-modulating compounds using <strong>the</strong><br />

claudin-4 reporter system.<br />

Conclusion<br />

The cell-based assay system using claudin-4 promoter reporter gene is very useful for<br />

identifying compounds modulating permeability in intestinal epi<strong>the</strong>lial cell barrier, and <strong>the</strong>


MONDAY<br />

compounds identified in this system could apply to an absorption enhancer in intestine and drug<br />

for inflammatory bowel disease.<br />

2143<br />

ILDR1 and ILDR2 recruit Tricellulin to tricellular tight junctions.<br />

T. Higashi 1 , M. Furuse 1 ; 1 Cell <strong>Biology</strong>, Grad Sch <strong>of</strong> Med, Kobe University, Kobe, Japan<br />

Tricellular tight junctions (tTJs) are specialized structures to obliterate <strong>the</strong> narrow extracellular<br />

space at tricellular contacts, where <strong>the</strong> corners <strong>of</strong> three epi<strong>the</strong>lial cells meet. LSR is a tTJassociated<br />

transmembrane protein containing a single Ig-like domain and recruits Tricellulin to<br />

tTJs to maintain a sufficient paracellular barrier function. LSR has two paralogs in mammalian<br />

genomes, ILDR1 and ILDR2. Here, we examined whe<strong>the</strong>r ILDR1 and ILDR2 have <strong>the</strong> same<br />

functions as LSR. Immun<strong>of</strong>luorescence microscopy <strong>of</strong> frozen sections <strong>of</strong> mouse tissues<br />

revealed that endogenous ILDR1 was localized at tTJs in several epi<strong>the</strong>lial cell types including<br />

<strong>the</strong> colon and kidney. ILDR2 was localized at <strong>the</strong> tTJs <strong>of</strong> <strong>the</strong> retinal pigment epi<strong>the</strong>lium and<br />

perineurium. Fur<strong>the</strong>rmore, we found that LSR, but not ILDR1 and ILDR2, was expressed in<br />

mouse EpH4 mammary epi<strong>the</strong>lial cells. It has been reported that Tricellulin is diffusely<br />

distributed throughout <strong>the</strong> lateral membrane in <strong>the</strong> LSR-knockdown EpH4 cells and <strong>the</strong><br />

exogenous expression <strong>of</strong> shRNA-resistant LSR rescues <strong>the</strong> tTJ-localization <strong>of</strong> Tricellulin. When<br />

ILDR1 or ILDR2 was exogenously expressed in <strong>the</strong> LSR-knockdown EpH4 cells, <strong>the</strong>y were<br />

localized at <strong>the</strong> tricellular contacts as well as <strong>the</strong> lateral membrane. In <strong>the</strong>se cells, Tricellulin<br />

was recruited to <strong>the</strong> tTJs. Taking <strong>the</strong>se observations toge<strong>the</strong>r, we conclude that LSR, ILDR1<br />

and ILDR2 share <strong>the</strong> common characteristic <strong>of</strong> tTJ-localization and Tricellulin-recruiting ability.<br />

2144<br />

Influence <strong>of</strong> fibroblasts on <strong>the</strong> synchronization <strong>of</strong> cardiomyocyte beating and community<br />

effect.<br />

T. Kaneko 1 , F. Nomura 1 , K. Yasuda 1 ; 1 Department <strong>of</strong> Biomedical Information, Institute <strong>of</strong><br />

Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan<br />

In <strong>the</strong> normal heart, cardiomyocytes are <strong>the</strong> dominant cell type in view <strong>of</strong> volume, and more<br />

than half <strong>of</strong> <strong>the</strong> cells consist <strong>of</strong> noncardiomyocytes, mainly fibroblasts. To elucidate <strong>the</strong> role <strong>of</strong><br />

fibroblasts on <strong>the</strong> synchronization <strong>of</strong> cardiomyocyte beating, we have made cardiomyocytefibroblast<br />

network model at <strong>the</strong> single-cell level in an agarose microchamber cultivation system.<br />

The synchronization phenomenon <strong>of</strong> two cardiomyocyte networks connected by fibroblasts<br />

showed (1) propagation velocity <strong>of</strong> electrophysiological signals decreased a magnitude<br />

depending on <strong>the</strong> increasing number <strong>of</strong> fibroblasts, not <strong>the</strong> lengths <strong>of</strong> fibroblasts; (2) fluctuation<br />

<strong>of</strong> interbeat intervals <strong>of</strong> <strong>the</strong> synchronized two cardiomyocyte network connected by fibroblasts<br />

did not always decreased, and was opposite from homogeneous cardiomyocyte networks; and<br />

(3) <strong>the</strong> synchronized cardiomyocytes connected by fibroblasts sometimes loses <strong>the</strong>ir<br />

synchronized condition and recovered to synchronized condition, in which <strong>the</strong> length <strong>of</strong><br />

asynchronized period was shorter less than 30 beats and was independent to <strong>the</strong>ir cultivation<br />

time, whereas <strong>the</strong> length <strong>of</strong> synchronized period increased according to cultivation time. The<br />

results indicated that fibroblasts can connect cardiomyocytes electrically but do not significantly<br />

enhance and contribute to beating interval stability and synchronization. This might also mean<br />

that an increase in <strong>the</strong> number <strong>of</strong> fibroblasts in heart tissue reduces <strong>the</strong> cardiomyocyte<br />

"community effect", which enhances synchronization and stability <strong>of</strong> <strong>the</strong>ir beating rhythms.


MONDAY<br />

2145<br />

Determining <strong>the</strong> <strong>Molecular</strong> Link Between Adherens Junctions and Tight Junctions.<br />

J. L. Maiers 1 , X. Peng 1 , A. S. Fanning 2 , K. A. DeMali 1 ; 1 Biochemistry, University <strong>of</strong> Iowa, Iowa<br />

City, IA, 2 University <strong>of</strong> North Carolina - Chapel Hill, NC<br />

Cell-cell adhesion is critical to <strong>the</strong> development and maintenance <strong>of</strong> multicellular organisms.<br />

Adhesion between epi<strong>the</strong>lial cells is mediated by three types <strong>of</strong> junctions: tight junctions,<br />

adherens junctions and <strong>the</strong> desmosomes. This study will focus on <strong>the</strong> adherens junctions and<br />

tight junctions because <strong>the</strong>se junctions are linked spatially and functionally, with <strong>the</strong> formation <strong>of</strong><br />

tight junctions dependent on <strong>the</strong> assembly <strong>of</strong> adherens junctions. Numerous pieces <strong>of</strong> data<br />

suggest that <strong>the</strong> assembly <strong>of</strong> tight junctions is linked to <strong>the</strong> assembly <strong>of</strong> adherens junctions.<br />

However, <strong>the</strong> molecular mechanism by which <strong>the</strong>se junctions are coupled is not known. α-<br />

Catenin, a cytoplasmic component <strong>of</strong> <strong>the</strong> adherens junction, has been implicated in <strong>the</strong> coupling<br />

process as cells lacking α-catenin exhibit impaired tight junction formation and abnormal<br />

localization <strong>of</strong> tight junction proteins. Fur<strong>the</strong>r support <strong>of</strong> this notion comes from data showing<br />

that α-catenin binds directly to <strong>the</strong> tight junction protein ZO-1. Based on <strong>the</strong>se observations, I<br />

tested whe<strong>the</strong>r α-catenin is required for linking tight junctions and adherens junctions, and if it<br />

serves this purpose through regulated binding to ZO-1. To test this possibility, I generated a<br />

powerful knockdown/addback system for studying this interaction in epi<strong>the</strong>lial cells. Using this<br />

system, I found that deletion <strong>of</strong> <strong>the</strong> alpha catenin C-terminus containing <strong>the</strong> putative binding site<br />

for ZO-1 delayed tight junction assembly and impaired paracellular resistance, while leaving<br />

adherens junction assembly and function intact. These data indicate that <strong>the</strong> α-catenin Cterminus<br />

is required for proper tight junction assembly and function. To determine if <strong>the</strong> effects<br />

<strong>of</strong> α-catenin on tight junctions require direct binding <strong>of</strong> ZO-1, I mapped <strong>the</strong> ZO-1 binding site on<br />

α-catenin and generated a full length α-catenin point mutant unable to bind ZO-1. This mutant is<br />

being introduced into epi<strong>the</strong>lial cells and tight junction assembly and integrity are being<br />

analyzed. Collectively, <strong>the</strong>se data are consistent with <strong>the</strong> notion that ZO-1 binding to α-catenin<br />

is required for coupling <strong>the</strong> assembly <strong>of</strong> tight junctions to adherens junctions.<br />

2146<br />

Gap Junction Assembly: Roles for <strong>the</strong> Formation Plaque and Regulation by <strong>the</strong> C-<br />

Terminus <strong>of</strong> Connexin43.<br />

R. G. Johnson 1 , J. K. Reynhout 2 , E. M. TenBroek 1,3 , B. J. Quade 1,4 , T. Yasumura 5 , K. V.<br />

Davidson 5 , J. D. Sheridan 1 , J. I. Nagy 6 , J. E. Rash 5,7 ; 1 Genetics, Cell <strong>Biology</strong> and Development,<br />

University <strong>of</strong> Minnesota, Minneapolis, MN, 2 <strong>Biology</strong>, Be<strong>the</strong>l University, St. Paul, MN, 3 Current<br />

address: Therapy, Research and Development, Medtronic, Inc., Fridley, MN, 4 Current address:<br />

Brigham and Women’s Hospital, Boston, MA, 5 Biomedical Sciences, Colorado State University,<br />

Fort Collins, CO, 6 Physiology, University <strong>of</strong> Manitoba, Winnipeg, MB, 7 Program in <strong>Molecular</strong>,<br />

Cellular and Integrative Neurosciences, Colorado State University, Fort Collins, CO<br />

To expand our understanding <strong>of</strong> gap junction site designation and initial assembly, we examined<br />

<strong>the</strong> molecular constituents, ultrastructural features, and effects <strong>of</strong> Cx43 truncation on <strong>the</strong><br />

development <strong>of</strong> “formation plaques” (FPs). FPs as apparent sites <strong>of</strong> gap junction development<br />

were monitored in experimentally disaggregated and reaggregated hepatoma cells or in HeLa or<br />

N2A cells transfected with full-length Cx43 or a C-terminal truncation <strong>of</strong> Cx43 (at residue 257).<br />

In freeze-fracture replicas, filipin (a probe for membrane cholesterol) labeled membrane areas<br />

surrounding FPs but not <strong>the</strong> FPs or gap junctions. Freeze-fracture replica immunolabeling<br />

(FRIL) methods detected Cx43 in dispersed 10-nm membrane particles that characterize FPs,<br />

providing fur<strong>the</strong>r evidence that FP membranes are distinct membrane “domains”, and that <strong>the</strong><br />

10-nm particles are gap junction precursors (i.e., connexin hemichannels) engaged in assembly.<br />

Analysis <strong>of</strong> FPs revealed a series <strong>of</strong> defined steps in assembly, including: a) pairing <strong>of</strong> FP


MONDAY<br />

membranes and partial narrowing <strong>of</strong> <strong>the</strong> extracellular space, b) accumulation <strong>of</strong> 10-nm particles<br />

within FPs, c) linkage <strong>of</strong> individual 10-nm particles, forming conical membrane attachments, d)<br />

aggregation <strong>of</strong> 10-nm particles within FPs, with fur<strong>the</strong>r reduction in <strong>the</strong> separation between FP<br />

membranes and e) lateral fusion <strong>of</strong> connexon aggregates. These observations fur<strong>the</strong>r advance<br />

current models <strong>of</strong> gap junction initiation and assembly. With longer cell reaggregation times, we<br />

found a significant increase in <strong>the</strong> packing density <strong>of</strong> particles in aggregates. With different<br />

regulatory sites residing in <strong>the</strong> C-terminus <strong>of</strong> Cx43, we tested whe<strong>the</strong>r <strong>the</strong> M257 truncation<br />

mutants had altered FP development and/or reduced assembly. FPs continued to develop in<br />

cells with truncated Cx43, although <strong>the</strong> density <strong>of</strong> 10-nm particles within developing gap junction<br />

aggregates failed to achieve control levels. Negative regulation <strong>of</strong> assembly, as measured by<br />

intercellular dye transfer, was evaluated by following <strong>the</strong> dramatic inhibition <strong>of</strong> gap junction<br />

assembly when protein kinase C (PKC) was activated with TPA treatment. This inhibition failed<br />

to occur in <strong>the</strong> M257 cells. The studies reported here demonstrate <strong>the</strong> importance <strong>of</strong> <strong>the</strong> Cterminus<br />

<strong>of</strong> Cx43 in gap junction assembly and regulation. By confocal immun<strong>of</strong>luorescence<br />

microscopy, we found that N-cadherin was associated with Cx36 in neuronal gap junctions <strong>of</strong><br />

rodent brain. Overall, <strong>the</strong>se studies validate a model for gap junction assembly that can now be<br />

evaluated with respect to <strong>the</strong> roles played by various connexin-interacting proteins, such as N-<br />

and E-cadherin, nectins, β-catenins, ZO-1, and AF6 [aka afadin] (work in progress).<br />

Supported by NIH GM-46277 (RGJ and JDS), NS-44395 (JER), and CIHR (JIN).<br />

2147<br />

The Role <strong>of</strong> Connexin43 in <strong>the</strong> Development <strong>of</strong> Skeletal Muscle using Cell and Animal<br />

Models <strong>of</strong> Oculodentodigital Dysplasia.<br />

P. A. Merrifield 1 , S. C. Qing 1 , P. Vecchio 1 , G. I. Fishman 2 , D. W. Laird 1 ; 1 Anatomy & Cell<br />

<strong>Biology</strong>, University <strong>of</strong> Western Ontario, London, ON, Canada, 2 Leon H. Charney Division <strong>of</strong><br />

Cardiology, New York University School <strong>of</strong> Medicine, New York, NY<br />

Skeletal muscle development involves <strong>the</strong> differentiation <strong>of</strong> myoblasts into myotubes; likely<br />

involving connexins (Cx) and <strong>the</strong> gap junctional exchange <strong>of</strong> secondary messengers and<br />

metabolites. Some patients with oculodentodigital dysplasia (ODDD), a rare primarily autosomal<br />

dominant disease caused by mutations in <strong>the</strong> gene encoding Cx43, become less ambulatory<br />

with aging and exhibit symptoms that may reflect defects in skeletal muscle development,<br />

maintenance, and repair. In this study, <strong>the</strong> role <strong>of</strong> Cx43 in skeletal muscle development was<br />

examined in differentiation-competent L6 myoblasts and two ODDD-linked Cx43-mutant mouse<br />

lines. Undifferentiated L6 myoblasts exhibited high levels <strong>of</strong> Cx43-based gap junctions which<br />

were inhibited by <strong>the</strong> co-expression <strong>of</strong> I130T and G60S mutants. Upon myotube formation,<br />

Cx43 was rapidly down-regulated and gap junctions were lost but myotubes continued to<br />

enlarge. Cx43-mutant mice heterozygous for <strong>the</strong> I130T mutation exhibited similar body weight,<br />

tibial length and muscle fiber size as littermate controls. While mutant mice heterozygous for <strong>the</strong><br />

G60S mutation also had similar tibial length <strong>the</strong>ir overall size and muscle fibre diameter were<br />

significantly smaller. Overall, Cx43 regulation may be important for initial myoblast differentiation<br />

but not linked to myotube growth and while our mutant mouse studies suggest that some ODDD<br />

patients are predicted to have smaller muscle fibres, this appears to be dependent on <strong>the</strong> site <strong>of</strong><br />

<strong>the</strong> Cx43 mutation or o<strong>the</strong>r unknown etiologies (Supported by grants from <strong>the</strong> CIHR to DWL and<br />

NSERC to PAM).


2148<br />

C-terminal interactions <strong>of</strong> Cx35 that regulate gap junction coupling.<br />

Y. Wang 1 , C. K. Mitchell 1 , A. Espejo 2 , M. T. Bedford 2 , J. O'Brien 1 ; 1 Department <strong>of</strong><br />

Ophthalmology & Visual Science, UTHSC Houston, Houston, TX, 2 Department <strong>of</strong><br />

Carcinogenesis, University <strong>of</strong> Texas M.D. Anderson Cancer Center, Houston, TX<br />

MONDAY<br />

Connexin 35/36 gap junctions in central nervous system neurons display a large degree <strong>of</strong><br />

plasticity. In retinal photoreceptors and AII amacrine cells, <strong>the</strong> plasticity in coupling strength is<br />

directly correlated to <strong>the</strong> phosphorylation states <strong>of</strong> two regulatory residues (Ser110 and Ser276,<br />

using numbering for Cx35). Coupling among retinal AII amacrine cells is reduced by dopamine<br />

D1 receptor activation through activation <strong>of</strong> PKA, downstream activation <strong>of</strong> protein phosphatase<br />

2A (PP2A) and dephosphorylation <strong>of</strong> Cx36. This regulation is replicated with ei<strong>the</strong>r Cx35 or<br />

Cx36 expressed in HeLa cells. In an earlier study (Ouyang at al. 2005, Mol Brain Res 135:1-11)<br />

we found that removal <strong>of</strong> <strong>the</strong> last 7 amino acids <strong>of</strong> <strong>the</strong> Cx35 C-terminus (Ser298ter) inverted <strong>the</strong><br />

effect <strong>of</strong> PKA activity on coupling. One hypo<strong>the</strong>sis that can account for this phenomenon is that<br />

<strong>the</strong> C-terminus <strong>of</strong> Cx35/36 is essential for an interaction with regulatory proteins, and that PP2A<br />

is part <strong>of</strong> a complex that does not associate with <strong>the</strong> mutant connexin. To examine this<br />

hypo<strong>the</strong>sis, we performed a protein microarray study and peptide pulldown experiments to<br />

examine interactions with <strong>the</strong> Cx35 C-terminus. We also examined regulation <strong>of</strong> coupling in a<br />

variety <strong>of</strong> C-terminal mutants expressed in HeLa cells. A 22 amino acid peptide corresponding<br />

to <strong>the</strong> tip <strong>of</strong> <strong>the</strong> Cx35 C-terminus bound to several different PDZ domains, including domains<br />

from MUPP1, NHERF2, and PDZK1. Phosphorylation <strong>of</strong> <strong>the</strong> C-terminus at Ser298 revealed<br />

binding to 3 different is<strong>of</strong>orms <strong>of</strong> 14-3-3 proteins. Truncation <strong>of</strong> <strong>the</strong> C-terminus at Ser298<br />

eliminated all <strong>of</strong> <strong>the</strong> interactions. These binding interactions were confirmed by pulldown<br />

experiments with recombinant PDZ and 14-3-3 proteins. In transfected HeLa cells, mutation <strong>of</strong><br />

serine 298 to alanine, which prevents S298 phosphorylation and should prevent 14-3-3 binding,<br />

also inverted regulation <strong>of</strong> coupling by PKA activity. In contrast, Cx35 with a C-terminal YFP tag,<br />

which should prevent PDZ binding, showed wild-type regulation by PKA activity. The results<br />

suggest that phosphorylation <strong>of</strong> Ser298 and recruitment <strong>of</strong> 14-3-3 proteins is involved in <strong>the</strong><br />

regulation <strong>of</strong> Cx35 coupling. 14-3-3 binding may facilitate assembly <strong>of</strong> a signaling complex<br />

containing PP2A or it may alter <strong>the</strong> sensitivity <strong>of</strong> Cx35 to dephosphorylation by PP2A.<br />

2149<br />

Matrix Stiffness Dictates Cytoskeletal Organization and Nuclear Morphology in 2D and<br />

3D.<br />

F. Rehfeldt 1,2 , T. Harada 2 , A. E. Brown 2 , A. Zajac 2 , S. Cai 2 , A. Zemel 3 , D. E. Discher 2 ; 1 3rd Inst.<br />

Physics - Biophysics, University Goettingen, Goettingen, Germany, 2 Biophysical Engineering<br />

Lab, University <strong>of</strong> Pennsylvania, Philadelphia, PA, 3 Hebrew University Jerusalem, Israel<br />

Physical aspects <strong>of</strong> <strong>the</strong> microenvironment such as matrix elasticity E can influence morphology,<br />

mechanics, and even <strong>the</strong> lineage choices <strong>of</strong> stem cells. An E-series <strong>of</strong> two types <strong>of</strong> hydrogels<br />

are ligand-functionalized here with collagen and used to elucidate wide-ranging cell,<br />

cytoskeletal, and nuclear responses to E in both two and three dimensional matrix geometries.<br />

Cross-linked hyaluronan-based hydrogels show that, within just hours <strong>of</strong> initial plating, cell<br />

adhesion, shape, and cytoskeletal order depend non-linearly on E over a broad range <strong>of</strong><br />

physiologically relevant elasticities. Force generation by non-muscle myosin IIs is required for<br />

<strong>the</strong> various cell responses. These include nuclear shape changes that can also be achieved in<br />

part by siRNA-knockdown <strong>of</strong> a key nuclear envelope gene, lamin-A/C, which is implicated in<br />

differentiation <strong>of</strong> s<strong>of</strong>t tissues such as fat and stiff tissues such as bone. The results lend support<br />

to an important matrix-myosin-nucleus pathway.


MONDAY<br />

2150<br />

Characterisation <strong>of</strong> tissue microenvironment by <strong>the</strong> direct culture <strong>of</strong> mesenchymal stem<br />

cells on tissue sections.<br />

W. Shen 1 , W. Y. Tong 2 , Y. Zhao 2 , C. F. Chan 3 , P. Chu 1 , W. K. Yeung 2 , Y. W. Lam 1 ; 1 City<br />

University <strong>of</strong> Hong Kong, Hong Kong, 2 Department <strong>of</strong> Orthopaedics and Traumatology, The<br />

University <strong>of</strong> Hong Kong, Hong Kong, 3 Department <strong>of</strong> Pediatircs & Adolescent Medicine, The<br />

University <strong>of</strong> Hong Kong, Hong Kong<br />

One ultimate goal <strong>of</strong> tissue engineering is to construct functional tissues and organs from<br />

isolated cells and fabricated materials. To achieve this, it is important to understand how<br />

extracellular environments affect and control cell behaviours. Although many studies have<br />

characterised <strong>the</strong> contributions <strong>of</strong> physical and biochemical factors in extracellular environments<br />

to cell physiology, <strong>the</strong>se works <strong>of</strong>ten fail to mimic <strong>the</strong> complexity <strong>of</strong> <strong>the</strong> native tissue<br />

microenvironment. We have developed a novel experimental system to evaluate <strong>the</strong> interactions<br />

<strong>of</strong> cells and extracellular microenvironment. We discovered that cryosections <strong>of</strong> Bovine Achilles<br />

tendon display intricate textural details on <strong>the</strong> surface, and hypo<strong>the</strong>sised that <strong>the</strong>se surfaces<br />

represent <strong>the</strong> internal tissue environments to which endogenous cells are exposed. By<br />

manipulating <strong>the</strong> orientation <strong>of</strong> sectioning, we could create section surfaces with highly different<br />

ultrastructures. Since <strong>the</strong>se sections were carved from <strong>the</strong> same piece <strong>of</strong> tendon, <strong>the</strong>ir<br />

biochemical compositions were identical. To assess how cells responded to <strong>the</strong>se surfaces, we<br />

directly cultured mammalian cancer cell lines and human mesenchymal stem cells (MSC), on<br />

<strong>the</strong>se sections. Morphology, adhesion and proliferation <strong>of</strong> cells were highly dependent on <strong>the</strong><br />

tissue architecture <strong>the</strong>y were exposed to. In particular, cells seeded on <strong>the</strong> longitudinal sections<br />

(LS) <strong>of</strong> tendon, but not cross sections (CS), adopted a highly elongated and aligned<br />

morphology. Time-lapse microscopy revealed that MSC on both LS and CS initially projected<br />

filopodia to all directions, but only cells on LS spread along one orientation. Remarkably, MSC<br />

cultured on LS, but not CS, expressed protein biomarkers characteristic <strong>of</strong> tenocytes,<br />

suggesting that LS contained biological cues that instructed MSC to commit to <strong>the</strong> tenogenic<br />

lineage. To delineate <strong>the</strong> nature <strong>of</strong> this signal, we prepared PDMS replicas using tendon<br />

sections as <strong>the</strong> mould. The resulting replicas, which faithfully copied <strong>the</strong> physical shape <strong>of</strong><br />

tendon sections, but not <strong>the</strong> biochemical composition, promoted a small degree <strong>of</strong> cell<br />

elongation, but failed to induce MSC differentiation into tenocyte-like cells. This suggests that<br />

although <strong>the</strong> physical structure <strong>of</strong> tissue microenvironment may be enough to modulate cell<br />

shape and morphology, specific biochemical molecules, when presented in a correct orientation,<br />

are required to drive stem cell differentiation. This study demonstrated how biophysical and<br />

biochemical information in <strong>the</strong> extracellular microenvironment intertwined to form a unique cell<br />

type specific niche that influences <strong>the</strong> fate <strong>of</strong> cells within a tissue. Direct culture <strong>of</strong> cells on<br />

sectioned tissues is a simple and useful model system for <strong>the</strong> study <strong>of</strong> this process.<br />

Regulation and Organization <strong>of</strong> <strong>the</strong> Genome<br />

2151<br />

Clusterin as a target gene <strong>of</strong> HIF-1alpha in prostate cancer cells.<br />

E. Shin 1 , S. Park 1 , J. Park 1 , D. Lee 1 , G. Roh 1 , H. Kim 1 , S. Kang 1 , G. Cho 1 , W. Choi 1 ;<br />

1 Department <strong>of</strong> Anatomy and Neurobiology, BK21 Biomedical Center, Gyeongsang National<br />

University, JINJU, Korea<br />

Clusterin (CLU) is a stress-response glycoprotein that increases expression by diverse<br />

apoptotic triggers in several human cancers such as prostate cancers. Previous studies showed<br />

that CLU expression was up-regulated in patho-physiological condition such as oxidative stress.


MONDAY<br />

HIF-1alpha (hypoxia inducible factor-1alpha) is a major regulator in oxygen homeostasis and<br />

transcriptional factor mediating <strong>the</strong> cellular response to hypoxia. In this study we evaluated<br />

whe<strong>the</strong>r CLU expression is regulated by HIF-1alpha in human prostate cancer cells exposed to<br />

Cobalt chloride (CoCl2). Interestingly, we observed that both CLU and HIF-1alpha were upregulated<br />

in mRNA and protein level. Also overexpression <strong>of</strong> HIF-1alpha up-regulates CLU<br />

expression in prostate cancer cells. Therefore, to evaluate <strong>the</strong> transcriptional regulation <strong>of</strong> CLU<br />

promoter by HIF-1alpha, we searched hypoxia response element (HRE) <strong>of</strong> Human CLU<br />

promoter sequence. Electrophoretic mobility shift assay (EMSA) and Chromatin<br />

Immunoprecipitation (ChIP) analyses demonstrated that HIF-1alpha binds <strong>the</strong> three putative<br />

HRE sites in CLU promoter. Reporter gene assay showed that three HRE sites were<br />

functionally active. Our results suggest that <strong>the</strong> transcription factor HIF-1alpha specifically binds<br />

to three HRE sites <strong>of</strong> CLU promotor and directly regulates <strong>the</strong> transcription <strong>of</strong> <strong>the</strong> human CLU<br />

gene in prostate cancer cells exposed to hypoxia.<br />

2152<br />

Phosphorylation <strong>of</strong> p53 by p38α regulates <strong>the</strong> expression <strong>of</strong> Gadd45β in cardiomyocytes.<br />

Y-A. Kim 1,2 , M-Y. Kim 1 , H. Yu 1,3 , S. Mishra 1 , J-H. Lee 2 , K. Choi 2 , J-H. Kim 2 , Y-S. Jung 1,2 ;<br />

1 College <strong>of</strong> Pharmacy, Ajou University, Suwon, Korea, 2 Brain Korea 21 for <strong>Molecular</strong> Science<br />

and Technology, Ajou University, Suwon, Korea, 3 Brain Korea 21 for Medical Sciences, School<br />

<strong>of</strong> Medicine, Ajou University, Suwon, Korea<br />

Both growth arrest and DNA damage-inducible 45β (Gadd45β) and p53 plays a critical<br />

mediatory role in cardiomyocyte apoptosis under ischemia/hypoxia. However, little is known<br />

about how Gadd45β expression is regulated under ischemia/hypoxia. In this study, we<br />

hypo<strong>the</strong>sized that Gadd45β is a direct target <strong>of</strong> p53 under ischemia/hypoxia in cardiomyocytes.<br />

Using actinomycin D, it was found that <strong>the</strong> expression <strong>of</strong> Gadd45β during hypoxia is regulated<br />

at <strong>the</strong> transcriptional level in rat heart-derived H9c2 cells. Silencing p53 gene abrogated <strong>the</strong><br />

induction <strong>of</strong> Gadd45β promoter luciferase reporter (Gadd45β-Luc) activity triggered by hypoxia,<br />

whereas overexpression <strong>of</strong> p53 enhanced activity <strong>of</strong> Gadd45β-Luc. Moreover, chromatin<br />

immunoprecipitation assay revealed that binding <strong>of</strong> p53 to Gadd45β promoter region during<br />

hypoxia, and direct binding between <strong>the</strong>m was confirmed by surface plasmon resonance<br />

imaging. We also observed <strong>the</strong> necessary role <strong>of</strong> p53 in inducing Gadd45β expressions in H9c2<br />

cells under hypoxia and in a rat myocardium <strong>of</strong> ischemia model. Fur<strong>the</strong>r studies showed that<br />

p38α was activated and responsible for <strong>the</strong> p53-dependent expressions <strong>of</strong> Gadd45β under<br />

hypoxia. We revealed that p38α induced <strong>the</strong> distinct temporal pr<strong>of</strong>iles <strong>of</strong> phosphorylation at<br />

Ser15 or Ser20 <strong>of</strong> p53, and mutation <strong>of</strong> ei<strong>the</strong>r site on p53 was sufficient to block <strong>the</strong> hypoxiainduced<br />

Gadd45β-Luc activity and Gadd45β expressions. Finally, p53-induced apoptosis during<br />

hypoxia was inhibited by Gadd45β gene silencing. These results provide first evidence that<br />

Gadd45β is a direct transcriptional target <strong>of</strong> p53 under ischemia/hypoxia and that <strong>the</strong> hypoxiap38α-p53-Gadd45β<br />

signaling module serves as an essential regulator <strong>of</strong> cardiomyocyte<br />

apoptosis. This work was supported by Basic Science Research Program through <strong>the</strong> National<br />

Research Foundation <strong>of</strong> Korea (NRF) funded by <strong>the</strong> Ministry <strong>of</strong> Education, Science and<br />

Technology (2011-0012659).<br />

2153<br />

Tramtrack69 Regulation <strong>of</strong> Notch Target Genes in Drosophila melanogaster.<br />

D. Eastman 1 , E. Hildebrand 2 ; 1 <strong>Biology</strong>, Connecticut College, New London, CT, 2 Connecticut<br />

College, New London, CT<br />

The Notch pathway is critical for determining many different cell types in developing animals. In<br />

Drosophila, <strong>the</strong> main targets <strong>of</strong> Notch signaling are <strong>the</strong> Enhancer <strong>of</strong> split (E(spl)) genes.


MONDAY<br />

Although all <strong>of</strong> <strong>the</strong>se genes are activated by Notch, <strong>the</strong>y have distinct expression patterns in<br />

developing embryos and larval imaginal discs. We are interested in determining <strong>the</strong> factors that<br />

are responsible for this differential expression. We previously identified sequences within <strong>the</strong><br />

promoters <strong>of</strong> <strong>the</strong>se genes that are conserved in at least 12 Drosophila species: some <strong>of</strong> <strong>the</strong>se<br />

are unique to individual genes and some are found upstream <strong>of</strong> a sub-set <strong>of</strong> <strong>the</strong> E(spl) genes.<br />

One <strong>of</strong> <strong>the</strong>se sequences is a putative site for <strong>the</strong> known transcriptional regulator, Tramtrack69<br />

(TTK69). We show that over- expression <strong>of</strong> TTK69 in eye discs down-regulates HLHmgamma<br />

expression. In addition, we have found that TTK69 inhibits activated Notch (ICN) induced E(spl)<br />

HLHmgamma expression in Drosophila tissue culture cells and that this affect is dependent on<br />

<strong>the</strong> presence <strong>of</strong> <strong>the</strong> BTB/POZ repressor domain at <strong>the</strong> TTK69 N-terminus. We are currently<br />

characterizing <strong>the</strong> interaction <strong>of</strong> TTK69 with <strong>the</strong> Notch activation complex and determining<br />

whe<strong>the</strong>r it inhibits Notch activation <strong>of</strong> o<strong>the</strong>r E(spl) genes.<br />

2154<br />

The transcriptional activation <strong>of</strong> melanocortin 2 receptor accessory protein by<br />

PPAR[gamma]2 in adipocyte.<br />

S. KIM 1 , S. Kwak 1 , Y-J. Kim 1 ; 1 Department <strong>of</strong> <strong>Biology</strong>, Kyung Hee University, Seoul, Korea<br />

Adrenocorticotropic hormone (ACTH) in rodents decreases body weight as well as <strong>the</strong> secretion<br />

<strong>of</strong> leptin. MC2R and Mrap are known as receptors <strong>of</strong> ACTH in adipocyte. PPARγ2 also plays<br />

roles in <strong>the</strong> transcriptional regulation in adipocyte such as adipogenesis and fatty acid betaoxidation.<br />

Nuclear receptors activated by PPARγ2 bind to target genes for <strong>the</strong>ir transcriptional<br />

activation. In <strong>the</strong> present study we investigated that <strong>the</strong> Mrap transcript is regulated by PPARγ2.<br />

After <strong>the</strong> differentiation <strong>of</strong> adipocyte, <strong>the</strong> expression pattern <strong>of</strong> Mrap was similar to that <strong>of</strong><br />

PPARγ2. In <strong>the</strong> presence <strong>of</strong> a PPAR agonist, <strong>the</strong> level <strong>of</strong> Mrap mRNA was highly increased. In<br />

addition, we identified putative PPRE site in <strong>the</strong> Mrap promoter by reporter assay, chromatin<br />

immunoprecipitation assay and <strong>the</strong> mutagenesis study. These results suggest that PPARγ2 is<br />

required for <strong>the</strong> transcriptional activity <strong>of</strong> Mrap during adipogenesis, which could be contributed<br />

to understand <strong>the</strong> molecular mechanism <strong>of</strong> lipolysis in adipocytes.<br />

2155<br />

Role <strong>of</strong> TopBP1 partner WDR18 in DNA damage checkpoint.<br />

J. Willis 1 , D. DeStephanis 1 , V. Gowda 1 , S. Yan 1 ; 1 Department <strong>of</strong> <strong>Biology</strong>, University <strong>of</strong> North<br />

Carolina at Charlotte, Charlotte, NC<br />

DNA damage checkpoint has significant implications for biological and pathological processes<br />

such as cancer and aging. Genomes <strong>of</strong> all living organisms are exposed to a variety <strong>of</strong> threats,<br />

but <strong>the</strong> genome’s DNA damage checkpoint functions as a surveillance mechanism, monitoring<br />

damage in a genome and directing cellular responses to such damage. If failing to activate DNA<br />

damage checkpoint when necessary, <strong>the</strong> result is unrepaired damage that leads to genomic<br />

instability and tumor formation. The main barriers to understanding this process are in<br />

identifying how checkpoints sense DNA damage and how <strong>the</strong> proteins relay <strong>the</strong> damage signal.<br />

Specifically, it is not known how <strong>the</strong> activated sensor kinase ATR (ATM and Rad3-related)<br />

phosphorylates its downstream effector protein Chk1 (Checkpoint kinase 1). In addition to <strong>the</strong><br />

critical roles in DNA replication initiation and DNA replication stress response, TopBP1 (DNA<br />

topoisomerase II β binding protein 1) plays an important role in DNA damage checkpoint<br />

signaling through its C-terminus region. However, <strong>the</strong> underlying mechanism is not known. We<br />

hypo<strong>the</strong>size that a TopBP1-interacting protein contributes to its role in DNA damage checkpoint<br />

signaling. The objective <strong>of</strong> this study is to reveal <strong>the</strong> molecular mechanism <strong>of</strong> how TopBP1 Cterminus<br />

contributes to DNA damage checkpoint signaling. Xenopus egg extracts, a reliable<br />

cell-free biochemical system, has been used for a wide variety <strong>of</strong> studies in cell cycle and


MONDAY<br />

checkpoint activation. Our data have shown that WDR18 (WD40 repeat-containing protein 18),<br />

a TopBP1-interacting protein, interacts with TopBP1 C-terminus, and <strong>the</strong>ir interaction is required<br />

for ATR activation <strong>of</strong> Chk1 in <strong>the</strong> response to double-stranded breaks. Fur<strong>the</strong>rmore,<br />

immunodepletion <strong>of</strong> endogenous WDR18 from Xenopus egg extract also compromises DNA<br />

damage checkpoint signaling. Taken toge<strong>the</strong>r, WDR18 plays an essential role in DNA damage<br />

checkpoint signaling via interacting with TopBP1 C-terminus.<br />

This work was supported, in part, by funds provided by The University <strong>of</strong> North Carolina at<br />

Charlotte and Wachovia foundation fund for faculty excellence.<br />

2156<br />

A genetic screen to identify interactions between cdc13-1and yku80 mutant alleles on<br />

yeast telomeres.<br />

L. Sanchez 1 , G. Guillaume 1 , S. Flores 1 , C. J. Hengartner 1 , L. R. Vega 1 ; 1 <strong>Biology</strong>, Barry<br />

University, Miami Shores, FL<br />

Telomeres are <strong>the</strong> physical ends <strong>of</strong> eukaryotic chromosomes that function to protect DNA ends<br />

from degradation and from end to end fusion. Telomeres consist <strong>of</strong> stretches <strong>of</strong> repeated C/Grich<br />

DNA ending with 3’ single stranded G-rich overhangs. The maintenance and function <strong>of</strong><br />

telomeres are facilitated by <strong>the</strong> enzyme telomerase and by accessory proteins such as Ku and<br />

Cdc13p. Cdc13p is an essential, G-strand binding protein that functions in telomere protection<br />

and in telomerase recruitment. cdc13-1 is a temperature sensitive allele <strong>of</strong> CDC13, that is<br />

defective for telomere end protection. Ku is a non-essential heterodimer composed <strong>of</strong> Ku70p<br />

and Ku80p. Ku plays multiple roles in DNA metabolism including: non-homologous end joining,<br />

recombination and end protection. Ku also interacts with TLC1, <strong>the</strong> RNA template <strong>of</strong> <strong>the</strong><br />

telomerase enzyme and has recently been shown to exhibit end-binding activity. This study<br />

examines <strong>the</strong> effect <strong>of</strong> mutations in yKU80 on cdc13-1 strains. We will also determine genetic<br />

interactions between yku80 mutants and Pif1p, a helicase that inhibits telomerase activity. We<br />

previously showed that cdc13-1 strains deleted for PIF1 display hyper-elongated telomeres and<br />

increased temperature resistance. Telomere elongation in cdc13-1, pif1∆ strains partially<br />

depends on <strong>the</strong> Ku-TLC1 interaction. Using a genetic library <strong>of</strong> yku80 mutations generated in A.<br />

Bertuch’s laboratory, we have introduced <strong>the</strong> yku80 alleles into <strong>the</strong> cdc13-1 background. The<br />

goals <strong>of</strong> <strong>the</strong>se experiments are to determine <strong>the</strong> effects on viability and telomere end protection<br />

<strong>of</strong> <strong>the</strong> various yku80 mutant alleles in cdc13-1 strains. We hope to identify a cdc13-1, yku80<br />

double mutant that mimics <strong>the</strong> phenotypes <strong>of</strong> cdc13-1, pif1∆ strains.<br />

Supported by NIH-NIGMS MBRS RISE Grant: 2R25 GM059244-10, Barry University and NIH-<br />

NIGMS/NCI MBRS SCORE grant, 5SC 2CA 138567 to LRV<br />

2157<br />

Function <strong>of</strong> Daxx at centromeres and pericentromeres.<br />

V. Morozov 1 , E. Gavrilova 2,3 , A. Ishov 1 ; 1 Anatomy & Cell biology, University <strong>of</strong> Florida,<br />

Gainesville, FL, 2 Institute <strong>of</strong> Cytology Russian Academy <strong>of</strong> Sciences, St. Petersburg, Russia,<br />

3 Faculty <strong>of</strong> <strong>Biology</strong> and Soil Sciences, St. Petersburg State University, St. Petersburg, Russia<br />

Heterochromatin architecture is essential for <strong>the</strong> proper orchestration <strong>of</strong> nuclear processes,<br />

while transcription from this part <strong>of</strong> genome is required for its own maintenance. Here we<br />

present first evidence that depletion <strong>of</strong> protein Daxx affects transcription <strong>of</strong> human<br />

heterochromatin, reducing accumulation <strong>of</strong> centromeric (CEN) RNA in normal conditions and<br />

pericentromeric (periCEN) RNA after heat shock (HS) application. Searching for <strong>the</strong> mechanism<br />

<strong>of</strong> Daxx-dependent regulation <strong>of</strong> heterochromatin transcription, we found that depletion <strong>of</strong> Daxx


MONDAY<br />

decreases incorporation <strong>of</strong> transcription-associated histone H3 variant, H3.3, into both CEN and<br />

periCEN.<br />

In normal conditions, Daxx is mostly accumulated at ND10/PML nuclear bodies, with minor<br />

association with CEN/periCEN in subpopulation <strong>of</strong> cells. HS changes this balance forcing very<br />

robust accumulation <strong>of</strong> Daxx on CEN/periCEN. Surprisingly, this transient redistribution <strong>of</strong> Daxx<br />

does not fur<strong>the</strong>r elevate incorporation <strong>of</strong> H3.3 that remained steady during HS and recovery.<br />

Instead, depletion <strong>of</strong> Daxx leads to HS-induced changes in <strong>the</strong> balance <strong>of</strong> epigenetic<br />

modifications at heterochromatin, most dramatically elevating levels <strong>of</strong> H3K4Me2 at periCEN.<br />

We propose dualistic function <strong>of</strong> Daxx-containing complexes at CEN/periCEN: 1) regulation <strong>of</strong><br />

H3.3 loading in normal conditions, and 2) protection <strong>of</strong> epigenetic status upon stress application,<br />

thus collectively guarding epigenetic identity <strong>of</strong> heterochromatin and genome integrity.<br />

2158<br />

Structural Requirements <strong>of</strong> <strong>the</strong> JIL-1 Tandem Kinase for Histone H3S10 Phosphorylation.<br />

Y. Li 1 , W. Cai 1 , C. Wang 1 , X. Bao 1 , H. Deng 1 , J. Girton 1 , J. Johansen 1 , K. M. Johansen 1 ;<br />

1 Biochemistry, Biophysics & <strong>Molecular</strong> <strong>Biology</strong>, Iowa State University, Ames, IA<br />

The JIL-1 tandem kinase in Drosophila localizes specifically to euchromatic interband regions <strong>of</strong><br />

polytene chromosomes and is <strong>the</strong> predominant kinase controlling <strong>the</strong> phosphorylation state <strong>of</strong><br />

histone H3S10 at interphase. JIL-1 can be divided into four main domains including a NH2terminal<br />

domain (NTD), two kinase domains (KDI and KDII), and a COOH-terminal domain<br />

(CTD). Our laboratory has previously determined that <strong>the</strong> CTD-domain <strong>of</strong> JIL-1 is sufficient for<br />

proper chromatin localization and for rescue <strong>of</strong> JIL- 1 null mutant chromosome morphology.<br />

Interestingly a JIL-1 construct without <strong>the</strong> CTD-domain has kinase activity for histone H3S10<br />

despite that it does not localize properly. However, <strong>the</strong> potential contributions from <strong>the</strong> NTD was<br />

not determined. Thus, in order to fur<strong>the</strong>r characterize <strong>the</strong> structural requirements <strong>of</strong> <strong>the</strong> JIL-1<br />

kinase for H3S10 phosphorylation, we generated a series <strong>of</strong> mutated JIL-1 constructs and<br />

expressed <strong>the</strong>m in a JIL-1 null mutant background. Our results show that a JIL-1 construct<br />

without <strong>the</strong> NTD localizes properly to chromatin and rescues mutant polytene chromosome<br />

morphology. However, in immunocytochemistry and immunoblot analyses with H3S10ph<br />

antibody no histone H3S10 phosphorylation could be detected, strongly suggesting that <strong>the</strong><br />

NTD domain is required for H3S10 kinase activity. Fur<strong>the</strong>rmore, we mutated ei<strong>the</strong>r or both<br />

kinase domains to render <strong>the</strong>m "kinase dead" and assessed <strong>the</strong>ir ability to phosphorylate<br />

H3S10. The results indicate that both kinase domains are required for kinase activity. In<br />

addition, using a LacI/lacO-repeat te<strong>the</strong>ring system we provide evidence that only JIL-1<br />

constructs with H3S10 phosphorylation activity have <strong>the</strong> capacity to induce a change in higher<br />

order chromatin structure from a condensed heterochromatin-like band state to a more open<br />

euchromatic interband state. These findings provide direct evidence that <strong>the</strong> epigenetic histone<br />

tail modification <strong>of</strong> H3S10 phosphorylation at interphase can function as a causative regulator <strong>of</strong><br />

chromatin structure. Supported by NIH grant GM62916.<br />

2159<br />

Epigenetic Regulation <strong>of</strong> MARCO in Tolerized Macrophages.<br />

J. Jing 1 , B. O’Connor 2 , B. Pederson 3 , E. Davidson 3 , I. V. Yang 3 , D. Schwartz 3 ;<br />

1 University <strong>of</strong> Colorado Denver, Denver, CO, 2 National Jewish Health, 3 University <strong>of</strong> Colorado<br />

Denver<br />

Macrophages play a key role in host defense against microbes through phagocytosis. MARCO<br />

(macrophage receptor with collagenous structure) is one <strong>of</strong> <strong>the</strong> scavenger receptors on <strong>the</strong> cell<br />

surface <strong>of</strong> macrophages that mediates <strong>the</strong> opsonin-independent phagocytosis. The goal <strong>of</strong> our<br />

study was to study <strong>the</strong> role <strong>of</strong> epigenetic regulation <strong>of</strong> MARCO in lipopolysaccharide (LPS) or


MONDAY<br />

lipotechoic acid (LTA)-induced tolerance. We first demonstrated that phagocytosis was<br />

increased in mouse bone marrow derived macrophage (MBMM) repeated stimulation by LPS or<br />

LTA. The expression <strong>of</strong> MARCO, but not o<strong>the</strong>r scavenger receptors, was also elevated,<br />

suggesting that MARCO plays a key role in <strong>the</strong> phagocytosis <strong>of</strong> tolerized macrophages. We<br />

comprehensively evaluated genome-wide DNA methylation by CHARM (comprehensive highthroughput<br />

arrays for relative methylation) and pr<strong>of</strong>iled <strong>the</strong> mRNA expression in tolerized<br />

macrophages. Although <strong>the</strong>re were no methylation changes associated with MARCO in<br />

tolerized compared to nontolerized macrophages, several transport related genes (SNX30,<br />

SNX24, Kif21b, Kif5, Kif6 and Kif9) have significant methylation changes and at least two fold<br />

mRNA expression change, indicating <strong>the</strong>se genes may be involved in regulating <strong>the</strong> transport <strong>of</strong><br />

MARCO to cells surface. We are in <strong>the</strong> process <strong>of</strong> confirming <strong>the</strong>se findings by examining<br />

expression <strong>of</strong> MARCO on <strong>the</strong> cell surface following siRNA-mediated inhibition <strong>of</strong> <strong>the</strong>se<br />

candidate genes. In addition, we are exploring whe<strong>the</strong>r H3K4me3 and H3K9me2 histone<br />

modifications contribute to increased expression <strong>of</strong> MARCO in tolerized macrophages.<br />

2160<br />

<strong>Molecular</strong> Basis for Interaction <strong>of</strong> let-7 microRNAs with Lin28.<br />

Y. Nam 1 , C. Chen 1 , R. Gregory 1 , J. Chou 1 , P. Sliz 1 ; 1 Harvard Medical School, Boston, MA<br />

MicroRNAs are small non-coding RNA molecules that regulate gene expression. Members <strong>of</strong><br />

<strong>the</strong> let-7 microRNA family control many cell fate determination genes to influence pluripotency,<br />

differentiation, and transformation. Lin28 is a specific, post-transcriptional inhibitor <strong>of</strong> let-7<br />

biogenesis. Moreover, Lin28 is a potent cell reprogramming factor, abnormally expressed in<br />

various cancers, and linked to various developmental traits in worms and mammals. Growing<br />

evidence suggests that Lin28 also binds certain mRNAs to regulate translation independent <strong>of</strong><br />

let-7.<br />

A major question regarding Lin28 activity is how it recognizes its RNA substrates, and also how<br />

it inhibits microRNA processing enzymes-Drosha and Dicer-while also recruiting a modification<br />

enzyme, TUTase, to target let-7 precursors to degradation. We report crystal structures <strong>of</strong> Lin28<br />

in complex with pre-element (preE) portions, or "terminal loop regions", <strong>of</strong> let-7d, let-7-f1, and<br />

let-7g precursors. The two folded domains <strong>of</strong> Lin28 recognize two distinct regions <strong>of</strong> <strong>the</strong> RNA;<br />

<strong>the</strong> terminal regions outside <strong>of</strong> <strong>the</strong>se domains are dispensable for inhibition <strong>of</strong> let-7 in vivo. We<br />

also show by NMR spectroscopy that <strong>the</strong> linker connecting <strong>the</strong> two folded domains is flexible, to<br />

accommodate Lin28 binding to diverse let-7 family members. Mutagenesis studies <strong>of</strong> protein<br />

and RNA components identified mutations in <strong>the</strong> binding interface that affect complex formation<br />

in vitro, Dicer activity in vitro, and processing <strong>of</strong> let-7 microRNAs in cultured cells.<br />

Our work underscores <strong>the</strong> importance <strong>of</strong> preEs in microRNA regulation, and we present a<br />

preferred preE sequence for binding to Lin28. Our model also provides a mechanistic<br />

explanation for <strong>the</strong> inhibitory effect <strong>of</strong> Lin28 on miRNA processing by Dicer; it fur<strong>the</strong>r suggests<br />

that induced folding in <strong>the</strong> CCHCx2:GGAG part <strong>of</strong> <strong>the</strong> complex directly influences downstream<br />

factor(s) important for let-7 regulation.


MONDAY<br />

2161<br />

The karyopherin Sal3 is required for nuclear import <strong>of</strong> <strong>the</strong> core RNA interference pathway<br />

protein Rdp1.<br />

J. Park 1 , S. I. Freitag 2 , P. Young 2 , T. C. Hobman 1 ; 1 Cell <strong>Biology</strong>, University <strong>of</strong> Alberta,<br />

Edmonton, AB, Canada, 2 <strong>Biology</strong>, Queen's University, Kingston, ON, Canada<br />

RNA-dependent RNA polymerase activity is required for RNA interference (RNAi) in many lower<br />

eukaryotes including <strong>the</strong> fission yeast S. pombe. Toge<strong>the</strong>r with Ago1 and Dcr1, <strong>the</strong> RNAdependent<br />

RNA polymerase Rdp1 mediates small RNA-dependent transcriptional- and<br />

posttranscriptional gene-silencing. Whereas <strong>the</strong> bulk <strong>of</strong> Rdp1 is localized to <strong>the</strong> nucleus, Ago1<br />

and Dcr1 are primarily cytoplasmic. This may reflect <strong>the</strong> fact that Rdp1 is required early in <strong>the</strong><br />

RNAi pathway to generate double strand RNA from transcripts that originate from centromeric<br />

loci. The relatively large size <strong>of</strong> Rdp1 (139.4 kD) precludes passive diffusion <strong>of</strong> <strong>the</strong> enzyme into<br />

<strong>the</strong> nucleus suggesting that karyopherin-dependent transport is involved in nuclear targeting <strong>of</strong><br />

this enzyme. In <strong>the</strong> present study, we report that <strong>the</strong> karyopherin Sal3 is required for nuclear<br />

import <strong>of</strong> Rdp1 in S. pombe. Loss <strong>of</strong> nuclear Rdp1 is associated with substantially reduced<br />

transcriptional gene-silencing, and surprisingly, post-transcriptional gene silencing which may<br />

occur in <strong>the</strong> cytoplasm, was also significantly affected. Toge<strong>the</strong>r, <strong>the</strong>se results identify Sal3 as a<br />

modulator <strong>of</strong> RNAi-dependent transcriptional gene silencing as well as a potential link between<br />

nuclear import and post-transcriptional gene-silencing.<br />

This work is supported by Natural Science and Engineering Research Council <strong>of</strong> Canada<br />

(NSERC) #rgpin 183820.<br />

Cell Polarity<br />

2162<br />

Siah regulation <strong>of</strong> Par3 controls neuronal cell adhesion during germinal zone exit.<br />

D. Solecki 1 , N. Trivedi 1 , J. Famulski 2 ; 1 St. Jude Children's Res Hosp, Memphis, TN, 2 University<br />

<strong>of</strong> Alberta, Edmonton, AB, Canada<br />

The brain’s circuitry is established by directed migration <strong>of</strong> neurons during development.<br />

Despite tremendous advances in our understanding <strong>of</strong> <strong>the</strong> substrates, guidance mechanisms<br />

and cytoskeletal elements required for neuronal migrations, remarkably little is known<br />

concerning <strong>the</strong> cell intrinsic machinery that initiates a neuron’s exit from <strong>the</strong>ir germinal zone<br />

(GZ) niche. We found that Seven in Absentia (Siah) is <strong>the</strong> first ubiquitin ligase to target Par3 for<br />

degradation and Siah-mediated proteosomal degradation <strong>of</strong> Par3 regulates <strong>the</strong> initiation <strong>of</strong><br />

radial migration as cerebellar granule neurons (CGNs) leave <strong>the</strong> external germinal layer (EGL).<br />

Both Par3 gain <strong>of</strong> function and Seven in Absentia loss <strong>of</strong> function spur precocious radial<br />

migration. Time-lapse imaging using a novel probe to measure neuronal cell contacts in live<br />

CGNs reveals that Par3 promotes adhesive interactions needed for germinal zone exit by<br />

recruiting <strong>the</strong> JAM-C epi<strong>the</strong>lial tight junction adhesion molecule to <strong>the</strong> neuronal cell surface. Our<br />

findings suggest a model in which post-translational modification <strong>of</strong> PAR complex function is a<br />

key mechanism regulating <strong>the</strong> exit <strong>of</strong> neuronal progenitors or immature neurons from a germinal<br />

zone niche. Moreover, <strong>the</strong>se results identify an unexpected parallel between epi<strong>the</strong>lial junction<br />

formation and <strong>the</strong> cell-cell interactions that occur during histogenesis in <strong>the</strong> developing<br />

vertebrate brain.


2163<br />

Cognitive deficits associated with CNS myelin dysfunction.<br />

A. Gow 1 , K. Meharas 1 ; 1 Genetics, Wayne State University, Detroit, MI<br />

MONDAY<br />

Cognitive dysfunction is typically considered a secondary symptom associated with mental<br />

illness. Studies suggest that mental illnesses <strong>the</strong>mselves are <strong>the</strong> cause <strong>of</strong> cognitive dysfunction,<br />

manifested as impairments in attention, memory, learning and executive function and lead to<br />

behavioral disorders. The pathophysiological processes that underlie cognitive dysfunction<br />

associated with behavioral changes have been debated for decades. Most studies focus on<br />

disrupted neural development or aberrant neurochemistry, however, an abundance <strong>of</strong> anecdotal<br />

clinical data and genetic evidence support a role for myelin and myelin-related genes in <strong>the</strong><br />

etiology <strong>of</strong> cognitive dysfunction and behavioral disorders. In <strong>the</strong> current study, we have<br />

generated a myelin mutant mouse in which expression <strong>of</strong> <strong>the</strong> tight junction protein, claudin 11,<br />

has been ablated in oligodendrocytes. The function <strong>of</strong> this protein in contributing to <strong>the</strong> electrical<br />

resistance <strong>of</strong> CNS myelin has been elucidated in molecular and ma<strong>the</strong>matical detail. Most<br />

importantly, <strong>the</strong> phenotype <strong>of</strong> <strong>the</strong>se knockout mice arises from changes to <strong>the</strong> passive<br />

membrane properties <strong>of</strong> small diameter myelinated fibers and not from degenerative processes.<br />

Accordingly, we are able to interpret behavioral changes in <strong>the</strong>se mutants in terms <strong>of</strong> cognitive<br />

dysfunction associated with slower conduction velocity. Our analyses <strong>of</strong> behavioral<br />

abnormalities resulting from slowed conduction will be presented.<br />

2164<br />

Cortical clustering <strong>of</strong> cell polarity regulators.<br />

J. Dodgson 1 , A. Chessel 1 , F. Vaggi 2 , S. Cox 3 , E. Rosten 3 , M. Yamamoto 4 , M. Geymonat 1 , Y.<br />

Dong 1 , J. Ahringer 1 , D. Albrecht 5 , M. Sato 4 , A. Csikasz-Nagy 2 , R. Carazo Salas 1 ; 1 Gurdon<br />

Institute, University <strong>of</strong> Cambridge, Cambridge, United Kingdom, 2 The Micros<strong>of</strong>t Research-<br />

University <strong>of</strong> Trento Centre for Computational Systems <strong>Biology</strong>, Trento, Italy, 3 Randall Division<br />

<strong>of</strong> Cell and <strong>Molecular</strong> Biophysics, Kings College London, United Kingdom, 4 Department <strong>of</strong><br />

Biophysics and Biochemistry, University <strong>of</strong> Tokyo, Japan, 5 Institute <strong>of</strong> Biochemistry, ETH Zurich,<br />

Zurich, Switzerland<br />

Cell polarity is a fundamental part <strong>of</strong> cell physiology. Despite <strong>the</strong> identification <strong>of</strong> many polarityregulating<br />

factors in diverse organisms, <strong>the</strong> microscopic structure and dynamics <strong>of</strong> <strong>the</strong> polarity<br />

machinery itself has been difficult to elucidate. We imaged frontally <strong>the</strong> cortical areas where <strong>the</strong><br />

polarity machinery accumulates in cells <strong>of</strong> <strong>the</strong> fission yeast Schizosaccharomyces pombe, using<br />

time-lapse and super-resolution (OMX structured-illumination and live-cell STORM) microscopy.<br />

Analysis <strong>of</strong> several key polarity factors reveals that <strong>the</strong>y localize to discrete cortical clusters<br />

('nodes'), resolvable to 50nm in diameter. Different polarity factors display different node<br />

distributions and dynamics at <strong>the</strong> cell cortex. Strikingly, we find that many factors thought to<br />

regulate polarity by interacting physically at <strong>the</strong> cortex – such as <strong>the</strong> factors Tea1p and Tea3p -<br />

do not necessarily co-localize <strong>the</strong>re, suggesting unexpected layers <strong>of</strong> regulation. Artificial<br />

dimerization <strong>of</strong> Tea1p and Tea3p leads to mislocalization <strong>of</strong> <strong>the</strong>se factors and cell polarity<br />

defects, suggesting that <strong>the</strong> segregation <strong>of</strong> polarity factors into adjacent but separate nodes<br />

may be crucial to <strong>the</strong>ir function. Similar nodes are found in Saccharomyces cerevisiae and<br />

Caenorhabditis elegans cells, suggesting conservation <strong>of</strong> this structural arrangement.


MONDAY<br />

2165<br />

Rapid cycles <strong>of</strong> phosphorylation and dephosphorylation on threonine 567 control ezrin<br />

localization and function in microvillus formation.<br />

R. Viswanatha 1 , P. Ohouo 1 , M. Smolka 1 , A. Bretscher 1 ; 1 Cornell University, Ithaca, NY<br />

Ezrin is a member <strong>of</strong> a group <strong>of</strong> conserved plasma membrane-actin cytoskeletal cross-linkers<br />

(called ERM proteins) that is negatively regulated by an intramolecular interaction between <strong>the</strong><br />

N- and C-termini. Ezrin is activated by threonine-567 phosphorylation near <strong>the</strong> C-terminus. In<br />

placental Jeg-3 cells at steady-state, we found that 50% <strong>of</strong> cellular ezrin is phosphorylated at<br />

threonine 567, and this phosphorylated pool is completely turned over within just 5 minutes. The<br />

N-terminus <strong>of</strong> ezrin is known to mediate its localization to <strong>the</strong> plasma membrane. Wild-type<br />

ezrin in Jeg-3 cells, however, is enriched on <strong>the</strong> microvillus membrane and virtually<br />

undetectable on o<strong>the</strong>r regions <strong>of</strong> <strong>the</strong> plasma membrane. Upon a brief treatment with a<br />

phosphatase inhibitor, by expressing a T567E phosphomimetic form <strong>of</strong> ezrin, or by expressing<br />

o<strong>the</strong>r constitutively open forms <strong>of</strong> ezrin, we find that this distribution is altered: ezrin “spills out”<br />

<strong>of</strong> <strong>the</strong> microvillar membrane and occupies o<strong>the</strong>r regions <strong>of</strong> <strong>the</strong> plasma membrane. To determine<br />

whe<strong>the</strong>r changing <strong>the</strong>se phospho-dynamics by stabilizing <strong>the</strong> phosphorylated form <strong>of</strong> ezrin<br />

would affect ezrin function, we performed siRNA-rescue experiments. Ezrin depletion in Jeg-3<br />

cell resulted in <strong>the</strong> loss <strong>of</strong> microvilli, and only wild-type, but nei<strong>the</strong>r T567A nor T567E ezrin<br />

restored <strong>the</strong>m. Taken toge<strong>the</strong>r, <strong>the</strong>se observations hint at <strong>the</strong> presence <strong>of</strong> constant, local kinase<br />

activity toward ezrin countering ubiquitous phosphatase activity, and this balance is essential for<br />

ezrin function. To determine how phosphorylation dynamics are regulated in Jeg-3 cells, we<br />

conducted a proteomic screen to identify kinases associated with ezrin. We identified several<br />

candidate kinases previously shown to phosphorylate <strong>the</strong> C-termini <strong>of</strong> ERMs in o<strong>the</strong>r systems.<br />

We are in <strong>the</strong> process <strong>of</strong> examining <strong>the</strong> distribution <strong>of</strong> <strong>the</strong>se kinases in Jeg-3 cells and <strong>the</strong>ir<br />

contribution to ezrin phospho-dynamics.<br />

2166<br />

Characterization <strong>of</strong> Banderuola, a novel regulator <strong>of</strong> asymmetric cell division in<br />

Drosophila melanogaster.<br />

F. Mauri 1 , J. L. Mummery-Widmer 1 , M. Yamazaki 1,2 , J. A. Knoblich 1 ; 1 Institute <strong>of</strong> <strong>Molecular</strong><br />

Biotechnology <strong>of</strong> <strong>the</strong> Austrian Academy <strong>of</strong> Sciences (IMBA), Vienna, Austria, 2 The Global COE<br />

program, Akita University School <strong>of</strong> Medicine, Akita, Japan<br />

Asymmetric cell division is a process whereby a progenitor cell can generate daughters with a<br />

different fate. The underlying molecular mechanisms have been widely studied in <strong>the</strong> Drosophila<br />

Sensory Organ Precursor (SOP) cells, that generate <strong>the</strong> 4 different cell types forming <strong>the</strong><br />

external sensory organs (bristles) through 2 rounds <strong>of</strong> asymmetric cell division. This is achieved<br />

through <strong>the</strong> partitioning <strong>of</strong> <strong>the</strong> protein Numb, that represses Notch signaling in <strong>the</strong> daughter cell<br />

where it is segregated.<br />

To gain a better insight <strong>of</strong> this process, we used tissue specific RNAi to score <strong>the</strong> effects <strong>of</strong> <strong>the</strong><br />

knockdown <strong>of</strong> genes on <strong>the</strong> morphology <strong>of</strong> <strong>the</strong> bristles. We have screened a library <strong>of</strong> 22,247<br />

transgenic Drosophila RNAi lines, and identified 130 genes potentially involved in asymmetric<br />

cell division. To identify <strong>the</strong> genes involved in <strong>the</strong> asymmetric localization <strong>of</strong> Numb, we<br />

developed a live time-lapse cell-imaging assay based on a GFP reporter. Among <strong>the</strong> 130<br />

candidates screened and not previously thought to play a role in asymmetric cell division, <strong>the</strong>re<br />

were three genes whose knockdown reproducibly affected Numb localization. We have<br />

confirmed <strong>the</strong> observed phenotypes by independent secondary RNAi lines and have started<br />

investigating <strong>the</strong> function <strong>of</strong> one <strong>of</strong> <strong>the</strong> identified genes, which encodes a previously<br />

uncharacterized protein. Loss-<strong>of</strong>-function studies suggest that <strong>the</strong> protein, which we named<br />

Banderuola, specifically acts in establishing an axis <strong>of</strong> cell polarity for asymmetric cell division.


MONDAY<br />

Co-Immunoprecipitation experiments show that Banderuola binds to Discs-large (Dlg), a<br />

membrane-associated guanylate kinase acting in many cell-polarity events.<br />

Our study has identified Banderuola, a new component <strong>of</strong> <strong>the</strong> machinery <strong>of</strong> asymmetric cell<br />

division. As its binding partner Dlg acts in several polarity events, including synaptogenesis and<br />

tumor formation, our studies may be relevant for a variety <strong>of</strong> biological processes.<br />

2167<br />

Secretory vesicles deliver Cdc42p to sites <strong>of</strong> polarized growth in S. cerevisiae.<br />

S. A. Dighe 1 , K. G. Kozminski 1,2 ; 1 <strong>Biology</strong>, University <strong>of</strong> Virginia, Charlottesville, VA, 2 Cell<br />

<strong>Biology</strong>, University <strong>of</strong> Virginia, Charlottesville, VA<br />

Establishment and maintenance <strong>of</strong> cell polarity in many eukaryotes depends upon activation <strong>of</strong><br />

<strong>the</strong> Rho GTPase Cdc42p. In <strong>the</strong> budding yeast S. cerevisiae, Cdc42p is essential for<br />

establishing and maintaining an axis <strong>of</strong> polarized growth that supports bud formation. In G1 <strong>of</strong><br />

<strong>the</strong> cell cycle, Cdc42p asymmetrically localizes on <strong>the</strong> plasma membrane (PM), at <strong>the</strong><br />

presumptive bud site, in an actin-independent manner. Maintenance <strong>of</strong> this axis <strong>of</strong> growth<br />

during bud formation, in S and G2, requires <strong>the</strong> actin-dependent localization <strong>of</strong> Cdc42p at <strong>the</strong><br />

bud tip. How Cdc42p is maintained at <strong>the</strong> bud tip is unclear, leading to divergent models <strong>of</strong><br />

Cdc42p and cell polarization (Wedlich-Soldner et al. 2003. Science 299:1231-1235; Layton et<br />

al. 2011. Current <strong>Biology</strong> 21:184-194). One model suggests that Cdc42p is delivered to <strong>the</strong> bud<br />

tip by secretory vesicles, replenishing Cdc42p that is lost at <strong>the</strong> bud tip through diffusion in <strong>the</strong><br />

plane <strong>of</strong> <strong>the</strong> PM or by endocytosis. Consistent with this model, we show Cdc42p associated with<br />

secretory vesicles both in vivo and in vitro. By immunogold labeling EM thin sections <strong>of</strong> whole<br />

cells, both wild-type and secretory mutants, we localized Cdc42p to vesicles 80-100nm in<br />

diameter, <strong>the</strong> signature size <strong>of</strong> secretory vesicles in S. cerevisiae, in addition to <strong>the</strong> PM and ER.<br />

We also show that Cdc42p co-fractionates with secretory vesicles and <strong>the</strong> secretory vesicle<br />

markers Sec4p, Bgl2p, Osh4p, and invertase. These results indicate that secretory vesicles can<br />

serve as a vehicle for <strong>the</strong> delivery <strong>of</strong> Cdc42p to sites <strong>of</strong> polarized growth. Supported by NSF<br />

0723342.<br />

2168<br />

Ral GTPases regulate biogenesis <strong>of</strong> epi<strong>the</strong>lial polarity.<br />

C. Hazelett 1 , C. Yeaman 1 ; 1 Anatomy & Cell <strong>Biology</strong>, University <strong>of</strong> Iowa, Iowa City, IA<br />

Tight junctions (TJs) are indispensible structures to epi<strong>the</strong>lial cells, and are responsible for<br />

regulation <strong>of</strong> paracellular diffusion and maintenance <strong>of</strong> cellular polarity that is crucial to human<br />

health. A detailed mechanistic understanding <strong>of</strong> how TJs form, however, is incomplete. TJ<br />

proteins assemble at <strong>the</strong> interface between apical and lateral membrane domains, but factors<br />

that target vesicles to <strong>the</strong>se sites during TJ formation have not been identified. TJ maturation is<br />

associated with establishment <strong>of</strong> polarity, and in vivo epi<strong>the</strong>lial cells form tubes with distinct<br />

lumenal and basal domains. Fur<strong>the</strong>rmore, polarized epi<strong>the</strong>lial cells generate a primary cilium on<br />

<strong>the</strong> apical surface. The Exocyst is required for ciliogenesis and cystogenesis, and under control<br />

<strong>of</strong> Ral GTPases, has been shown to direct vesicle trafficking to basolateral membranes. We<br />

investigated <strong>the</strong> roles <strong>of</strong> Ral GTPases in TJ assembly and ciliogenesis, and also in <strong>the</strong> more<br />

complex process <strong>of</strong> cystogenesis. We show that RalA and RalB serve opposing functions during<br />

<strong>the</strong> former two processes, and both are indispensible for <strong>the</strong> latter. Silencing expression <strong>of</strong> RalA<br />

or RalB altered TJ gate function in opposite ways during initial establishment <strong>of</strong> TJs in MDCK<br />

cells; RalA knockdown abrogated <strong>the</strong> trans-epi<strong>the</strong>lial resistance (TER) overshoot while RalB<br />

knockdown greatly exaggerated it. However, TJ gate function, as assayed by transcellular lipid<br />

diffusion, was unaffected by ei<strong>the</strong>r knockdown. Immun<strong>of</strong>luorescence and detergent solubility<br />

studies revealed Ral-specific differences in TJ composition. Levels <strong>of</strong> some TJ components


MONDAY<br />

were reduced in RalA knockdown cells when compared to controls, but were increased in TJs <strong>of</strong><br />

RalB knockdown cells. Additional data showed that both Ral GTPases exert effects on TJ<br />

formation through binding to <strong>the</strong> Exocyst. Fur<strong>the</strong>rmore, RalB was necessary for endocytosis <strong>of</strong><br />

<strong>the</strong> junctional protein E-cadherin. RalA knockdown also decreased ciliogenesis, while RalB<br />

knockdown led to an increase <strong>of</strong> ciliated cells. Finally, formation <strong>of</strong> a central lumen during<br />

cystogenesis in three-dimensional Matrigel cultures was dependent on expression <strong>of</strong> both<br />

GTPases. Our working model is that RalA facilitates trafficking <strong>of</strong> TJ and ciliary constituents to<br />

<strong>the</strong> plasma membrane via <strong>the</strong> Exocyst, while RalB regulates endocytosis and/or recycling <strong>of</strong><br />

<strong>the</strong>se components. This and future work will significantly increase our understanding <strong>of</strong> how<br />

<strong>the</strong>se GTPases function toge<strong>the</strong>r, and in collaboration with <strong>the</strong> Exocyst, to regulate epi<strong>the</strong>lial<br />

polarization.<br />

2169<br />

Out <strong>of</strong> <strong>the</strong> loop: The role <strong>of</strong> Bem1 in actin-independent polarization in S. cerevisiae.<br />

S. E. Smith 1,2 , R. Li 1,2 ; 1 Stowers Institute for Medical Research, Kansas City, MO, 2 Dept <strong>of</strong><br />

<strong>Molecular</strong> and Integrative Physiology, University <strong>of</strong> Kansas Medical Center, Kansas City, KS<br />

Cell polarization is vital for a range <strong>of</strong> important processes across <strong>the</strong> spectrum <strong>of</strong> life, from cell<br />

migration in human development to polarized cell division in budding yeast, but <strong>the</strong> mechanisms<br />

<strong>of</strong> polarization are still not well understood. A key player is <strong>the</strong> small GTPase Cdc42, <strong>the</strong><br />

“master regulator” <strong>of</strong> polarity. In budding yeast, polarization <strong>of</strong> Cdc42 requires nei<strong>the</strong>r spatial<br />

cues nor cytoskeletal structures, but occurs via a dynamic, diffusion-based process. One model<br />

consists <strong>of</strong> a positive feedback loop in which a small, stochastic accumulation <strong>of</strong> active Cdc42-<br />

GTP recruits <strong>the</strong> adaptor molecule Bem1, which <strong>the</strong>n recruits <strong>the</strong> activator <strong>of</strong> Cdc42, Cdc24,<br />

resulting in an increased local active pool and fur<strong>the</strong>r recruitment. In this work, we disrupted<br />

individual steps in <strong>the</strong> proposed feedback loop by abrogating <strong>the</strong> ability <strong>of</strong> Bem1 to interact with<br />

ei<strong>the</strong>r Cdc42 or Cdc24. We <strong>the</strong>n investigated <strong>the</strong> effect <strong>of</strong> <strong>the</strong> disruption on diffusion-based<br />

polarization. As expected, we found that abrogation <strong>of</strong> <strong>the</strong> Bem1-Cdc24 interaction resulted in a<br />

complete loss <strong>of</strong> polarization. However, we were surprised to find that Cdc42 polarization was<br />

still robust when <strong>the</strong> Cdc42-Bem1 interaction was disrupted, even though Bem1 did not localize<br />

to <strong>the</strong> polar cap. This finding invalidates <strong>the</strong> current feedback loop model, and suggests that<br />

Bem1 may have a location-independent role in activating Cdc24.<br />

2170<br />

Does ciliary autotomy in <strong>the</strong> kidney contribute to PKD?<br />

C. Yeaman 1 , N. Ma 1 ; 1 Anatomy & Cell <strong>Biology</strong>, University <strong>of</strong> Iowa, Iowa City, IA<br />

Primary cilia are active signaling centers present on most differentiated cell types. Cells may<br />

lose <strong>the</strong>ir cilia ei<strong>the</strong>r by resorption into <strong>the</strong> cell, or shedding (autotomy) from <strong>the</strong> cell surface.<br />

Autotomy is an ancient process, but its physiological significance in mammals is unclear. We<br />

have shown that diverse agents trigger ciliary shedding in renal epi<strong>the</strong>lial cells both in vitro and<br />

in vivo. This occurs by a distinct mechanism than that described for ciliary resorption, as<br />

inhibitors <strong>of</strong> resorption fail to block autotomy and intact cilia are recovered in <strong>the</strong> media <strong>of</strong><br />

cultured cells and in <strong>the</strong> urine <strong>of</strong> mice following treatment with autotomy-inducing agents.<br />

Shedding <strong>of</strong> primary cilia promotes dramatic changes to epi<strong>the</strong>lial cells, including compositional<br />

and functional changes to tight junctions, induction <strong>of</strong> partial epi<strong>the</strong>lial-to-mesenchymal<br />

transition and dysregulated cell cycle control. A full-length cilium is not required to trigger<br />

autotomy-induced epi<strong>the</strong>lial remodeling. It occurs in cells that express stunted pro-cilia, such as<br />

non-differentiated epi<strong>the</strong>lial cells and cells lacking essential ciliogenesis components (e.g.<br />

IFT88). We hypo<strong>the</strong>size that autotomy liberates <strong>the</strong> maternal centriole from its te<strong>the</strong>red state at<br />

<strong>the</strong> apical plasma membrane, <strong>the</strong>reby facilitating its differentiation into a signal-generating


MONDAY<br />

centrosome. In renal epi<strong>the</strong>lial cells, ciliary autotomy is dependent on <strong>the</strong> catalytic activity <strong>of</strong><br />

Nek8 and <strong>the</strong> Exocyst vesicle te<strong>the</strong>ring complex. jck mice, which develop autosomal-recessive<br />

polycystic kidney disease and harbor a mutation in <strong>the</strong> NEK8 gene, exhibit substantially<br />

elevated levels <strong>of</strong> ciliary shedding in vivo. Therefore, we propose that ciliary autotomy is a<br />

physiologic phenomenon in <strong>the</strong> kidney, and that dysregulated autotomy may contribute to PKD<br />

by altering trans-epi<strong>the</strong>lial fluid transport, tissue morphogenesis and growth regulation.<br />

Cancer Cell <strong>Biology</strong> I<br />

2171<br />

Evaluating <strong>the</strong> effect <strong>of</strong> South African herbal extracts on breast cancer cells.<br />

M. Choene 1 , L. Motadi 1 ; 1 Genetics, University <strong>of</strong> <strong>the</strong> Witwatersrand, Johannesburg, South Africa<br />

South African plants have been used for generations by traditional healers due to <strong>the</strong>ir antibacterial,<br />

anti-viral, anti-fungal and anti-amoebic properties. Now with <strong>the</strong> emergence <strong>of</strong> natural<br />

product research in <strong>the</strong> science fraternity, a lot <strong>of</strong> <strong>the</strong>se indigenous plants have been showing<br />

great potential as an alternative cancer treatment to orthodox treatments. For our research, our<br />

aim was to investigate <strong>the</strong> anti-breast cancer activities <strong>of</strong> three South African plant extracts:<br />

Kedrostis Foetidissima, Euphorbia Mauritanica and Elytropappus Rhinocerotis. We looked at<br />

<strong>the</strong> effect <strong>of</strong> <strong>the</strong>se extracts on breast cancer cells since it is one <strong>of</strong> <strong>the</strong> leading cancers in<br />

females in South Africa. The herbal extracts were screened for <strong>the</strong>ir cytotoxicity in two breast<br />

cancer cell lines MCF-7 and YMB-1 by means <strong>of</strong> an MTT (3-(4,5-dimethylthiazol-2-yl)-2,5diphenyl<br />

tetrazolium bromide) assay. MTT is a simple colorimetric assay used to measure cell<br />

cytotoxicity, proliferation or viability. The cells were treated with varying concentrations (10, 30,<br />

50 and 100µg/ml) <strong>of</strong> <strong>the</strong> extracts, <strong>the</strong>n <strong>the</strong> MTT assay was conducted to determine <strong>the</strong> IC50<br />

(concentration <strong>of</strong> extract that causes 50% cell death) <strong>of</strong> <strong>the</strong>se herbal extracts on <strong>the</strong> cell lines.<br />

We found that Elytropappus Rhinocerotis had no significant effect on both cancer cell lines,<br />

Kedrostis Foetidissima exhibited cytotoxicity on both cell lines with 48% cytotoxicity at 100µg/ml<br />

(IC50 > 100µg/ml) on MCF-7 cells and 40% cytotoxicity at 100µg/ml (IC50 > 100µg/ml) on YMB-<br />

1 cells. Euphorbia Mauritanica showed 53% cytotoxicity at 100µg/ml and 37% cytotoxicity at 50<br />

µg/ml (50µg/ml 100 µg/ml) on MCF-7 but had no significant effect on YMB-1. For those<br />

extracts which exhibited cytotoxicity on <strong>the</strong> breast cancer cells, we <strong>the</strong>n aimed to investigate if<br />

<strong>the</strong> cells were dying due to apoptosis and not necrosis. We <strong>the</strong>n employed <strong>the</strong> use <strong>of</strong> a flow<br />

cytometer using Annexin V, which can detect cells that are alive, undergoing apoptosis and<br />

necrosis (Annexin V has a strong calcium dependent affinity to phosphatidylserine residues,<br />

which are normally hidden in <strong>the</strong> cytoplasmic plasma membrane but translocate to <strong>the</strong> cell<br />

surface during apoptosis, acting as a probe to detect apoptosis). It was <strong>the</strong>n observed that <strong>the</strong><br />

herbal extracts inducing death via apoptosis and not necrosis. We can conclude that <strong>the</strong> herbal<br />

extracts K. Foetidissima and E. Mauritanica exhibit some level <strong>of</strong> cytotoxicity on breast cancer<br />

cells. It is also promising that <strong>the</strong>y cause cells to undergo apoptosis showing great potential in<br />

<strong>the</strong>ir use in breast cancer natural product research and drug design sector.<br />

2172<br />

Traditional herb in cancer.<br />

L. R. Motadi 1 , A. Thafeni 1 , M. Choene 1 ; 1 School <strong>of</strong> <strong>Molecular</strong> <strong>Biology</strong>, University <strong>of</strong> <strong>the</strong><br />

Witwatersrand, Johannesburg, South Africa<br />

Euphorbia mauritanica and Kedrostis hirtella extracts can induce anti-proliferative activities in<br />

cancer cells.


MONDAY<br />

In recent times a significant proportion <strong>of</strong> <strong>the</strong> drugs used to treat cancer are ei<strong>the</strong>r natural<br />

products or compounds discovered based on <strong>the</strong> study <strong>of</strong> natural products and <strong>the</strong>ir interaction<br />

with cellular targets. While <strong>the</strong> use <strong>of</strong> syn<strong>the</strong>tic combinatorial libraries has enhanced <strong>the</strong> pace <strong>of</strong><br />

drug discovery, <strong>the</strong> extraordinary structural diversity intrinsic to natural products assures that<br />

<strong>the</strong>y will continue to find utility in defining novel cellular targets for <strong>the</strong>rapeutic intervention, and<br />

also as drug candidates and structural leads for chemical optimization. With our increased<br />

understanding <strong>of</strong> <strong>the</strong> molecular mechanisms underlying cancer progression and cell death, this<br />

work aims to target E. mauritanica and K. hirtella as new <strong>the</strong>rapeutic targets for treatment in<br />

cancer in particular Breast and Lung cancer. Accordingly, <strong>the</strong> study was aimed to investigate<br />

possible molecular mechanisms that are associated with <strong>the</strong> potential anti-carcinogenic property<br />

<strong>of</strong> this agr<strong>of</strong>ield weed. MCF-7 and A549 cells were exposed to different concentrations (0-100<br />

mg/ml) <strong>of</strong> <strong>the</strong> crude methanolic extract <strong>of</strong> E. mauritanica and K. hirtella to evaluate <strong>the</strong>ir growth<br />

inhibitory and apoptosis inducing effects. The extract elicited a dose- and time-dependent<br />

inhibition <strong>of</strong> cell proliferation, followed by a concomitant decrease in cell viability. The observed<br />

cytotoxicity was linked to <strong>the</strong> induction <strong>of</strong> apoptosis as determined by Flow cytometer Annexin V<br />

features known to be associated with <strong>the</strong> advent <strong>of</strong> apoptosis. Real time quantitative RT-PCR<br />

analyses <strong>of</strong> Bax, Bcl-2, RBBP6 and p53 exhibited aberrant expression pr<strong>of</strong>iles <strong>of</strong> <strong>the</strong>se genes<br />

under various treatment conditions. Taken toge<strong>the</strong>r, <strong>the</strong> data suggest that <strong>the</strong> crude methanolic<br />

extract <strong>of</strong> E. mauritanica and K. hirtella contains bioactive compounds that may be beneficial in<br />

<strong>the</strong> treatment <strong>of</strong> malignant growths. However, <strong>the</strong> mechanisms behind this activity still need to<br />

be researched.<br />

2173<br />

A natural small molecule YCG185 inhibits angiogenesis both in vitro and in vivo.<br />

Y. Kim 1 , H. Jung 1 , H. Kwon 1 ; 1 Chemical Genomics National Research Laboratory, TRCP,<br />

Department <strong>of</strong> Biotechnology, College <strong>of</strong> Life Science & Biotechnology, Yonsei University,<br />

Seoul, Korea<br />

Angiogenesis, a formation <strong>of</strong> new blood vessels from pre-existing ones, plays a critical role in<br />

normal and pathological phenotypes including solid tumor growth and metastasis. Accordingly,<br />

<strong>the</strong> development <strong>of</strong> new anti-angiogenic agents has been considered as an efficient strategy for<br />

<strong>the</strong> treatment <strong>of</strong> cancer and o<strong>the</strong>r human diseases related with angiogenesis. To identify such<br />

small molecules, 300 crude extracts <strong>of</strong> natural plants were subjected to cell-based screening<br />

toward <strong>the</strong> proliferation <strong>of</strong> human umbilical vein endo<strong>the</strong>lial cells (HUVECs). As <strong>the</strong> result, <strong>the</strong><br />

active principle <strong>of</strong> YCG185 was identified as a new anti-angiogenic agent. YCG185 inhibited <strong>the</strong><br />

proliferation <strong>of</strong> HUVECs at IC50 <strong>of</strong> 6.6 μg/mL without showing any toxicity to <strong>the</strong> cells. YCG185<br />

significantly suppressed in vitro angiogenesis such as VEGF-induced tube formation and<br />

chemoinvasion. Moreover, <strong>the</strong> compound inhibits chorioallantoic membrane in vivo<br />

angiogenesis at non-toxic doses. In addition, YCG185 decreased <strong>the</strong> expression levels <strong>of</strong><br />

hypoxia inducible factor-1a and its target gene, VEGF, in a dose-dependent manner. Taken<br />

toge<strong>the</strong>r, <strong>the</strong>se results demonstrate that YCG185 could be a novel natural small molecule<br />

targeting angiogenesis.<br />

2174<br />

Genetic Modification <strong>of</strong> Breast Cancer Gene Targets Using ZFN Technology Reveal<br />

Differential Responses to Drug Sensitivity.<br />

G. L. Davis 1 , C. Corman 1 , L. Daley 1 , G. Pegg 1 , H. Zakeri 1 , Z. Zhang 1 , G. Wemh<strong>of</strong>f 1 ; 1 Sigma<br />

Aldrich, St. Louis, MO<br />

Breast cancer tumorigenesis is a complex disease where multiple signaling pathways<br />

participate in cell proliferation and invasion. The heterogeneity <strong>of</strong> this disease presents a


MONDAY<br />

challenge in developing <strong>the</strong>rapuetic treatments because patients respond to <strong>the</strong>rapies with<br />

varying degrees <strong>of</strong> sensitvity 1,2. For example, previous studies have demonstrated that<br />

activation <strong>of</strong> <strong>the</strong> MEK/MAPK pathway, loss <strong>of</strong> cell-cell adhesion, and enahnced epi<strong>the</strong>lial-tomesenchymal<br />

transition confer resistance to <strong>the</strong> breast cancer <strong>the</strong>rapeutic, gefitinib 2,3,4.<br />

With <strong>the</strong> advent <strong>of</strong> zinc finger nuclease (ZFN) technology, it is possible to generate cancer<br />

relevant mutations in one or more endogenous genes. Zinc finger nucleases have been<br />

optimized to target specific genes where <strong>the</strong>y induce a double-strand break adjacent to <strong>the</strong>ir<br />

binding site. Double strand-breaks are ei<strong>the</strong>r repaired by non-homologous end joining (NHEJ) or<br />

homology dependent repair (HDR). As a result, cell lines harbor insertions, deletions, or<br />

integrations within <strong>the</strong> targeted gene <strong>of</strong> interest.<br />

We have utilized zinc finger nuclease technology to target and disrupt endogenous breast<br />

cancer relevant genes, SYK, ESR2, BCR, and APC, in <strong>the</strong> near normal mammary epi<strong>the</strong>lial line,<br />

MCF10a. These gene targets play a pivotal role in cellular proliferation, migration and cell<br />

adhesion and may <strong>the</strong>reby play a contributing role in gefitinib sensitivity 5,6,7,8,9,10. In this<br />

study, we aim to investigate how loss <strong>of</strong> SYK, ESR2, BCR, and APC gene function, in ZFN<br />

engineered cell lines, affect sensitivity towards <strong>the</strong> tyrosine kinasae inhibitor, gefitinib.<br />

2175<br />

Gene Specific Regulation <strong>of</strong> NFκB-Dependent Anti-Apoptotic Genes in Metastatic<br />

Prostate Cancer Cells by Bortezomib.<br />

S. Manna 1 , B. Singha 1 , T-P. Chang 1 , S. Sanacora 1 , I. Vancurova 1 ; 1 Biological Sciences, St.<br />

John's University, Queens, NY<br />

Prostate cancer is <strong>the</strong> third most common cause <strong>of</strong> death from cancer in men <strong>of</strong> all ages. One <strong>of</strong><br />

<strong>the</strong> critical factors in progression to <strong>the</strong> metastatic prostate cancer is <strong>the</strong> increased activity <strong>of</strong> <strong>the</strong><br />

transcription factor NFκB, which induces syn<strong>the</strong>sis <strong>of</strong> anti-apoptotic genes, such as Bcl-2, cIAP-<br />

1 and cIAP-2, thus promoting inhibition <strong>of</strong> apoptosis and resistance to chemo<strong>the</strong>rapy.<br />

Bortezomib is a potent 26S proteasome inhibitor that has been used for <strong>the</strong> treatment <strong>of</strong><br />

multiple myeloma and mantle cell lymphoma, and has shown promising results in several o<strong>the</strong>r<br />

types <strong>of</strong> cancer, including <strong>the</strong> metastatic prostate cancer. However, <strong>the</strong> molecular mechanisms<br />

<strong>of</strong> bortezomib function in prostate cancer cells are not fully understood. In this study, we<br />

investigated <strong>the</strong> mechanism <strong>of</strong> bortezomib function in <strong>the</strong> metastatic prostate cancer PC-3 cells.<br />

Our data demonstrate that in PC-3 cells, bortezomib induces translocation <strong>of</strong> <strong>the</strong> NFκB<br />

inhibitory protein, IκBα, from <strong>the</strong> cytoplasm to <strong>the</strong> nucleus, thus inhibiting <strong>the</strong> constitutive NFκB<br />

activity in <strong>the</strong>se cells. However, our results indicate that <strong>the</strong> regulation <strong>of</strong> NFκB activity by <strong>the</strong><br />

bortezomib-induced nuclear IκBα is gene specific. While <strong>the</strong> expression <strong>of</strong> NFκB-dependent<br />

anti-apoptotic genes cIAP-1 and cIAP-2 is inhibited by bortezomib, expression <strong>of</strong> Bcl-2 is not<br />

suppressed. Differences in <strong>the</strong> transcriptional regulation by <strong>the</strong> bortezomib-induced nuclear<br />

IκBα might hold <strong>the</strong> key for development <strong>of</strong> more effective <strong>the</strong>rapies for <strong>the</strong> advanced forms <strong>of</strong><br />

prostate cancer and o<strong>the</strong>r cancers characterized by <strong>the</strong> constitutive activation <strong>of</strong> NFκB and<br />

resistance to chemo<strong>the</strong>rapy.<br />

Funded by: NIH grants GM079581 and AI085497 to I.V.


MONDAY<br />

2176<br />

The neuroprotective efficacies and anticancer properties <strong>of</strong> various botanical extracts,<br />

Rue chalpanesis and Hibiscus sabdariffa, against neurotoxic-induced Parkinson's<br />

disease and pancreatic adenocarcinoma in vitro.<br />

T. Zeleke 1 , H. Tsegaye 1 , A. Bettica 1 ; 1 <strong>Biology</strong>, Manhattanville College, Purchase, NY<br />

Polyphenolic antioxidants have demonstrated anticarcinogenic activities by inhibiting or delaying<br />

<strong>the</strong> oxidation process caused by free radicals. Many extracts, containing anthocyanins,<br />

flavonoids, or phenolic compounds, have been shown to be strong antioxidants and possible<br />

neuroprotective agents. Common neurodegenerative disorders, such as Parkinson's disease<br />

(PD), may show neuronal cell loss as an accumulation <strong>of</strong> reactive oxygen species (ROS) due to<br />

increased oxidative stress. Roselle extract from <strong>the</strong> tropical plant Hibiscus sabdariffa contains<br />

primarily anthocyanins and protocatechuic acid, which can exhibit antioxidant, anti-inflammatory<br />

and antitumorigenic properties. Rue extract, from <strong>the</strong> tropical plant Ruta chalepensis, may also<br />

protect cells from oxidative stress-induced free radicals. The planetary structures <strong>of</strong> acridone, a<br />

major constituent in <strong>the</strong> Rue extract, can be inserted between a double stranded DNA and<br />

interfere with cellular machinery, contributing to its antitumor properties. For plant extractions,<br />

dried plant samples <strong>of</strong> R. chalapensis are centrifuged, vortexed, and agitated in <strong>the</strong> cold for 24<br />

hours. H. sabdariffa (10g) is boiled in water, followed by filtration. To determine inhibition <strong>of</strong><br />

growth rate in pancreatic adenocarcinoma cell lines, PANC-1 cells are seeded at a<br />

concentration <strong>of</strong> 6x104 cells/well in 96-well plates. After seeding and adherence to <strong>the</strong> well<br />

plate, all cells are allowed one to two population doubling times (PDT) before and after<br />

treatments. Single drug 24 hour treatments for dose response curves in ten fold dilutions in<br />

water determine effective IC50 values for Ruta and Hibiscus extracts. To determine <strong>the</strong><br />

neuroprotective efficacies <strong>of</strong> <strong>the</strong>se extracts with relationship to an oxidative stress-induced in<br />

vitro PD model, pretreatments <strong>of</strong> SK-N-SH neuroblastoma cells with <strong>the</strong>se extracts against <strong>the</strong><br />

neurotoxic insults, MPP+ and 6-OHDA were undertaken. The cells are pretreated in 96-well<br />

plates to formulate dose-response curves, with each <strong>of</strong> <strong>the</strong> serially-diluted extracts added for 24<br />

hours before a potent dose <strong>of</strong> MPP+, a mitochondrial complex I inhibitor, or 6-OHDA, a potential<br />

ROS generator. The cells are exposed to each insult first for 24 hours for posttreatment studies,<br />

followed by <strong>the</strong> extracts for 24 hours. Cotreatments with each extract and <strong>the</strong> neurotoxic insults<br />

were performed to elucidate possible preconditioning or gene activation versus actual<br />

mechanisms <strong>of</strong> action. Cell viability/ proliferation <strong>of</strong> triplicate data sets are assessed ei<strong>the</strong>r<br />

through <strong>the</strong> MTT mitochondrial (for PANC-1 cells) or Neutral Red lysosomal assay (for<br />

neuroblastoma cells). All data are statistically analyzed using a one-way ANOVA with Tukey’s<br />

post hoc tests. Preliminary dose responses for R. chalepensis and H. sabdariffa extracts show<br />

effective doses <strong>of</strong> 10-4-10-6 g/ml for SK-N-SH cells. The neuroprotective efficacies <strong>of</strong> <strong>the</strong><br />

botanical extracts against <strong>the</strong> neurotoxin-induced PD model may indicate <strong>the</strong>ir successful use in<br />

<strong>the</strong> prevention or treatment <strong>of</strong> Parkinson’s disease. Effective IC50 values may lead to possible<br />

chemopreventive strategies for certain cancer cell types as well.<br />

2177<br />

Gefitinib resistance <strong>of</strong> cancer cells correlated with TM4SF5-mediated<br />

epi<strong>the</strong>lialmesenchymal transition.<br />

M. Lee 1 , J. W. Lee 1,2 ; 1 Department <strong>of</strong> Pharmacy, Reseach Inst <strong>of</strong> Pharmaceutical Sciences, Cell<br />

Dynamics Res Ctr, College <strong>of</strong> P, Seoul National University, Seoul, Korea, 2 Cancer Research<br />

Institute, College <strong>of</strong> Medicine, Seoul National University, Seoul, Korea<br />

Although cancers can be initially treated with <strong>the</strong> epidermal growth factor receptor (EGFR)<br />

inhibitor, gefitinib, continued gefitinib <strong>the</strong>rapy does not benefit <strong>the</strong> survival <strong>of</strong> patients due to<br />

acquired resistance through EGFR mutations, c-Met amplification, or epi<strong>the</strong>lial-mesenchymal


MONDAY<br />

transition (EMT). It is <strong>of</strong> fur<strong>the</strong>r interest to determine whe<strong>the</strong>r mesenchymal-like, but not<br />

epi<strong>the</strong>lial-like, cancer cells can become resistant to gefitinib by bypassing EGFR signaling and<br />

acquiring alternative routes <strong>of</strong> proliferative and survival signaling. Here we examined whe<strong>the</strong>r<br />

gefitinib resistance <strong>of</strong> cancer cells can be caused by transmembrane 4 L six family member 5<br />

(TM4SF5), which has been shown to induce EMT via cytosolic p27Kip1 stabilization. Gefitinibresistant<br />

cells exhibited higher and/or sustained TM4SF5 expression, cytosolic p27Kip1<br />

stabilization, and mesenchymal phenotypes, compared with gefitinib-sensitive cells. Conversion<br />

<strong>of</strong> gefitinib-sensitive to -resistant cells by introduction <strong>of</strong> <strong>the</strong> T790M EGFR mutation caused<br />

enhanced and sustained expression <strong>of</strong> TM4SF5, phosphorylation <strong>of</strong> p27Kip1 Ser10<br />

(responsible for cytosolic location), loss <strong>of</strong> E-cadherin from cell-cell contacts,and gefitinibresistant<br />

EGFR and survival signaling activities. Additionally, TM4SF5 overexpression lessened<br />

<strong>the</strong> sensitivity <strong>of</strong> NSCLC cells to gefitinib. Suppression <strong>of</strong> TM4SF5 or p27Kip1 in gefitinibresistant<br />

cells via <strong>the</strong> T790M EGFR mutation or TM4SF5 expression rendered <strong>the</strong>m gefitinibsensitive,<br />

displaying more epi<strong>the</strong>lial-like and less mesenchymal-like characteristics. Toge<strong>the</strong>r,<br />

<strong>the</strong>se results indicate that TM4SF5-mediated EMT may have an important function in <strong>the</strong><br />

gefitinib resistance <strong>of</strong> cancer cells. [This work was supported by NRF by senior researchers<br />

program (Leap research, 2011-0001160) and Global Frontier Project grant (NRF-M1AXA002-<br />

2010-0029778), and a grant <strong>of</strong> <strong>the</strong> Korean Health Technology R&D Project (A100727),<br />

MHWFA, Korea to JW Lee].<br />

Key words: epi<strong>the</strong>lial-mesenchymal transition; drug resistance; EGFR; lung cancer;<br />

tetraspanin<br />

2178<br />

Bioavailability Enhancing Strategy for Oral Administration <strong>of</strong> Doxorubicin.<br />

Y. ZHAO 1 , M. L. Forrest 1 ; 1 Pharmaceutical Chemistry, <strong>the</strong> University <strong>of</strong> Kansas, Lawrence, KS<br />

Doxorubicin (DOX) has been used as an anticancer drug to treat a variety <strong>of</strong> cancers. Due to<br />

<strong>the</strong> first pass effect, DOX has been only administrated intravenously. Recent studies have<br />

shown that is<strong>of</strong>lavonoid, i.e. quercetin, can improve oral bioavailability <strong>of</strong> doxorubicin by<br />

blocking <strong>the</strong> activity <strong>of</strong> p-glycoprotein. In addition, quercetin is absorbed primarily by gut lymph.<br />

Our hypo<strong>the</strong>sis is that oral administrated doxorubicin-quercetin prodrug (DQ) will improve oral<br />

bioavailability and potentially enhance <strong>the</strong> absorption <strong>of</strong> DOX in gut. We have successfully<br />

syn<strong>the</strong>sized DQ. The IC50 <strong>of</strong> DOX, quercetin and DQ in 4T1.2 cells were 5.7 µM, 49.2 µM and<br />

14.04 µM, respectively. Both free DOX and DQ were orally fed to Sprague Dawley female rats.<br />

Pharmacokinetic study showed that 1h after drug administration, only DQ, but not free DOX,<br />

gave an active form <strong>of</strong> DOX in <strong>the</strong> blood serum. This study showed that <strong>the</strong> doxorubicinquercetin<br />

prodrug strategy can be used to improve oral bioavailability <strong>of</strong> DOX.<br />

2179<br />

Induction <strong>of</strong> Intracellular Superoxide in Chronic Myelogenous Leukemia Cells Leads to<br />

an Antioxidant Response Downstream <strong>of</strong> NFE2L2.<br />

L. F. Gemta 1 , F. Alvarez-Calderon 1,2 , K. Hansen 1 , J. DeGregori 1,3 ; 1 Biochemistry and <strong>Molecular</strong><br />

Genetics, University <strong>of</strong> Colorado - Anschutz Medical Campus, Aurora, CO, 2 Medical Scientist<br />

Training Program, University <strong>of</strong> Colorado - Anschutz Medical Campus, Aurora, CO 3 <strong>Molecular</strong><br />

<strong>Biology</strong>, University <strong>of</strong> Colorado - Anschutz Medical Campus, Aurora, CO<br />

Chronic myelogenous leukemia (CML) is a myeloproliferative disorder characterized by <strong>the</strong><br />

BCR-ABL fusion gene. Bcr-Abl tyrosine kinase inhibitors including Imatinib have been used to<br />

treat CML in patients. These inhibitors fail to eliminate all types <strong>of</strong> Bcr-Abl+ leukemia especially<br />

in advance and blast crisis phases. In this study, we identified that inhibiting mitochondrial


MONDAY<br />

metabolism with oligomycin A, an inhibitor <strong>of</strong> mitochondria ATP-synthase, sensitizes CML cells<br />

to Imatinib. The combination <strong>of</strong> 1 µM Imatinib and 5-10 nM oligomycin A resulted in elimination<br />

<strong>of</strong> CML cells without inhibition <strong>of</strong> mitochondrial respiration. Although Annexin V/Propidium<br />

Iodine analysis showed that most <strong>of</strong> <strong>the</strong> cells died by apoptosis, <strong>the</strong> exact mechanism by which<br />

<strong>the</strong> combination <strong>the</strong>rapy successful eliminated <strong>the</strong>se cancer cells is unknown since <strong>the</strong> doses <strong>of</strong><br />

OA used are ~100-fold lower than <strong>the</strong> IC50. The level <strong>of</strong> HIF-1α and downstream targets <strong>of</strong> Bcr-<br />

Abl including p-Akt, p-ERK, and p-STAT5 were not affected by <strong>the</strong> treatment. The combination<br />

<strong>the</strong>rapy increased intracellular superoxide levels and induced expression <strong>of</strong> <strong>the</strong> antioxidant<br />

response downstream <strong>of</strong> NFE2L2 expression. Normal and cancerous cells protect <strong>the</strong>mselves<br />

from <strong>the</strong> damage caused by high levels <strong>of</strong> superoxide by activating <strong>the</strong>ir antioxidant responses.<br />

These responses include <strong>the</strong> induction <strong>of</strong> <strong>the</strong> transcription factor nuclear factor (erythroidderived<br />

2)-like 2 (NFE2L2), which in turns regulates <strong>the</strong> expression <strong>of</strong> its downstream target<br />

genes including superoxide dismutase-2 (SOD2), gamma-glutamyltransferase 1 (GGT1),<br />

glutamate-cysteine ligase catalytic subunit (GCLC), and heme oxygenase (decycling) 1<br />

(HMOX1). The combination <strong>the</strong>rapy inhibits mitochondrial metabolism without inhibiting<br />

mitochondrial respiration and leads to increases in superoxide levels and compensatory<br />

increases in antioxidant responses.<br />

2180<br />

Genome-Wide RNAi Screen for Tetraploid-Specific Lethality in Cancer Cells.<br />

T. S. Kuroda 1 , R. K. Dagher 1 , D. Pellman 1 ; 1 Pediatric Oncology, Dana-Farber Cancer Institute,<br />

Howard Hughes Medical Institute, Boston, MA<br />

Cancer cells are <strong>of</strong>ten genetically unstable, resulting in aneuploid genomes. The consequences<br />

<strong>of</strong> aneuploidy are poorly understood, but it has been suggested to play roles in tumor<br />

aggressiveness, chemo<strong>the</strong>rapy resistance, and metastasis.<br />

One important route to an aneuploid genome may be through doubling <strong>of</strong> <strong>the</strong> genome, leading<br />

to unstable tetraploid cells. Many cell division errors ultimately manifest as a failure <strong>of</strong><br />

cytokinesis. Especially in cells defective in p53 function, tetraploid cells proliferate, but generate<br />

both high rates <strong>of</strong> whole chromosome aneuploidy and defects in <strong>the</strong> maintenance <strong>of</strong><br />

chromosome structural integrity. Our laboratory has demonstrated that in p53-null primary cells,<br />

tetraploidy promotes tumor development and <strong>the</strong> resulting tumors display markedly abnormal<br />

genomes (Fujiwara T. et al. Nature 2005, 437: 1043). It is now known that several human<br />

cancer-inducing mutations-- e.g. RB loss or loss <strong>of</strong> <strong>the</strong> adenomatous polyposis coli tumor<br />

suppressor-- also predispose cytokinesis failure and tetraploidy. These findings highlight a need<br />

to understand in detail <strong>the</strong> physiological changes that accompany tetraploidy, which might<br />

provide with a new strategy for cancer <strong>the</strong>rapeutics.<br />

To gain an understanding <strong>of</strong> physiological alterations associated with tetraploidy in cancer cells,<br />

we have established tetraploid cell lines derived from a near diploid human colorectal cancer<br />

cell line HCT116. Compared to diploid cells, anaphase FISH showed that tetraploid cells have a<br />

higher rate <strong>of</strong> chromosome mis-segregation—with levels similar to chromosomally unstable<br />

cancer cells.<br />

Using <strong>the</strong>se paired tetraploid and diploid cell lines, we have performed a genome-wide RNAi<br />

screen to identify genes whose knockdown selectively affect <strong>the</strong> viability <strong>of</strong> chromosomally<br />

unstable tetraploid cells. After secondary validation <strong>of</strong> <strong>the</strong> screen, we have identified 61<br />

tetraploid-selective lethality genes. Consistent with <strong>the</strong> known p53-dependent barrier to<br />

proliferation <strong>of</strong> tetraploid and aneuploid cells, and validating <strong>the</strong> overall approach, one<br />

tetraploid-selective lethality gene is MDM2, an E3 ligase for p53.


MONDAY<br />

2181<br />

Growth Inhibitory Effects <strong>of</strong> Death Receptor 5 Agonist, Drozitumab alone and in<br />

Combination with <strong>the</strong> Novel Spirocyclic Lignan Ramonanin A.<br />

K. J. Chavez 1 , B. Stone 1 , S. Lipkowitz 1 ; 1 Laboratory <strong>of</strong> Cellular and <strong>Molecular</strong> <strong>Biology</strong>, National<br />

Institutes <strong>of</strong> Health, National Cancer Insititute, Be<strong>the</strong>sda, MD<br />

Clinically, breast cancers can be divided into distinct subtypes that express estrogen receptors<br />

(ER), progesterone receptors (PR), those that have amplification <strong>of</strong> HER2/Neu, and those that<br />

lack expression <strong>of</strong> ER or PR and lack amplification <strong>of</strong> Her2/Neu (so called triple negative breast<br />

cancer or TNBC). TNBC has a poor outcome compared to <strong>the</strong> o<strong>the</strong>r subtypes <strong>of</strong> breast cancers.<br />

Previously, we have shown that a recombinant GST fusion protein with TNF-related Apoptosis<br />

Inducing Ligand (GST-TRAIL) selectively kills triple-negative breast cancer cells with<br />

mesenchymal features by activation <strong>of</strong> TRAIL receptor 2 (TRAIL-R2). In this study we have<br />

characterized <strong>the</strong> growth inhibitory effects <strong>of</strong> a clinically relevant agonistic antibody to TRAIL-<br />

R2, referred to as Drozitumab. To determine <strong>the</strong> selectivity <strong>of</strong> Drozitumab, we chose a panel <strong>of</strong><br />

15 breast cancer cell lines including 3 ER/PR positive cell lines, 4 HER2/Neu amplified cell<br />

lines, and 8 triple-negative cell lines. Treatment <strong>of</strong> <strong>the</strong>se cells with Drozitumab selectively killed<br />

TNBC cell lines with mesenchymal features. ER positive, HER2/Neu amplified and TNBC cell<br />

lines with epi<strong>the</strong>lial features were resistant to Drozitumab induced cell death. Drozitumab<br />

induced caspase activation (measured by activation <strong>of</strong> <strong>the</strong> initiator caspase 8, activation <strong>of</strong> <strong>the</strong><br />

downstream caspases 3/7, and PARP cleavage) and <strong>the</strong> toxicity <strong>of</strong> Drozitumab was blocked by<br />

<strong>the</strong> pan-caspase inhibitor z-VAD-FMK. This was identical to <strong>the</strong> pattern seen with GST-TRAIL.<br />

Cross-linking with an anti-FC antibody enhanced <strong>the</strong> efficacy <strong>of</strong> Drozitumab as shown by <strong>the</strong><br />

more rapid appearance <strong>of</strong> cleaved caspase 8, greater caspase 3/7 activity, and more rapid<br />

PARP cleavage. Fur<strong>the</strong>rmore, we show that a novel family <strong>of</strong> Spirocyclic lignans from<br />

Guaiacum sp. (Ramonanins) enhances Drozitumab -induced apoptosis in MBA-MD231and<br />

HCC38 triple-negative breast cancer cell lines. In conclusion, our data indicates that Drozitumab<br />

selectively inhibits growth <strong>of</strong> mesenchymal phenotype triple-negative breast cancer cell lines in<br />

a caspase dependent fashion and that cell death is enhanced in <strong>the</strong> presence <strong>of</strong> Ramonanin A.<br />

This data provides evidence that TRAIL-R2 targeted <strong>the</strong>rapies may have <strong>the</strong>rapeutic potential in<br />

<strong>the</strong> treatment <strong>of</strong> TNBC with mesenchymal features.<br />

2182<br />

The effective inhibition <strong>of</strong> growth rate and induction <strong>of</strong> apoptosis in adherent and<br />

suspension cancer cell lines by <strong>the</strong> administration <strong>of</strong> various Panax (Ginseng) species.<br />

N. Yousaf 1 , A. Luiso 1 , T. Zeleke 1 , A. Bettica 1 ; 1 <strong>Biology</strong>, Manhattanville College, Purchase, NY<br />

Ginseng (Panax) is a perennial plant with fleshy roots that contains Rg1, Rg2, Rf and o<strong>the</strong>r<br />

ginsenoside groups. A popular traditional medicine for centuries, ginseng is believed to enhance<br />

memory, stimulate <strong>the</strong> immune system, reduce cholesterol levels, and fight depression and<br />

insomnia. Various Panax species, such as Korean, American, and Chinese Red or White<br />

Ginseng, have been shown to enhance memory and stimulate <strong>the</strong> immune system. Ginseng<br />

extracts have also been considered as adaptogens, natural substances that aid in homeostatic<br />

balance. The red ginseng, in its various forms, differs from white ginseng in its processing. The<br />

Korean ginseng, grown in cooler climates, has higher ginsenosides/saponin count than Chinese<br />

ginseng. Panax red ginseng, ei<strong>the</strong>r Chinese or Korean, is considered to be more potent than <strong>the</strong><br />

American ginseng, Panax quinquefolium. To determine <strong>the</strong> anticancer effects <strong>of</strong> <strong>the</strong> various<br />

ginseng species, <strong>the</strong> inhibition <strong>of</strong> growth rate and <strong>the</strong> induction <strong>of</strong> apoptosis is determined for<br />

both an adherent pancreatic adenocarcinoma cell line (PANC-1) and a suspension B-cell<br />

leukemic cell line (SUP-B15). B cell chronic lymphocytic leukemia (B-CLL) has long been<br />

considered a disease in which B-CLL cells accumulate due to a presumed defect in <strong>the</strong>ir


MONDAY<br />

apoptotic mechanism. Previous morphologic observations suggest that B-CLL cells are resting<br />

cells with rarely detected cell cycle activity in <strong>the</strong> blood. For single and combination treatments,<br />

cells will be seeded at a density <strong>of</strong> 5x104 cells/well in 96-well plates. For suspension cultures,<br />

round bottom 96 well plates are used. When seeding round bottom well plates, <strong>the</strong> cells are<br />

allowed to sediment, forming a relatively loose monolayer. All plated cells are allowed one to<br />

two population doubling times (PDT) before treating. Single drug 24 hour treatments for dose<br />

response curves in ten fold dilutions determine effective IC50 values for each <strong>of</strong> <strong>the</strong> ginseng<br />

species. Results from <strong>the</strong>se preliminary dose response studies determine whe<strong>the</strong>r consecutive<br />

(24 hours <strong>of</strong> one species followed by 24 hours <strong>of</strong> second species at effective doses) or additive<br />

drug trials (1/2 effective dose <strong>of</strong> each species toge<strong>the</strong>r for 24 hours), is considered. The<br />

treatment course is followed by 1-2 PDT and assayed by one <strong>of</strong> two different methods. All<br />

treatment plates are repeated in triplicate. To determine inhibition <strong>of</strong> growth rate and cell<br />

viability, <strong>the</strong> MTT cell proliferation assay is performed with adequate washing. The optical<br />

density is read between 490-550nm and adjusted for background. The Caspase-3 colorimetric<br />

protease assay quantifies caspases that recognize <strong>the</strong> amino acid sequence DEVD, and since<br />

caspase 3 is an effector caspase, <strong>the</strong> level <strong>of</strong> apoptosis under <strong>the</strong> test conditions can be<br />

determined. All data are statistically analyzed using a one-way ANOVA with Tukey’s post hoc<br />

tests. Treatments with American Ginseng showed protection in <strong>the</strong> nanogram ranges. Korean<br />

Ginseng was more effective individually and, in combination treatments, <strong>the</strong> Chinese<br />

Red/Chinese White Ginseng extracts were most effective overall. Considering <strong>the</strong> growth rate<br />

inhibition and induction <strong>of</strong> apoptosis on adherent and suspension cancer cultures, ginseng<br />

species are potential anticancer agents.<br />

2183<br />

Is<strong>of</strong>orm specific function <strong>of</strong> <strong>the</strong> formin FMNL2.<br />

C. Péladeau 1 , A. Heibein 1 , G. L. Lahaie 1 , S. Copeland 1 , J. Copeland 1 ; 1 Cellular and <strong>Molecular</strong><br />

Medicine, University <strong>of</strong> Ottawa, Ottawa, ON, Canada<br />

Cancer cell metastasis is induced by actin-dependent cell migration and is consequently<br />

affected by <strong>the</strong> regulated function <strong>of</strong> cytoskeletal remodelling proteins. Formins, a highly<br />

conserved family <strong>of</strong> actin remodelling proteins, have been shown to provoke motility during<br />

metastasis and invasion. Indeed, formin-like 2 (FMNL2) has been shown to increase motility<br />

and facilitate invasion when overexpressed in colorectal cancer cells (CRC). Fur<strong>the</strong>rmore,<br />

knocking down <strong>the</strong> expression <strong>of</strong> FMNL2 inhibits amoeboid-style invasion by melanoma cells in<br />

vitro. FMNL2 mRNA is subject to alternative splicing and results from our lab, and o<strong>the</strong>rs,<br />

suggest that <strong>the</strong> resulting encoded proteins are likely to differ in <strong>the</strong>ir regulation, subcellular<br />

localization and activity. The main objectives <strong>of</strong> this study are to identify <strong>the</strong> FMNL2 splice forms<br />

expressed in various cancer cell lines and determine if FMNL2 activity is required by <strong>the</strong>se cells<br />

for invasion. In this study we have used RT-PCR to identify four FMNL2 splice forms (ITM, YHY,<br />

PMR and TQS) expressed in non-invasive (SW480) and invasive (SW620) colorectal cancer<br />

cells, as well as in highly invasive A375 melanoma cells. Preliminary data from cell-based<br />

assays suggest that <strong>the</strong>re are functional differences between our identified FMNL2 is<strong>of</strong>orms in<br />

terms <strong>of</strong> both auto-regulation and <strong>the</strong>ir effects on actin dynamics. In addition, our qPCR data<br />

suggests that an “invasive” is<strong>of</strong>orm <strong>of</strong> FMNL2 may be preferentially expressed in highly invasive<br />

cells. The data obtained in this study will lead to fur<strong>the</strong>r understanding <strong>of</strong> <strong>the</strong> role <strong>of</strong> FMNL2 in<br />

invasion and metastasis as well as identify specific targets for <strong>the</strong> development <strong>of</strong> future antimetastatic<br />

<strong>the</strong>rapies.


2184<br />

Pr<strong>of</strong>ilin-1 downregulation promotes transendo<strong>the</strong>lial migration <strong>of</strong> breast cancer cells<br />

through increased VEGF secretion.<br />

M. Joy 1 , P. Roy 2 ; 1 Bioengineering, University <strong>of</strong> Pittsburgh, 2 Bioengineering and Pathology,<br />

University <strong>of</strong> Pittsburgh, Pittsburgh, PA<br />

MONDAY<br />

Transendo<strong>the</strong>lial migration <strong>of</strong> cancer cells is necessary for both intravasation and extravasation<br />

steps <strong>of</strong> tumor metastasis. Clinical correlation and xenograft studies from our laboratory have<br />

revealed that downregulation in <strong>the</strong> expression <strong>of</strong> pr<strong>of</strong>ilin-1 (Pfn1: an actin-binding protein) is<br />

associated with increased metastatic potential <strong>of</strong> breast cancer cells. The aim <strong>of</strong> <strong>the</strong> present<br />

study was to evaluate <strong>the</strong> effect <strong>of</strong> loss <strong>of</strong> Pfn1 expression on transendo<strong>the</strong>lial migration <strong>of</strong><br />

breast cancer cells. We found that stably silencing Pfn1 expression through shRNA increases<br />

<strong>the</strong> ability <strong>of</strong> MDA-MB-231 breast cancer cells to transmigrate through a confluent monolayer <strong>of</strong><br />

human vascular endo<strong>the</strong>lial cells (VEC), and this can be rescued by re-expression <strong>of</strong> an RNAiresistant<br />

version <strong>of</strong> Pfn1. Immunostaining for beta-catenin (a component <strong>of</strong> adherens junctions)<br />

showed that Pfn1 knockdown facilitates tumor cell-induced disruption <strong>of</strong> cell-cell junctions in<br />

VEC. Biochemical analyses <strong>of</strong> conditioned media derived from MDA-231 cells revealed that<br />

secretion <strong>of</strong> several factors that promote tumor metastasis including VEGF (a potent disruptor <strong>of</strong><br />

endo<strong>the</strong>lial cell-cell junctions) is significantly increased upon loss <strong>of</strong> Pfn1 expression. Silencing<br />

VEGF expression abrogates <strong>the</strong> stimulatory effect <strong>of</strong> Pfn1 KD on transendo<strong>the</strong>lial migration thus<br />

suggesting that Pfn1 downregulation promotes transendo<strong>the</strong>lial migration <strong>of</strong> breast cancer cells<br />

through increased VEGF secretion. These findings set <strong>the</strong> stage for future in vivo studies<br />

examining Pfn1’s role in regulating tumor cell dissemination into <strong>the</strong> vasculature.<br />

2185<br />

Microarray analysis <strong>of</strong> differentially expressed genes in MDA-MB-231 breast cancer cells<br />

treated with <strong>the</strong> laminin-derived peptide C16.<br />

E. S. Santos 1 , V. M. Freitas 1 , M. S. Menezes 1 , G. M. Machado-Santelli 1 , R. G. Jaeger 1 ;<br />

1<br />

Departament <strong>of</strong> Cell and Developmental <strong>Biology</strong>, Institute <strong>of</strong> Biomedical Sciences <strong>of</strong> The<br />

University <strong>of</strong> Sao Paulo, Sao Paulo, Brazil<br />

Human breast cancer constitutes a worldwide health care problem. During breast cancer<br />

development and progression, tumor cells are engaged in a complex interplay with <strong>the</strong><br />

surrounding microenvironment. In this scenario, extracellular matrix molecules play important<br />

roles influencing malignant behavior. An increasing number <strong>of</strong> evidences have shown that<br />

peptides derived from laminin cleavage are involved in tumor progression. Among <strong>the</strong>m, C16<br />

(KAFDITYVRLKF), derived from laminin-111 gamma-1 chain, is a cell-adhesive peptide that<br />

increases cell migration and invasion, enhances metastasis, and promotes angiogenesis. This<br />

prompted us to analyze whe<strong>the</strong>r C16 would regulate gene expression in cells derived from<br />

human breast cancer (MDA-MB-231). Cells were treated with C16 or scrambled peptide control<br />

(FKLRVYTIDFAK) for 24 hours. After treatment, gene expression was analyzed by microarray.<br />

Total RNA was extracted, biotin-labeled cDNA was generated and hybridized to Human Gene<br />

1.0 ST Array (Affymetrix). GeneChips were stained and scanned. Expression Console s<strong>of</strong>tware<br />

converted gene fluorescence intensities into numerical values. We found that C16 regulates<br />

expression <strong>of</strong> 80 genes in MDA-MB-231, including genes directly involved in processes<br />

associated with cancer. Among those genes, FGFR3, GPNMB and SPOCK1 expression was<br />

increased by C16 treatment, as confirmed by quantitative Real-Time PCR. FGFR3 encodes a<br />

tyrosine kinase receptor, which regulates various biological processes, including cell migration<br />

and angiogenesis. GPNMB encodes a transmembrane glycoprotein involved in angiogenesis<br />

and promotion <strong>of</strong> migration, invasion and bone metastasis <strong>of</strong> breast cancer cells. SPOCK1, on<br />

<strong>the</strong> o<strong>the</strong>r hand, encodes a proteoglycan associated with cell adhesion and increased migration.


MONDAY<br />

To correlate gene expression with function, we analyzed whe<strong>the</strong>r C16 would regulate invasion,<br />

proliferation and apoptosis in MDA-MB-231 cells. Functional results also indicated that C16<br />

significantly increases MDA-MB-231 cell invasion, as shown by assays in Boyden chambers<br />

coated with Matrigel. However, no effect on cell proliferation and apoptosis was observed. We<br />

concluded that laminin-derived peptide C16 regulates gene expression and enhances invasion<br />

<strong>of</strong> metastatic MDA-MB-231 breast cancer cells.<br />

2186<br />

Laminin-derived peptide C16 induces invasion and invadopodia activity in human oral<br />

squamous cell carcinoma and fibrosarcoma cells.<br />

A. S. Siqueira 1 , R. Rafael 1 , M. P. Pinto 1 , V. M. Freitas 1 , R. G. Jaeger 1 ; 1 Cell and Developmental<br />

<strong>Biology</strong>, Institute <strong>of</strong> Biomedical Sciences - University <strong>of</strong> Sao Paulo, Sao Paulo, Brazil<br />

Our Laboratory has been studying <strong>the</strong> effects <strong>of</strong> laminin-derived peptides in tumor biology.<br />

Here, we addressed whe<strong>the</strong>r peptide C16 (KAFDITYVRLKF, gamma 1 chain) would stimulate<br />

invasion activity in cell lines derived from oral squamous cell carcinoma (OSCC) and<br />

fibrosarcoma (HT1080), two malignant tumors with different origins and poor prognosis. Using<br />

Boyden chambers coated by Matrigel, we observed that C16 increased invasion rate in both cell<br />

lines compared to scrambled control peptide (C16SX). Tumor cells that actively invade<br />

surrounding tissues depend on invadopodia to degrade extracellular matrix barriers.<br />

Invadopodia are actin-rich membrane protrusions associated with proteolytic activity. Therefore,<br />

we analyzed <strong>the</strong> role <strong>of</strong> C16 on invadopodia activity <strong>of</strong> OSCC and HT1080 cells, through a<br />

fluorescent substrate degradation assay. Measurement <strong>of</strong> digestion spots (black areas in<br />

fluorescent background) showed that C16 significantly increased invadopodia activity <strong>of</strong> both<br />

neoplastic cell lines compared to C16SX. Invadopodia dynamics in C16-treated cells was fu<strong>the</strong>r<br />

explored using time-lapse 4D fluorescence microscopy. Time-lapse videos <strong>of</strong> living cells<br />

transfected with GFP-cortactin and cultured on fluorescent gelatin were acquired at 5 min<br />

interval (total 1-2 hours). Invadopodia extensions through <strong>the</strong> fluorescent substrate were<br />

evaluated with acquisition <strong>of</strong> at least ten Z sections per time point, using a piezoelectric device<br />

coupled to <strong>the</strong> microscope objective. Measurements <strong>of</strong> degraded areas showed that C16<br />

increased invadopodia activity over time compared to C16SX. We also searched for signaling<br />

pathways related to C16 effects in OSCC and HT1080 cells. This peptide increased p-ERK<br />

expression in both cell lines compared to control.Fur<strong>the</strong>rmore, inhibition <strong>of</strong> ERK signaling<br />

pathway with MEK inhibitor U0126 decreased C16-related invasion and invadopodia activity.<br />

These results demonstrated that C16 effects in <strong>the</strong>se cell lines may be related to ERK 1/2<br />

pathway. We conclude that C16 increases invasion and invadopodia activity in cells derived<br />

from oral squamous cell carcinoma and fibrosarcoma, probably through ERK signaling pathway.<br />

Support: FAPESP (08/57103-8; 09/17336-6) and CNPq (304986/2009-7).<br />

2187<br />

ADAMTS-1 regulating migration, invasion, and invadopodia formation in breast cancer<br />

cells.<br />

V. M. Freitas 1 , J. B. Amaral 1 , E. S. Santos 1 , R. G. Jaeger 1 , F. R. Mangone 2 , M. A. Nagai 2 , G. M.<br />

Machado-Santelli 1 ; 1 Department <strong>of</strong> Cell and Developmental <strong>Biology</strong>, Institute <strong>of</strong> Biomedical<br />

Sciences <strong>of</strong> The University <strong>of</strong> Sao Paulo, Sao Paulo, Brazil, 2 Department <strong>of</strong> Radiology, School<br />

<strong>of</strong> Medicine <strong>of</strong> The University <strong>of</strong> Sao Paulo, Sao Paulo, Brazil<br />

ADAMTS-1 (a disintegrin and metalloprotease with thrombospondin motifs) is a member <strong>of</strong> <strong>the</strong><br />

ADAMTS family <strong>of</strong> metalloproteases. This enzyme is related to pathological processes such as<br />

inflammation and cancer. In spite <strong>of</strong> ADAMTS-1 biological relevance, <strong>the</strong> mechanisms<br />

underlying its involvement in tumor biology remain elusive. Here we sought to investigate <strong>the</strong>


MONDAY<br />

role played by ADAMTS-1 regulating migration and invasion <strong>of</strong> malignant mammary cell lines<br />

(MDA-MB-231 and MCF7). We observed variable levels <strong>of</strong> ADAMTS1 mRNA expression in a<br />

series <strong>of</strong> primary breast tumors. MDA-MB-231 cells with reduced expression <strong>of</strong> ADAMTS-1<br />

exhibited increase in migration and invasion. Cancer cells depend on invadopodia to degrade<br />

and invade surrounding extracellular matrix. Invadopodia are membrane protrusions with<br />

enzymes required for extracellular matrix components (ECM) degradation. Silencing <strong>of</strong><br />

ADAMTS-1 by siRNA increased invadopodia activity in MDA-MB-231 cells, as shown by<br />

fluorescent degradation substrate assays. Cortactin and MT1-MMP are important invadopodia<br />

proteins. Immunoblot <strong>of</strong> cells transfected with ADAMTS-1 siRNA showed augmentation <strong>of</strong><br />

cortactin and MT1-MMP expression compared to controls. Regulatory mechanisms underlying<br />

ADAMTS-1 effects may be related to VEGF, a growth factor involved in migration and invasion.<br />

MDA-MB-231 cells with silenced ADAMTS-1 showed increase <strong>of</strong> VEGF concentration in <strong>the</strong><br />

conditioned medium compared to control. This conditioned medium also induced HUVEC<br />

tubulogenesis. Fur<strong>the</strong>rmore, MBA-MB-231 showed high expression <strong>of</strong> VEGF receptor<br />

(VEGFR2) compared to MCF7 cells. We may assume that ADAMTS-1 effects in tumor<br />

invasiveness may be related to VEGF and VEGFR expression. We suggest that decrease <strong>of</strong><br />

ADAMTS-1 stimulates migration, invasion and invadopodia formation in breast cancer cells.<br />

These effects are probably regulated by VEGF and VEGFR.<br />

2188<br />

Analysis <strong>of</strong> endo<strong>the</strong>lium mimicry by <strong>the</strong> CSC-like cells derived from iPS cells.<br />

S-I. Matsuda 1 , A. Mizutani 1 , T. Kasai 1 , A. Satoh 1 , T. Kudoh 1 , L. Chen 1,2 , M. Seno 1 ; 1 Graduate<br />

School <strong>of</strong> Natural Science and Technology, Okayama University, Okayama, Japan, 2 Tianjin<br />

Central Hospital <strong>of</strong> Bynecology Obstetrics, China<br />

Cancer stem cells (CSCs) are capable <strong>of</strong> self-renewal and differentiation, which has been<br />

considered to be responsible for tumor initiation and to contribute to cancer resistance. In<br />

addition, CSC-like cells in glioblastoma have also been demonstarted to mimic or differentiate<br />

into vascular endo<strong>the</strong>lium. Eradication <strong>of</strong> <strong>the</strong>se CSCs is considered critical part <strong>of</strong> successful<br />

anti-cancer treatment, but detailed analysis <strong>of</strong> CSCs is still slow because <strong>of</strong> <strong>the</strong>ir rarity and<br />

difficulty <strong>of</strong> maintenance in culture.<br />

Recently, we have established stable CSC-like cells from mouse induced pluripotent stem<br />

(miPS) cells (Mizutani et al., 2011 ASCB meeting), which were cultured in <strong>the</strong> conditioned<br />

medium <strong>of</strong> several kinds <strong>of</strong> mouse cancer cell lines P19, Lewis lung carcinoma (LLC), B16 and<br />

MC.E12. In particular, miPS cells converted to CSC-like cells in <strong>the</strong> conditioned medium <strong>of</strong><br />

Lewis lung carcinoma (miPS-LLCcm) cells exhibited extensive angiogenesis in vivo. In this<br />

study, we evaluated <strong>the</strong>se miPS-LLCcm cells for <strong>the</strong> potency to mimic vascular endo<strong>the</strong>lium.<br />

First we inquire about <strong>the</strong>ir stemness by assessing <strong>the</strong> expression <strong>of</strong> stemness marker genes,<br />

Oct3/4, Sox2, Klf4 and Nanog. Compared with miPS, <strong>the</strong>se markers except Oct3/4 tend to be<br />

up-regulated in <strong>the</strong> obtained CSC-like cells implying some difference from normal stem cells.<br />

Simultaneously, <strong>the</strong> expression <strong>of</strong> vascular endo<strong>the</strong>lial markers, VE-cadherin and VEGFR2,<br />

were also found upregulated in miPS-LLCcm and miPS-B16cm cells. However, only miPS-<br />

LLCcm cells exhibited tube formation on MatrigelTM, which is consistent with <strong>the</strong> results <strong>of</strong><br />

tumorigenicity in vivo. Since this in vitro tube formation was observed in <strong>the</strong> absence <strong>of</strong> VEGF,<br />

VEGFR2 might not directly be involved in <strong>the</strong> angiogenesis. When miPS-LLCcm cells were<br />

cultured in <strong>the</strong> presence <strong>of</strong> puromycin, <strong>the</strong> tube formation was not observed. This impotency<br />

was recovered when puromycin was removed from <strong>the</strong> culture media. Taking <strong>the</strong>se into<br />

consideration miPS-LLCcm cells should have potential <strong>of</strong> differentiation, which is responsible for<br />

vasculogenic mimicry in vivo, maintaining <strong>the</strong> heterogeneous population in <strong>the</strong> culture. This<br />

speculation was supported by <strong>the</strong> flow cytometoric analysis for GFP expression in miPS-LLCcm<br />

cells showing approximately 30 % was GFP positive. Thus, miPS-LLCcm cells in this study


MONDAY<br />

have successfully been shown to have CSC-like characters, such as cancer stemness,<br />

malignant behavior and capacity to mimic vascular endo<strong>the</strong>lium. Our CSC-like cells might be a<br />

good model to explain <strong>the</strong> mechanism <strong>of</strong> tumor angiogensis in detail not limiting in glioblastoma.<br />

2189<br />

The HT29 and DLD1 colorectal cancer cell lines contain cancer stem cells.<br />

C. B. Penny 1 , B. L. Milner 1 , V. E. Gibbon 1 , P. Ruff 1 ; 1 Oncology, University <strong>of</strong> Witwatersrand Med<br />

Sch, Johannesburg, South Africa<br />

Cancer stem cells (CSCs) similar to embryonic stem cells are defined by <strong>the</strong>ir abilities to selfrenew<br />

and additionally retain <strong>the</strong> ability to form tumours ectopically. They have been identified in<br />

several human malignancies, recently being associated with functional properties <strong>of</strong> drug<br />

resistance, invasion and migration. This resistance to conventional chemo<strong>the</strong>rapies may lead to<br />

tumour relapse and <strong>the</strong> arisal <strong>of</strong> new tumours. Using confocal microscopy, we have identified<br />

putative colon cancer stem cells (CSC) in <strong>the</strong> HT29 and DLD-1 colon cancer derived cell lines.<br />

These lines representative <strong>of</strong> stage 3 colorectal adenocarcinoma, co-express <strong>the</strong> cell surface<br />

markers, CD133 (prominin 1) and epi<strong>the</strong>lial cell adhesion marker (EpCam). Upon FACS<br />

analysis <strong>of</strong> <strong>the</strong>se cell lines, using CD133 (PE-Miltenyi Biotech) and EpCam antibodies (FITC-BD<br />

Biosciences), two distinct cell populations were gated; within <strong>the</strong> HT29 cell line, 64.63% <strong>of</strong> gated<br />

events were positive for EpCam and 35.30% expressed both EpCam and CD133; and in <strong>the</strong><br />

DLD1 cell line, 46.77% <strong>of</strong> gated events were positive for EpCam expression and 52.83% for<br />

both markers. Subsequently, magnetic cell separation was used to isolate pure populations <strong>of</strong><br />

<strong>the</strong>se cells from each cell line, with a CD133 antibody labeled with superparamagnetic particles<br />

(Miltenyi Biotech). The purified CD133+ cells were cultured in StemPro human embryonic stem<br />

cell medium (serum free) (GIBCO), to minimize <strong>the</strong> possibility <strong>of</strong> differentiation. The cells were<br />

grown both as adherent cultures and as three dimensional spheroids, <strong>the</strong>se being more<br />

representative <strong>of</strong> in vivo tumours. To fur<strong>the</strong>r investigate <strong>the</strong> “stemness” <strong>of</strong> <strong>the</strong>se cells, <strong>the</strong><br />

expression <strong>of</strong> <strong>the</strong> pluripotency markers Nanog, Sox 2 and Oct3/4 were assessed by confocal<br />

microscopy. All <strong>of</strong> <strong>the</strong>se transcription factors essential to stem cell identity are present in <strong>the</strong><br />

CD133+ cells. In summary, <strong>the</strong> CSCs isolated here, represent a novel population <strong>of</strong> cells for<br />

evaluating cancer <strong>the</strong>rapies directed against pluripotent cells.<br />

2190<br />

Floating spherical cell colonies from human adenocarcinoma <strong>of</strong> <strong>the</strong> lung are enriched in<br />

stem-like phenotypic markers.<br />

A. Tesei 1 , C. Arienti 1 , G. Paganelli 1 , A. Pasini 2 , G. Brigliadori 1 , D. Calistri 1 , E. Giordano 2 , W.<br />

Zoli 1 ; 1 Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori<br />

(I.R.S.T.), Meldola (FC), Italy, 2 Laboratory <strong>of</strong> Cellular and <strong>Molecular</strong> Engineering, University <strong>of</strong><br />

Bologna - Campus <strong>of</strong> Cesena, Cesena (FC), Italy<br />

It is presently widely accepted that tumor growth rely on a niche <strong>of</strong> so-called cancer stem cells<br />

(CSCc), or tumor-initiating cells (TICs), endowed with self-renewal properties. We recently<br />

showed that a subset <strong>of</strong> normal lung epi<strong>the</strong>lial-specific stem cells might reside within <strong>the</strong><br />

bronchoalveolar duct junction (Tesei A., et al. Cell Proliferation 2009; 42: 298-398). The aim <strong>of</strong><br />

<strong>the</strong> present work was to isolate and characterize CSCc/TICs from a human adenocarcinoma<br />

cell line (RAL) that we originally established at <strong>the</strong> Istituto Scientifico Romagnolo per lo Studio e<br />

la Cura dei Tumori (I.R.S.T.) at Meldola (Gasperi-Campani A., et al., Cancer Genetics and<br />

Cytogenetics 1998; 107:11–20) and from surgical human tissue samples. Single-cell<br />

suspensions derived from <strong>the</strong> RAL cell line were cultured in ultralow attachment plates in a<br />

serum-free medium. Neoplastic lung tissue samples, derived from patients affected by<br />

adenocarcinoma at different stages, were mechanically and enzymatically dissociated before


MONDAY<br />

expansion in culture. Expression <strong>of</strong> stemness related genes in cell clusters was evaluated with<br />

RT-PCR and Real Time RT-PCR. Single-cell suspensions gave rise to floating spherical<br />

colonies within about ten days. Lung-spheres obtained by our originally established lung<br />

adenocarcinoma cell line RAL, resulted enriched in CD133 and OCT-3/4 transcripts. RT-PCR<br />

analysis <strong>of</strong> tumor spheres obtained from surgical tissue samples showed a higher expression <strong>of</strong><br />

BCRP-1, CD133, BMI-1, OCT-3/4, Lef-1, CD44 and Slug, when compared to original tissue. A<br />

gradual loss <strong>of</strong> CD133 expression was evident as <strong>the</strong> tumors become more undifferentiated.<br />

Evaluation <strong>of</strong> <strong>the</strong> level <strong>of</strong> CpG methylation in OCT-3/4 and CD133 gene promoters was also<br />

performed by means <strong>of</strong> bisulfite sequencing in cells growing in floating spherical RAL cell<br />

colonies and in <strong>the</strong> original adherent cell culture. They both showed a constant hypermethylated<br />

status suggesting that this level <strong>of</strong> transcriptional control is not involved in modulating <strong>the</strong><br />

expression <strong>of</strong> <strong>the</strong>se stem-like phenotypic markers.<br />

2191<br />

Generation <strong>of</strong> Cancer Stem Cell Model from Mouse iPS Cells.<br />

A. Mizutani 1 , S-I. Matsuda 1 , T. Kasai 1 , T. Kudoh 1 , L. Chen 2 , M. Seno 1 ; 1 Okayama University,<br />

Okayama, Japan, 2 Tianjin Central Hospital <strong>of</strong> Bynecology Obstetrics, China<br />

Objective; Heterotopic transplantation <strong>of</strong> embryonic stem cells and induced pluripotent stem<br />

(iPS) cells have been shown to form teratoma, a benign tumor exhibiting heterogeneous but<br />

normal phenotype <strong>of</strong> differentiated cells, but not teratocarcinoma. Since <strong>the</strong> stem cells niche has<br />

been thought to play a significant role for <strong>the</strong> maintenance and <strong>the</strong> differentiation <strong>of</strong> stem cells,<br />

we hypo<strong>the</strong>sized that cancer niche could drive stem cells into malignant cells, particularly into<br />

cancer stem cells (CSCs). The presence <strong>of</strong> CSCs in tumor has been considered as a reason for<br />

a poor prognosis. Until now, no model <strong>of</strong> cancer stem cell line is established as an effective tool<br />

to study CSC. To investigate CSC in detail, we tried to generate cancer cells from mouse iPS<br />

(miPS) cells, in which green fluorescent protein (GFP) has been inserted into <strong>the</strong> 5â€<br />

untranslated region <strong>of</strong> <strong>the</strong> Nanog gene.<br />

Methods and Results;A ccording to our hypo<strong>the</strong>sis, miPS cells were cultured in <strong>the</strong> conditioned<br />

medium <strong>of</strong> various cancer derived cells (P19, LLC, B16, and MC.E12) for 4 weeks. The survived<br />

population <strong>of</strong> <strong>the</strong> cells was evaluated for tumorigenicity in nude mice. The miPS cells cultured in<br />

<strong>the</strong> conditioned medium from Lewis lung carcinoma cells (miPS-LLCcm) developed tumor with<br />

rapid growth and exhibited remarkable angiogenesis. Histology <strong>of</strong> <strong>the</strong> tumor derived from miPS-<br />

LLCcm showed <strong>the</strong> malignancy, while that <strong>of</strong> teratoma derived from miPS cells showed no<br />

malignancy. Morphologically stem-like cells expressing GFP was observed in <strong>the</strong> primary<br />

culture <strong>of</strong> <strong>the</strong> malignant tumor derived from miPS-LLCcm cells. These cells were found to grow<br />

as spheroid with GFP expression in suspension culture. The spheroid cells were also found to<br />

be highly tumorigenic in nude mice. The expression <strong>of</strong> <strong>the</strong> stem cell marker genes, such as<br />

Nanog, Cripto, Oct3/4, Sox2, Klf4, were detectable in miPS-LLCcm cells as much as in miPS<br />

cells.<br />

Conclusion; In this study, we cultured miPS cells in <strong>the</strong> conditioned mediun <strong>of</strong> cancer derived<br />

cells, which was regarded as cancer niche converting <strong>the</strong> normal stem cell into CSC cells. The<br />

resultant cells formed spheroids in suspension culture and developed malignant tumor in vivo<br />

exhibiting extensive angiogenesis. In addition, <strong>the</strong>se cells showed a capacity <strong>of</strong> self-renewal<br />

and expression <strong>of</strong> <strong>the</strong> markers associated with stem cell properties <strong>of</strong> undifferentiated state.<br />

Thus we concluded <strong>the</strong>se cells as a model <strong>of</strong> CSC. This model should critically be helpful for <strong>the</strong><br />

CSC study and for <strong>the</strong> development <strong>of</strong> effective cancer <strong>the</strong>rapies.


Cell <strong>Biology</strong> <strong>of</strong> Microbes and Parasites<br />

2192<br />

<strong>Molecular</strong> mechanism underlying MinE-induced membrane deformation.<br />

Y-L. Shih 1,2 ; 1 Institute <strong>of</strong> Biological Chemistry, Academa Sinica, Taipei, Taiwan, 2 Institute <strong>of</strong><br />

Biochemical Sciences, National Taiwan University, Taiwan<br />

MONDAY<br />

The Min system <strong>of</strong> E. coli mediates placement <strong>of</strong> <strong>the</strong> cell division septum at midcell. We recently<br />

reported that MinE can directly associate with <strong>the</strong> cell membrane through its N-terminal domain.<br />

An interesting observation is <strong>the</strong> ability <strong>of</strong> MinE to induce membrane tubule formation in vitro, a<br />

property similar to several eukaryotic proteins involved in membrane trafficking, such as<br />

dynamin and Bar-domain proteins. To investigate <strong>the</strong> mechanism underlying <strong>the</strong> MinE-induced<br />

membrane deformation, we used time-lapse fluorescence microscopy to characterize <strong>the</strong><br />

membrane deformation in vitro. We recorded massive tubulation from giant liposomes and<br />

initiation <strong>of</strong> membrane tubule formation and lipid clustering on <strong>the</strong> supported lipid bilayers.<br />

Fur<strong>the</strong>rmore, we found that residues 2-9 are capable <strong>of</strong> forming an amphipathic helix <strong>of</strong> 1-2<br />

helical turns when interact with <strong>the</strong> membrane.We used <strong>the</strong> tryptophan blue shift assays to<br />

determine <strong>the</strong> helical face that interacts with <strong>the</strong> membranes. CD spectral analyses suggested<br />

that conformational changes in MinE 1-31 occurred in <strong>the</strong> presence <strong>of</strong> liposomes. We conclude<br />

that <strong>the</strong> alpha helix <strong>of</strong> MinE 2-9 appears to work in concert with <strong>the</strong> charge residues R10/K11/K12<br />

to mediate <strong>the</strong> membrane association <strong>of</strong> MinE. It is known that insertion <strong>of</strong> an amphipathic helix<br />

into <strong>the</strong> membranes can induce local curvature changes that are able to destabilize <strong>the</strong> cell<br />

membranes and initiate membrane tubule formation. In conclusion, MinE not only shares<br />

common structural motifs to proteins involved in membrane trafficking, but is also active in<br />

inducing membrane deformation. We <strong>the</strong>refore uncovered <strong>the</strong> mechanism <strong>of</strong> how MinE may<br />

sculpt <strong>the</strong> membrane curvature.<br />

2193<br />

Biochemical and functional characterization <strong>of</strong> GlpQ homolog <strong>of</strong> Xenorhabdus<br />

nematophila.<br />

P. Singh 1,2 , R. Bhatnagar 1 , N. Banerjee 2 ; 1 School <strong>of</strong> Biotechnology, Jawaharlal Nehru<br />

University, Delhi, India, 2 Insect Resistance Group, International Centre for Genetic Engineering<br />

and Biotechnology, New Delhi, India<br />

Xenorhabdus nematophila is a gram negative bacteria living in symbiotic association with a soil<br />

nematode <strong>of</strong> <strong>the</strong> genus Steinernema and as a pathogen stage in insect hosts. The bacteria<br />

produces different exoenzymes which help in degradation <strong>of</strong> macromolecules to provide<br />

nutrients necessary for development <strong>of</strong> both <strong>the</strong> bacteria and <strong>the</strong> nematode. In <strong>the</strong> present<br />

study, we have characterized an extracellular enzyme glycerophosphodiester<br />

phosphodiesterase (GDPD) encoded by glpQ gene <strong>of</strong> X. nematophila. The DNA sequence<br />

encoding <strong>the</strong> enzyme was cloned, expressed and purified. We used <strong>the</strong> recombinant protein for<br />

biochemical characterization. The enzyme was able to hydrolyse glycerophosphodiesters like<br />

glycerophosphorylcholine, glycerophosphoethanolamine and to a lesser extent serine and<br />

inositol esters <strong>of</strong> phospholipids. The enzyme is <strong>the</strong>rmostable at 50°C for upto 4 hours, retaining<br />

85% <strong>of</strong> initial activity. It has an optimum pH <strong>of</strong> 9.5. The enzyme activity was stimulated by<br />

Ca 2+ ions. Activity pr<strong>of</strong>ile <strong>of</strong> GDPD in different growth phases <strong>of</strong> X. nematophila showed<br />

maximum activity in mid log phase. The enzyme was largely localised in periplasm and<br />

extracellular medium while negligible activity was detected in cytosol. It is also displayed on <strong>the</strong><br />

cell surface <strong>of</strong> <strong>the</strong> bacteria, as treatment with pronase abolished all <strong>the</strong> enzyme activity from <strong>the</strong><br />

outer membrane preparation. Based on <strong>the</strong> crystal structure <strong>of</strong> GlpQ <strong>of</strong> E. coli a homology<br />

model <strong>of</strong> Xenorhabdus GlpQ was created. The residues Glu-59, Asp-61 and His-74 located in


MONDAY<br />

<strong>the</strong> active site <strong>of</strong> <strong>the</strong> enzyme were changed to alanine using site directed mutagenesis. Enzyme<br />

activity was abolished in all <strong>the</strong> three protein variants validating <strong>the</strong> crucial role <strong>of</strong> <strong>the</strong>se residues<br />

in <strong>the</strong> active site <strong>of</strong> <strong>the</strong> enzyme. Since <strong>the</strong> bacteria is exposed to different unfavourable<br />

environments in <strong>the</strong> hosts so we determined glpQ pr<strong>of</strong>ile in nutritional stress conditions.<br />

Transcriptional analysis <strong>of</strong> glpQ specific mRNA has shown upregulation <strong>of</strong> <strong>the</strong> gene in response<br />

to phosphate starvation. Similarly enzyme activity was increased by 1.6 folds when <strong>the</strong> cells<br />

were starved for phosphate upto 7 hours. To fur<strong>the</strong>r analyse it with <strong>the</strong> growth phase <strong>of</strong> bacteria<br />

it was shown that upregulation was mainly associated with <strong>the</strong> early log phase <strong>of</strong> <strong>the</strong> bacteria.<br />

Stationary phase cells did not respond to phosphate starvation. Bioinformatic analysis <strong>of</strong> <strong>the</strong><br />

upstream region <strong>of</strong> glpQ showed presence <strong>of</strong> two promoters. This was confirmed by primer<br />

extension analysis which clearly indicated promoters P1 and P2 about ~80 and ~565<br />

nucleotides upstream <strong>of</strong> <strong>the</strong> start codon <strong>of</strong> <strong>the</strong> gene. To explore <strong>the</strong> function <strong>of</strong> both <strong>the</strong><br />

promoters in phosphate regulation, lacZ fusion constructs were prepared and promoter activity<br />

was determined by β-galactosidase assay which indicated that promoter located 80 bp<br />

upstream <strong>of</strong> <strong>the</strong> start codon has a role in phosphate regulation <strong>of</strong> glpQ gene.<br />

2194<br />

The role <strong>of</strong> a bacterial SMC in chromosome segregation.<br />

M. Schwartz 1 , L. Shapiro 1 ; 1 Developmental <strong>Biology</strong>, Stanford University, Stanford, CA<br />

The bacterial genome must be completely replicated and segregated prior to <strong>the</strong> completion <strong>of</strong><br />

cell division in order to ensure that each daughter cell receives one complement <strong>of</strong> <strong>the</strong> genome.<br />

The Structural Maintenance <strong>of</strong> Chromosomes (SMC) protein is conserved from bacteria to<br />

humans and is thought to play important roles in chromosome organization and segregation.<br />

We have identified a point mutation (SMC-E1076Q) in <strong>the</strong> ATPase domain <strong>of</strong> Caulobacter SMC<br />

that disrupts chromosome segregation and cell division. This mutation caused a dominantnegative<br />

phenotype in which DNA replication was able to proceed, but segregation was<br />

inhibited, resulting in an accumulation <strong>of</strong> parS centromeres at one pole. Surprisingly, DNA was<br />

still deployed throughout <strong>the</strong> cell and <strong>the</strong> cellular positions <strong>of</strong> o<strong>the</strong>r chromosomal loci were in <strong>the</strong><br />

wild-type order relative to <strong>the</strong> parS centromere. Loss <strong>of</strong> ATP hydrolysis, ei<strong>the</strong>r by this point<br />

mutation or by <strong>the</strong> utilization <strong>of</strong> non-hydrolyzable ATP caused abnormally stable binding <strong>of</strong> SMC<br />

to DNA in vitro. We propose that SMC spuriously links <strong>the</strong> duplicated chromosomes<br />

immediately after passage <strong>of</strong> <strong>the</strong> replication fork when <strong>the</strong> two chromosomes are in very close<br />

proximity. In wild type cells, ATP hydrolysis opens <strong>the</strong> SMC dimer, freeing one chromosome to<br />

segregate to <strong>the</strong> opposite pole. SMC is more likely to trap DNA from a single chromosome once<br />

<strong>the</strong> replicated chromosomes are physically separated, <strong>the</strong>reby condensing <strong>the</strong> chromosome. In<br />

this model, <strong>the</strong> loss <strong>of</strong> ATP hydrolysis causes <strong>the</strong> SMC-E1076Q dimer to remain bound to both<br />

chromosomes, inhibiting segregation. The linking <strong>of</strong> <strong>the</strong> two chromosomes can be reinforced by<br />

additional mutant dimers binding elsewhere on <strong>the</strong> neighboring chromosomes.<br />

2195<br />

Binding <strong>of</strong> recombinant soluble Influenza A hemagglutinins to cell surface sialic acids <strong>of</strong><br />

cultured cells and tissue sections.<br />

A-K. Sauer 1 , M. Bohm 1 , C. Schwegmann-Weßels 1 , G. Herrler 1 ; 1 Institute <strong>of</strong> Virology, University<br />

<strong>of</strong> Veterinary Medicine Hannover, Hannover, Germany<br />

The major viral envelope glycoprotein <strong>of</strong> influenza A viruses is <strong>the</strong> hemagglutinin (HA) which is<br />

responsible for host cell recognition and virus entry. It is syn<strong>the</strong>sized as an uncleaved precursor<br />

in <strong>the</strong> host cell secretory pathway where it is posttranslationally modified and forms trimers. The<br />

receptor determinants for influenza A viruses are sialic acids <strong>of</strong> glycolipds and/or glycoproteins.<br />

Avian influenza strains preferentially bind to sialic acids linked to galactose in α2,3-conformation


MONDAY<br />

whereas human strains prefer α2,6-linked sialic acids. To distinguish between both forms, <strong>the</strong><br />

plant lectins MAA and SNA, respectively, are used in many studies. Due to <strong>the</strong> huge diversity <strong>of</strong><br />

oligosaccharide structures and <strong>the</strong> different hemagglutinin subtypes, those two plant lectins are<br />

not sufficient to characterize <strong>the</strong> binding properties <strong>of</strong> influenza hemagglutinins.<br />

To get a closer insight into <strong>the</strong> cellular interaction partners <strong>of</strong> influenza A viruses we generated<br />

soluble hemagglutinins that can be used as lectins for detection <strong>of</strong> those sialoglycoconjugates<br />

that are recognized by influenza viruses. Connection <strong>of</strong> <strong>the</strong> ectodomain to <strong>the</strong> Fc-component <strong>of</strong><br />

a human IgG and to a modified GCN4 leucin zipper motif as trimerization domain resulted in<br />

chimeric proteins that are secreted into cell culture supernatant after transfection and were<br />

purified by FPLC. The binding <strong>of</strong> <strong>the</strong> soluble HAs can be demonstrated by immun<strong>of</strong>luorescencebased<br />

assays..<br />

These recombinant soluble HAs <strong>of</strong> <strong>the</strong> avian subtypes H7 and H9 were found to bind to <strong>the</strong><br />

surface <strong>of</strong> different cell lines, trachea sections <strong>of</strong> chicken and turkey and sections <strong>of</strong> <strong>the</strong> porcine<br />

lung. On all permanent mammalian cell lines tested, <strong>the</strong> H9 shows stronger binding compared<br />

to <strong>the</strong> H7 protein. On trachea sections <strong>of</strong> chicken and turkey a 2fold higher amount <strong>of</strong> H9 is<br />

needed to establish an equal binding pattern compared to that <strong>of</strong> H7 despite <strong>the</strong> presence <strong>of</strong><br />

both sialic acid linkage types as indicated by <strong>the</strong> lectin stain. Porcine lung sections show more<br />

SNA staining on <strong>the</strong> surface whereas MAAII stains cells in basal layers <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium. Only<br />

H9 binds to porcine lung sections but not as widely distributed over <strong>the</strong> surface as <strong>the</strong> SNA<br />

stain.<br />

These experiments show that a lectin staining alone cannot sufficiently depict <strong>the</strong> distribution <strong>of</strong><br />

<strong>the</strong> cellular interaction partners <strong>of</strong> influenza A viruses. Soluble hemagglutinins promise to be a<br />

valuable tool to visualize potential influenza binding sites on cells and tissues.<br />

2196<br />

Adaptation <strong>of</strong> avian influenza viruses <strong>of</strong> <strong>the</strong> subtype H9N2 to avian and human<br />

respiratory epi<strong>the</strong>lial cells.<br />

M. Erdt 1 , M. Bohm 1 , H. Petersen 2 , C. Winter 1,2 , S. Rautenschlein 2 , C. Schwegmann-Weßels 1 , G.<br />

Herrler 1 ; 1 Institute <strong>of</strong> Virology, University <strong>of</strong> Veterinary Medicine Hannover, Foundation,<br />

Hannover, Germany, 2 Clinic for Poultry, University <strong>of</strong> Veterinary Medicine Hannover, Foundation<br />

Influenza viruses initiate infection by binding <strong>of</strong> <strong>the</strong> haemagglutinin (HA) to sialic acid residues<br />

presented by cell surface components. Viruses isolated from different host species may differ in<br />

<strong>the</strong>ir preference for <strong>the</strong> type <strong>of</strong> sialic acid, e.g. N-acetylneuraminic acid (Neu5Ac), or for a<br />

linkage type connecting <strong>the</strong> sialic acid molecule to <strong>the</strong> neighboring sugar. Influenza viruses from<br />

mammalian hosts prefer α-2,6-linked sialic acids whereas most avian influenza viruses<br />

prefentially recognize Neu5Ac attached via an α-2,3-linkage to galactose. Avian viruses <strong>of</strong> <strong>the</strong><br />

H9 subtype are an exception, because several strains <strong>of</strong> H9 influenza viruses have been shown<br />

to favor α-2,6-linked sialic acids. This finding was surprising because in some avian species α-<br />

2,6-linked sialic acids are expressed in very low amounts, at least in <strong>the</strong> respiratory epi<strong>the</strong>lium,<br />

a primary target for influenza virus infection<br />

We analyzed <strong>the</strong> adaptation <strong>of</strong> an egg-grown avian influenza virus <strong>of</strong> <strong>the</strong> subtype H9N2 (strain<br />

A/chicken/SaudiArabia/CP7/1998) to <strong>the</strong> respiratory epi<strong>the</strong>lium <strong>of</strong> chicken and turkeys. Tracheal<br />

organ cultures (TOC) served as a culture system which preserves <strong>the</strong> setting <strong>of</strong> epi<strong>the</strong>lial cells<br />

as present in <strong>the</strong> avian trachea.<br />

The parental virus was passaged four times in ei<strong>the</strong>r chicken or turkey TOCs. The progress <strong>of</strong><br />

infection was monitored by determining <strong>the</strong> viral titer induced by this virus at various time points<br />

post infection. In <strong>the</strong> course <strong>of</strong> <strong>the</strong> passages, <strong>the</strong> virus underwent an adaptation process<br />

indicated by an increase in <strong>the</strong> viral titer. Infection <strong>of</strong> chicken TOCs by <strong>the</strong> chicken TOCadapted<br />

virus resulted in a 3.5-fold increase in <strong>the</strong> viral titer compared to <strong>the</strong> egg-grown virus.


MONDAY<br />

By contrast, infection <strong>of</strong> turkey TOCs with <strong>the</strong> turkey TOC-adapted virus was characterized by a<br />

nearly 10-fold increase in viral titer.<br />

Fur<strong>the</strong>rmore, Calu-3 cells, a permanent human cell line with features <strong>of</strong> <strong>the</strong> bronchiolar<br />

epi<strong>the</strong>lium when grown under air-liquid interface conditions (ALI), was infected with <strong>the</strong>se avian<br />

viruses. The influence <strong>of</strong> several passages in <strong>the</strong>se cells was monitored and analysis <strong>of</strong> cell<br />

culture supernatants showed that avian H9N2 viruses in Calu-3 cells reached higher titers than<br />

in <strong>the</strong> TOC system. In <strong>the</strong> future <strong>the</strong>se experiments should be extended to primary human<br />

respiratory epi<strong>the</strong>lial cells.<br />

2197<br />

Peritoneal Macrophages Employ Predominantly αxβ2 But Not αMβ2 For Candida albicans<br />

Recognition.<br />

S. Jawhara 1 , D. Soloviev 1 ; 1 <strong>Molecular</strong> Cardiology, Cleveland Clinic Lerner Research Institute,<br />

Cleveland, OH<br />

Candida albicans is an opportunistic yeast pathogen that represents <strong>the</strong> fourth most frequent<br />

cause <strong>of</strong> nosocomial bloodstream infections in <strong>the</strong> United States. Recently, we demonstrated<br />

that <strong>the</strong> integrin αMβ2 is <strong>the</strong> principal leukocyte receptor for <strong>the</strong> filamentous form <strong>of</strong> C. albicans.<br />

In <strong>the</strong> present study, we explored <strong>the</strong> role <strong>of</strong> ano<strong>the</strong>r β2 integrin, αxβ2 (CD11c/CD18, p150,95),<br />

in <strong>the</strong> model <strong>of</strong> antifungal defenses. Although αxβ2 is highly homologous to <strong>the</strong> αMβ2, its role<br />

remains unclear in <strong>the</strong> immune response to fungal infections. Employing αx-knock out (KO)<br />

mice in <strong>the</strong> murine model <strong>of</strong> systemic candidiasis, we first demonstrated that mice deficient in<br />

αxβ2 had significantly increased susceptibility to <strong>the</strong> invasive fungal infection. We observed that<br />

C. albicans induced mortality in αx-KO mice more rapidly (2- to 3-fold faster) than when <strong>the</strong><br />

same inoculum was used in wild-type mice. Notably, <strong>the</strong> fungal burden was significantly higher<br />

in both <strong>the</strong> brain and kidneys <strong>of</strong> αx-KO than wild-type mice. Subsequently, <strong>the</strong> histopathology<br />

examination <strong>of</strong> both kidneys and brain from C. albicans-infected αx-KO mice showed a higher<br />

presence <strong>of</strong> filamentous growth and tissue damage than in wild-type mice. Elimination <strong>of</strong> αxβ2<br />

significantly impedes peritoneal macrophages recruitment to <strong>the</strong> place <strong>of</strong> infection and <strong>the</strong>ir<br />

capacity for anti-fungal activity, while having virtually no effect on <strong>the</strong> neutrophil`s function. In<br />

contrast, αM-elimination mostly affected <strong>the</strong> antifungal activity <strong>of</strong> neutrophils but not that <strong>of</strong><br />

peritoneal macrophages. Additionally, αxβ2 elimination increased <strong>the</strong> expression <strong>of</strong> interleukin-<br />

6, but not <strong>the</strong> tumor necrosis factor alpha (TNFα). Taken toge<strong>the</strong>r, <strong>the</strong>se data indicate that αxβ2<br />

is essential for C. albicans elimination by macrophages and plays a role in innate antifungal<br />

immunity.<br />

2198<br />

The role <strong>of</strong> TrpM7 in Salmonella pathogenesis.<br />

A. M. Young 1 , J. E. McCombs 1 , S. Van Engelenburg 1 , W. W. Blakeslee 1 , A. E. Palmer 1 ;<br />

1 Chemistry and Biochemistry, University <strong>of</strong> Colorado Boulder, Boulder, CO<br />

Salmonella subspecies are enteropathogenic gram-negative bacteria responsible for food-borne<br />

illness in humans ranging from gastroenteritis to typhoid fever. The infection <strong>of</strong> a range <strong>of</strong> host<br />

cells by Salmonella is driven by <strong>the</strong> injection <strong>of</strong> bacterial effector proteins through a needle-like<br />

type III secretion system (T3SS). Effector proteins hijack host cell machinery and intracellular<br />

signaling pathways. These interactions promote invasion <strong>of</strong> <strong>the</strong> cell through cytoskeleton<br />

rearrangement and <strong>the</strong> engulfment <strong>of</strong> <strong>the</strong> bacteria into a Salmonella containing vacuole (SCV).<br />

Salmonella invasion is known to induce an elevation in host cell Ca 2+ , however <strong>the</strong> molecular<br />

mechanism <strong>of</strong> this event has remained undefined. Fluctuations in Ca 2+ activate a diverse array<br />

<strong>of</strong> downstream signaling processes including gene transcription, actin polymerization, and<br />

endocytosis. Given <strong>the</strong>se roles, it is likely that Salmonella manipulates and utilizes host cell Ca 2+


MONDAY<br />

signaling during invasion. In this work, we investigate <strong>the</strong> origin and nature <strong>of</strong> <strong>the</strong> Ca2 + elevation<br />

by examining Salmonella invasion <strong>of</strong> non-phagocytic cells at <strong>the</strong> single cell level. Localized Ca 2+<br />

elevation was observed to result from influx through a transient receptor potential cation channel<br />

(TRPM7) at sites <strong>of</strong> invasion. This study identifies a novel mechanism by which TRPM7 is<br />

activated by a Salmonella effector, SopB, through its ability to modulate phosphoinositides,<br />

indicating that TRPM7 can be gated by rare signaling phospholipids.<br />

2199<br />

Defining <strong>the</strong> roles <strong>of</strong> inclusion membrane microdomain proteins in chlamydial<br />

pathogenesis.<br />

J. Mital 1 , E. Lutter 1 , T. Hackstadt 1 ; 1 Rocky Mountain Labs/NIH, Hamilton, MT<br />

Chlamydia trachomatis is <strong>the</strong> leading cause <strong>of</strong> sexually transmitted disease in <strong>the</strong> Western<br />

world and <strong>of</strong> infectious blindness in <strong>the</strong> world. Chlamydiae are obligate intracellular bacteria that<br />

reside within a membrane bound vacuole termed <strong>the</strong> inclusion, which does not fuse with <strong>the</strong><br />

endosomal pathway but selectively interacts with vesicular pathways to acquire lipids and<br />

nutrients from <strong>the</strong> host cell. The inclusion is extensively modified by insertion <strong>of</strong> chlamydial<br />

proteins (Incs), which are ideally positioned to mediate host-pathogen interactions. There is little<br />

functional information regarding Inc proteins. Once internalized, <strong>the</strong> nascent inclusion<br />

repositions itself at <strong>the</strong> centrosome in a process that is dependent upon chlamydial protein<br />

syn<strong>the</strong>sis, microtubules and dynein. Inclusion repositioning is independent <strong>of</strong> <strong>the</strong> cargo linker<br />

p50 dynamitin, indicating that a bacterial effector may directly link <strong>the</strong> inclusion to dynein. We<br />

previously described a subset <strong>of</strong> Incs (B, 101, 222, 850) that colocalize with host Src-family<br />

kinases in microdomains in <strong>the</strong> inclusion membrane. These microdomains are closely apposed<br />

to host centrosomes, indicating a probable role in microtubule interaction. Knockout and<br />

inhibitor studies show that Src family kinases are involved in <strong>the</strong> microtubule dependent<br />

processes <strong>of</strong> inclusion repositioning, sphingomyelin trafficking and are essential for normal<br />

chlamydial development. Here we report studies <strong>of</strong> <strong>the</strong> functional roles <strong>of</strong> <strong>the</strong> microdomain Incs<br />

in chlamydial pathogenesis. When ectopically expressed in HeLa cells, Inc850 consistently<br />

colocalizes with host centrosomes. Inc850 contains a dynein light chain binding domain and has<br />

been shown by yeast two-hybrid assays to interact with dynein light chain subunits. This<br />

suggests that inclusion microdomains comprised <strong>of</strong> chlamydial and host proteins are directly<br />

involved in microtubule dependent trafficking <strong>of</strong> <strong>the</strong> nascent chlamydial inclusion. The full role <strong>of</strong><br />

<strong>the</strong>se microdomains in pathogenic processes remains to be elucidated.<br />

2200<br />

The PDZ1 domain <strong>of</strong> MAGI-1 rescues CAR Ex8 from MAGI-1 mediated degradation and<br />

augments adenoviral infection.<br />

A. O. Kolawole 1 , R. Yan 1 , P. Sharma 1 , K. J. Exc<strong>of</strong>fon 1 ; 1 Biological Sciences, Wright State<br />

University, Dayton, OH<br />

A major factor in virus entry into cells is localization and abundance <strong>of</strong> <strong>the</strong> primary receptor. The<br />

Coxsackievirus and adenovirus receptor (CAR) is <strong>the</strong> primary receptor for group B<br />

coxsackievirus and many serotypes <strong>of</strong> adenovirus. In most epi<strong>the</strong>lia, a seven exon is<strong>of</strong>orm <strong>of</strong><br />

CAR (CAR Ex7 ) is exclusively localized at <strong>the</strong> basolateral surface where it behaves as a<br />

homophilic adhesion protein and is inaccessible for viral infection. However, in welldifferentiated<br />

human airway epi<strong>the</strong>lia, we recently discovered an alternatively spliced, low<br />

abundance eight exon is<strong>of</strong>orm <strong>of</strong> CAR (CAR Ex8 ) that is localized apically, where it may initiate<br />

apical viral infection. The two is<strong>of</strong>orms differ only in <strong>the</strong> last 26 (CAR Ex7 ) or 13 (CAR Ex8 ) amino<br />

acids <strong>of</strong> <strong>the</strong> cytoplasmic domain, which suggests that some intracellular interactions may differ.<br />

One such differential interaction involves MAGI-1, an essential PDZ domain containing protein


MONDAY<br />

with 6 distinct PDZ domains (PDZ0-5), known to be involved in cell polarization and cancer. The<br />

molecular basis <strong>of</strong> <strong>the</strong> CAR-MAGI-1 interaction was investigated by adenovirus infection,<br />

immunocytochemistry, MAGI-1 siRNA knockdown, yeast two hybrid assay, coimmunoprecipitation,<br />

in vitro translation, and fluorescence resonance energy transfer. In nonpolarized<br />

cells, MAGI-1 caused a loss <strong>of</strong> CAR Ex8 at <strong>the</strong> protein level and reduced adenovirus<br />

infection. In contrast, MAGI-1 siRNA knockdown increased adenovirus infection, suggesting that<br />

endogenous MAGI-1 regulates CAR Ex8 levels. The key PDZ-based interactions between CAR<br />

and MAGI-1 were <strong>the</strong>n identified and confirmed. Both CAR is<strong>of</strong>orms strongly interacted with<br />

PDZ3. CAR Ex8 also interacted with PDZ1. Co-expression <strong>of</strong> CAR Ex8 with isolated MAGI-1 PDZ3<br />

significantly reduced CAR Ex8 cell surface expression and adenovirus infection but did not reduce<br />

total CAR Ex8 protein levels. These data suggest that MAGI-1 PDZ3 is responsible for retaining<br />

CAR Ex8 within <strong>the</strong> cell but degradation <strong>of</strong> CAR Ex8 likely requires MAGI-1 interacting<br />

proteins/domains.Co-expression <strong>of</strong> <strong>the</strong> isolated PDZ1 domain <strong>of</strong> MAGI-1 with CAR Ex8 did not<br />

affect adenovirus infection. However, PDZ1 was able to rescue CAR Ex8 from MAGI-1-mediated<br />

degradation, <strong>the</strong>reby supporting adenovirus infection via CAR Ex8 in <strong>the</strong> presence <strong>of</strong> MAGI-1.<br />

These data suggest that molecules directed at facilitating or inhibiting <strong>the</strong> interaction between<br />

PDZ1 and CAR Ex8 may be able to regulate <strong>the</strong> levels <strong>of</strong> CAR Ex8 and hence increase or decrease<br />

epi<strong>the</strong>lial susceptibility to adenoviral infection.<br />

2201<br />

The VP1 Subunit <strong>of</strong> JC Polyomavirus Drives Trafficking to <strong>the</strong> ER for Productive Viral<br />

Infection.<br />

A. Derdowski 1 , C. Nelson 1 , M. Maginnis 1 , B. O'Hara 1 , W. Atwood 1 ; 1 <strong>Molecular</strong> <strong>Biology</strong>, Cell<br />

<strong>Biology</strong> and Biochemistry, Brown University, Providence, RI<br />

There are at least nine different human polyomaviruses. Two <strong>of</strong> <strong>the</strong>se, JC polyomavirus (JCV)<br />

and BK polyomavirus (BKV), are long-established as human pathogens in immunocompromised<br />

hosts causing progressive multifocal leukoencephalopathy (PML) and nephropathy,<br />

respectively. The remaining human polyomaviruses have all been discovered within <strong>the</strong> last<br />

decade, establishing this class <strong>of</strong> viruses as emerging opportunistic pathogens. Polyomaviruses<br />

are coated with 72 pentamers <strong>of</strong> <strong>the</strong> viral attachment protein VP1. VP1 is responsible for<br />

attachment to <strong>the</strong> host cell surface and implicated in trafficking to <strong>the</strong> ER and nucleus for<br />

productive infection. The VP1 subunit <strong>of</strong> JCV was bacterially expressed and purified by FPLC.<br />

VP1 pentamers were <strong>the</strong>n directly labeled and viral entry and trafficking were analyzed by<br />

confocal microscopy in real-time. We demonstrate for <strong>the</strong> first time that <strong>the</strong> JCV VP1 pentameric<br />

subunit drives viral trafficking through Rab 5 and 7-positive vesicles to <strong>the</strong> ER for productive<br />

infection, and interference <strong>of</strong> this trafficking with pharmacological inhibitors reduces infection.<br />

This work not only highlights <strong>the</strong> importance <strong>the</strong> viral VP1 pentamer in polyomavirus trafficking,<br />

but also establishes a viral trafficking model for emerging polyomaviruses that lack cell culture<br />

systems.<br />

2202<br />

Inhibition <strong>of</strong> Rab5 activation by Exotoxin S <strong>of</strong> Pseudomonas aeruginosa.<br />

S. Mustafi 1 , N. Rivero 1 , M. Barbieri 1 ; 1 Florida International University, Miami, FL<br />

Pseudomonas aeruginosa is a human pathogen that frequently causes severe hospital-acquired<br />

infections. Exotoxins secreted from Type III secretion system <strong>of</strong> Pseudomonas aeruginosa are<br />

critical molecules to disrupt mammalian epi<strong>the</strong>lial cell membrane trafficking and signaling. One<br />

<strong>of</strong> <strong>the</strong>m also known as Exotoxin S, is a bi functional protein which shows ADP ribosyltransferase<br />

activity and targets several intracellular proteins, including Ras and Rab small GTPases. Here,<br />

we demonstrate that Rab5, but not Rab4 and Rab7, play a critical role during early steps <strong>of</strong> <strong>the</strong>


MONDAY<br />

invasion <strong>of</strong> Pseudomonas aeruginosa. We show that invasion <strong>of</strong> live Pseudomonas aeruginosa,<br />

but not heat inactivated Pseudomonas aeruginosa, down-regulates <strong>of</strong> Rab5 activity in<br />

macrophages. In addition, invasion <strong>of</strong> live Pseudomonas aeruginosa lacking Exotoxin S was<br />

unable to inactivate Rab5. Fur<strong>the</strong>rmore, we also found that overexpression <strong>of</strong> Rin1 partially<br />

reverses <strong>the</strong> inactivation <strong>of</strong> Rab5 during invasion <strong>of</strong> live Pseudomonas aeruginosa. These<br />

observations are consistent with a model, in which <strong>the</strong> presence <strong>of</strong> Exotoxin S may interfere<br />

with <strong>the</strong> Rab5 cycle activity in macrophage<br />

2203<br />

The ATPase activity <strong>of</strong> EccC is required for ESX-1 secretion in Mycobacterium<br />

tuberculosis.<br />

D. Dovala 1 , O. Rosenberg 1 , J. Cox 1 ; 1 University <strong>of</strong> California, San Francisco, San Francisco, CA<br />

The human pathogen Mycobacterium tuberculosis (Mtb) utilizes a specialized type VII secretion<br />

system, ESX-1, to circumvent host defenses during macrophage infection. ESX-1 mutants are<br />

severely attenuated, highlighting <strong>the</strong> importance <strong>of</strong> this secretion system. While many<br />

substrates and components <strong>of</strong> ESX-1 have been identified, <strong>the</strong> mechanistic details <strong>of</strong> how <strong>the</strong>se<br />

substrates are pumped into <strong>the</strong> host cell remain elusive. The Mtb proteins EccCa and EccCb<br />

contain several putative FtsK-like ATPase domains and form a complex which is required for<br />

ESX-1 secretion. FtsK domains in o<strong>the</strong>r proteins are known to exert force on substrates, and<br />

thus may serve as <strong>the</strong> pump in <strong>the</strong> ESX-1 translocon. We show that <strong>the</strong> EccC complex forms an<br />

ATPase with three functional ATPase domains. We show that all ATPase domains are required<br />

for successful secretion <strong>of</strong> ESX-1 substrates from Mtb, and genetic approaches have identified<br />

<strong>the</strong> roles <strong>of</strong> each domain in complex formation and substrate binding.<br />

2204<br />

Host Innate Receptor Regulation is Mediated by Influenza Virus Proteins.<br />

D. J. Vigerust 1,2 , V. L. Shepherd 1,2 ; 1 VanderbIlt University SOM, Nashville, TN, 2 Department <strong>of</strong><br />

Veteran Affairs, Nashville, TN<br />

Influenza virus A (IVA) infection is a significant cause <strong>of</strong> morbidity and mortality each year.<br />

Interactions between <strong>the</strong> influenza virus and cells <strong>of</strong> <strong>the</strong> innate immune system – specifically<br />

macrophages – may exacerbate acute symptomology and immune evasion and disease<br />

outcome, but <strong>the</strong> interaction between influenza and <strong>the</strong> macrophage remains poorly understood.<br />

Host-pathogen dynamics may prevent <strong>the</strong> proper function <strong>of</strong> <strong>the</strong> macrophage in <strong>the</strong> clearance <strong>of</strong><br />

harmful pathogens. Our current hypo<strong>the</strong>sis is that host innate defense molecules such as <strong>the</strong><br />

cell surface mannose receptor (MR) may be utilized as a receptor <strong>of</strong> entry for viruses such as<br />

influenza, and that molecules such as MR and MHC-I are targeted for down-regulation by<br />

influenza A viruses leading to dysregulation <strong>of</strong> macrophage function. In previous work from our<br />

laboratory <strong>the</strong> HIV-1 Nef protein was shown to interact with <strong>the</strong> MR tail to down-regulate <strong>the</strong><br />

surface receptor expression. The interaction between <strong>the</strong> MR and Nef was localized to two<br />

specific regions <strong>of</strong> Nef. Sequence comparison <strong>of</strong> <strong>the</strong> known interacting domains <strong>of</strong> <strong>the</strong> HIV-1<br />

Nef protein and <strong>the</strong> sequence <strong>of</strong> influenza A NS1 and PB1 revealed similarity in several motifs<br />

that indicate a potential for MR regulation by influenza. Our initial data suggest that infection <strong>of</strong><br />

MR-expressing cells results in a significant decrease in surface expression <strong>of</strong> MR by 48 hours<br />

post-infection. Fur<strong>the</strong>r, in vitro transient transfection <strong>of</strong> macrophages with influenza proteins<br />

leads to removal <strong>of</strong> MR from <strong>the</strong> surface at levels approaching 30% for NS1 and PB1. Removal<br />

<strong>of</strong> MHC-I from <strong>the</strong> cell surface was also seen as early as 5 hours post-transfection. These data<br />

support <strong>the</strong> hypo<strong>the</strong>sis that influenza proteins regulate two important host innate defense<br />

molecules in a fashion that is similar to that seen with HIV-1 Nef.


New Technologies and Frontiers<br />

MONDAY<br />

2205<br />

High-throughput cellular assays using a well-less plate format.<br />

G. Quinones 1 , K. Nicholes 1 , M. Lye 2 , N. Kim 2 , J-P. Stephan 1 ; 1 Genentech, South San Francisco,<br />

CA, 2 Curiox Biosystems, Singapore<br />

Cellular assays represent a great opportunity for researchers to test various molecules in a<br />

more biologically relevant context than biochemical assays. Many <strong>of</strong> <strong>the</strong>se cell-based assays<br />

could be adapted for high-throughput and high content screening assays, providing<br />

investigators <strong>the</strong> opportunity to interrogate large number <strong>of</strong> samples and conditions. Despite<br />

significant technical advances made during <strong>the</strong> last few years, high-throughput/high content<br />

cellular assays still suffer from key limitations in working with a large number and variety <strong>of</strong> cell<br />

lines. A key limitation is <strong>the</strong> use <strong>of</strong> suspension cell lines, especially when multi-step staining<br />

procedures are required. Here, we demonstrate that <strong>the</strong> use <strong>of</strong> a well-less plate system which<br />

significantly improves <strong>the</strong> flexibility <strong>of</strong> our high content screening platform. The well-less format<br />

utilizes surface tension to maintain <strong>the</strong> cell population on <strong>the</strong> glass surface <strong>of</strong> plate in 2.5mm<br />

diameter drops, while a gentle buffer exchange allows <strong>the</strong> cells to remain on <strong>the</strong> plate surface<br />

throughout a variety <strong>of</strong> experimental procedures. We performed cell viability experiments in <strong>the</strong><br />

presence <strong>of</strong> <strong>the</strong> antimitotic agent monomethyl auristatin E (MMAE) on <strong>the</strong> suspension cell lines<br />

U-937 and K-562 in immun<strong>of</strong>luorescence and chemiluminescence viability assays, showing that<br />

in both assays we achieve an EC50 <strong>of</strong> 4.4uM and 9.1uM respectively. We also performed<br />

immun<strong>of</strong>luorescence-based, protein-protein interaction assays using a large library <strong>of</strong> single<br />

transmembrane, multi-transmembrane and secreted proteins expressed in COS7 cells. The<br />

expressed library was incubated with tagged bait proteins to interrogate novel protein-protein<br />

interactions at <strong>the</strong> cell surface. This resulted in <strong>the</strong> successful expression and binding <strong>of</strong> several<br />

known ligands to <strong>the</strong>ir respective receptors, i.e. PD-1 binding to PD-L1, NGF to NGFR, PVRL2<br />

to PVRIG, and HVEM binding to BTLA. We also examined changes in cell morphology using<br />

both established and primary human cell lines infected with fluorescent organelle-specific<br />

baculovirus constructs. Live imaging <strong>of</strong> <strong>the</strong> infected cells revealed alterations in <strong>the</strong> trafficking<br />

patterns <strong>of</strong> EEA1-labeled early endosomes in <strong>the</strong> presence <strong>of</strong> <strong>the</strong> dynamin inhibitor Dynasore.<br />

We also observed changes in <strong>the</strong> actin and tubulin cytoskeletal structure in <strong>the</strong> presence <strong>of</strong><br />

Cytochalasin D and Blebbistatin. The added flexibility in terms <strong>of</strong> cell lines and readouts enables<br />

<strong>the</strong> screening platform to truly perform high-throughput and high content assays to examine a<br />

wide variety <strong>of</strong> cellular processes.<br />

2206<br />

Solutions for <strong>the</strong> preparation <strong>of</strong> samples for integrated correlative microscopy.<br />

M. A. Karreman 1,2 , A. V. Agronskaia 1 , E. G. Van Donselaar 2 , K. E. Vocking 2 , H. C. Gerritsen 1 ;<br />

1 <strong>Molecular</strong> Biophysics, Utrecht University, Utrecht, Ne<strong>the</strong>rlands, 2 Biomolecular Imaging, Utrecht<br />

University, Utrecht, Ne<strong>the</strong>rlands<br />

The Integrated Laser and Electron Microscope is a novel tool for correlative microscopy,<br />

combining a fluorescence microscope (FM) and a Transmission Electron Microscope (TEM)<br />

within one set-up. Here, a specially designed laser scanning fluorescence microscope is<br />

mounted into one <strong>of</strong> <strong>the</strong> side ports <strong>of</strong> a conventional Tecnai 12 TEM. First, <strong>the</strong> region <strong>of</strong> interest<br />

is identified based on fluorescence labeling <strong>of</strong> <strong>the</strong> sample. This region can <strong>the</strong>n effortlessly be<br />

relocated in <strong>the</strong> TEM and subsequently investigated at high resolution. Due to <strong>the</strong> integrated<br />

nature <strong>of</strong> this set-up, <strong>the</strong> sample (eg. sections <strong>of</strong> cells on a grid) needs to be suitable both for<br />

FM and TEM. Unfortunately, <strong>the</strong> heavy metal stain employed to contrast <strong>the</strong> biological material<br />

for TEM has a quenching affect on <strong>the</strong> fluorophore required for FM imaging. Fur<strong>the</strong>rmore, most


MONDAY<br />

fluorophores are optimized for an aqueous environment and lose a large part <strong>of</strong> <strong>the</strong>ir brightness<br />

in a dry environment, which is needed for <strong>the</strong> TEM.<br />

Here, we demonstrate an optimized sample preparation protocol for iLEM. By creating a<br />

distance between <strong>the</strong> metal stain and <strong>the</strong> fluorophore, <strong>the</strong> quenching effect can be prevented.<br />

This is achieved by performing a block staining, so <strong>the</strong> biological material in <strong>the</strong> section is<br />

already contrasted. The fluorescent label on <strong>the</strong> section is <strong>the</strong>n physically separated from <strong>the</strong><br />

stain in <strong>the</strong> section. We show that <strong>the</strong> intensity and <strong>the</strong> lifetime <strong>of</strong> <strong>the</strong> fluorophore are not<br />

affected by <strong>the</strong> heavy metal stain bound to <strong>the</strong> biological material in <strong>the</strong> section. Fur<strong>the</strong>rmore,<br />

<strong>the</strong> choice <strong>of</strong> fluorophores for iLEM imaging is critical. Although in wet conditions Alexa 488<br />

fluoresces brighter than <strong>the</strong> TRITC dye, in a dry environment <strong>the</strong> opposite is <strong>the</strong> case. We have<br />

found that <strong>the</strong> lifetime and intensity <strong>of</strong> Alexa 488 is severely reduced in a dry environment<br />

whereas <strong>the</strong> TRITC dye is unaffected.<br />

Therefore, we can conclude that <strong>the</strong> sample preparation can be optimized by physically<br />

separating <strong>the</strong> heavy metal stain from <strong>the</strong> fluorophore. Also, <strong>the</strong> choice <strong>of</strong> fluorophores is <strong>of</strong> high<br />

importance due to <strong>the</strong>ir different properties in a wet or dry environment.<br />

2207<br />

Imaging promoter activity with Intracellular MultiAptamer Genetic tag (IMAGEtag).<br />

J. Ray 1 , I. Shin 1 , M. Zhao 2 , M. Ilgu 1 , V. Gupta 3 , J. Beasley 3 , L. Peng 2 , G. Kraus 3 , M. Nilsen-<br />

Hamilton 1 ; 1 Biochemistry, Biophysics and <strong>Molecular</strong> <strong>Biology</strong>, Iowa State University, Ames, IA,<br />

2 Optical Sciences, The University <strong>of</strong> Arizona, Tucson, AZ, 3 Chemistry, Iowa State University,<br />

Ames, IA<br />

A detailed understanding <strong>of</strong> <strong>the</strong> processes involved in gene transcription is necessary to answer<br />

many fundamental biological questions. Imaging techniques have been developed in <strong>the</strong> last<br />

decade with <strong>the</strong> capability <strong>of</strong> exploring <strong>the</strong> elements <strong>of</strong> gene expression and monitoring <strong>the</strong><br />

fates <strong>of</strong> selected mRNAs. In this study we describe an aptamer-based system to monitor<br />

promoter activity in living cells and in real time, which we have named as IMAGEtag<br />

(Intracellular MultiAptamer Genetic tag). We have cloned and expressed a string <strong>of</strong> multiaptamers<br />

downstream <strong>of</strong> <strong>the</strong> GAL1 promoter in Saccharomyces cerevisiae and have incubated<br />

<strong>the</strong>se cells with fluorescent-labeled aptamer target molecules followed by induction <strong>of</strong> <strong>the</strong><br />

promoter. This technology uses FRET (Förster resonance energy transfer) to tell us that when<br />

<strong>the</strong> promoter is active and provides a dynamic transcriptional pr<strong>of</strong>ile <strong>of</strong> <strong>the</strong> cells. In this work we<br />

have used three different types <strong>of</strong> multiaptamers (viz. PDC aptamer, tobramycin aptamer and<br />

neomycin aptamer) with <strong>the</strong>ir respective ligands, which have been individually labeled with cy3<br />

and cy5 dyes. With this noninvasive technology we are able to measure gene expression in real<br />

time <strong>of</strong> individual living cells. This allows an evaluation <strong>of</strong> <strong>the</strong> intercellular variation in gene<br />

expression. We have also used this method to measure <strong>the</strong> rate <strong>of</strong> transcriptional elongation.<br />

The IMAGEtag system has <strong>the</strong> potential <strong>of</strong> being used to track <strong>the</strong> real-time levels <strong>of</strong> gene<br />

expression at <strong>the</strong> cellular level in a broad range <strong>of</strong> cell types.


MONDAY<br />

2208<br />

In Situ Microscopic Visualization and Relative Quantification <strong>of</strong> Inorganic Polyphosphate<br />

Stores by 4',6-Diamidino-2-Phenylindole (DAPI)-stainning.<br />

F. M. Gomes 1,2 , I. Ramos 1,2 , W. Girard-Dias 2 , E. Machado 1,3 , K. Miranda 2,3 ; 1 Laboratorio de<br />

Entomologia Medica, Instituto de Bi<strong>of</strong>ísica Carlos Chagas Filhos, UFRJ, Rio de Janeiro, Brazil,<br />

2 Laboratorio de Ultraestrutura Celular Hertha Meyer, Instituto de Bi<strong>of</strong>isica Carlos Chagas Filho,<br />

UFRJ, Rio de Janeiro, Brazil, 3 Laboratorio de Biotechnologia, Diretoria de Programas,<br />

INMETRO, Rio de Janeiro, Brazil<br />

Inorganic polyphosphate (polyP) is a widely distributed biological polymer composed <strong>of</strong><br />

phosphate residues linked by phosphoanhydride bonds. They play key biological roles such as<br />

phosphate and energy reservoir, metal homeostasis, regulation <strong>of</strong> transcription factors and<br />

transcription fidelity, as well as regulation <strong>of</strong> several enzyme activities [1]. Never<strong>the</strong>less, polyP<br />

has remained poorly studied mainly due to <strong>the</strong> limited methods for polyP quantification and in<br />

situ visualization. 4',6-diamidino-2-phenylindole (DAPI) is a commonly used nuclei stain that<br />

binds to double stranded DNA, exhibiting a fluorescence emission maximum around 450 nm.<br />

Interestingly, it has been shown that polyP is able to shift DAPI emission maximum to a higher<br />

wavelength around 525-550 nm displaying a greenish-yellowish color distinct from <strong>the</strong> blue<br />

nuclei-signals [2]. This DAPI-polyP interaction has been shown to be specific and to generate<br />

proper quantum yield for microscopic observation. Never<strong>the</strong>less, although a few studies have<br />

focused on <strong>the</strong> spectroscopic properties <strong>of</strong> analytical grade sodium polyphosphate-DAPI<br />

fluorescence, <strong>the</strong>re are no consistent reports concerning methods <strong>of</strong> polyP visualization using<br />

DAPI. In addition, <strong>the</strong> lack <strong>of</strong> a general protocol for polyP staining hinders its wider utilization as<br />

a polyP-visualization tool. In <strong>the</strong> present study, we report a systematic evaluation <strong>of</strong> different<br />

protocols <strong>of</strong> DAPI-staining for <strong>the</strong> detection <strong>of</strong> polyP stores in different biological models.<br />

Suspensions <strong>of</strong> subcellular fractions <strong>of</strong> insect eggs and parasites containing polyP granules<br />

showed a clean DAPI-polyP fluorescence after brief incubation with DAPI, without previous<br />

treatments. Staining <strong>of</strong> Poly P stores in intact cells usually required aldehyde fixation and/or<br />

detergent permeabilization, as exemplified in DAPI-polyP fluorescence obtained from Eimeria<br />

parasites. DAPI-polyP signals were also detected from cryosections <strong>of</strong> mild-fixed, OCTembedded<br />

midgut <strong>of</strong> insects. In addition, a DAPI-polyP fluorimetric method was applied to<br />

quantify polyP mobilization during early egg development <strong>of</strong> <strong>the</strong> insect Rhodnius prolixus. The<br />

method was validated by <strong>the</strong> parallel measurement using a more widely accepted polyP<br />

quantification protocol that uses a recombinant yeast exopolyphosphatase. Taken toge<strong>the</strong>r, our<br />

results strongly support <strong>the</strong> broader use <strong>of</strong> DAPI as a valuable tool for both polyP visualization<br />

and quantification in different biological models. Never<strong>the</strong>less, one should take into account that<br />

sample preparation may vary according to <strong>the</strong> model used and that care should be taken when<br />

localizing and quantifying polyP from different biological models.<br />

[1] Rao, N.N., M.R. Gómez-García, and A. Kornberg, Inorganic polyphosphate: essential for<br />

growth and survival. Annual review <strong>of</strong> biochemistry, 78, 605-647 (2009) .<br />

[2] Aschar-Sobbi, R., et al., High Sensitivity, Quantitative Measurements <strong>of</strong> Polyphosphate<br />

Using a New DAPI-Based Approach. Journal <strong>of</strong> Fluorescence, 18 (5), 859-866 (2008).


MONDAY<br />

2209<br />

Teachable tool for standardization <strong>of</strong> human induced pluripotent stem cell colony<br />

selection from live cell microscopy image sequences.<br />

S-J. J. Lee 1 , H. Lai 1 , W. T. Hendriks 2 , Z. A. Kenyon 1 , S. V. Alworth 1 , C. Nakada 3 , L. Daheron 4 ,<br />

Y. Kiyota 3 , L. L. Rubin 2 , C. A. Cowan 2 ; 1 DRVision Technologies LLC, Bellevue, WA,<br />

2 Department <strong>of</strong> Stem Cell and Regenerative <strong>Biology</strong>, Harvard Stem Cell Institute, Cambridge,<br />

MA, 3 Instruments Company, Nikon, Yokohama-city, Kanagawa,Japan 4 Harvard University,<br />

Harvard Stem Cell Institute, Cambridge, MA<br />

In a 2006 breakthrough study, Yamanaka and Takahashi proved that somatic cells could be<br />

reprogrammed to an embryonic-like state using four transcription factors. The induced<br />

pluripotent stem cell (iPSC) field has since progressed at a remarkable pace impacting basic<br />

research, drug discovery, bioproduction, drug screening, personalized medicine and cell<br />

<strong>the</strong>rapy. One <strong>of</strong> <strong>the</strong> key challenges facing <strong>the</strong> industry is that iPSC, or iPSC-like colony<br />

selection remains technically challenging, and thus state-<strong>of</strong>-<strong>the</strong>-art protocols in a broad range <strong>of</strong><br />

iPSC applications are <strong>of</strong>ten <strong>of</strong> low yield and un-predictable quality. Tools and techniques for <strong>the</strong><br />

standardization <strong>of</strong> colony selection in real time systems, such as microscopy incubators are<br />

particularly valuable, to reduce <strong>the</strong> variability <strong>of</strong> outcomes and lower costs. In particular, tools<br />

that can predict future outcomes are useful because it allows productive colonies to be selected.<br />

We developed a teachable image recognition tool that can be taught to perform <strong>the</strong><br />

quantitative analysis and classification <strong>of</strong> phase contrast and fluorescence images<br />

automatically. To enable <strong>the</strong> standardization <strong>of</strong> iPSC colony selection we created an image<br />

analytic by teaching. The analytic performs fully automated colony detection, colony feature<br />

measurement and iPSC colony classification in human iPSCs. Importantly, <strong>the</strong> classification<br />

step in <strong>the</strong> analytic is updateable by teaching. This allows <strong>the</strong> tool to be iteratively improved<br />

over time as <strong>the</strong> assay protocol is improved and colony true outcomes are determined by<br />

monitoring <strong>the</strong> complete re-programming process.<br />

The objective <strong>of</strong> this initial study was to validate our tool for <strong>the</strong> prediction <strong>of</strong> human<br />

iPSC colony outcomes. Registered images covering <strong>the</strong> whole well were acquired on <strong>the</strong> Nikon<br />

BioStation CT and stitched in our tool to create a large (~20,000 x 20,000 pixels) composite<br />

image sequence <strong>of</strong> <strong>the</strong> whole well include phase constrast and a viral-GFP construct that is<br />

silenced upon full reprogramming. We validated <strong>the</strong> tool by predicting week four outcomes using<br />

image frames up to week three. Ground truth is established by a loss <strong>of</strong> <strong>the</strong> GFP reporter. Our<br />

hypo<strong>the</strong>sis is that <strong>the</strong> prediction using images up to week three can accurately predict each<br />

colonies' week four outcome as quantified by a loss in GFP fluorescence. We test <strong>the</strong><br />

hypo<strong>the</strong>sis by assessing <strong>the</strong> sensitivity and specificity <strong>of</strong> <strong>the</strong> week three prediction accuracy.<br />

We conclude that <strong>the</strong> hypo<strong>the</strong>sis is supported with high prediction accuracy. Future studies will<br />

look towards real time colony selection and future outcome (e.g. differentiation) prediction.<br />

2210<br />

The Use <strong>of</strong> Discrete Polyethylene Glycol Linkers to Reduce Non-Specific Staining in<br />

Immun<strong>of</strong>luorescence.<br />

R. S. Brody 1 , M. Vermillion 1 , A. A. Rampersaud 1 , P. D. Davis 2 , D. Moothard 3 ; 1 QuantaBiodesign,<br />

Columbus, OH, 2 QuantaBiodesign, Powell, OH, 3 American Qualex, San Clemente, CA<br />

The objective <strong>of</strong> this study was to develop fluorescein conjugated antibodies for tissue and cell<br />

imaging that are brighter and show less non-specific staining than standard fluorescein<br />

isothiocyanate (FITC) conjugates. This objective was achieved by conjugating fluorescein to an<br />

antibody through a discrete polyethylene glycol (dPEG®) linker. This study will be extended to


MONDAY<br />

<strong>the</strong> use <strong>of</strong> dPEG® linkers to conjugate proteins with o<strong>the</strong>r fluorescent dyes with different<br />

excitation and emission wavelengths.<br />

METHODS.Fluorescent antibody conjugates were prepared by attaching fluorescein (FL) to one<br />

end <strong>of</strong> a dPEG® linker that contains 12 ethylene glycol units and a terminal Nhydroxysuccinimide<br />

ester (FL-dPEG®12-NHS). FL-dPEG®12-NHS was conjugated to goat<br />

anti-rabbit IgG (GAR) and goat anti-mouse IgG (GAM) using standard NHS ester reaction<br />

conditions followed by purification over size exclusion columns. The same antibodies were also<br />

labeled with FITC for comparison. Cell based images were made using paraformaldehyde-fixed<br />

MCF-7 cells that were treated with 0.3% Triton X-100 and <strong>the</strong>n incubated with a primary mouse<br />

monoclonal antibody against a golgi protein (GM130) followed by incubation with ei<strong>the</strong>r FLdPEG®12-GAM<br />

or FITC-GAM. Cell nuclei were labeled with DRAQ5 and imaging was done<br />

with a Zeiss 510 scanning laser confocal microscope. Fixed tissue sections were investigated<br />

by incubating <strong>the</strong>m with a rabbit monoclonal antibody against a cancer bio-marker protein<br />

followed by incubation with ei<strong>the</strong>r FL-dPEG®12-GAR or FITC-GAR. Fluorescence was<br />

detected using a Nikon TE2000-E inverted epi-fluorescent microscope. For both <strong>the</strong> cell and<br />

tissue analyses, <strong>the</strong> intensity <strong>of</strong> <strong>the</strong> specific labeling and <strong>the</strong> fluorescence background for each<br />

conjugate was evaluated.<br />

RESULTS. At high incorporation levels, fluorescein that is attached to an antibody through a<br />

dPEG12® linker shows much less self-quenching than fluorescein attached to an antibody via<br />

a FITC label. The fluorescence <strong>of</strong> FL-dPEG®12 labeled antibodies in solution increases<br />

almost linearly with fluorescein incorporation until approximately 20 fluorescein molecules are<br />

attached to each antibody. FL-dPEG®12-GAM was found to bind specifically to <strong>the</strong> target<br />

mouse monoclonal in <strong>the</strong> MCF-7 cells, giving images with moderate intensities and low<br />

backgrounds. FITC labeled GAM gave images with similar intensities but much higher<br />

backgrounds.<br />

Cancerous cells could be clearly imaged in tissue sections using FL-dPEG®12-GAR. In<br />

contrast, FITC-GAR gave very high backgrounds with similar sections that made detection <strong>of</strong><br />

cancerous cells problematic.<br />

2211<br />

Computational Quantitative Image Analysis <strong>of</strong> Tissue Elongation in <strong>the</strong> Drosophila<br />

Embryo.<br />

M. Westacott 1 , A. Zommer 1 , T. Blankenship 1 , D. Loerke 1 ; 1 University <strong>of</strong> Denver (DU), Denver,<br />

CO<br />

Elongation <strong>of</strong> <strong>the</strong> Drosophila embryonic epi<strong>the</strong>lium is driven by cell intercalation, which causes<br />

<strong>the</strong> tissue to narrow in one dimension and leng<strong>the</strong>n in <strong>the</strong> o<strong>the</strong>r. The remodeling <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lial<br />

sheet during this process is a powerful system to illuminate <strong>the</strong> role <strong>of</strong> cell-cell adhesion and<br />

membrane traffic in generating <strong>the</strong> necessary forces for re-organization <strong>of</strong> <strong>the</strong> cellular<br />

architecture, and 4-D in vivo imaging <strong>of</strong> <strong>the</strong> Drosophila embryo allows us to measure cell<br />

interface remodeling dynamics on <strong>the</strong> second timescale. We have developed an automated<br />

computational assay to quantitatively measure <strong>the</strong> dynamics <strong>of</strong> epi<strong>the</strong>lial sheet remodeling. This<br />

assay is based on <strong>the</strong> segmentation <strong>of</strong> cells/interfaces to yield a 'skeletonized' representation <strong>of</strong><br />

<strong>the</strong> sheet, <strong>the</strong> tracking <strong>of</strong> features across planes and in time, and <strong>the</strong> in silico reconstitution <strong>of</strong><br />

<strong>the</strong> 3D spatial structure and network topology <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lial sheet and its temporal<br />

remodeling. This assay allows quantitative measurement <strong>of</strong> interface dynamics and cellular<br />

reorganization, as well as an unbiased measurement <strong>of</strong> protein localization asymmetries within<br />

<strong>the</strong> epi<strong>the</strong>lial sheet in multi-color imaging experiments. We have measured <strong>the</strong> temporal<br />

dynamics <strong>of</strong> interface contraction/elongation, and observe <strong>the</strong> propagation <strong>of</strong> remodeling


MONDAY<br />

dynamics both laterally within <strong>the</strong> sheet and in <strong>the</strong> vertical direction (from apical to basolateral<br />

plane).<br />

2212<br />

A simple, versatile method for GFP-based single molecule localization microscopy.<br />

J. Ries 1 , C. Kaplan 1 , E. Platonova 1 , H. Eghlidi 1 , H. Ewers 1 ; 1 Institute <strong>of</strong> Biochemistry, ETH<br />

Zurich, Zurich, Switzerland<br />

Single molecule localization-based superresolution microscopy methods such as PALM or<br />

STORM, have been breakthrough techniques <strong>of</strong> <strong>the</strong> last years. Until now however, <strong>the</strong>y require<br />

special fluorescent proteins to be cloned or high-affinity antibodies to be generated for specific<br />

labeling. On <strong>the</strong> o<strong>the</strong>r hand, many laboratories will have most <strong>of</strong> <strong>the</strong>ir constructs in GFP form<br />

and entire genomes are available as functional GFP-fusion proteins.<br />

Here, we report a method that makes all <strong>the</strong>se constructs available for superresolution<br />

microscopy by targeting GFP with tiny, high-affinity antibodies coupled to blinking dyes. It thus<br />

combines <strong>the</strong> molecular specificity <strong>of</strong> genetic tagging with <strong>the</strong> high photon yield <strong>of</strong> organic dyes<br />

and minimal linkage error.<br />

Direct STORM on microtubules labeled with our novel antibodies showed that indeed <strong>the</strong><br />

linkage error was minimal, whereas <strong>the</strong> large size <strong>of</strong> standard antibodies resulted in an<br />

additional error <strong>of</strong> >10 nm in immunolabeling.<br />

The brightness <strong>of</strong> our labels enabled us to perform rapid time-lapse dSTORM and sptPALM on<br />

living neurons expressing <strong>the</strong> outer membrane protein GPI-GFP. Three-dimensional dSTORM<br />

on microtubules using <strong>the</strong> bi-plane approach allowed us to distinguish overlapping microtubules<br />

with an axial separation <strong>of</strong> ~100 nm.<br />

Using a budding yeast GFP-tag genomic library we could readily image several GFP-tagged<br />

proteins targeted to specific intracellular locations.<br />

In summary, targeting <strong>of</strong> GFP-labeled constructs with tiny antibodies provides fast and simple<br />

access to superresolution microscopy <strong>of</strong> virtually any known protein in cells. Since for several<br />

organisms <strong>the</strong> entire genome is available as GFP-tagged constructs, all <strong>the</strong>se proteins are<br />

immediately accessible without <strong>the</strong> requirement for cloning or <strong>the</strong> generation <strong>of</strong> antibodies.<br />

Finally, due to a simple one-step labeling protocol, our technique opens <strong>the</strong> door to highthroughput<br />

localization analysis <strong>of</strong> entire genomes at <strong>the</strong> nanoscopic level in cells.<br />

2213<br />

Next Generation Sequencing Analysis <strong>of</strong> <strong>the</strong> Evolutionary Impact <strong>of</strong> Ploidy.<br />

A. Selmecki 1 , P. A. Richmond 2 , R. Dowell 2,3 , D. Pellman 1 ; 1 Department <strong>of</strong> Pediatric Oncology,<br />

Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 2 <strong>Molecular</strong>, Cellular,<br />

Developmental <strong>Biology</strong>, University <strong>of</strong> Colorado, Boulder, CO, 3 Bi<strong>of</strong>rontiers Institute, University <strong>of</strong><br />

Colorado<br />

The role <strong>of</strong> polyploidy—increased copy number <strong>of</strong> chromosomes relative to an organism’s<br />

accepted euploidy—on cells under environmental stress is poorly understood. Ploidy is common<br />

in cancer so it is important to understand how ploidy impacts evolution. We performed an<br />

evolution experiment on strains <strong>of</strong> Saccharomyces cerevisiae with different ploidy (haploid,<br />

diploid, and tetraploid) under selection for growth on a low-glucose, high raffinose media. After<br />

240 generations, <strong>the</strong> parental strain and seven <strong>of</strong> <strong>the</strong> surviving strains (2 diploid and 5<br />

tetraploids) were sequenced via next generation sequencing technology. We analyzed <strong>the</strong><br />

sequenced strains to identify mutations causal for survival and compared <strong>the</strong> mutations<br />

between <strong>the</strong> strains <strong>of</strong> different ploidy. Preliminary results reveal more mutations in <strong>the</strong><br />

tetraploid strains relative to <strong>the</strong>ir diploid counterparts. In addition, we have benchmarked


MONDAY<br />

variation-calling s<strong>of</strong>tware with both “in silico” generated syn<strong>the</strong>tic datasets, as well as our<br />

evolved strains for calling SNPs and indels at higher ploidy (N > 2).<br />

2214<br />

An automated method for tracking punctate objects in live cell imaging data.<br />

A. Ma 1 , J. Metz 2 ; 1 Albert Einstein Coll Med, Bronx, NY, 2 Albert Einstein College <strong>of</strong> Medicine,<br />

Bronx, NY<br />

Continuing advances in fluorescence optical microscopy are yielding an unprecedented surge in<br />

live cell imaging data, leaving manual analysis intractable. We have developed a general and<br />

model-free algorithm for automatic tracking <strong>of</strong> single-particle objects in crowded and noisy<br />

environments in live cell time-lapse imaging data. In this algorithm, we divided objects in <strong>the</strong><br />

imaging data into two categories: those with unique image patterns and those without. We first<br />

use multi-scale bidirectional template matching procedure to identify objects with unique image<br />

patterns. From object linkings made for <strong>the</strong>se objects, we can extract valuable information <strong>of</strong> <strong>the</strong><br />

dynamics <strong>of</strong> <strong>the</strong> system. Such information will be used as inputs to <strong>the</strong> dynamic model based<br />

procedure that we use to track <strong>the</strong> objects without uniquely recognizable image patterns. The<br />

versatility and robustness <strong>of</strong> <strong>the</strong> method were demonstrated by both syn<strong>the</strong>tic and experimental<br />

time lapsed imaging data.<br />

2215/L124<br />

Enabling <strong>Biology</strong> with <strong>the</strong> Structural <strong>Biology</strong> Knowledgebase.<br />

M. Gabanyi 1 , J. Westbrook 1 , D. Micallef 1 , Y-P. Tao 1 , R. Shah 1 , W. McLaughlin 2 , T. Schwede 3 , P.<br />

Adams 4 , W. Minor 5 , H. Berman 1 ; 1 Chemistry and Chemical <strong>Biology</strong>, Rutgers University,<br />

Piscataway, NJ, 2 The Commonwealth Medical College, Scranton, PA, 3 Swiss Institute <strong>of</strong><br />

Bioinformatics, University <strong>of</strong> Basel, Switzerland, 4 Physical Biosciences, Lawrence Berkeley<br />

National Labs, 5 University <strong>of</strong> Virginia Medical School, Charlottesville, VA<br />

The PSI Structural <strong>Biology</strong> Knowledgebase (SBKB, http://sbkb.org) is a scientific search portal<br />

that returns comprehensive biological, structural, and methodological information about<br />

proteins. Information related to 3D protein structures and <strong>the</strong> sequences targeted by worldwide<br />

structural genomics efforts are combined with links to open biological resources to give an<br />

integrated view <strong>of</strong> a protein. Using <strong>the</strong> information found on <strong>the</strong> SBKB, researchers can make<br />

informed decisions about <strong>the</strong>ir projects and what to do next. It is created by <strong>the</strong> Protein<br />

Structure Initiative in collaboration with <strong>the</strong> Nature Publishing Group, and features <strong>the</strong> latest<br />

research and technology advances each month to enable researchers in a broad range <strong>of</strong><br />

biomedical fields.<br />

2216<br />

Identification <strong>of</strong> Ca2+/CaM as a target protein <strong>of</strong> a new hypoxia-inducible factor 1-alpha<br />

inhibiting small molecule using phage display biopanning.<br />

B. Kim 1 , K. Lee 2 , H. Jung 1 , H. Kwon 1 ; 1 Chemical Genomics National Research Laboratory,<br />

TRCP, Department <strong>of</strong> Biotechnology, College <strong>of</strong> Life Science and Biotechnology, Yonsei<br />

University, Seoul, Korea, 2 Dongguk University, Seoul, Korea<br />

Hypoxia inducible factor-1 alpha (HIF-1α) is an essential transcription factor subunit that<br />

regulates expression <strong>of</strong> proteins involving in angiogenesis and metastasis such as VEGF.<br />

Accordingly, HIF-1α represents an emerging <strong>the</strong>rapeutic target for development <strong>of</strong> novel<br />

anticancer drugs. We recently developed a new syn<strong>the</strong>tic small molecule inhibiting HIF-1α<br />

(YCG-6), which results in decrease <strong>of</strong> VEGF mRNA expression. Here, <strong>the</strong> anti-proliferative and<br />

<strong>the</strong> anti-angiogenic activity <strong>of</strong> <strong>the</strong> compound on human umbilical vascular endo<strong>the</strong>lial cells


MONDAY<br />

(HUVECs) were validated through MTT assay and in vitro tube formation and chemoinvasion<br />

assays. Fur<strong>the</strong>rmore, identification <strong>of</strong> <strong>the</strong> target protein <strong>of</strong> <strong>the</strong> compound was conducted by<br />

using phage display biopanning. As <strong>the</strong> result, calmodulin (CaM) was isolated as a plausible<br />

target protein and Ca 2+ is required for binding <strong>of</strong> <strong>the</strong> compound to CaM. Moreover, direct<br />

interaction <strong>of</strong> <strong>the</strong> CaM and compound was confirmed by Surface Plasmon Resonance analysis.<br />

Virtual molecular docking study suggested that <strong>the</strong> compound acts as a CaM antagonist by<br />

binding to hydrophobic pocket <strong>of</strong> <strong>the</strong> protein. Moreover, <strong>the</strong> cell proliferation was arrested by <strong>the</strong><br />

compound as like o<strong>the</strong>r CaM antagonizers did. The effect <strong>of</strong> YCG-6 on Ras/ERK signaling and<br />

prostate specific antigen expression in LNCaP androgen-sensitive cells also clearly reveal that<br />

YCG-6 acts as HIF-1α inhibitor by targeting CaM.<br />

2217<br />

Exploring Atomic Force Microscopy for Single Cell Manipulations.<br />

R. Afrin 1 , U. S. Zohora 2 , H. Uehara 2 , S-I. Machida 1 , T. Watanabe-Nakayama 2 , M. Saito 1 , A.<br />

Ikai 1 ; 1 Innovation Laboratory, Tokyo Institute <strong>of</strong> Technology, Yokohama, Japan, 2 Life Science,<br />

Tokyo Institute <strong>of</strong> Technology, Yokohama, Japan<br />

The atomic force microscope (AFM) is a versatile tool for imaging, force measurement and<br />

manipulation <strong>of</strong> proteins, DNA and living cells basically at <strong>the</strong> single molecular level. In <strong>the</strong><br />

cellular level manipulation, extraction and identification <strong>of</strong> mRNA’s from defined loci <strong>of</strong> a cell,<br />

insertion <strong>of</strong> plasmid DNA and pulling <strong>of</strong> membrane proteins, for example, have been reported<br />

from our group [1]. We used AFM as a novel tool to create holes at defined loci on <strong>the</strong> cell<br />

membrane for visualization <strong>of</strong> intracellular structure through <strong>the</strong> hole and for targeted gene<br />

delivery into living cells as <strong>the</strong> ultimate purpose <strong>of</strong> <strong>the</strong> application <strong>of</strong> nano-medicine. Targeted<br />

gene delivery was successfully performed by inserting an AFM probe that was coated with <strong>the</strong><br />

Monster Green Fluorescent Protein phMGFP Vector for transfection <strong>of</strong> <strong>the</strong> cell. Following<br />

targeted transfection, <strong>the</strong> gene expression <strong>of</strong> GFP was observed and confirmed by <strong>the</strong><br />

fluorescence microscope with high accuracy. Using this AFM probe technology as a fishing<br />

nano-device we successfully harvested intra-cellular mRNA’s and study mRNA expression in<br />

single living cells without damage to <strong>the</strong> cells and analyzed by PCR method. For this, an AFM<br />

tip was inserted into a living cell to extract mRNAs, which were <strong>the</strong>n analyzed for <strong>the</strong> number <strong>of</strong><br />

adhered mRNA through multiplication by RT-PCR and quantitative PCR [2]. We performed<br />

quantitative measurement <strong>of</strong> mRNA at different loci within individual living cells before and after<br />

stimulation with fetal bovine serum. Before activation, mRNAs were found mostly near <strong>the</strong><br />

nucleus, whereas after activation, <strong>the</strong>y were also found in <strong>the</strong> frontal regions <strong>of</strong> locomotive cells.<br />

The results agreed with previous experiments <strong>of</strong> FISH [3]. Asymmetric localizations <strong>of</strong> cellular<br />

proteins and mRNAs are important for cell functions such as division, differentiation and<br />

development. The localization <strong>of</strong> speci?c mRNA generates cell polarity by controlling <strong>the</strong><br />

translation sites <strong>of</strong> speci?c proteins and <strong>the</strong>reby restricting <strong>the</strong>ir locations to appropriate cellular<br />

regions. This method can also be applied to study such important aspects <strong>of</strong> mRNA distribution<br />

within living cells.<br />

References:<br />

1. Afrin R. et al., (2009) J. Mol. Recognit. 22:363-372.<br />

2. Uehara H. et al., (2009) Methods Mol Biol. 544:599-608.<br />

3. Kislauskis EH et al., (1997) J. Cell Biol. 136:1263-1270 .


MONDAY<br />

2218<br />

High Throughput Passive Rheology Assay for Cancer Cell Mechanics.<br />

L. D. Osborne 1 , J. Cribb 1 , V. Swaminathan 2 , R. Spero 1 , E. O'Brien 1 , R. Taylor 3 , R. Superfine 1 ;<br />

1 Dept. <strong>of</strong> Physics & Astronomy, The University <strong>of</strong> North Carolina at Chapel Hill, Chapel Hill, NC,<br />

2 Laboratory <strong>of</strong> Cell and Tissue Morphodynamics, National Heart, Lung, and Blood Institute,<br />

3 Dept. <strong>of</strong> Computer Science, The University <strong>of</strong> North Carolina at Chapel Hill<br />

While <strong>the</strong> dynamic nature <strong>of</strong> cellular structure enables a cell to sense and respond to<br />

environmental forces, this complexity introduces a significant degree <strong>of</strong> variability in <strong>the</strong><br />

mechanical properties <strong>of</strong> individual cells within a population. Understanding <strong>the</strong> composition <strong>of</strong><br />

cell populations is critical as cells become cancerous and <strong>the</strong>ir mechanics change due to<br />

alterations in cytoskeletal structure. To address this need, we are developing a high throughput<br />

system that utilizes passive microbead rheology to characterize cancer cell mechanics. Here,<br />

we report on <strong>the</strong> progress towards a parallel array <strong>of</strong> 12 independently functioning imaging<br />

systems capable <strong>of</strong> ga<strong>the</strong>ring mechanical measurements for a 96-well specimen plate in under<br />

10 minutes. Using our single imaging system prototype, we show that <strong>the</strong> <strong>the</strong>rmal diffusion <strong>of</strong><br />

1um beads connected to integrin surface receptors via fibronectin can distinguish ovarian<br />

cancers with varying metastatic potentials. With a sampling <strong>of</strong> over 500 beads, we report moduli<br />

differences between cancer cells and within cancer cell populations. Additionally, we show<br />

differences in cell moduli caused by substrate stiffness. Our results support previously<br />

documented work describing <strong>the</strong> inverse relationship between mechanical stiffness and invasion<br />

behavior. This demonstrates <strong>the</strong> value <strong>of</strong> our high throughput passive rheology assay as a<br />

screening tool for studying specific signaling pathways involved in mechanotransduction.<br />

2219 WITHDRAWN<br />

2220<br />

Toxic metal detection in foodstuff. Syn<strong>the</strong>tic biology approach used to create<br />

biosensors.<br />

A. Bartos 1,2 , B. Wang 1 , M. Buck 1 , J. Schumacher 1 ; 1 Imperial College London, London, United<br />

Kingdom, 2 Technical University <strong>of</strong> Lodz, Poland<br />

The objective <strong>of</strong> <strong>the</strong> presented research was to explore possibilities <strong>of</strong> using redesigned E.coli<br />

genetic regulatory systems to quantify foodborne metals for analytical purposes. Trace amounts<br />

<strong>of</strong> exogenous metals are known food contaminants. Metallic ions such as mercury, nickel, led,<br />

copper, cadmium, mercury etc. are amongst most toxic agents found in commercially available<br />

food products. Adsorbed and accumulated over years <strong>the</strong>y lead to chronic health disorders.<br />

Ordinarily used instrumental chemistry methods (graphite furnace atomic absorption<br />

spectrometry, inductively coupled plasma atomic emission spectroscopy, inductively coupled<br />

plasma mass spectrometry) can go down to ng/dm3 in <strong>the</strong>ir limits <strong>of</strong> detection, but have a set <strong>of</strong><br />

drawbacks. We designed a microbiological transcription-responsive biosensor to identify trace<br />

amounts <strong>of</strong> toxic metals. Such recombinant senses a number <strong>of</strong> metals, resulting in transcription<br />

<strong>of</strong> genes regulated by inducible protein factors: ZraR in <strong>the</strong> case <strong>of</strong> Zn2+, Pb2+; CusR for Cu2+,<br />

NikR for Ni2+ etc. We have combined <strong>the</strong> promoter sequences <strong>of</strong> GlnK, ZraP, NorV, PspA,<br />

PrpB, CusC to <strong>the</strong> coding sequence <strong>of</strong> GFP to obtain metal responsive fluorescent outputs.<br />

Conducted research provides scope and prospects <strong>of</strong> recombinant E.coli as biosensors, with<br />

emphasis <strong>of</strong> detection limits confined by cells responsibility to molecular signalling and viability<br />

to grow in toxic environment. NCM3722, MC1061 and Top10 strains were induced at<br />

OD600=0.05 and, separately, at OD600=0.4 with 10 metal solutions known to activate <strong>the</strong><br />

corresponding signalling pathways. Results were obtained with <strong>the</strong> use <strong>of</strong> plate reader in <strong>the</strong><br />

form <strong>of</strong> fluorescence and OD600 measurements over time past induction. They indicate


MONDAY<br />

successful use <strong>of</strong> bacterial cultures to sense metals with detection limits below <strong>the</strong> milimolar<br />

range.<br />

2221<br />

Integrated Laser and Electron Microscopy: in search <strong>of</strong> <strong>the</strong> causes <strong>of</strong> FSHD Muscle<br />

Dystrophy.<br />

M. Karreman 1,2 , E. van Donselaar 2 , S. Agronskaia 1 , W. Voorhout 3 , T. Verrips 2 , H. Gerritsen 1 ;<br />

1 Science Faculty, <strong>Molecular</strong> Biophysics, Utrecht, Ne<strong>the</strong>rlands, 2 Dept. <strong>of</strong> <strong>Biology</strong>, Biomolecular<br />

Imaging, Utrecht, Ne<strong>the</strong>rlands, 3 Life Sciences Division, FEI Company, Eindhoven, Ne<strong>the</strong>rlands<br />

Facio Scapulo Humeral Dystrophy (FSHD) is <strong>the</strong> third most common type <strong>of</strong> muscle dystrophy,<br />

and affects 1:20,000 people per year worldwide. Currently, much is known about <strong>the</strong> genetic<br />

background <strong>of</strong> this disease, and how FSHD affects <strong>the</strong> patients. However, <strong>the</strong> cellular<br />

processes involved in FSHD are hi<strong>the</strong>rto not identified.<br />

We set out to study <strong>the</strong> cellular causes and effects <strong>of</strong> FSHD with a novel tool for correlative<br />

microscopy. The Integrated Laser and Electron Microscope (iLEM) combines a fluorescence<br />

microscope (FM) and a transmission electron microscope (TEM) within one set-up. This allows<br />

for <strong>the</strong> localization <strong>of</strong> regions <strong>of</strong> interest over a large field <strong>of</strong> view with <strong>the</strong> FM, and subsequent<br />

ultrastructural analysis <strong>of</strong> <strong>the</strong>se areas with <strong>the</strong> TEM. Since both microscopes are integrated<br />

within one set-up, <strong>the</strong> correlation between <strong>the</strong> FM and <strong>the</strong> TEM is extremely fast and very<br />

accurate.<br />

FSHD is known to be a heterogeneous disease; its expression can vary from muscle to muscle<br />

and from cell to cell. Ultrastructural studies <strong>of</strong> affected cells will help us to gain more insight in<br />

<strong>the</strong> processes involved in this disease. The iLEM is employed to navigate to <strong>the</strong> FSHD affected<br />

regions, so that <strong>the</strong>se areas can subsequently be studied at high resolution. We aimed to<br />

localize FSHD affected cells with <strong>the</strong> iLEM in muscle biopsies from patients. At a certain stage<br />

<strong>of</strong> muscular dystrophy, creatine kinase forms crystalline structures in <strong>the</strong> mitochondria. This<br />

protein was used as a marker to navigate to <strong>the</strong> FSHD affected cells in <strong>the</strong> biopsy; next <strong>the</strong><br />

crystalline structures and <strong>the</strong>ir cellular context was studied at high resolution.<br />

In conclusion; due to <strong>the</strong> heterogeneity <strong>of</strong> <strong>the</strong> expression <strong>of</strong> FSHD, <strong>the</strong> iLEM is a unique tool to<br />

rapidly navigate to <strong>the</strong> cells <strong>of</strong> interest and study <strong>the</strong> effects <strong>of</strong> this disease with high resolution.<br />

2222<br />

A novel single chain Rac biosensor for <strong>the</strong> study <strong>of</strong> tumor cell migration.<br />

Y. Moshfegh 1 ; 1 Albert Einstein College <strong>of</strong> Medicine, Bronx, NY<br />

The p21 Rho family <strong>of</strong> small GTPases are heavily involved in cell motility and migration.<br />

Specifically, Rac is critical for cell protrusion at <strong>the</strong> leading edge, and has been shown to be<br />

overexpressed in several types <strong>of</strong> tumors. However, <strong>the</strong> exact role <strong>of</strong> Rac in relation to <strong>the</strong> o<strong>the</strong>r<br />

Rho GTPases still remains unclear, because previous approaches for <strong>the</strong> study <strong>of</strong> <strong>the</strong>se<br />

molecules are limited due to <strong>the</strong>ir lack <strong>of</strong> sub-cellular spatiotemporal resolution. By developing<br />

genetically encoded, single-chain fluorescent biosensors, we are able to bypass this limitation<br />

and visualize protein activation as well as localization in real time, making this innovation a<br />

necessary and powerful tool for <strong>the</strong> study <strong>of</strong> small G-proteins and cell migration.<br />

To study <strong>the</strong> function <strong>of</strong> <strong>the</strong> Rac during cell protrusion, and also its relationship to <strong>the</strong> o<strong>the</strong>r Rho<br />

GTPases, we developed a novel, genetically-encoded fluorescent Rac1 biosensor, based on an<br />

intra-molecular design. This new biosensor is a massive improvement over <strong>the</strong> previousgeneration<br />

Rac biosensor because <strong>the</strong>re is an equimolar distribution <strong>of</strong> FRET donor and


MONDAY<br />

acceptor and this produces a more accurate readout. Our design maintains <strong>the</strong> correct Cterminal<br />

lipid modification <strong>of</strong> full-length Rac1, enabling proper interaction with upstream<br />

regulators, including GEFs, GAPs, and GDIs. We constructed this new single-chain biosensor<br />

by modifying <strong>the</strong> placement <strong>of</strong> <strong>the</strong> binding domain in relation to <strong>the</strong> first fluorescent protein, to<br />

achieve <strong>the</strong> proper orientation for optimal interaction between <strong>the</strong> GTPase and its binding<br />

domain. In addition, we incorporated an internal autoinhibitory mechanism into our design, to<br />

modulate <strong>the</strong> binding domain affinity.<br />

This new biosensor will allow <strong>the</strong> real-time visualization <strong>of</strong> <strong>the</strong> spatiotemporal dynamics <strong>of</strong> Rac<br />

in live carcinoma cells. In particular, we will be able to explore <strong>the</strong> molecular basis for <strong>the</strong><br />

mechanism <strong>of</strong> Rac, Rho, and Cdc42 coordination at <strong>the</strong> leading edge.<br />

2223<br />

An inducible, reversible system for <strong>the</strong> rapid and complete destruction <strong>of</strong> proteins in<br />

mammalian cells.<br />

D. Fachinetti 1 , A. J. Holland 2 , D. W. Cleveland 3 ; 1 Department <strong>of</strong> Cellular and <strong>Molecular</strong><br />

Medicine, Ludwig Institute for Cancer Research, La Jolla, CA, 2 Department <strong>of</strong> Cellular and<br />

<strong>Molecular</strong> Medicine, University <strong>of</strong> California, Ludwig Institute for Cancer Research, La Jolla, CA,<br />

3 Department <strong>of</strong> Cellular and <strong>Molecular</strong> Medicine, University <strong>of</strong> California, Ludwig Institute for<br />

Cancer Research<br />

Inducible degradation in living cells is a powerful approach for identifying <strong>the</strong> function <strong>of</strong> a<br />

specific protein or protein complex. Here we demonstrate that an auxin-inducible degron (AID)<br />

system is capable <strong>of</strong> controlling <strong>the</strong> stability <strong>of</strong> AID-tagged proteins that are in ei<strong>the</strong>r nuclear or<br />

cytoplasmic compartments and even when incorporated into protein complexes. Induced<br />

degradation occurs rapidly after addition <strong>of</strong> auxin with protein half-life reduced to as little as 9<br />

minutes and proceeding to completion with first order kinetics. Auxin-inducible, degron-mediated<br />

instability is demonstrated to be rapidly reversible. Induced degradation is shown to initiate and<br />

continue in all cell cycle phases, including mitosis, making it especially useful for identifying<br />

function(s) <strong>of</strong> proteins <strong>of</strong> interest during specific points in <strong>the</strong> mammalian cell cycle.<br />

2224<br />

An Application Specific Micr<strong>of</strong>luidic Format for Dynamic Live Cell Studies.<br />

M. Allen 1 , P. Hung 1 , T. Gaige 1 , P. Lee 1 ; 1 CellASIC Corp., Hayward, CA<br />

Studying living mammalian cells in vitro is critical to understanding basic biology, signaling<br />

pathways, drug effects, and disease models. While <strong>the</strong> tools and techniques for cellular analysis<br />

have advanced rapidly over <strong>the</strong> past decades, <strong>the</strong> cell culture platform has remained largely<br />

undeveloped since <strong>the</strong> Petri dish. In light <strong>of</strong> growing evidence that <strong>the</strong> cellular culture<br />

environment is critical for sustaining physiologically relevant phenotype and behavior, we have<br />

developed a micr<strong>of</strong>luidic cell culture platform to control various cellular microenvironment<br />

parameters including reagent perfusion, temperature, and gas composition, all through a<br />

universal control system used in tandem with different application specific micr<strong>of</strong>luidic plates.<br />

Each plate fits a standard microplate format containing four independent micro-fabricated PDMS<br />

culture chambers which can be addressed by up to 6 different reagents. The micr<strong>of</strong>luidic plate<br />

houses all necessary solutions in simple layout <strong>of</strong> wells that are readily accessed during<br />

experimental setup using a pipette. A 170-micron thick glass coverslip bottom enables high<br />

magnification imaging on an inverted microscope. Dynamic environment changes within plates<br />

are controlled through s<strong>of</strong>tware that integrates <strong>the</strong> universal control system with real-time image<br />

analysis to form a closed feedback loop. The utility <strong>of</strong> <strong>the</strong> platform was demonstrated with a<br />

number <strong>of</strong> applications. First, automated immunostaining was demonstrated using HT-1080<br />

fibrosarcoma cells. Cells were cultured in <strong>the</strong> micr<strong>of</strong>luidic plate to <strong>the</strong> desired density, <strong>the</strong>n


MONDAY<br />

automatically washed, fixed, permeablized, blocked, and fluorescently stained with antibodies<br />

using a series <strong>of</strong> solutions and programmed exposure times, significantly simplifying <strong>the</strong><br />

traditional protocol. Second, promyelocytic HL-60 cells differentiated into granulocytes were<br />

tracked to show a chemotactic response to a dynamic chemical gradient for live cell migration<br />

studies. Third, real-time transfection optimization was performed using HeLa cervical carcinoma<br />

cells revealing expression <strong>of</strong> GFP-tagged tubulin via live cell fluorescence microscopy. Fourth,<br />

3D cell culture was demonstrated with MCF10A mammary epi<strong>the</strong>lial cells cultured in Matrigel<br />

with continuous perfusion to track acinus formation. This application specific design philosophy<br />

promises to bring dynamic cell microenvironment control within reach for a large range <strong>of</strong> usergenerated<br />

cell biology applications to complement current advanced cellular analysis<br />

techniques.<br />

2225<br />

Use <strong>of</strong> Short Tandem Repeats for Rapid Au<strong>the</strong>ntication <strong>of</strong> Human Cell Lines.<br />

T. Reid 1 , K. Wilson 2 , C. Kraemer 3 , M. Baird 4 ; 1 DNA Diagnostics Center, Cincinnati, OH,<br />

2 University <strong>of</strong> Cincinnati, OH 3 DNA Diagnostics Center, 4 DNA Diagnostics Ctr, Fairfield, OH<br />

Human cell lines are used worldwide in biological and biomedical research, and accurate data<br />

interpretation depends on unambiguous identity <strong>of</strong> a cell line relative to its original source.<br />

Contamination and misidentification <strong>of</strong> human cell cultures has been identified as a longstanding<br />

issue within <strong>the</strong> scientific community, and steps are now being taken to address <strong>the</strong><br />

problem.<br />

Short-tandem-repeat (STR) DNA pr<strong>of</strong>iling provides an accurate, reliable, and standardized<br />

method for au<strong>the</strong>ntication <strong>of</strong> human identity. Currently, <strong>the</strong> ATCC is considering proposed<br />

standards for cell line au<strong>the</strong>ntication utilizing human STR typing, and many journals have<br />

adopted <strong>the</strong> NIH-recommended policy and mandate cell line au<strong>the</strong>ntication as a requirement for<br />

publication.<br />

Here we demonstrate <strong>the</strong> validation and use <strong>of</strong> two different STR systems to au<strong>the</strong>nticate<br />

human cell lines as well as detect contamination by mouse feeder cells and o<strong>the</strong>r human cell<br />

lines. Threshold levels for detection <strong>of</strong> complete pr<strong>of</strong>iles as well as contamination are compared<br />

between <strong>the</strong> systems, and typical typing issues specific to cell lines are discussed.<br />

2226<br />

Monitoring Changes in NF-kB Pathway Regulation Using Highly Sensitive Multipex<br />

Bioluminescent Reporter Assays.<br />

D. Hughes 1 , M. dobbs 1 , J. Narahari 1 , J. Choi 1 , A. Deshpande 1 , B. Webb 1 ; 1 Therm<strong>of</strong>isher<br />

Scientific, Rockford, IL<br />

The study <strong>of</strong> complex cellular signaling pathways requires powerful and specific tools to monitor<br />

changes in gene activation or repression. In order to accurately monitor <strong>the</strong>se processes,<br />

reporter gene assays are commonly used. We have developed a series <strong>of</strong> next generation<br />

multiplexed luciferase reporters for studying gene regulation. These reporters were developed<br />

to improve <strong>the</strong> sensitivity and convenience <strong>of</strong> conventional luciferase reporter systems. First, we<br />

have used two naturally secreted luciferase genes, Gaussia luciferase from <strong>the</strong> Marine copepod<br />

Gaussia princeps and Cypridina luciferase from <strong>the</strong> Marine ostracod Cypridina noctiluca to<br />

develop a novel dual secreted reporter system. This Gaussia/Cypridina dual system enables<br />

monitoring transcriptional regulation <strong>of</strong> two promoters within tissue culture media without <strong>the</strong><br />

need for cell lysis using sequential addition <strong>of</strong> luciferase substrates. Importantly, both Gaussia<br />

luciferase and Cypridina luciferase are considerably brighter than traditional Firefly luciferase<br />

reporters. Second, we have utilized a mutant form <strong>of</strong> <strong>the</strong> Italian Firefly Luciferase from Luciola


MONDAY<br />

cruciata that has a red-shifted emission spectrum to develop a dual luciferase assay with<br />

Gaussia luciferase in which <strong>the</strong> light output <strong>of</strong> <strong>the</strong> two luciferases are spectrally resolvable. This<br />

Gaussia/Red Firefly dual spectral assay allows simultaneous monitoring <strong>of</strong> two promoters in a<br />

single read assay through addition <strong>of</strong> both substrates and <strong>the</strong>n spectral interrogation <strong>of</strong> <strong>the</strong><br />

resulting light output. In <strong>the</strong> present study, we utilized both techniques, multiplexing by spectral<br />

separation using Gaussia/Red Firefly, and multiplex assays relying on sequential addition <strong>of</strong><br />

substrates two secretory luciferases, Gaussia/Cypridina to monitor changes in NFkB promoter<br />

activity in response to small molecule agonists. Our results demonstrate <strong>the</strong> utility <strong>of</strong> dual<br />

secreted luciferase assays for sensitive real time monitoring <strong>of</strong> NFkB reporter activity in <strong>the</strong><br />

media and simultaneous detection <strong>of</strong> spectrally resolvable luciferases using filter based<br />

detection.<br />

2227<br />

Optimization <strong>of</strong> Protein Purification Using Small-Scale Separation Columns.<br />

R. Tobias 1 , L. Hoang 1 , C. Suh 1 , D. Gjerde 1 ; 1 PHYNEXUS, INC., San Jose, CA<br />

Biophysical and functional characterization <strong>of</strong> <strong>the</strong>rapeutic candidates requires that proteins are<br />

well purified and enriched post-expression. Currently <strong>the</strong> process for adequate preparation<br />

requires that sufficient quantities <strong>of</strong> material be scaled up and processed in a time consuming<br />

manner using expensive chromatography equipment. As improvements in functional and<br />

analytical assays increase throughput and reduce <strong>the</strong> quantity <strong>of</strong> protein required for analysis,<br />

<strong>the</strong> availability <strong>of</strong> efficient small-volume protein purification methods would be <strong>of</strong> high value to<br />

researchers in earlier stages <strong>of</strong> drug discovery and development. Recent advances in <strong>the</strong> area<br />

<strong>of</strong> miniaturized, high-throughput tools for purification, enrichment and desalting <strong>of</strong> proteins<br />

eliminate bottlenecks associated with traditional protein purification processes. By performing<br />

high-performance functional protein separations on small samples in parallel, it is now possible<br />

to obtain more relevant data in a completely automated format. Data from protein separations in<br />

micro-scale chromatography columns is presented here along with optimized conditions<br />

enabling functional and analytical characterization <strong>of</strong> <strong>the</strong>rapeutic proteins purified by this unique<br />

format.<br />

2228<br />

Alternative Digital Micr<strong>of</strong>luidic Devices for Cell Assays.<br />

N. Thorne 1 , B. Demchak 1 , L. Pontiggia 1 , S. Freire 1 ; 1 University <strong>of</strong> <strong>the</strong> Sciences, Philadelphia, PA<br />

Digital micr<strong>of</strong>luidics (DMF) is a technique for transport <strong>of</strong> droplets by an electric potential applied<br />

to an array <strong>of</strong> electrodes, and has appeared as an alternative to <strong>the</strong> conventional fluid transport<br />

in microchannels. DMF enables automation and control <strong>of</strong> droplets unparalleled by any o<strong>the</strong>r<br />

technique <strong>of</strong> fluid transport, without <strong>the</strong> need for pumps, tubes, or valves (1). In particular, we<br />

are interested in developing DMF platforms to study ciliary motion.<br />

Usually, <strong>the</strong> fabrication <strong>of</strong> DMF devices requires access to well-equipped facilities and <strong>the</strong><br />

training <strong>of</strong> students in a variety <strong>of</strong> techniques (e.g, metal deposition, photolithography, etc.). This<br />

is <strong>of</strong>ten expensive and time consuming, limiting <strong>the</strong> access to this technique, particularly for<br />

researchers at undergraduate institutions.<br />

We developed an alternative method for fabrication <strong>of</strong> DMF devices (2). One key aspect is that<br />

miniaturized interelectrode gaps, thought as essential for optimal performance, are not a<br />

requirement for droplet actuation; altoge<strong>the</strong>r, <strong>the</strong> strategy enables fast fabrication <strong>of</strong> robust and<br />

reliable devices at low costs.


MONDAY<br />

However, this technique requires relatively high voltages for DMF operation (typically 500<br />

VRMS), which contrasts with <strong>the</strong> low voltages (~120 VRMS) used in conventional, clean-room<br />

based devices. This has been a point <strong>of</strong> criticism, with <strong>the</strong> claim that high voltages might be<br />

detrimental to biological specimens.<br />

Tetrahymena <strong>the</strong>rmophila (SB 255) cells, well suited for research purposes due to <strong>the</strong> large<br />

number <strong>of</strong> cilia, were used for tests. Droplets (10 µL) containing cells were actuated on DMF<br />

devices. We will present results showing that no changes were observed in <strong>the</strong> vitality and<br />

proliferation <strong>of</strong> <strong>the</strong>se cells after transported in <strong>the</strong> alternative DMF devices. This corroborates<br />

previous results showing that <strong>the</strong> voltage across <strong>the</strong> droplet in a DMF device is an insignificant<br />

fraction <strong>of</strong> <strong>the</strong> total applied voltage, indicating very little, if any, effects on cells (3).<br />

We are currently evaluating non-specific adsorption <strong>of</strong> analytes to surfaces, which effects device<br />

performance. We have chosen a simple and well-established assay, cell deciliation with<br />

dibucaine. We will show that <strong>the</strong> minimum concentration <strong>of</strong> dibucaine for a complete and almost<br />

instantaneous loss <strong>of</strong> cilia is close to <strong>the</strong> used in conventional, <strong>of</strong>f-chip deciliation (1.3 mM)(4).<br />

This indicates little non-specifc adsorption <strong>of</strong> dibucaine to surfaces in this assay.<br />

Since adsorption to surfaces might be analyte dependent, we are also looking for collaborations<br />

to develop o<strong>the</strong>r assays <strong>of</strong> interest, to fur<strong>the</strong>r characterize <strong>the</strong> devices. However, <strong>the</strong>se<br />

preliminary results pave <strong>the</strong> way to on-chip deciliation. Future studies will focus on<br />

characterizing motion <strong>of</strong> extracted cilia, directly on a micr<strong>of</strong>luidic device. This work has been<br />

done by undergraduate students (Biological Sciences and Physics).<br />

References:<br />

1 Fair, R. B. "Digital micr<strong>of</strong>luidics: is a true lab-on-a-chip possible?" Micr<strong>of</strong>luidics and<br />

Nan<strong>of</strong>luidics 2007, 3, 245-281.<br />

2 Thorne, N.; Lamberto, M.; Mazza, A.; Freire, S. L. S. "An Alternative Strategy for<br />

Fabrication <strong>of</strong> Robust and Flexible Digital Micr<strong>of</strong>luidic Devices" Pacific Northwest Journal <strong>of</strong><br />

Undergraduate Research and Creative Activities 2011, 2<br />

3 Barbulovic-Nad, I.; Yang, H.; Park, P. S.; Wheeler, A. R. "Digital micr<strong>of</strong>luidics for cellbased<br />

assays" Lab Chip 2008, 8, 519-526.<br />

4 Thompson, G. A., Jr.; Baugh, L. C.; Walker, L. F. "Nonlethal deciliation <strong>of</strong> Tetrahymena<br />

by a local anes<strong>the</strong>tic and its utility as a tool for studying cilia regeneration" J Cell Biol 1974, 61,<br />

253-257.<br />

2229<br />

Deciphering <strong>the</strong> Intermediate Filament Structure(s) using cryo Electron Microscopy and<br />

Novel Metal Labels.<br />

R. Kirmse 1 , C. Bouchet-Marquis 1 , M. Pagratis 1 , A. Hoenger 1 ; 1 MCDB, University <strong>of</strong> Colorado at<br />

Boulder, Boulder, CO<br />

Intermediate filaments (IFs) are critical building blocks <strong>of</strong> <strong>the</strong> cell´s cytoskeleton. Toge<strong>the</strong>r with<br />

actin filaments and microtubules <strong>the</strong>y are responsible for various cell functions such as<br />

mechanical stress resistance, signal transduction, and material transport. In contrast to actin<br />

and tubulin a detailed 3D structure <strong>of</strong> IF proteins and grown filaments remains elusive.<br />

Crystallization <strong>of</strong> IF proteins for example is extremely difficult caused by <strong>the</strong>ir elongated shape.<br />

Regardless, cryo electron microscopy and tomography provides <strong>the</strong> opportunity to observe<br />

proteins in 3D. By rapid freezing we are able to keep <strong>the</strong> IFs hydrated and as close to <strong>the</strong>ir<br />

native state as possible. Subsequently cryo tomography allows reconstructing <strong>the</strong> 3D structure


MONDAY<br />

<strong>of</strong> <strong>the</strong> observed sample in great detail. In addition we are developing new labeling methods<br />

utilizing cloneable high-density markers to fur<strong>the</strong>r label <strong>the</strong> IF substructure. These markers will<br />

enable us to draw conclusion about <strong>the</strong> basic underlying arrangements <strong>of</strong> <strong>the</strong> IF proteins in <strong>the</strong><br />

mature filaments.


TUESDAY, <strong>DECEMBER</strong> 6- <strong>Late</strong> <strong>Abstracts</strong> 3<br />

Actin Cytoskeleton<br />

TUESDAY<br />

2230<br />

Depletion <strong>of</strong> Arp2/3 complex reveals critical role <strong>of</strong> lamellipodia in haptotaxis, but not<br />

chemotaxis.<br />

C. Wu 1 , J. E. Bear 2,3 ; 1 Cell and Developmental <strong>Biology</strong>, UNC-Chapel Hill, Chapel Hill, NC,<br />

2 UNC-Chapel Hill, Chapel Hill. CA 3 HHMI<br />

Lamellipodia are sheet-like, leading edge protrusions in firmly adherent cells that contain<br />

Arp2/3-generated dendritic actin networks. Although lamellipodia are widely believed to be<br />

critical for cell motility, this notion has not been rigorously tested. Using fibroblasts derived from<br />

Ink4a/Arf-deficient mice, we generated a stable line depleted <strong>of</strong> Arp2/3 complex that lacks<br />

lamellipodia. This line shows defective random cell motility and relies on a filopodia-based<br />

protrusion system. Utilizing a micr<strong>of</strong>luidic gradient generation system, we tested <strong>the</strong> role <strong>of</strong><br />

Arp2/3 complex and lamellipodia in directional cell migration. Surprisingly, <strong>the</strong>se cells respond<br />

normally to shallow gradients <strong>of</strong> PDGF indicating that lamellipodia are not required for<br />

chemotaxis. Conversely, <strong>the</strong>se cells cannot respond to a gradient <strong>of</strong> extracellular matrix<br />

(haptotaxis). Consistent with this finding, cells depleted <strong>of</strong> Arp2/3 fail to globally align focal<br />

adhesions suggesting that one principle function <strong>of</strong> lamellipodia is to organize cell-matrix<br />

adhesions in a spatial coherent manner.<br />

2231<br />

A Peptide Mimetic <strong>of</strong> Heat Shock Protein Beta 6 Alters 3t3 Migration through a C<strong>of</strong>ilin-<br />

Dependent Mechanism.<br />

C. Smoke 1 , D. N. Derkach 1 , D. Eng 1 , J. Uhlenkamp 1 , K. B. Perkins 1 , M. R. Sheller 1 ; 1 Capstone<br />

Therapeutics, Tempe, AZ<br />

Prior research has shown that AZX100, a 24-amino acid peptide mimetic <strong>of</strong> Heat Shock Protein<br />

β6 (HSPβ6), reduces filamentous actin and focal adhesions in my<strong>of</strong>ibroblasts, cells instrumental<br />

in <strong>the</strong> process <strong>of</strong> dermal scar healing. It has also been shown that HSPβ6 binds to 14-3-3,<br />

leading to dephosphorylation <strong>of</strong> c<strong>of</strong>ilin. Dephosphorylated c<strong>of</strong>ilin severs filamentous actin and<br />

increases <strong>the</strong> pool <strong>of</strong> globular actin. Investigating <strong>the</strong> manner in which AZX100 affects c<strong>of</strong>ilin<br />

dephosphorylation, alters actin dynamics and influences migratory behavior in my<strong>of</strong>ibroblasts<br />

could elucidate <strong>the</strong> role <strong>of</strong> AZX100 in scar healing.<br />

The present study used 3T3 fibroblasts treated with transforming growth factor β-1 (TGFβ-1)<br />

alone or in <strong>the</strong> presence <strong>of</strong> varying concentrations <strong>of</strong> AZX100 for one hour. Fibroblasts were<br />

treated with TGFβ-1 to induce differentiation to my<strong>of</strong>ibroblast-like cells. Cell lysates were<br />

resolved by 2-dimensional gel electrophoresis to determine <strong>the</strong> c<strong>of</strong>ilin-to-phospho-c<strong>of</strong>ilin<br />

expression levels. AZX100 treatment <strong>of</strong> <strong>the</strong> 3T3 cells significantly increased <strong>the</strong> percent <strong>of</strong><br />

dephosphorylated c<strong>of</strong>ilin compared to cells treated with TGFß-1 alone (73% vs. 40%<br />

respectively; p =0.006). Additionally, live cell imaging was performed to study <strong>the</strong> effect <strong>of</strong><br />

various doses <strong>of</strong> AZX100 on <strong>the</strong> actin cytoskeleton. Treatment with 1 and 10 μM AZX100<br />

produced <strong>the</strong> greatest change in <strong>the</strong> actin cytoskeleton as measured by overall reduction in cell<br />

area. Doses <strong>of</strong> AZX100 greater than 10 μM and lower than 1 μM did not significantly alter <strong>the</strong><br />

actin cytoskeleton compared to control cells. Finally, my<strong>of</strong>ibroblast migration was analyzed<br />

through <strong>the</strong> Electric Cell-substrate Impedence Sensing (ECIS) system, which measured<br />

changes in resistance over time. AZX100 treatment resulted in a dose-dependent reduction <strong>of</strong><br />

migration over one hour compared to TGFβ-1 alone, with a maximal reduction <strong>of</strong> 53%. In


TUESDAY<br />

conclusion, AZX100 increases c<strong>of</strong>ilin dephosphorylation inmigratory cells. The associated<br />

migration pattern was also altered, which may be due to morphological changes resulting from<br />

<strong>the</strong> liberation <strong>of</strong> dephosphorylated c<strong>of</strong>ilin from 14-3-3.<br />

2232<br />

Biophysical Linkage between Local Mechanical Properties and Global Cellular<br />

Responses in Living Fibroblasts: Actin Cytoskeleton Perspectives.<br />

H-C. Harn 1 , Y-K. Wang 2 , H-H. Lin 3 , C-M. Cheng 4 , M-J. Tang 1,3 ; 1 Institute <strong>of</strong> Basic Medical<br />

Sciences, National Cheng Kung University, Tainan, Taiwan, 2 Dept. <strong>of</strong> Medicine, Skeleton-Joint<br />

Research Center, National Cheng Kung University, 3 Department <strong>of</strong> Physiology, College <strong>of</strong><br />

Medicine, National Cheng Kung University, Tainan, Taiwan, 4 Institute <strong>of</strong> Nanoengineering and<br />

Microsystems, National Tsing Hua University, Hsinchu, Taiwan<br />

Each organ in our body has different mechanical properties—ranging from 200 Pa (brain) to 310<br />

MPa (Achilles tendon)—serving as <strong>the</strong> structural-based scaffolding and <strong>the</strong> inherent force<br />

source <strong>of</strong> mechanical stimulation for single cells. The cellular response under mechanical<br />

stimulation is highly tuned to influence a cell’s structure provided by <strong>the</strong> cytoskeleton. The<br />

cytoskeleton governs a diversity <strong>of</strong> cellular behaviors such as cell division, motility, and<br />

morphology. Solon et al. have observed that fibroblasts adjusted <strong>the</strong>ir spreading areas and actin<br />

cytoskeleton organization in order to match <strong>the</strong> change <strong>of</strong> an external environment. We<br />

<strong>the</strong>refore hypo<strong>the</strong>size that actin cytoskeleton could be a key determinant to <strong>the</strong> changes in cell’s<br />

mechanical properties in response to different mechanical stimuli. In this study, we attempted to<br />

explore <strong>the</strong> mechanical properties <strong>of</strong> an individual actin filament in living fibroblasts (NRK49F),<br />

and to determine its contributions to <strong>the</strong> elasticity <strong>of</strong> <strong>the</strong> cell as a whole with external physical<br />

stimuli. We used a biological atomic force microscope to (i) scan a living fibroblast in a liquid<br />

environment, (ii) indent around <strong>the</strong> periphery in a living fibroblast, and (iii) indent on top <strong>of</strong> <strong>the</strong><br />

nucleus in a living fibroblast to determine Young’s moduli <strong>of</strong> specific locations, respectively. We<br />

also constructed cell’s 3D images via confocal fluorescent microscopy, in order to probe <strong>the</strong><br />

morphological responses <strong>of</strong> actin cytoskeleton with mechanical stimuli. The results showed that<br />

a fibroblast changed its local and global elasticities under substratum stiffness. The organization<br />

<strong>of</strong> actin filaments—<strong>the</strong> number <strong>of</strong> actin filaments at a specific location that we probed in a living<br />

fibroblast—were strongly affected by <strong>the</strong> change with substrate stiffness. Fur<strong>the</strong>rmore, even for<br />

a single fibroblast seeded on a stiffer substrate showed a distinct difference (20 and 200 kPa) in<br />

<strong>the</strong> elasticity <strong>of</strong> actin filaments; this could correspond to <strong>the</strong> variations in <strong>the</strong> thickness <strong>of</strong> actin<br />

filaments and <strong>the</strong> distribution <strong>of</strong> actin filaments around <strong>the</strong> cell periphery. This study connects<br />

<strong>the</strong> local mechanical properties <strong>of</strong> actin filaments to <strong>the</strong> overall elasticity <strong>of</strong> a living cell in<br />

response to mechanical stimulation. It highlights <strong>the</strong> important contribution <strong>of</strong> actin filaments<br />

could play in regulating <strong>the</strong> behavior and function <strong>of</strong> a cell.<br />

2233<br />

Stereocilia F-actin polymerization is coupled to <strong>the</strong> mechanotransduction machinery in<br />

<strong>the</strong> auditory hair bundles.<br />

V. Michel 1,2 , E. Caberlotto 1 , D. Weil 1 , C. Petit 1,3 ; 1 Pasteur Institut-INSERM, Paris, France,<br />

2 Université Pierre et Marie Curie, Paris, France, 3 Collège de France, Paris, France<br />

In <strong>the</strong> hair bundles <strong>of</strong> <strong>the</strong> auditory hair cells, <strong>the</strong> mechanotransducer channels are gated by tiplinks,<br />

extracellular filaments that interconnect <strong>the</strong> stereocilia and stretch from <strong>the</strong> tips <strong>of</strong><br />

stereocilia in <strong>the</strong> short and middle rows to <strong>the</strong> sides <strong>of</strong> neighboring, taller stereocilia. Tip-links<br />

consist <strong>of</strong> cadherin-23 and protocadherin-15, products <strong>of</strong> <strong>the</strong> Usher syndrome type 1 genes<br />

USH1D and USH1F, respectively. In this study, we found that upon <strong>the</strong> post-natal<br />

disappearance <strong>of</strong> <strong>the</strong> tip-links, caused by <strong>the</strong> deletion <strong>of</strong> one <strong>of</strong> its components, cadherin-23, or


TUESDAY<br />

<strong>of</strong> one <strong>of</strong> its anchoring proteins, sans, stereocilia from <strong>the</strong> short and medium but not <strong>of</strong> <strong>the</strong> high<br />

stereocilia row undergo a dramatic reduction in length. This indicates that <strong>the</strong><br />

mechanotransduction machinery has a positive effect on F-actin polymerization. The loss <strong>of</strong> <strong>the</strong><br />

tip-links and <strong>the</strong> resulting loss <strong>of</strong> functional MET channels is what stops <strong>the</strong> renewal <strong>of</strong> <strong>the</strong> short<br />

and middle rows <strong>of</strong> stereocilia in <strong>the</strong>se mutant mice. We can conclude that <strong>the</strong> tip-link plays an<br />

unsuspected, direct or indirect role on <strong>the</strong> undergoing actin polymerization processes in <strong>the</strong><br />

different stereocilia rows, which ensures <strong>the</strong> development and <strong>the</strong> dynamic maintenance <strong>of</strong> <strong>the</strong><br />

hair bundle’s staircase pattern.<br />

2234<br />

Mutual regulations <strong>of</strong> actin cytoskelton remodeling and peroxisome proliferator-activated<br />

receptor-γ on early adipocyte differentiation.<br />

H. Nobusue 1 , N. Onishi 2 , Y. Oki 1 , T. Shimizu 2 , T. Chiyoda 2 , H. Saya 2 , K. Kano 1 ; 1 Laboratory <strong>of</strong><br />

Cell and Tissue <strong>Biology</strong>, College <strong>of</strong> Bioresource Sciences, Nihon University, Fujisawa, Japan,<br />

2 Division <strong>of</strong> Gene Regulation, Institute for Advanced Medical Research, Keio University School<br />

<strong>of</strong> Medicine, Shinjuku-ku, Japan<br />

Adipocyte differentiation at an early stage is directly regulated by gene expression <strong>of</strong> a master<br />

regulator such as peroxisome proliferator-activated receptor-γ (PPARγ). During adipogenesis<br />

actin cytoskelton remodeling defines important events <strong>of</strong> <strong>the</strong> differentiation process; this is<br />

characterized by <strong>the</strong> conversion <strong>of</strong> filamentous actin from stress fibers to cortical actin<br />

structures. Here, we examined <strong>the</strong> regulatory relationship between actin cytoskelton remodeling<br />

and PPARγ in early adipocyte differentiation. Depending on adipogenic induction, actin stress<br />

fibers were immediately disrupted prior to <strong>the</strong> expression <strong>of</strong> PPARγ. And treatment with<br />

cytoskeletal fixation agent phalloidin maintained actin fiber structures even after adipogenic<br />

induction, and caused a down-regulation <strong>of</strong> PPARγ. In addition, ectopic expression <strong>of</strong> activated<br />

RhoA, which is known to promote <strong>the</strong> formation <strong>of</strong> actin stress fibers, inhibited <strong>the</strong> disruption <strong>of</strong><br />

actin stress fibers and PPARγ expression after adipogenic induction, and <strong>the</strong>se effects were<br />

recovered by treatment with <strong>the</strong> RhoA kinase inhibitor Y-27632. Moreover, treatment <strong>of</strong> <strong>the</strong> actin<br />

polymerization inhibitor cytochalasin D in active RhoA-expressing cells caused <strong>the</strong> disruption <strong>of</strong><br />

actin stress fibers, and as a result, PPARγ expression was rescued. On <strong>the</strong> o<strong>the</strong>r hand,<br />

introduction <strong>of</strong> <strong>the</strong> PPARγ-specific shRNA resulted in maintained <strong>the</strong> high-level expression <strong>of</strong><br />

fibronectin (FN) matrix and inhibited <strong>the</strong> reorganization into adipocyte-specific cortical actin<br />

structures during adipocyte differentiation. And transfection <strong>of</strong> FN-specific siRNA in PPARγknockdown<br />

cells after adipogenic induction caused <strong>the</strong> formation <strong>of</strong> cortical actin structures. In<br />

conclusion, our findings indicate that actin stress fiber disruption directly induces PPARγ<br />

expression, and that PPARγ regulates <strong>the</strong> reorganization into adipocyte-specific cortical actin<br />

structures through <strong>the</strong> down-regulation <strong>of</strong> FN matrix, and provide a novel insight into <strong>the</strong> mutual<br />

regulatory mechanisms between actin cytoskelton remodeling and adipogenic transcription<br />

factors in early adipocyte differentiation.<br />

2235<br />

The Carboxy Termini <strong>of</strong> Human Myopodin Is<strong>of</strong>orms Influences Non-Muscle Myosin II-<br />

Dependent Myopodin Subcellular Localization.<br />

F. Kai 1 , R. Duncan 1,2 ; 1 Department <strong>of</strong> Microbiology, Dalhousie University, Halifax, NS, Canada,<br />

2 Department <strong>of</strong> Biochemistry & <strong>Molecular</strong> <strong>Biology</strong> and Department <strong>of</strong> Pediatrics, Dalhousie<br />

University, Halifax, NS, Canada<br />

More than 80% <strong>of</strong> patients with invasive prostate cancer have deletions within <strong>the</strong>ir myopodin<br />

gene, suggesting that myopodin is a reliable predictive marker <strong>of</strong> prostate cancer metastasis.<br />

However, <strong>the</strong> functions <strong>of</strong> myopodin in cancer metastasis remain unknown. Biochemical studies


TUESDAY<br />

have revealed that myopodin is an actin-binding protein. Since actin rearrangement is an<br />

essential process involved in cell migration, it has been postulated that myopodin might<br />

suppress cancer metastasis by modulating <strong>the</strong> actin cytoskeleton. Presently, five human<br />

myopodin (hMYO) splicing variants, each with a unique amino and/or carboxy-termini, have<br />

been identified. However, it is unclear whe<strong>the</strong>r <strong>the</strong> unique sequence <strong>of</strong> each is<strong>of</strong>orm<br />

differentially regulates myopodin function. To examine if <strong>the</strong>se splicing variants function<br />

distinctly, we ectopically expressed myopodin is<strong>of</strong>orms in invasive prostate cancer cells (PC3)<br />

and benign prostatic hyperplasia cells (BPH-1). Immun<strong>of</strong>luorescent staining revealed that <strong>the</strong><br />

different myopodin is<strong>of</strong>orms induce distinct actin structures within <strong>the</strong> cell body. Each is<strong>of</strong>orm<br />

interacts distinctly with <strong>the</strong>se actin structures, but none <strong>of</strong> <strong>the</strong> is<strong>of</strong>orms colocalize with actin near<br />

<strong>the</strong> cell periphery. Truncation analysis indicated <strong>the</strong> unique carboxy-terminal sequences<br />

regulate <strong>the</strong> distinct localization <strong>of</strong> myopodin is<strong>of</strong>orms with <strong>the</strong> actin cytoskeleton. Fur<strong>the</strong>rmore,<br />

<strong>the</strong> restrictive subcellular localization <strong>of</strong> some myopodin is<strong>of</strong>orms was abrogated by inhibitors<br />

that affect downstream effectors in <strong>the</strong> activated RhoA pathway (i.e. <strong>the</strong> ROCK inhibitor Y-<br />

27632 and <strong>the</strong> non-muscle myosin II inhibitor blebbistatin). Taken toge<strong>the</strong>r, <strong>the</strong>se results<br />

indicate <strong>the</strong> carboxy-termini <strong>of</strong> <strong>the</strong> human myopodin is<strong>of</strong>orms regulate <strong>the</strong>ir ability to<br />

differentially alter and co-localize with actin cytoskeletal structures in a non-muscle myosin IIdependent<br />

manner.<br />

FuiBoon Kai is supported by a trainee award from The Beatrice Hunter Cancer Research<br />

Institute with funds provided by The Terry Fox Foundation Strategic Health Research Training<br />

Program in Cancer Research at <strong>the</strong> Canadian Institutes <strong>of</strong> Health Research.<br />

2236<br />

Surface-bound VASP controls Arp2/3 complex-dependent actin gel growth.<br />

P. Noguera 1 , A. Lamora 1 , J. Plastino 1 ; 1 Physico-chimie Curie (PCC) - UMR 168, Institut Curie,<br />

Paris, France<br />

The Drosophila Enabled/Vasodilator Stimulated Phosphoprotein (Ena/VASP) protein family is<br />

involved in numerous actin based processes such as filopodia formation and lamellipodia<br />

protrusion, but <strong>the</strong> precise function and mode <strong>of</strong> action <strong>of</strong> Ena/VASP proteins remain<br />

controversial. To address this question, we use an in vitro assay composed <strong>of</strong> beads coated<br />

with actin polymerization activators, incubated in a protein mix that supports <strong>the</strong> growth <strong>of</strong> an<br />

actin gel on <strong>the</strong> bead surface. Over time, <strong>the</strong> symmetrical gel breaks open to form a comet tail<br />

that propels <strong>the</strong> bead forward, mimicking in simplified conditions <strong>the</strong> dynamics <strong>of</strong> <strong>the</strong> actin<br />

cytoskeleton at <strong>the</strong> plasma membrane <strong>of</strong> moving cells. Here we use beads that activate <strong>the</strong><br />

actin-related protein (Arp) 2/3 complex, an actin polymerization nucleator, and additionally<br />

recruit wild-type and mutant forms <strong>of</strong> murine VASP to <strong>the</strong> bead surface. We show that VASP<br />

increases <strong>the</strong> rate <strong>of</strong> actin comet growth. We fur<strong>the</strong>r show that this activity requires <strong>the</strong> F-actin<br />

binding capacity <strong>of</strong> VASP, its tetramerization domain and its pr<strong>of</strong>ilin recruiting site, but that <strong>the</strong><br />

G-actin binding site is dispensable. When VASP is not targeted to <strong>the</strong> bead surface, it has no<br />

effect on bead speed, despite <strong>the</strong> fact that it is present throughout <strong>the</strong> actin comet. In ano<strong>the</strong>r<br />

series <strong>of</strong> experiments, we provide evidence that VASP focuses polymerization near <strong>the</strong> bead<br />

surface in <strong>the</strong> absence <strong>of</strong> capping protein via its F-actin binding capacity. Overall <strong>the</strong>se results<br />

shed light on <strong>the</strong> molecular mechanism <strong>of</strong> Ena/VASP proteins, and on how <strong>the</strong>se proteins enter<br />

into <strong>the</strong> actin dynamics equation in vivo.


TUESDAY<br />

2237<br />

Rho-GTPase mediated regulation and membrane targeting <strong>of</strong> human leukocyte formins<br />

FMNL1 and FMNL2.<br />

S. Kuehn 1 , J. Block 2 , K. Rottner 2 , M. Geyer 1 ; 1 Department <strong>of</strong> Physical Biochemistry, Max Planck<br />

Institute <strong>of</strong> <strong>Molecular</strong> Physiology, Dortmund, Germany, 2 Institute <strong>of</strong> Genetics, University <strong>of</strong><br />

Bonn, Bonn, Germany<br />

Dynamic actin filament structures required for cell motility and migration are regulated by<br />

nucleation and elongation factors, such as members <strong>of</strong> <strong>the</strong> formin family. The large,<br />

multidomain proteins FMNL1 and FMNL2 belong to <strong>the</strong> subgroup <strong>of</strong> Diaphanous-related formins<br />

(DRFs). The FH2 domain-mediated actin assembly activity <strong>of</strong> DRFs is inhibited by an<br />

intramolecular interaction between <strong>the</strong> N-terminal regulatory region and <strong>the</strong> C-terminal DAD<br />

domain. This inhibition is released via binding <strong>of</strong> Rho-GTPases to <strong>the</strong> GBD and FH3 domain.<br />

Additional c<strong>of</strong>actors might be required for full activation, localization or membrane targeting.<br />

We here show that human FMNL2N interacts specifically with GppNHp-bound Cdc42 in <strong>the</strong><br />

micromolar affinity range. Cdc42 binds to FMNL2N via switch I, switch II as well as <strong>the</strong> insert<br />

helix that is unique for Rho-GTPases. In FMNL2, binding to Cdc42 involves <strong>the</strong> GBD and all<br />

armadillo repeats <strong>of</strong> <strong>the</strong> FH3 domain. The structure thus provides for <strong>the</strong> first time an effectorspecific<br />

binding <strong>of</strong> <strong>the</strong> Rho-specific insert helix. We could determine crucial residues, which can,<br />

if mutated, switch <strong>the</strong> specificity <strong>of</strong> FMNL2 for Cdc42 to Rac1. We identified a basic insert that<br />

disrupts <strong>the</strong> canonical FH3 repeat fold. This insert binds acidic phospholipids and negatively<br />

charged liposomes in vitro, suggesting plasma membrane association through electrostatic<br />

interactions. Additionally, FMNL2 is N-terminally myristoylated, a fatty acid modification that<br />

drives membrane binding. Toge<strong>the</strong>r with <strong>the</strong> C-terminal prenylation <strong>of</strong> <strong>the</strong> GTPase, a large<br />

membrane binding surface <strong>of</strong> <strong>the</strong> dimeric formin complex assembles that sustains membrane<br />

association during migration. In vivo experiments reveal that both, <strong>the</strong> basic insert and <strong>the</strong><br />

myristoyl moiety, are responsible for plasma membrane targeting <strong>of</strong> <strong>the</strong> actin polymerization<br />

factor.<br />

2238<br />

Watching <strong>the</strong> motions <strong>of</strong> <strong>the</strong> cell wall syn<strong>the</strong>sis machinery and underlying cytoskeleton<br />

in B. subtilis with high precision particle tracking.<br />

E. C. Garner 1 , X. Zhuang 2 , W. Wang 3 , R. Bernard 4 , D. Rudner 4 , T. MItchison 5 ; 1 Harvard<br />

University, Cambridge, MA, 2 Chemistry and Chemical biology, 3 Physics, Harvard University,<br />

Cambridge, MA, 4 Microbiology, Harvard University, Cambridge, MA, 5 Systems <strong>Biology</strong>, Harvard<br />

University, Cambridge, MA<br />

Rod-shaped bacteria elongate by <strong>the</strong> action <strong>of</strong> cell-wall syn<strong>the</strong>sis complexes linked to<br />

underlying dynamic MreB filaments, but how <strong>the</strong>se proteins function to allow continued<br />

elongation as a rod remains unknown. To understand how <strong>the</strong> movement <strong>of</strong> <strong>the</strong>se filaments<br />

relates to cell wall syn<strong>the</strong>sis, we characterized <strong>the</strong> dynamics <strong>of</strong> MreB and <strong>the</strong> cell wall<br />

elongation machinery using high-resolution particle tracking in Bacillus subtilis. We found that<br />

both MreB and <strong>the</strong> elongation machinery move in linear paths across <strong>the</strong> cell, moving at similar<br />

rates (~20nm / second) and angles to <strong>the</strong> cell body, suggesting <strong>the</strong>y function as single<br />

complexes. These proteins move circumferentially around <strong>the</strong> cell, principally perpendicular to<br />

its length. We find that <strong>the</strong> motions <strong>of</strong> <strong>the</strong>se complexes are independent, as <strong>the</strong>y can pause and<br />

reverse, and also as nearby complexes move independently in both directions across one<br />

surface <strong>of</strong> <strong>the</strong> cell. Inhibition <strong>of</strong> cell wall syn<strong>the</strong>sis with antibiotics or depletions in <strong>the</strong> cell wall<br />

syn<strong>the</strong>sis machinery blocked MreB movement, suggesting that <strong>the</strong> cell wall syn<strong>the</strong>tic machinery<br />

is <strong>the</strong> motor in this system. We propose that bacteria elongate by <strong>the</strong> uncoordinated,


TUESDAY<br />

circumferential movements <strong>of</strong> syn<strong>the</strong>tic complexes that span <strong>the</strong> plasma membrane and insert<br />

radial hoops <strong>of</strong> new peptidoglycan during <strong>the</strong>ir transit.<br />

2239 WITHDRAWN<br />

2240<br />

Computational models <strong>of</strong> embryonic wound healing.<br />

M. A. Wyczalkowski 1 , V. D. Varner 1 , L. A. Taber 1 ; 1 Biomedical Engineering, Washington<br />

University in St. Louis, St. Louis, MO<br />

The objective <strong>of</strong> this work is to characterize experimentally <strong>the</strong> healing <strong>of</strong> circular and slash<br />

wounds in epi<strong>the</strong>lia <strong>of</strong> early stage chick embryos and to develop a finite element model <strong>of</strong><br />

wound healing dynamics.<br />

Early stage chick embryos were harvested and cultured ex ovo. Circular and slash wounds<br />

(~100um in size) were made in <strong>the</strong> blastoderm and allowed to heal. The healing process was<br />

captured with time lapse microscopy, and wound area and shape were characterized as<br />

functions <strong>of</strong> time. The contractile response <strong>of</strong> <strong>the</strong> tissue to wounding was implemented in a finite<br />

element model, and <strong>the</strong> results were critically compared to experimental results for both wound<br />

types.<br />

The embryonic wounds displayed a two-phase behavior: rapid wound healing lasting about a<br />

minute, followed by a more gradual healing to full closure within tens <strong>of</strong> minutes. This behavior,<br />

as well as more complicated wound geometry trends (e.g., aspect ratio), were reproduced by<br />

<strong>the</strong> model in which 1) wounding induces a rapid isotropic contraction <strong>of</strong> <strong>the</strong> tissue surrounding<br />

<strong>the</strong> wound followed by 2) a slower formation and contraction <strong>of</strong> a relatively thin fiber ring<br />

bordering <strong>the</strong> wound edge.<br />

The computational model is able to capture <strong>the</strong> non-trivial healing dynamics <strong>of</strong> wounds in<br />

embryonic epi<strong>the</strong>lia, lending support to a hypo<strong>the</strong>sis where wound healing is governed by two<br />

distinct cellular mechanisms.<br />

2241<br />

Nuclear protein MARVELD1 regulates activity <strong>of</strong> Rho GTPases and inhibits cell adhesion<br />

and cell motility.<br />

S. Wang 1 , J. Hu 1 , F. Han 1 , M. Shi 1 , J. He 1 , S. Liu 1 , L. Yue 1 , Y. Li 1 ; 1 Harbin Institute <strong>of</strong><br />

Technology, Harbin, China<br />

A number <strong>of</strong> cytoplasmic factors regulate cell adhesion on an extracellular matrix (ECM), which<br />

plays a crucial role in cell motility, invasion, cell signal transduction and gene expression.<br />

Activity <strong>of</strong> Rho GTPase regulates F-actin dynamics. The focal adhesions and associated F-actin<br />

structures reorganization led to <strong>the</strong> alteration <strong>of</strong> focal adhesion anchored actin stress fibers.<br />

However, its regulation via nuclear proteins remains obscure. In this study, we identified a<br />

nuclear protein, MARVELD1 that could suppress cell spreading and movement. Overexpression<br />

<strong>of</strong> MARVELD1 in A549 cells decreased <strong>the</strong> expression level <strong>of</strong> integrin β1 and<br />

vinculin, and fur<strong>the</strong>r led to dephosphorylation <strong>of</strong> FAK at Tyr 397. Also, knockdown <strong>of</strong><br />

MARVELD1 in HeLa cells and H520 cells could upregulate integin β1 and vinculin, and promote<br />

FAK Tyr397 phosphoylation. Moreover, we found that activity <strong>of</strong> Cdc42/Rac1/RhoA was<br />

regulated. And MARVELD1 partially colocalized with SC35 and interacted with nuclear cap<br />

binding protein subunit 2, CBP20. In addition, we explored that pre-mRNA processing <strong>of</strong> several<br />

focal adhesion proteins, including vinculin and actinin, was affected by MARVELD1.


TUESDAY<br />

Our studies demonstrate that <strong>the</strong> nuclear protein MARVELD1 plays a role in pre-mRNA<br />

processing <strong>of</strong> some important FAC genes and regulates activity <strong>of</strong> Rho GTPases, and <strong>the</strong>reby<br />

inhibits cell adhesion and cell motility. These studies provide a novel regulatory mechanism <strong>of</strong><br />

cell-ECM adhesion by nuclear protein in tumor cells.<br />

<strong>Molecular</strong> Motors<br />

2242<br />

The Tail region inhibits <strong>the</strong> motor activity <strong>of</strong> human myosin VIIA at molecular and cellular<br />

level.<br />

T. Sakai 1 , R. Ikebe 1 , M. Ikebe 1 ; 1 Microbiology and Physiological Systems, University <strong>of</strong><br />

Massachusetts Medical school, Worcester, MA<br />

Myosin VIIA is a responsible gene for human Usher Syndrome type 1B, which causes hearing<br />

and visual loss, and plays a role in human auditory function. We previously reported that<br />

Drosophila myosin VIIA is a monomer and <strong>the</strong> tail domain inhibits <strong>the</strong> actin activated ATPase<br />

activity under low Ca2+ condition. However, it is unknown whe<strong>the</strong>r <strong>the</strong> tail-inhibition mechanism<br />

is operating in cells. Ano<strong>the</strong>r critical issue is whe<strong>the</strong>r or not mammalian myosin VIIA is regulated<br />

by similar mechanism since it has been known that regulation mechanism <strong>of</strong> myosin is <strong>of</strong>ten<br />

quite different between vertebrate myosin and its non-vertebrate counterpart. To address <strong>the</strong>se<br />

questions, we studied <strong>the</strong> regulation mechanism <strong>of</strong> human myosin VIIA (HuM7A) at both<br />

molecular and cellular level.<br />

We studied <strong>the</strong> inhibitory function <strong>of</strong> <strong>the</strong> tail by examining <strong>the</strong> actin-activated ATPase activity <strong>of</strong><br />

HuM7A. The C-terminal deletion (-40 amino acids) <strong>of</strong> <strong>the</strong> full-length HuM7A significantly<br />

increased <strong>the</strong> ATPase activity. This result suggested that <strong>the</strong> C-terminal end <strong>of</strong> tail region<br />

inhibits <strong>the</strong> motor activity. The result is similar to <strong>the</strong> study <strong>of</strong> Drosophila myosin VIIA,<br />

suggesting that tail-inhibition mechanism is also operating in HuM7A.<br />

Our earlier study showed that <strong>the</strong> dimer formation <strong>of</strong> GFP-HuM7A HMM or GFP-HuM7A fulllength<br />

induces <strong>the</strong> filopodia tip localization <strong>of</strong> human myosin VIIA in cells, suggesting that<br />

HuM7A moves along actin bundles in filopodia to <strong>the</strong> tips. Using this system, we examined <strong>the</strong><br />

inhibitory activity <strong>of</strong> HuM7tail in cells. We co-expressed <strong>the</strong> tail with <strong>the</strong> forced-dimer <strong>of</strong> HuM7A<br />

HMM, and studied <strong>the</strong> effect <strong>of</strong> <strong>the</strong> tail on HuM7AHMM translocation to <strong>the</strong> filopodial tips.<br />

Expression <strong>of</strong> <strong>the</strong> entire tail significantly inhibited <strong>the</strong> translocation <strong>of</strong> HuM7A HMM to <strong>the</strong><br />

filopodial tips. We also found that translocation <strong>of</strong> <strong>the</strong> forced-dimer <strong>of</strong> full-length HuM7A is<br />

significantly attenated and this was facilitated by deletion <strong>of</strong> <strong>the</strong> C-terminal 40 amino acids.<br />

These results are consistent with <strong>the</strong> tail-inhibition <strong>of</strong> <strong>the</strong> ATPase activity in vitro. Present results<br />

suggested that <strong>the</strong> tail-induced regulatory mechanism is common between vertebrate and nonvertebrate<br />

myosin VIIA and is operating in vivo.<br />

2243<br />

Myosin-Va is implicated in cell adhesion and spreading.<br />

A. Ramao 1 , G. P. Nader 1,2 , E. M. Espreafico 1 ; 1 Cellular and <strong>Molecular</strong> <strong>Biology</strong>, FMRP-USP,<br />

Ribeirão Preto, Brazil, 2 Pathology and Cell <strong>Biology</strong>, Columbia University, New York, NY<br />

The main objective <strong>of</strong> this work was to assess <strong>the</strong> role <strong>of</strong> myosin-Va in cell adhesion and<br />

spreading. Here we show that MYO5A mutant melanoma cells exhibit deficient adhesion and<br />

spreading abilities, and lack <strong>the</strong> usual organization <strong>of</strong> <strong>the</strong> actin cytoskeleton. Myosin-Va<br />

expression in <strong>the</strong>se cells restored actin bundles, induced lamelipodial/filopodial growth and led<br />

to redistribution <strong>of</strong> focal adhesions towards <strong>the</strong> cell cortex. It is well known that depolymerization<br />

<strong>of</strong> microtubules in quiescent fibroblasts leads to an intense formation <strong>of</strong> focal adhesions


TUESDAY<br />

followed by dissociation <strong>of</strong> <strong>the</strong>se structures after microtubule repolymerization. To investigate a<br />

possible role for myosin-Va in <strong>the</strong> formation step or dissociation <strong>of</strong> focal adhesions, human<br />

fibroblasts from a healthy individual (control) or from a patient with deleterious mutation in <strong>the</strong><br />

MYO5A gene were left 48 hours in quiescence and treated with nocodazole for 4 hours.<br />

Confocal microscopy analysis <strong>of</strong> treated cells stained for <strong>the</strong> focal adhesion marker, talin,<br />

showed no differences between mutant and wild-type cells in <strong>the</strong> ability <strong>of</strong> focal adhesion<br />

formation nor in <strong>the</strong> disassemble <strong>of</strong> focal adhesions observed concomitantly to microtubule<br />

regrowth, 60 minutes after washing out <strong>the</strong> nocodazole. The same assay was performed with<br />

myosin-Va knocked-down WM793 human melanoma cells. Knock-down was mediated by a<br />

lentiviral vector carrying a short-hairpin RNA targeted to MYO5A mRNA (shMYO5A) or, as<br />

control, an irrelevant shRNA against bacterial LacZ. We observed that, after treatment with<br />

nocodazole, lamelipodia from WM793-shLacZ cells had numerous peripheral focal adhesions,<br />

as expected, whereas most <strong>of</strong> myosin-Va knocked-down cells ra<strong>the</strong>r showed ruffled edges and<br />

lacked focal adhesions. These data suggest that myosin-Va is required for formation <strong>of</strong> focal<br />

adhesions in melanocytic cells, but not in fibroblasts. Our laboratory has previously showed that<br />

<strong>the</strong> MYO5A-null fibroblasts used here overexpress myosin-Vb and Vc. Therefore, future studies<br />

should be conducted to answer whe<strong>the</strong>r in fibroblasts <strong>the</strong> formation <strong>of</strong> focal adhesions is<br />

compensated by one <strong>of</strong> <strong>the</strong> o<strong>the</strong>r two myosin-V paralogs, and also to uncover <strong>the</strong> mechanisms<br />

by which myosin-V functions in focal adhesion formation in melanocytic cells.<br />

Financial support: FAPESP, CNPq, CAPES, FAEPA<br />

2244<br />

Characterization <strong>of</strong> <strong>the</strong> kinesin KIF9 in mammalian cell cycle progression.<br />

J. E. Hoke 1 , M. E. Rivera 2 , A. M. Billow 1 , L. Alsina 1 , N. J. Quintyne 1 ; 1 Harriet L. Wilkes Honors<br />

College, Florida Atlantic University, Jupiter, FL, 2 Florida Atlantic University-Honors Coll, Jupiter,<br />

FL<br />

The kinesin family <strong>of</strong> microtubule motors is divided into subfamilies based upon structure and<br />

function. KIF9 is <strong>the</strong> founding member <strong>of</strong> <strong>the</strong> Kinesin-9 family, a largely uncharacterized group<br />

<strong>of</strong> kinesins. It was originally identified by sequence homology to o<strong>the</strong>r kinesins and shown to<br />

interact with <strong>the</strong> Ras-like GTPase Gem (Piddini et al., 2001). Subsequent studies have indicated<br />

that KIF9 is vital for flagellar movement in Trypanasoma brucei, likely via interaction with dynein<br />

(Demonchy et al., 2009) as well as podosome regulation (Cornfine et al., 2011). Additionally,<br />

Kinesin-9 family members have also been proposed to function in proper microtubule organizing<br />

center (MTOC) positioning and timely mitotic entry. We have examined KIF9 function in<br />

mammalian cells by using siRNA-mediated knockdown and overexpression. Our analysis has<br />

focused on normal progression through S phase and subsequent transition to, and progression<br />

through, mitosis. We have seen several effects on cell cycle progression. First, a transfected,<br />

synchronized population <strong>of</strong> cells shows a notable delay in mitotic entry. Second, <strong>the</strong>re is a delay<br />

in normal S phase progression. Third, we see that <strong>the</strong>re is a change in mitotic index and<br />

decrease in <strong>the</strong> number <strong>of</strong> anaphase and telophase cells. Finally, we see an increase in <strong>the</strong> rate<br />

<strong>of</strong> multinuclearity, a hallmark <strong>of</strong> failure <strong>of</strong> cytokinesis. Taken toge<strong>the</strong>r, we propose that KIF9 is<br />

required for normal entry and completion <strong>of</strong> mitosis, possibly via regulation <strong>of</strong> <strong>the</strong> contractile<br />

ring.


2245<br />

Kinesin-5 promotes disassembly <strong>of</strong> long kinetochore microtubules.<br />

E. S. Tubman 1 , A. Claas 1 , D. J. Odde 1 ; 1 Biomedical Engineering, University <strong>of</strong> Minnesota,<br />

Minneapolis, MN<br />

TUESDAY<br />

Kinesin-5 is a plus end directed, homotetrameric motor protein that is well known for its role in<br />

mitosis. It is widely accepted that <strong>the</strong> motor protein crosslinks antiparallel microtubules, where it<br />

walks towards <strong>the</strong> plus end <strong>of</strong> each, sliding <strong>the</strong>m apart and creating a bipolar spindle.<br />

Additionally, kinesin-5 in budding yeast plays a role in maintaining a spatial gradient <strong>of</strong><br />

kinetochores (protein complexes that link chromosomes to dynamically assembling microtubule<br />

plus ends) along <strong>the</strong> metaphase spindle. Kinesin-5 establishes this gradient by promoting<br />

disassembly <strong>of</strong> long kinetochore microtubules. We are interested in determining if kinesin-5 also<br />

controls <strong>the</strong> spatial gradient <strong>of</strong> kinetochore microtubule assembly in o<strong>the</strong>r organisms. To test<br />

this hypo<strong>the</strong>sis we used <strong>the</strong> infectious yeast Candida Albicans imaged by confocal fluorescence<br />

microscopy to measure <strong>the</strong> fluorescence distribution <strong>of</strong> <strong>the</strong> tubulin in control cells and mutant<br />

cells, where one <strong>of</strong> <strong>the</strong> two copies <strong>of</strong> <strong>the</strong> KIP1 gene (kinesin-5 in Candida) was knocked out. In<br />

control cells, a bilobed fluorescence distribution indicated that <strong>the</strong> ends <strong>of</strong> <strong>the</strong> kinetochore<br />

microtubules are drawn to an attractor zone located between <strong>the</strong> spindle pole and <strong>the</strong> spindle<br />

equator, consistent with <strong>the</strong> distribution in budding yeast. In mutant cells, we found a<br />

homogeneous fluorescence distribution, indicating a loss <strong>of</strong> <strong>the</strong> attractor zone. Our initial results<br />

here suggest that kinesin-5 acts as a kinetochore microtubule length-dependent disassembly<br />

promoter in C. Alibcans to mediate <strong>the</strong> proper alignment <strong>of</strong> chromosomes across <strong>the</strong> spindle<br />

equator during metaphase.<br />

2246<br />

Casein Kinase 2 Reverses Tail-Independent Inactivation <strong>of</strong> Kinesin-1.<br />

J. Xu 1 , B. Reddy 2 , P. Anand 2 , Z. Shu 2 , S. Cermelli 2 , M. Mattson 2 , S. Tripathy 2 , M. Hoss 2 , N.<br />

James 2 , S. King 3 , L. Huang 2 , L. Bardwell 2 , S. Gross 2 ; 1 University <strong>of</strong> California, Merced, Merced,<br />

CA, 2 University <strong>of</strong> California, Irvine, Irvine, CA, 3 University <strong>of</strong> Central Florida, Orlando, FL<br />

Kinesin-1 is a plus-end microtubule-based motor, and defects in kinesin-based transport are<br />

linked to diseases including neurodegeneration. Kinesin can auto-inhibit via a direct head-tail<br />

interaction, but is believed to be active o<strong>the</strong>rwise. Here we report a tail-independent inactivation<br />

<strong>of</strong> kinesin, reversible by <strong>the</strong> disease-relevant signaling protein, casein kinase 2 (CK2). The<br />

majority <strong>of</strong> initially active kinesin (native or tail-less) loses its ability to bind/interact with<br />

microtubules in vitro, and CK2 reverses this inactivation (~ 4-fold) without altering kinesin’s<br />

single motor properties. This activation pathway does not require motor phosphorylation, and is<br />

independent <strong>of</strong> head-tail autoinhibition. In cultured mammalian cells, reducing CK2 expression,<br />

but not kinase activity, decreases <strong>the</strong> force required to stall lipid droplet transport, consistent<br />

with a reduction in <strong>the</strong> number <strong>of</strong> active motors. These results provide <strong>the</strong> first direct evidence <strong>of</strong><br />

a protein kinase up-regulating kinesin-based transport, and suggest a novel pathway for<br />

regulating <strong>the</strong> activity <strong>of</strong> cargo-bound kinesin.<br />

2247<br />

Protein-Protein Interactions within Dynactin’s Shoulder/Sidearm.<br />

S. A. Ketcham 1 , T. A. Schroer 1 ; 1 <strong>Biology</strong>, Johns Hopkins University, Baltimore, MD<br />

Dynactin is a multi-subunit complex that acts as a cargo adaptor and processivity enhancer for<br />

<strong>the</strong> microtubule-based motors dynein and kinesin-2. Dynactin interacts with <strong>the</strong>se motors<br />

through <strong>the</strong> p150Glued subunit, which along with dynamitin and p24 comprise <strong>the</strong><br />

shoulder/sidearm. Disruption <strong>of</strong> dynactin with <strong>the</strong> chaotropic salt KI yields two subcomplexes,


TUESDAY<br />

<strong>the</strong> shoulder, which contains p24 and dynamitin, and <strong>the</strong> shoulder/sidearm, which contains p24,<br />

dynamitin and p150Glued. Although <strong>the</strong> shoulder/sidearm is released from dynactin as a stable<br />

subcomplex, <strong>the</strong> protein-protein interactions that determine its structure remain undefined. Our<br />

current work suggests that p24 serves as a bridge between dynamitin and p150Glued.<br />

Recombinant p24 is insoluble, but solubility can be restored by stepwise dialysis out <strong>of</strong><br />

denaturant in <strong>the</strong> presence <strong>of</strong> ei<strong>the</strong>r dynamitin or a fragment <strong>of</strong> p150Glued. Without ei<strong>the</strong>r<br />

binding partner, p24 remains insoluble when subjected to this procedure. We have taken<br />

advantage <strong>of</strong> <strong>the</strong> necessity <strong>of</strong> a binding partner for p24 solubility to explore p24 structure.<br />

Renaturation <strong>of</strong> a series <strong>of</strong> p24 fragments with dynamitin or p150Glued, coupled with gel<br />

filtration and velocity sedimentation to analyze <strong>the</strong> complexes that form, has allowed us to map<br />

<strong>the</strong> domains <strong>of</strong> p24 necessary for its interactions with its binding partners. These results are<br />

beginning to yield a better understanding <strong>of</strong> <strong>the</strong> structure <strong>of</strong> <strong>the</strong> shoulder/sidearm and will be<br />

exploited to gain insight into how <strong>the</strong> shoulder/sidearm anchors to <strong>the</strong> Arp1 minifilament.<br />

Cell Division<br />

2248<br />

Cellular Size Checkpoints During G1/S- and G2/M-Transition Mediate Contact Inhibition<br />

Of Proliferation.<br />

S. J. Streichan 1 , C. R. Hoerner 1 , D. Holzer 1 , T. Schneidt 1 , L. Hufnagel 1 ; 1 Cell <strong>Biology</strong> &<br />

Biophysics Unit, European <strong>Molecular</strong> <strong>Biology</strong> Laboratory (EMBL), Heidelberg, Germany<br />

Tissue and organ development is characterized by tightly controlled patterns <strong>of</strong> coordinated cell<br />

growth, cell division, and cell rearrangements. Non-cancerous epi<strong>the</strong>lial cells in culture show a<br />

similar behavior in terms <strong>of</strong> regulated proliferation: initially, <strong>the</strong>y proliferate at high rate and<br />

compensate for <strong>the</strong>ir reduced size after cell division by cell growth. Then, after reaching<br />

confluence cells undergo size-reductive divisions and <strong>the</strong> rate <strong>of</strong> cell division drops markedly, a<br />

phenomenon called contact inhibition <strong>of</strong> proliferation (CI). However, it remains elusive how cells<br />

coordinate growth and division and how during CI <strong>the</strong> cell cycle <strong>of</strong> individual cells is regulated<br />

according to <strong>the</strong> cell density in <strong>the</strong> cell collective.<br />

Here we use fluorescent time-lapse live cell imaging, quantitative image analysis, and<br />

biophysical modeling to study <strong>the</strong> link between cell growth and cell proliferation in Madin Darby<br />

canine kidney (MDCK) cells. Based on <strong>the</strong> predictions <strong>of</strong> our physical description we perform<br />

biophysical tests including active and passive manipulation <strong>of</strong> cell size. First, we find that cells<br />

have a critical size when entering S phase. Second, reduced cell size after reentry into <strong>the</strong> cell<br />

cycle results in prolonged G2 phase. Third, direct manipulation <strong>of</strong> cell size by stretching <strong>of</strong> a<br />

contact-inhibited cell collective is sufficient to reverse CI as cells reenter <strong>the</strong> cell cycle and<br />

progress through mitosis.<br />

We suggest that mechanical constraints in tissues may delay cell cycle reentry and progression<br />

by activating size checkpoints acting during <strong>the</strong> G1/S- and G2/M-transition.<br />

2249<br />

Concentration-dependent Correlation <strong>of</strong> Cell Cycle and Apoptosis Impacts <strong>of</strong> Anti-Cancer<br />

Compounds.<br />

K. Tran 1 , K. Gillis 1 , A. Khan 1 , J. Clor 1 , K. Tyagarajan 1 ; 1 EMD Millipore, Millipore Corporation,<br />

Hayward, CA<br />

Recent research has demonstrated <strong>the</strong> delicate relationship between <strong>the</strong> opposing processes <strong>of</strong><br />

cell cycle progression and apoptosis. Several anti-cancer compounds have been demonstrated<br />

to cause apoptosis and subsequently have been shown to cause arrests in specific phases <strong>of</strong>


TUESDAY<br />

<strong>the</strong> cell cycle. The study <strong>of</strong> cell cycle impacts in addition to apoptosis can thus bring meaningful<br />

understanding <strong>of</strong> <strong>the</strong> diverse effects <strong>of</strong> cellular modulators and compound treatment and provide<br />

deeper understanding <strong>of</strong> <strong>the</strong> inter-relationship between cell cycle and apoptosis. In this study,<br />

we evaluated a series <strong>of</strong> anti-cancer compounds with known apoptotic activity to determine <strong>the</strong><br />

sequence <strong>of</strong> impact on cell cycle versus apoptosis induction for multiple cell types. Jurkat and<br />

HeLa cells were treated with multiple concentrations <strong>of</strong> compounds and <strong>the</strong> impact on cell cycle<br />

and apoptosis (based on Annexin V binding) on different dosages <strong>of</strong> treatment were evaluated.<br />

Our studies demonstrate that multiple compounds could modulate different cell cycle phases<br />

depending on <strong>the</strong> concentration and cell type used. Fur<strong>the</strong>r <strong>the</strong> sequence <strong>of</strong> impacts on cell<br />

cycle and apoptosis could be very different. Our results here demonstrate that treatment with<br />

Etoposide, a DNA topoisomerase II inhibitor at low concentrations results in <strong>the</strong> arrest <strong>of</strong> cells in<br />

<strong>the</strong> G2M stage under conditions where little apoptosis is observed. With increased<br />

concentration <strong>of</strong> etoposide, increase in <strong>the</strong> % <strong>of</strong> cells in <strong>the</strong> S phase <strong>of</strong> cell cycle is observed<br />

along with increased apoptosis. Thus for etoposide, cell cycle impacts were observed at<br />

significantly lower concentration than impacts on apoptosis. Staurosporine demonstrated both<br />

an arrest in G2/M phase with concurrent impacts on apoptosis at low concentrations, higher<br />

concentrations showed increase in <strong>the</strong> percentage <strong>of</strong> cells in <strong>the</strong> G0/G1 phase. Gambogic acid,<br />

a potential anti-cancer agent resulted in significant apoptosis and death but no impacts on cell<br />

cycle were observed. Nocodazole demonstrated impacts on cell cycle and apoptosis<br />

concurrently. Parallel analysis <strong>of</strong> apoptosis data along with cell cycle effects will be critical for<br />

understanding <strong>the</strong> linkage between <strong>the</strong>se processes and provide for a more complete<br />

understanding <strong>of</strong> impacts <strong>of</strong> modulators <strong>of</strong> cell cycle and cell death and <strong>the</strong> design <strong>of</strong> effective<br />

cellular study models.<br />

2250<br />

Phospho-regulation <strong>of</strong> <strong>the</strong> anillin-related scaffolding protein Mid1 in fission yeast<br />

cytokinesis.<br />

A. K. DeWitt 1 , E. Kranz 1 , J. Phelan 1 , B. Nader 1 , K. L. Gould 2 , D. M. Clifford Hart 1 ; 1 Cell and<br />

<strong>Molecular</strong> <strong>Biology</strong>, Grand Valley State University, Allendale, MI, 2 Department <strong>of</strong> Cell and<br />

Developmental <strong>Biology</strong>, Howard Hughes Medical Institute & Vanderbilt University School <strong>of</strong><br />

Medicine, Nashville, TN<br />

During cytokinesis, physical separation <strong>of</strong> one cell into two identical cells occurs through<br />

constriction <strong>of</strong> a protein-rich ring structure, called <strong>the</strong> contractile ring. The contractile ring is<br />

composed <strong>of</strong> F-actin, type II myosin and more than 100 cytokinetic ring proteins. In fission<br />

yeast, <strong>the</strong> anillin-related protein Mid1 plays a critical role in organizing <strong>the</strong> early steps <strong>of</strong><br />

contractile ring formation and functions as a scaffold to bridge <strong>the</strong> cell cortex with <strong>the</strong> contractile<br />

ring. Cells lacking mid1 form <strong>of</strong>f-centered, highly disorganized ring structures and exhibit severe<br />

cytokinesis defects. Coincident with its cortical accumulation, Mid1 becomes hyperphosphorylated.<br />

Our previous research demonstrates that cyclin-dependent kinase, Cdc2, and<br />

<strong>the</strong> polo-like kinase, Plo1, directly phosphorylate Mid1. In addition to consensus phosphorylation<br />

motifs for Plo1 and Cdc2, Mid1 contains several RXXS motifs, which fit <strong>the</strong> phosphorylation<br />

consensus sequence for Sid2 kinase. Sid2 is <strong>the</strong> most downstream kinase in <strong>the</strong> Septation<br />

Initiation Network signaling cascade, which signals from <strong>the</strong> spindle pole body to trigger<br />

constriction <strong>of</strong> <strong>the</strong> contractile ring. To identify specific sites <strong>of</strong> phosphorylation, a combination <strong>of</strong><br />

site-directed mutagenesis (serine to non-phosphorylatable alanine) with mass spectroscopy and<br />

two-dimensional phosphopeptide mapping were used. Both methods identified multiple Sid2<br />

phosphorylation sites within Mid1. To analyze <strong>the</strong> significance <strong>of</strong> Sid2 phosphorylation in vivo,<br />

phospho-site mutants were generated at <strong>the</strong> endogenous mid1 locus and examined for cell<br />

division defects. Interestingly, phospho-site mutants displayed cell division defects, including<br />

sensitivity to low dose latrunculin A and disorganized actin localization. While phospho-site


TUESDAY<br />

mutants maintained <strong>the</strong> mitotic spindle checkpoint, <strong>the</strong> majority <strong>of</strong> cells exhibited severe polarity<br />

phenotypes and Mid1 protein levels increased when compared to checkpoint activated cells<br />

expressing wild-type Mid1. Given that Mid1 departure from <strong>the</strong> contractile ring coincides with<br />

Sid2 relocalization to <strong>the</strong> division site, Sid2 may temporally regulate <strong>the</strong> interaction <strong>of</strong> Mid1 with<br />

<strong>the</strong> membrane or o<strong>the</strong>r contractile ring components; our initial experiments favor Sid2 regulating<br />

Mid1 dissociation from <strong>the</strong> cell cortex.<br />

2251<br />

Regulation <strong>of</strong> Myosin-II Dynamics and its Implication in <strong>the</strong> Assembly <strong>of</strong> <strong>the</strong> Cytokinesis<br />

Machine in S. cerevisiae.<br />

C. Wloka 1,2 , E. Bi 1 ; 1 Cell and Developmental <strong>Biology</strong>, University <strong>of</strong> Pennsylvania, Philadelphia,<br />

PA, 2 Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin, Germany<br />

Cytokinesis in animal and fungal cells utilizes a contractile actomyosin ring (AMR) to power <strong>the</strong><br />

ingression <strong>of</strong> <strong>the</strong> cleavage furrow. The AMR consists <strong>of</strong> myosin-II and actin filaments. However,<br />

how <strong>the</strong> AMR is assembled and disassembled during <strong>the</strong> cell cycle is not clearly understood in<br />

any organism. We have analyzed <strong>the</strong> dynamics <strong>of</strong> 13 cytokinesis proteins during <strong>the</strong> cell cycle,<br />

including <strong>the</strong> sole myosin-II, Myo1, in <strong>the</strong> budding yeast Saccharomyces cerevisiae by<br />

fluorescence recovery after photo-bleaching. We found that Myo1 is dynamic until <strong>the</strong> onset <strong>of</strong><br />

anaphase when it becomes progressively immobile. Fur<strong>the</strong>r analysis indicates that <strong>the</strong><br />

immobility <strong>of</strong> Myo1 during cytokinesis does not require its motor domain and actin filaments but<br />

requires <strong>the</strong> putative assembly domain near its C-terminus, which regulates Myo1 localization<br />

and cytokinesis. Thus, <strong>the</strong> putative higher-order assembly is important for Myo1 localization,<br />

dynamics, and its role in cytokinesis. These data also indicate that <strong>the</strong> AMR is progressively<br />

disassembled during its constriction without high turnover.<br />

We also found that proteins involved in AMR assembly and septum formation, such as Iqg1<br />

(IQGAP), H<strong>of</strong>1 (a F-BAR protein), and Inn1 are immobile whereas those involved in actin ring<br />

assembly (formin and tropomyosin) are highly dynamic during cytokinesis. Strikingly, <strong>the</strong><br />

immobility <strong>of</strong> all examined proteins depends on <strong>the</strong> presence <strong>of</strong> Myo1. This and <strong>the</strong> observation<br />

that Myo1 is required for actin ring assembly suggest that Myo1 plays a scaffolding role in <strong>the</strong><br />

assembly <strong>of</strong> <strong>the</strong> cytokinesis machine.<br />

2252<br />

Orbit/CLASP plays a role as linker connecting cortical microtubules and actomyosin ring<br />

in Drosophila male meiosis.<br />

D. Kitazawa 1 , D. Hayashi 1 , C. Miyauchi 1 , Y. Inoue 1 ; 1 Kyoto Institute <strong>of</strong> Technology, Insect<br />

Biomedical Research Center, Kyoto, Japan<br />

Microtubules (MTs) near <strong>the</strong> cell cortex are essential for continuous constriction <strong>of</strong> <strong>the</strong><br />

contractile ring in cytokinesis. However, <strong>the</strong> way in which MTs interact with <strong>the</strong> contractile<br />

apparatus has not been completely unveiled. In previous studies, we have shown that Orbit is<br />

required for stabilization <strong>of</strong> <strong>the</strong> MT populations essential for cytokinesis. Therefore, it is possible<br />

to speculate that Orbit plays an important role in cleavage furrow (CF) initiation in Drosophila. A<br />

body <strong>of</strong> evidence indicating that Orbit possesses a binding activity to both MTs and F-Actin in<br />

Drosophila and Mammalian cells implies ano<strong>the</strong>r role in initiation <strong>of</strong> cytokinesis. In this study, we<br />

carefully examined localization <strong>of</strong> GFP-tagged Orbit in Drosophila male meiosis. We showed<br />

that Orbit changes its localization from around <strong>the</strong> spindle matrix towards prospective CF<br />

regions at late anaphase. It continuously stayed on <strong>the</strong> ring in <strong>the</strong> middle <strong>of</strong> <strong>the</strong> cell. Although<br />

formation <strong>of</strong> <strong>the</strong> Orbit ring depends on both MTs and F-Actin, Orbit maintains its localization on<br />

<strong>the</strong> CF even in <strong>the</strong> absence <strong>of</strong> Myosin II. We showed that Orbit directly binds to F-Actin and <strong>the</strong><br />

heavy chain <strong>of</strong> Myosin II on <strong>the</strong> CF. Surprisingly, ectopic localization <strong>of</strong> Orbit on <strong>the</strong> cortex


TUESDAY<br />

induces contractile ring formation in situ and it was followed by CF ingression. Accumulation <strong>of</strong><br />

Orbit on <strong>the</strong> CF is dependent on MTs and F-Actin. In order to look for transport proteins<br />

conveying <strong>the</strong> Orbit toward <strong>the</strong> CF, we <strong>the</strong>n performed knockdown experiments to examine<br />

whe<strong>the</strong>r all known MT-based motors and proteins displaying a genetic interaction with Orbit are<br />

required for cytokinesis. Among several genes that we have shown are critical for cytokinesis,<br />

we especially focused on CLIP190 and its relationship with Orbit. We found that CLIP190 is<br />

required for transport <strong>of</strong> Orbit to <strong>the</strong> prospective CF site independent <strong>of</strong> <strong>the</strong> MT binding domain<br />

<strong>of</strong> Orbit. Our current data suggests that Orbit plays a central role in contractile ring formation as<br />

a linker connecting MTs with Actomyosin ring.<br />

2253<br />

Fission yeast cyclin-dependent kinase, Cdc2, regulates <strong>the</strong> Mid1 scaffolding protein<br />

during cytokinesis.<br />

J. B. Hackett 1 , J. Phelan 1 , K. L. Gould 2 , D. M. Clifford Hart 1 ; 1 Cell and <strong>Molecular</strong> <strong>Biology</strong>, Grand<br />

Valley State University, Allendale, MI, 2 Howard Hughes Medical Institute, Department <strong>of</strong> Cell<br />

and Developmental <strong>Biology</strong>, Vanderbilt University School <strong>of</strong> Medicine, Nashville, TN<br />

Cytokinesis is <strong>the</strong> final stage <strong>of</strong> cell division and utilizes a highly conserved actin-myosin<br />

contractile ring that forms medially around <strong>the</strong> cell to pinch <strong>the</strong> cell membranes toge<strong>the</strong>r<br />

resulting in physical separation <strong>of</strong> one cell into two. The conserved anillin homolog, Mid1, is a<br />

foundational part <strong>of</strong> <strong>the</strong> fission yeast contractile ring, functioning as <strong>the</strong> initial organizer and anchoring<br />

<strong>the</strong> structure to <strong>the</strong> cell membrane. Mid1 mutants show severe defects in contractile ring assembly and<br />

medial placement <strong>of</strong> <strong>the</strong> division site. Coincident with contractile ring formation, Mid1 is hyperphosphorylated<br />

and our previous research identified at least two mitotic kinases that directly<br />

phosphorylate Mid1. Mid1 phosphorylation by <strong>the</strong> cyclin-dependent kinase Cdc2 at Threonine 517<br />

facilitates association between <strong>the</strong> polo-like kinase, Plo1, and <strong>the</strong> C-terminus <strong>of</strong> Mid1. To identify<br />

additional Cdc2 phosphorylation sites within Mid1, we implemented a combination <strong>of</strong> mutagenesis and<br />

two-dimensional phosphopeptide mapping. Mutation <strong>of</strong> Serine 28 to a non-phosphorylatable alanine<br />

residue reduced phosphorylation <strong>of</strong> Mid1 N-terminus and this site fits a consensus motif for Plo1<br />

binding. Fission yeast cells expressing Mid1 phospho-site mutations show sensitivity to low dose<br />

latrunculin A treatment and bypass <strong>the</strong> mitotic spindle checkpoint. Surprisingly, phosphosite mutants<br />

with an activated mitotic spindle checkpoint continue through mitosis but accumulate as septated cells.<br />

This research highlights a regulatory role for Plo1 and Mid1 in early and late stages <strong>of</strong> cytokinesis.<br />

2254<br />

Centrosomal Protein 55 Stability Is Negatively Regulated BY P53 Through POLO-Like<br />

Kinase 1.<br />

Y-C. Chang 1,2 , P. Ouyang 3,4 ; 1 Graduate Institute <strong>of</strong> Clinical Medicine Sciences, Chang Gung<br />

University, Guei-San, Taiwan, 2 Nuclear Medicine and <strong>Molecular</strong> Imaging Center, Chang Gung<br />

Memorial Hospital, Guei-San, Taiwan, 3 Anatomy, Chang Gung University, Guei-Shan, Taiwan,<br />

4 <strong>Molecular</strong> Medicine Research Center, Chang Gung University, Guei-San, Taiwan<br />

Centrosomal protein 55 (Cep55), which is localized to <strong>the</strong> centrosome in interphase cells and<br />

recruited to <strong>the</strong> midbody during cytokinesis, is a regulator required for <strong>the</strong> completion <strong>of</strong> cell<br />

abscission. Upregulation <strong>of</strong> Cep55 and inactivation <strong>of</strong> p53 occur in <strong>the</strong> majority <strong>of</strong> human<br />

cancers, raising <strong>the</strong> possibility <strong>of</strong> a link between <strong>the</strong>se two genes. In this study, we evaluated<br />

<strong>the</strong> role <strong>of</strong> p53 in Cep55 regulation. We demonstrated that Cep55 expression levels are well<br />

correlated with cancer cell growth rate, and that p53 is able to negatively regulate Cep55 protein<br />

and promoter activity. Downregulation <strong>of</strong> expression <strong>of</strong> Cep55 was accompanied by repression<br />

<strong>of</strong> polo-like kinase 1 (Plk1) levels due to p53 induction. Overexpression <strong>of</strong> Plk1 and knockdown


TUESDAY<br />

<strong>of</strong> p53 expression both enhanced <strong>the</strong> post-translational protein stability <strong>of</strong> Cep55. BI 2356, a<br />

selective Plk1 inhibitor, however, prevented Cep55 accumulation in p53 knockdown cells, while<br />

persistently keeping Plk1 levels elevated. Our results, <strong>the</strong>refore, indicate <strong>the</strong> existence <strong>of</strong> a p53-<br />

Plk1-Cep55 axis in which p53 negatively regulates expression <strong>of</strong> Cep55 through Plk1 which, in<br />

turn, is a positive regulator <strong>of</strong> Cep55 protein stability.<br />

2255<br />

Cleavage furrow organization requires PIP2 mediated recruitment <strong>of</strong> anillin.<br />

J. Liu 1 , D. Ceccareli 2 , G. Fairn 3 , F. Sicheri 1,2 , A. Wilde 1 ; 1 <strong>Molecular</strong> Genetics, University <strong>of</strong><br />

Toronto, Toronto, ON, Canada, 2 SLRI, Toronto, Canada, 3 Hospital for Sick Children, Toronto,<br />

ON, Canada<br />

Anillin is a key component <strong>of</strong> <strong>the</strong> cytokinetic machinery that is essential for <strong>the</strong> successful<br />

completion <strong>of</strong> cytokinesis. Anillin is a multi-domain protein that has <strong>the</strong> potential to link <strong>the</strong><br />

actomyosin and septin cytoskeletons. It also has a PH domain involved in targeting anillin to <strong>the</strong><br />

cleavage furrow. We sought to examine <strong>the</strong> mechanism by which <strong>the</strong> PH domain targets anillin<br />

to <strong>the</strong> cleavage furrow. We find that <strong>the</strong> PH domain <strong>of</strong> anillin interacts with phosphatidylinositol<br />

phosphate lipids (PIPs), including PI(4,5)P2, which is enriched in <strong>the</strong> furrow. Reduction <strong>of</strong><br />

cellular PI(4,5)P2 or mutations in <strong>the</strong> PH domain <strong>of</strong> anillin that specifically disrupt <strong>the</strong> interaction<br />

with PI(4,5)P2, interfere with <strong>the</strong> localization <strong>of</strong> anillin to <strong>the</strong> furrow. Reduced expression <strong>of</strong><br />

anillin disrupts symmetric furrow ingression in <strong>the</strong> middle <strong>of</strong> <strong>the</strong> cell. However, symmetrical<br />

furrow ingression and division plane stability can be rescued in anillin depleted cells by targeting<br />

ectopically expressed anillin to <strong>the</strong> furrow using an alternate PI(4,5)P2 binding module. These<br />

data demonstrate an anillin-PI(4,5)P2 interaction targets anillin to <strong>the</strong> cleavage furrow and is<br />

required for stable, symmetrical ingression <strong>of</strong> <strong>the</strong> furrow during cytokinesis.<br />

2256<br />

Sauron is <strong>the</strong> Drosophila orthologue <strong>of</strong> <strong>the</strong> oncogene GOLPH3.<br />

S. Sechi 1 , V. Mattei 2 , G. D. Raffa 3 , G. Belloni 3 , G. Colotti 1 , M. G. Giansanti 1 ; 1 Istituto di Biologia<br />

e Patologia molecolari c/o Università Sapienza, CNR, Roma, Italy, 2 Consorzio Universitario<br />

Sabina Universitas Rieti, Rieti, Italy, 3 Dipartimento di Biologia e Biotecnologie, Università<br />

Sapienza, Roma, Italy<br />

Drosophila male meiosis provides an excellent cell system for <strong>the</strong> molecular dissection <strong>of</strong><br />

cytokinesis. Drosophila spermatocytes are considerably larger than most somatic cells and <strong>the</strong><br />

spindle assembly checkpoint is not stringent in spermatocytes, allowing <strong>the</strong> characterization <strong>of</strong><br />

genes whose products are required for multiple stages <strong>of</strong> cell division in addition to cytokinesis.<br />

In <strong>the</strong> course <strong>of</strong> a large screen for mutants defective in spermatocyte cytokinesis, we have<br />

isolated sauron (sau), a male sterile mutant with defects in both central spindle formation and<br />

contractile ring assembly. A P element lethal insertion (l(2)s5379) in <strong>the</strong> CG7085 gene failed to<br />

complement <strong>the</strong> sau 1 mutation. DNA sequencing <strong>of</strong> <strong>the</strong> sau 1 allele revealed a single point<br />

mutation in CG7085, resulting in a Glu-Lys amino acid substitution at <strong>the</strong> C-terminus <strong>of</strong> <strong>the</strong><br />

predicted protein. These results toge<strong>the</strong>r indicate that sau encodes a polypeptide <strong>of</strong> 294 amino<br />

acids which is 70% identical to human GOLPH3 (alias Gpp34, Gmx33, MIDAS). GOLPH3 is a<br />

Golgi protein which has recently been recognized as a potent oncogene amplified in many<br />

human cancers and implicated in cellular transformation via changes in <strong>the</strong> activity <strong>of</strong> mTOR.<br />

We have generated transgenic flies that express GFP-Sau under <strong>the</strong> control <strong>of</strong> tubulin<br />

promoter. The GFP-Sau protein rescues <strong>the</strong> defects <strong>of</strong> sau 1 , confirming that <strong>the</strong> cytokinesis<br />

phenotype is <strong>the</strong> consequence <strong>of</strong> alterations in <strong>the</strong> Drosophila GOLPH3 ortholog. We have also<br />

raised polyclonal antibodies against Sauron and started immun<strong>of</strong>luorescence analysis in fixed<br />

spermatocytes. Immunostaining <strong>of</strong> primary spermatocytes revealed that <strong>the</strong> endogenous protein


TUESDAY<br />

is enriched in <strong>the</strong> Golgi <strong>of</strong> prophase spermatocytes and accumulates at <strong>the</strong> cleavage site during<br />

early telophase suggesting an early role in cytokinesis.<br />

2257<br />

Microtubule-free cytokinesis in vertebrate somatic cells: <strong>the</strong> roles <strong>of</strong> Plk1 and Aurora B<br />

kinase.<br />

E. H. Hinchcliffe 1 ; 1 Hormel Institute, University <strong>of</strong> Minnesota, Austin, MN<br />

Cytokinetic furrow formation requires <strong>the</strong> initiation <strong>of</strong> cortical contractility, and its restriction to<br />

<strong>the</strong> cell’s equator. Following anaphase onset spindle microtubules (MTs) and <strong>the</strong> mitotic kinases<br />

Aurora B (Aur B) and Polo-like kinase 1 (Plk1) are thought to drive furrow formation, but how<br />

<strong>the</strong>se activities are coordinated remains poorly understood. Here we use live-cell imaging and a<br />

temperature-based spindle MT depolymerization/re-growth assay to examine cytokinesis in<br />

vertebrate somatic cells. Spindle MTs disassembled in ei<strong>the</strong>r early anaphase (


2260<br />

Delivery <strong>of</strong> <strong>the</strong> cytokinetic signal to <strong>the</strong> plasma membrane – analysis <strong>of</strong> <strong>the</strong> RhoGEF<br />

Ect2.<br />

K-C. Su 1 , T. Takaki 1 , M. Petronczki 1 ; 1 Cell Division and Aneuploidy, Cancer Research UK -<br />

London Research Institute, Clare Hall, Potters Bar, England<br />

TUESDAY<br />

Cytokinesis is <strong>the</strong> final stage <strong>of</strong> cell division and leads to <strong>the</strong> birth <strong>of</strong> two individual daughter<br />

cells. During <strong>the</strong> process <strong>of</strong> cytokinesis, <strong>the</strong> ingression <strong>of</strong> cleavage furrow divides <strong>the</strong> cytoplasm<br />

<strong>of</strong> <strong>the</strong> mo<strong>the</strong>r cell.<br />

In animal cells, cytokinesis is controlled by activation <strong>of</strong> <strong>the</strong> small GTPase RhoA, which initiates<br />

<strong>the</strong> formation <strong>of</strong> <strong>the</strong> contractile ring at <strong>the</strong> equatorial cell cortex during anaphase. It is well<br />

established that <strong>the</strong> mitotic spindle determines <strong>the</strong> position and activity <strong>of</strong> <strong>the</strong> contractile<br />

machinery at <strong>the</strong> plasma membrane. However, <strong>the</strong> precise mechanism by which <strong>the</strong><br />

microtubule-associated signalling complexes control <strong>the</strong> cytokinetic machinery at <strong>the</strong> membrane<br />

is poorly understood.<br />

The conserved RhoGEF protein epi<strong>the</strong>lial cell transforming sequence 2 (Ect2) is essential for<br />

cleavage furrow formation and RhoA activation at <strong>the</strong> equatorial cortex. Ect2, through its Nterminal<br />

tandem BRCT domains, interacts with <strong>the</strong> centralspindlin subunit MgcRacGAP leading<br />

to Ect2 recruitment to <strong>the</strong> spindle midzone. In its C-terminal region, Ect2 contains a guanine<br />

nucleotide exchange factor domain and a pleckstrin homology domain, <strong>the</strong> detailed function <strong>of</strong><br />

ei<strong>the</strong>r remain to be understood. We are using genetic, cell biological, and biochemical assays to<br />

address how Ect2 initiates cytokinesis at <strong>the</strong> plasma membrane in a spatially and temporally<br />

controlled manner.<br />

Our analysis will provide new insights into <strong>the</strong> molecular mechanism that delivers <strong>the</strong> cytokinetic<br />

signal to <strong>the</strong> equatorial cortex during anaphase in animal cells.<br />

2261<br />

Control <strong>of</strong> Mitotic Exit Network (MEN) signaling in S. cerevisiae meiosis.<br />

M. Attner 1 , A. Amon 1,2 ; 1 David H. Koch Institute for Integrative Cancer Research, Dept <strong>of</strong><br />

<strong>Biology</strong>, Massachusetts Inst Technology, Cambridge, MA, 2 Howard Hughes Medical Institute<br />

When cells exit from mitosis, cyclin dependent kinases (CDKs) are inactivated. In budding<br />

yeast, Cdc14, a protein phosphatase that is released from its inhibitor in <strong>the</strong> nucleolus during<br />

anaphase, triggers CDK inactivation. Cdc14 release is controlled by <strong>the</strong> mitotic exit network<br />

(MEN), an essential GTPase signaling cascade. How CDK activity is regulated in meiosis is less<br />

clear. During meiosis, a diploid cell undergoes two rounds <strong>of</strong> chromosome segregation following<br />

one round <strong>of</strong> DNA replication, resulting in <strong>the</strong> formation <strong>of</strong> four haploid gametes. At <strong>the</strong> meiosis<br />

I-meiosis II transition, CDK activity is thought to be low enough to allow <strong>the</strong> meiosis I spindle to<br />

dissemble but high enough to prevent DNA re-replication. To understand how <strong>the</strong> cell achieves<br />

this balance at <strong>the</strong> meiosis I-meiosis II transition, we have characterized <strong>the</strong> mitotic exit network<br />

(MEN) in meiosis. We have found that <strong>the</strong> MEN is not required and not active at <strong>the</strong> meiosis Imeiosis<br />

II transition. However, <strong>the</strong> MEN becomes active during anaphase II and is required for<br />

timely exit from meiosis II. We fur<strong>the</strong>r show that MEN signaling in meiosis is regulated differently<br />

from MEN signaling in mitosis. Whereas localization to <strong>the</strong> spindle pole body (SPB) is critically<br />

important for MEN signaling in mitosis, meiotic MEN signaling relies on <strong>the</strong> regulated binding <strong>of</strong><br />

downstream MEN components and does not depend on SPB components. Taken toge<strong>the</strong>r, our<br />

data suggest that exit from meiosis I and II are regulated by different mechanisms than exit from<br />

mitosis, which may help establish permissive conditions for <strong>the</strong> meiotic divisions.


TUESDAY<br />

2262<br />

A midzone-based ruler adjusts chromosome compaction to anaphase spindle length.<br />

G. E. Neurohr 1,2 , A. Naegeli 2 , I. Titos 1 , D. Theler 2 , B. Greber 2 , Y. Barral 2 , M. Mendoza 1 ; 1 Center<br />

<strong>of</strong> Genomic Regulation (CRG), Barcelona, Spain, 2 Institute <strong>of</strong> Biochemistry, ETH Zurich, Zurich,<br />

Switzerland<br />

For successful partitioning <strong>of</strong> <strong>the</strong> genetic material during cell division, mitotic chromosomes<br />

must condense enough to be segregated by <strong>the</strong> fully elongated spindle, while spindles must<br />

elongate enough to segregate <strong>the</strong> bulkiest chromosome. Although both maximal spindle length<br />

and chromosome size can vary even within a given species, we know little about how cells cope<br />

with <strong>the</strong>se changes to ensure faithful segregation <strong>of</strong> <strong>the</strong> genetic material under such conditions.<br />

We have managed to fuse <strong>the</strong> two longest chromosomes <strong>of</strong> <strong>the</strong> budding yeast S. cerevisiae to<br />

generate a yeast strain containing an oversized chromosome. The long arm <strong>of</strong> this chromosome<br />

exceeds <strong>the</strong> longest wild type arm by 50%. Remarkably, <strong>the</strong>se cells were able to cope with <strong>the</strong><br />

presence <strong>of</strong> <strong>the</strong> long chromosome without delaying anaphase or increasing spindle length, and<br />

without slowing down <strong>the</strong>ir growth rate. Instead, cells adapted through increasing <strong>the</strong><br />

condensation <strong>of</strong> <strong>the</strong> compound chromosome specifically. Consistently, cells carrying <strong>the</strong> fused<br />

chromosomes became more sensitive to loss <strong>of</strong> condensin activity. In addition we observed that<br />

length-dependent stimulation <strong>of</strong> condensation took place during anaphase and depended on <strong>the</strong><br />

spindle midzone, aurora/Ipl1 activity, and phosphorylation <strong>of</strong> histone H3 on Ser10. We propose<br />

that <strong>the</strong> spindle midzone functions as a ruler that adapts <strong>the</strong> condensation <strong>of</strong> long chromosome<br />

arms to spindle length to promote <strong>the</strong>ir faithful segregation during anaphase, regardless <strong>of</strong><br />

variations in chromosome or spindle length.<br />

2263<br />

Identifying Novel Kinases Involved in Mitotic Progression Using Kinome-wide Screens.<br />

H. Chen 1 , H. Ma 1 , Y. Tsang 1 , C. Cheung 1 , S-S. Ng 1 , J. Zhang 1 , R. Poon 1 ; 1 Division <strong>of</strong> Life<br />

Science, Hong Kong University <strong>of</strong> Science and Technology, Hong Kong, China<br />

The main purpose <strong>of</strong> mitosis is to segregate sister chromatids into two nascent cells, such that<br />

each daughter cell inherits one complete set <strong>of</strong> chromosomes. Protein phosphorylation is one <strong>of</strong><br />

<strong>the</strong> main post-translational regulation mechanisms <strong>of</strong> mitosis. A number <strong>of</strong> protein kinases such<br />

as cyclin-dependent kinases, Aurora kinases, Polo-like kinases (PLKs), NIMA-related kinases,<br />

and spindle assembly checkpoints kinases BUB1, BUBR1 and MPS1, are known to be involved<br />

in <strong>the</strong> control <strong>of</strong> various mitotic processes. Dysfunction <strong>of</strong> phosphorylation events regulated by<br />

<strong>the</strong>se protein kinases would cause abnormal chromosome segregation as well as genome<br />

instability. However, <strong>the</strong> whole repertoire <strong>of</strong> mitotic kinases probably remains to be deciphered.<br />

In this study, we sought to identify novel mitotic kinases with RNAi approaches. Mouse<br />

fibroblasts NIH3T3 modified with histone H2B-GFP expression was transfected with a mouse<br />

kinome siRNA library to deplete individual kinases. Novel kinase candidates essential for<br />

mitosis progression were identified according to an increased mitotic index. Subsequently, <strong>the</strong><br />

candidate mitotic kinases were compared with a previous screen in Drosophila. Several<br />

matched candidates from <strong>the</strong> mouse and Drosophila screens were chosen for fur<strong>the</strong>r analysis in<br />

human cancer cells. Approaches including flow cytometry, live cell imaging, and immunostaining<br />

were applied to confirm <strong>the</strong> functional impact <strong>of</strong> <strong>the</strong>se candidates in mitosis. These results<br />

indicate that in addition to <strong>the</strong> set <strong>of</strong> well-studied kinases, several novel protein kinases may<br />

also play critical role in mitosis.


TUESDAY<br />

2264<br />

A stochastic model <strong>of</strong> kinetochore–microtubule attachment accurately describes<br />

chromosome segregation.<br />

G. Gay 1 , T. Cour<strong>the</strong>oux 1 , C. Reyes, 1 , S. Tournier 1 , Y. P. Gachet 1 ; 1 LBCMCP, CNRS, Toulouse,<br />

France<br />

Erroneous attachments <strong>of</strong> spindle microtubules to kinetochores are frequent in early mitosis.<br />

Most <strong>of</strong> <strong>the</strong>se are corrected before <strong>the</strong> onset <strong>of</strong> anaphase by a mechanism involving <strong>the</strong> protein<br />

kinase Aurora B which destabilizes kinetochore microtubules in <strong>the</strong> absence <strong>of</strong> tension between<br />

sister chromatids. Here, we describe a minimal ma<strong>the</strong>matical model <strong>of</strong> mitotic chromosome<br />

segregation based on stochastic attachment and detachment <strong>of</strong> kinetochore microtubules. The<br />

model accurately reproduces <strong>the</strong> process and timing <strong>of</strong> correct chromosome bi-orientation and<br />

segregation seen in fission yeast. Prevention <strong>of</strong> attachment defects requires both appropriate<br />

kinetochore orientation and an Aurora B-like activity. The model also reproduces abnormal<br />

chromosome segregation behavior (due, for example, to inhibition <strong>of</strong> Aurora B). It predicts that,<br />

in metaphase, merotelic attachment is prevented by a kinetochore orientation effect and<br />

corrected by an Aurora B-like activity whereas in anaphase it is corrected through unbalanced<br />

forces applied to <strong>the</strong> kinetochore. These unbalanced forces are sufficient to prevent aneuploidy<br />

and <strong>the</strong> ‘cut’ phenotype.<br />

2265<br />

High Throughput imaging <strong>of</strong> cell based assays and cellular models using microplate<br />

cytometry.<br />

P. Wylie 1 , D. Caracino 2 , D. Onley 1 , W. Gaisford 1 ; 1 TTP Labtech Ltd, Royston, United Kingdom,<br />

2 TTP Labtech, Cambridge, MA<br />

Microscope-based, high-content instruments are used for many cell based assays in high<br />

content screening (HCS). Using Acumen eX3 ® with its wide field objective lens and “on-<strong>the</strong>-fly”<br />

laser scanning capabilities, it is possible to rapidly scan entire wells in a wide range <strong>of</strong> cell<br />

based assays relevant to <strong>the</strong> drug discovery industry including cell cycle analysis, cell surface<br />

markers and cell migration.<br />

In this poster, data is shown demonstrating <strong>the</strong> use <strong>of</strong> Acumen to assess <strong>the</strong> effect <strong>of</strong> inhibitory<br />

compounds on <strong>the</strong> mitotic index and cell cycle. In addition, with its high depth <strong>of</strong> field, it has also<br />

shown to be capable <strong>of</strong> scanning larger organisms such as C.elegans and even Zebrafish to<br />

provide information on multi-cellular drug interactions at an early screening stage. For efficient<br />

and rapid resolution most assays require <strong>the</strong> use <strong>of</strong> higher resolutions which entail lengthy read<br />

times, using single colours and analyzing only a small percentage <strong>of</strong> <strong>the</strong> total number <strong>of</strong> cells in<br />

a well to keep plate read times at a minimum. Recent studies also demonstrate that Acumen is<br />

capable <strong>of</strong> rapidly analysing complex cellular or animal models, such as angiogenic tube<br />

formation, C. elegans or drosophila larvae, thus enabling <strong>the</strong> development <strong>of</strong> whole organism or<br />

tissue based assays models for HCS.<br />

Built in s<strong>of</strong>tware <strong>of</strong>fers <strong>the</strong> flexibility <strong>of</strong> exporting whole well open source TIFF images for batch<br />

processing by third party image analysis s<strong>of</strong>tware packages. This new screening paradigm<br />

represents a major breakthrough in how microplate cytometry can be applied to complex cellular<br />

models since rapid cytometric analysis can now be combined with image-processing<br />

methodology.


2266<br />

Condensin structures chromosomal DNA through topological links.<br />

S. Cuylen 1 , J. Metz 1 , C. H. Haering 1 ; 1 Cell <strong>Biology</strong> & Biophysics Unit, EMBL, Heidelberg,<br />

Germany<br />

TUESDAY<br />

The multisubunit condensin complex is essential for <strong>the</strong> structural organization <strong>of</strong> eukaryotic<br />

chromosomes during <strong>the</strong>ir segregation by <strong>the</strong> mitotic spindle, but <strong>the</strong> mechanistic basis for its<br />

function is not understood. To address how condensin binds to and structures chromosomes,<br />

we have isolated from Saccharomyces cerevisiae cells circular minichromosomes linked to<br />

condensin. We find that ei<strong>the</strong>r linearization <strong>of</strong> minichromosome DNA or proteolytic opening <strong>of</strong><br />

<strong>the</strong> ring-like structure formed through <strong>the</strong> connection <strong>of</strong> <strong>the</strong> two ATPase heads <strong>of</strong> condensin’s<br />

structural maintenance <strong>of</strong> chromosomes (SMC) heterodimer by its kleisin subunit eliminates<br />

<strong>the</strong>ir association. This suggests that condensin rings encircle chromosomal DNA. We fur<strong>the</strong>r<br />

show that release <strong>of</strong> condensin from chromosomes by ring opening in dividing cells<br />

compromises <strong>the</strong> partitioning <strong>of</strong> chromosome regions distal to centromeres. Condensin hence<br />

forms topological links within chromatid arms that provide <strong>the</strong> arms with <strong>the</strong> structural rigidity<br />

necessary for <strong>the</strong>ir segregation.<br />

2267<br />

Antioxidants rescue carcinogen induced mitotic defects in both chromosomally stable<br />

and unstable cells.<br />

I. S. Griffin 1 , T. J. Yates 1 , N. J. Quintyne 1 ; 1 Honors College, Florida Atlantic University, Jupiter,<br />

FL<br />

Tumor cells are characterized by an increase in genomic instability, brought about by both<br />

chromosomal rearrangement and chromosomal instability. Both <strong>of</strong> <strong>the</strong>se broad changes can be<br />

induced by exposure to carcinogens. During mitosis, cells can exhibit lagging chromosomes,<br />

multipolar spindles or anaphase bridges, all <strong>of</strong> which contribute to genomic rearrangement. We<br />

have studied <strong>the</strong> link betweenexposure to carcinogen and prevalence <strong>of</strong> mitotic defect in both<br />

chromosomally stable and unstable cell lines. We have exposed UPCI:SCC103 oral cancer<br />

cells, MES-SA uterine cancer cells and RPE normal retinal cells to vinyl chloride,tamoxifen and<br />

aflatoxin at varying concentrations. When treated with carcinogens, we see a preferential<br />

increase in <strong>the</strong> frequency <strong>of</strong> lagging chromosomes, although rates <strong>of</strong> o<strong>the</strong>r mitotic defects are<br />

also higher. In addition, we have examined <strong>the</strong> effects <strong>of</strong> antioxidants on decreasing <strong>the</strong><br />

frequency <strong>of</strong> mitotic defect: analysis on chromosomally unstable cells with or without carcinogen<br />

treatment as well as chromosomally stable cells after carcinogen treatment indicates that Betacarotene,<br />

vitamin C and vitamin E are all effective at reducingrates <strong>of</strong> mitotic defect. We are<br />

also investigating whe<strong>the</strong>r pre-treatment with <strong>the</strong> antioxidants will weaken <strong>the</strong> effects <strong>of</strong><br />

carcinogen exposure in <strong>the</strong>se cell lines.<br />

2268<br />

Bod1 inhibits Protein Phosphatase 2A-B56 during mitosis.<br />

I. M. Porter 1 , K. Schleicher 1 , J. R. Swedlow 1 ; 1 Wellcome Trust Centre for Gene Regulation and<br />

Expression, University <strong>of</strong> Dundee, Dundee, UK<br />

Bod1 is a small, 22 kDa, protein that is required for resolution <strong>of</strong> syntelic attachments and<br />

proper chromosome alignment during mitosis and proper phosphorylation <strong>of</strong> MCAK by Aurora B<br />

in mitosis (Porter et al., 2007). Here we demonstrate that Bod1 has sequence similarity to Ensa<br />

and Arpp-19, two proteins recently identified as inhibitors <strong>of</strong> PP2A-B55, which are required for<br />

proper entry into mitosis (Mochida et al., 2010; Gharbi-Ayachi et al., 2010). We have now shown<br />

that Bod1 binds to <strong>the</strong> PP2A-B56 holo-enzyme during mitosis and is required for mitotic


TUESDAY<br />

progression. Binding <strong>of</strong> Ensa and Arpp-19 to PP2A-B55 requires a conserved Asp residue and<br />

phosphorylation by Greatwall kinase at a conserved Ser adjacent to <strong>the</strong> Asp. The Asp residue is<br />

conserved in Bod1 (D98), whereas <strong>the</strong> Ser is not, but a consensus CDK1 site (T95) is present<br />

immediately upstream. We show that Bod1 is phosphorylated in vitro by CDK1 kinase.<br />

Expression <strong>of</strong> Bod1 D98A or T95A mutants fail to rescue Bod1 depletion whereas <strong>the</strong> T95E<br />

mutations rescues as well as expression <strong>of</strong> wt Bod1. siRNA depletion <strong>of</strong> Bod1 from HeLa cells<br />

increases localisation <strong>of</strong> PP2A-B56 to kinetochores and loss <strong>of</strong> Sgo1 and Plk1 from<br />

kinetochores. Overall, <strong>the</strong>se data show that when phosphorylated by CDK1, Bod1 binds PP2A-<br />

B56 and suggest that this binding inhibits <strong>the</strong> activity <strong>of</strong> PP2A. In <strong>the</strong> presence <strong>of</strong> hyper-active<br />

kinetochore-associated PP2A, Plk1 activity is compromised and phosphorylation <strong>of</strong> downstream<br />

targets is decreased. Toge<strong>the</strong>r Bod1, Ensa and Arpp-19 form a new class <strong>of</strong> PP2A inhibitors.<br />

2269<br />

Daxx and USP7: novel regulators <strong>of</strong> genomic stability, mitosis and taxanes sensitivity.<br />

S. Giovinazzi 1 , V. Morozov 1 , W. C. Reinhold 2 , A. M. Ishov 1,3 ; 1 Anatomy and Cell <strong>Biology</strong>,<br />

University <strong>of</strong> Florida, Gainesville, FL, 2 Genomics & Bioinformatics Group, National Cancer<br />

Institute, NIH, Be<strong>the</strong>sda, MD, 3 Shands Cancer Center, University <strong>of</strong> Florida, Gainesville, FL<br />

Daxx is a multifunctional protein that plays a pivotal role in both physiological and pathological<br />

cellular processes. We previously demonstrated that cells with low levels <strong>of</strong> Daxx have reduced<br />

sensitivity to taxanes, powerful chemo<strong>the</strong>rapeutic agents, by persisting in a pro-metaphase<br />

block that allows cells to escape taxane-induced cell death. In this study we dissected <strong>the</strong><br />

mechanisms <strong>of</strong> Daxx-dependent taxanes resistance that also suggests function <strong>of</strong> this protein in<br />

mitotic progression. We show that Daxx interacts and cooperates with Ubiquitin Specific<br />

processing Protease-7 (USP7) to regulate mitosis. We demonstrate that depletion <strong>of</strong> USP7<br />

promotes stabilization <strong>of</strong> cyclin B, aneuploidy and mitotic anomalies, as it was previously<br />

observed for Daxx. We fur<strong>the</strong>r demonstrate that USP7 depletion results in reduced stability <strong>of</strong><br />

<strong>the</strong> mitotic E3 ubiquitin ligase Checkpoint with Forkhead and RING finger (CHFR).<br />

Consequently cells depleted by USP7 accumulate CHFR substrate, Aurora A kinase that has a<br />

crucial role in mitotic progression. We conclude that Daxx and USP7 are necessary to regulate<br />

proper execution <strong>of</strong> mitosis and <strong>the</strong>ir effects are at least partially mediated by CHFR and Aurora<br />

A kinase. Results from colony formation assay and in silico analysis show that USP7 expression<br />

negatively correlates with response to taxanes in cancer cell lines indicating that this protein can<br />

be used as predictive factor for taxanes response in cancer patients.<br />

2270<br />

Effects <strong>of</strong> actin and myosin inhibitors on PtK2 spindle length changes induced by laser<br />

microbeam irradiations across <strong>the</strong> spindle.<br />

R. Sheykhani 1 , M. W. Berns 2 , A. Forer 1 ; 1 York University, Toronto, ON, Canada, 2 University <strong>of</strong><br />

California San Diego, San Diego, CA<br />

Cutting spindle microtubules across <strong>the</strong> entire half spindle using a 532 nm pulsed picosecond<br />

laser microbeam causes <strong>the</strong> irradiated spindle pole to move inward (N. Baker PhD <strong>the</strong>sis,<br />

University <strong>of</strong> California, San Diego 2010). This usually is followed by <strong>the</strong> inward movement <strong>of</strong><br />

<strong>the</strong> un-irradiated pole. We tested whe<strong>the</strong>r <strong>the</strong> inward movement <strong>of</strong> <strong>the</strong> poles might be due to <strong>the</strong><br />

function <strong>of</strong> actin and myosin: we pharmacologically poisoned <strong>the</strong> actin-myosin system using<br />

anti-actin drugs Cytochalasin D (CD), Latrunculin B (LatB), or Jasplakinolide (JASP), or antimyosin<br />

drugs BDM and Y-27632, and <strong>the</strong>n irradiated across <strong>the</strong> entire half spindle. All <strong>the</strong>se<br />

drugs altered <strong>the</strong> inwards movements <strong>of</strong> both poles. In some cells nei<strong>the</strong>r pole moved, in some<br />

cells only one pole moved, in some cells movements <strong>of</strong> <strong>the</strong> poles were delayed, and in only a<br />

few cells did <strong>the</strong> poles move as far as in non-treated cells. This suggests that, as in Xenopus


TUESDAY<br />

cells (Woolner et al., 2008), <strong>the</strong>re is an interaction between actin, myosin and microtubules to<br />

control spindle length, though in our case it might be that actin/myosin forces are antagonistic to<br />

microtubule forces.<br />

2271<br />

HDAC3 regulates NudC acetylation in mitosis.<br />

C. Chuang 1 , J. Pan 2 , D. H. Hawke 3 , S-H. Lin 3 , L-Y. Yu-Lee 1,2 ; 1 <strong>Molecular</strong> and Cellular <strong>Biology</strong>,<br />

Baylor College <strong>of</strong> Medicine, Houston, TX, 2 Medicine, Baylor College <strong>of</strong> Medicine, Houston, TX,<br />

3 Department <strong>of</strong> <strong>Molecular</strong> Pathology, MD Anderson Cancer Center, Houston, TX<br />

Mitosis is a highly regulated process in which errors can lead to genomic instability, a hallmark<br />

<strong>of</strong> cancer. During this phase <strong>of</strong> <strong>the</strong> cell cycle, transcription is silent and RNA translation is<br />

globally inhibited. Thus, mitosis is largely driven by post-translational modification <strong>of</strong> proteins,<br />

including phosphorylation, methylation, ubiquitination, and sumoylation. Recent proteomics<br />

studies suggest that protein acetylation is as prevalent as protein phosphorylation. However,<br />

very little is known about <strong>the</strong> role <strong>of</strong> acetylation in mitotic progression.<br />

Using anti-acetyl-lysine immunoprecipitation <strong>of</strong> mitotic HeLa cell lysates followed by mass<br />

spectrometry, we identified 51 unique non-histone proteins, including proteins involved in RNA<br />

binding and processing, and cell cycle regulation (1). One cell cycle protein identified is NudC, a<br />

highly-conserved dynein/dynactin associated factor that plays a role in mitosis and cytokinesis. I<br />

confirmed that NudC is acetylated in mitosis by immunoprecipitation followed by immunoblot.<br />

Moreover, NudC acetylation in mitosis increased upon treatment with <strong>the</strong> HDAC inhibitor<br />

apicidin that shows specificity towards HDAC3, suggesting that NudC acetylation is regulated by<br />

HDAC3.<br />

Interestingly, comparing unperturbed HeLa cells to mitotically-enriched HeLa cells, NudC is<br />

more acetylated outside <strong>of</strong> mitosis. Utilizing a double thymidine block and release protocol, I<br />

enriched for HeLa cells in S, G2, early mitosis (prometaphase-like), late mitosis (anaphase), and<br />

G1. I found that NudC associates with HDAC3 throughout mitosis, which may explain its lower<br />

level <strong>of</strong> acetylation during mitosis. Immun<strong>of</strong>luorecense staining confirmed that NudC and<br />

HDAC3 co-localize during early mitosis.<br />

Mass spectrometry analysis <strong>of</strong> NudC immunoprecipitated from unperturbed HeLa cell lysate<br />

identified one acetylation site on NudC at K39. I generated NudC K39 acetyl-deficient (K39R)<br />

and acetyl-mimetic (K39Q) mutants and will decipher <strong>the</strong> role <strong>of</strong> NudC K39 acetylation in mitotic<br />

progression. Additionally, how NudC acetylation/deacetylation regulates its association with<br />

various mitotic regulators to promote mitotic progression are under analysis.<br />

These studies provide a framework from which to address how dynamic<br />

acetylation/deacetylation regulates <strong>the</strong> function and/or localization <strong>of</strong> proteins that drive mitotic<br />

progression and cell division. Understanding acetylation <strong>of</strong> cell cycle protein networks in cell<br />

cycle regulation is likely to reveal new paradigms for anti-cancer <strong>the</strong>rapy.<br />

1. Chuang et al. J. Proteome Res. 2010, 9, 4554-4564.


2272<br />

Attenuation <strong>of</strong> mitotic RanGTP gradient in normal somatic cells.<br />

K. Hasegawa 1 , S. Ryu 1 , P. Kalab 1 ; 1 LCMB, National Cancer Institute, NIH, Be<strong>the</strong>sda, MD<br />

TUESDAY<br />

Studies in variety <strong>of</strong> organisms showed that mitotic segregation <strong>of</strong> chromosomes requires<br />

regulation <strong>of</strong> spindle assembly factors (SAFs) by small GTPase Ran. Essential feature <strong>of</strong> such<br />

regulation is thought to be <strong>the</strong> chromosome-centered concentration gradient <strong>of</strong> free RanGTP<br />

which arises in mitotic cells due to chromosome binding <strong>of</strong> RCC1, <strong>the</strong> guanine nucleotide<br />

exchange factor for Ran and cytoplasmic localization <strong>of</strong> RanGAP1. Close to chromosomes,<br />

RanGTP releases spindle assembly factors (SAFs) from <strong>the</strong>ir inhibitory complexes with<br />

importins, thus providing a spatial clue to mitotic spindle assembly. Using live cell microscopy<br />

with Forster resonance energy transfer (FRET) biosensors, we observed that in contrast to<br />

HeLa, normal human primary cells divide in <strong>the</strong> absence <strong>of</strong> a steep mitotic RanGTP gradient or<br />

a gradient <strong>of</strong> activated SAFs. Consistent with proposed Ran functions in mitotic spindle<br />

assembly, <strong>the</strong> absence <strong>of</strong> a steep mitotic RanGTP gradient in primary cells correlated with<br />

extended prometaphase, although no mitotic defects were observed in such cells. As expected,<br />

we found that RCC1 abundance and its binding to chromatin were among <strong>the</strong> key factors<br />

controlling <strong>the</strong> steepness <strong>of</strong> mitotic RanGTP gradient. These results show that a steep RanGTP<br />

gradient is not required for mitosis in all normal cells and suggest that attenuated mitotic Ran<br />

function may be characteristic to homeostatic cell proliferation <strong>of</strong> normal tissues in vivo.<br />

2273<br />

<strong>Molecular</strong> mechanisms governing extrinsic forces in mitotic spindle organization.<br />

M. Kwon 1,2 , M. Bagonis 2 , G. Danuser 2 , D. Pellman 1,2 ; 1 HHMI/Pediatric Oncology, Dana-Farber<br />

Cancer Institute, Boston, MA, 2 Cell <strong>Biology</strong>, Harvard Medical School, Boston, MA<br />

Precise control <strong>of</strong> centrosome number is crucial for bipolar spindle assembly and accurate<br />

segregation <strong>of</strong> chromosomes to daughter cells. Cancer cells <strong>of</strong>ten contain supernumerary<br />

centrosomes that create <strong>the</strong> potential for catastrophic multipolar divisions. Despite <strong>the</strong> presence<br />

<strong>of</strong> extra centrosomes, many cancer cells successfully divide because <strong>of</strong> mechanisms that<br />

suppress multipolar mitoses by clustering <strong>the</strong>ir extra centrosomes. Using functional genomics<br />

and follow-up studies, we have previously identified key pathways contributing to centrosome<br />

organization in cancer cells. In addition to spindle intrinsic forces, we found that cell adhesion<br />

patterns can determine <strong>the</strong> fate <strong>of</strong> mitosis whe<strong>the</strong>r <strong>the</strong>y divide into two (bipolar) or more<br />

(multipolar). Yet, <strong>the</strong> mechanism by which actin and adhesion-dependent forces to control <strong>the</strong><br />

fidelity <strong>of</strong> mitosis remain unclear at <strong>the</strong> molecular level. One model proposes that sites <strong>of</strong> strong<br />

cell matrix adhesion are imprinted as an actin rich structure called retraction fibers during<br />

mitosis, and <strong>the</strong> retraction fibers serve as cortical cues to concentrate force generators and<br />

regulators to pull centrosomes. By imaging cancer cells plated on fibronectin micropatterns,<br />

here we demonstrate that Myo10, an unconventional myosin identified from our genome-wide<br />

screen, is an essential component linking retraction fiber-mediated forces to astral microtubules<br />

(MTs) during spindle organization. Myo10 specifically localizes along and to <strong>the</strong> tips <strong>of</strong> <strong>the</strong><br />

retraction fibers. When cells with extra centrosomes were plated on Y-shaped fibronectin<br />

micropatterns, control cells divide tripolar toward Y-adhesion axis; however, cells depleted <strong>of</strong><br />

Myo10 undergo bipolar division independent <strong>of</strong> adhesion and retraction fiber positions,<br />

suggesting that Myo10 is a force coupler from retraction fibers to centrosomes. We have<br />

hypo<strong>the</strong>sized that intrinsic properties <strong>of</strong> MTs adjacent to retraction fibers are altered to favor<br />

stable interaction between astral MTs and cell cortex. To uncover heterogeneity <strong>of</strong> MT dynamics<br />

in relation to retraction fibers, we have performed live cell imaging <strong>of</strong> GFP-EB3 with high spatiotemporal<br />

resolution in cells plated on fibronectin micropatterns where retraction fiber positions<br />

are precisely manipulated. We have developed a system that quantifies and visualize MT


TUESDAY<br />

dynamics in different subregions <strong>of</strong> cells by an automated computer program capable <strong>of</strong><br />

tracking GFP-EB3 comets. This approach uncovered heterogeneity <strong>of</strong> MT dynamics around<br />

retraction fiber regions. Interestingly, our analyses have revealed that Myo10 is in part<br />

responsible for promoting <strong>the</strong> long-lived and long distance travelling MTs preferentially at<br />

adhesion sites. Taken toge<strong>the</strong>r, <strong>the</strong>se results suggest that Myo10 is a key adhesion-dependent<br />

regulator that confers heterogeneity <strong>of</strong> mitotic cortex by coupling cell geometry/adhesion to<br />

mitotic fidelity and genome stability.<br />

2274<br />

Terminating Mitotic Checkpoint Surveillance at <strong>the</strong> Anaphase Onset.<br />

M. D. Vazquez Novelle 1 , M. Petronczki 1 ; 1 Cancer Research UK (Clare Hall Labs), Potters Bar,<br />

England<br />

The mitotic checkpoint monitors <strong>the</strong> attachment <strong>of</strong> kinetochores to microtubules and delays<br />

anaphase onset until all sister kinetochores have become attached to opposite poles. Correct<br />

bipolar attachment leads to kinetochore deformation and tension that silences <strong>the</strong> checkpoint.<br />

What prevents mitotic checkpoint reactivation when sister centromeres are split and tension is<br />

lost at anaphase onset? Aurora-B kinase, <strong>the</strong> catalytic subunit <strong>of</strong> <strong>the</strong> chromosomal passenger<br />

protein complex (CPC), acts as a sensor at inner centromeres for <strong>the</strong> status <strong>of</strong> attachment.<br />

Phosphorylation <strong>of</strong> Aurora-B targets at erroneously attached kinetochores elicits <strong>the</strong> correction<br />

<strong>of</strong> <strong>the</strong>se attachments and <strong>the</strong> activation <strong>of</strong> <strong>the</strong> mitotic checkpoint. At anaphase <strong>the</strong> CPC leaves<br />

<strong>the</strong> centromeres and relocates to <strong>the</strong> spindle midzone. This Cdk1-controlled iconic translocation<br />

might prevent <strong>the</strong> checkpoint from re-engaging after anaphase onset. To test this hypo<strong>the</strong>sis we<br />

experimentally retained Aurora-B and <strong>the</strong> CPC at <strong>the</strong> centromere throughout anaphase in<br />

human cells. Preventing CPC translocation caused <strong>the</strong> untimely recruitment <strong>of</strong> mitotic<br />

checkpoint proteins (Bub1, BubR1 and Mps1) to kinetochores at anaphase in an Aurora-B<br />

kinase activity dependent manner. Our results suggest that <strong>the</strong> relocalization <strong>of</strong> <strong>the</strong> CPC, an<br />

evolutionarily conserved event in eukaryotes, is a key mechanism that prevents mitotic<br />

checkpoint activation at anaphase. However, <strong>the</strong> retention <strong>of</strong> Aurora-B at centromeres after<br />

anaphase onset does not suffice to trigger a full mitotic checkpoint engagement in response to<br />

<strong>the</strong> loss <strong>of</strong> tension due to sister chromatid splitting. Mad1 and Mad2 recruitment to kinetochores<br />

is suppressed after anaphase onset independently <strong>of</strong> CPC relocalization. What regulates <strong>the</strong>se<br />

events at <strong>the</strong> metaphase-to-anaphase transition in human cells? Experiments in yeast, flies and<br />

frogs suggest that Cdk1 activity is required for mitotic checkpoint activity and <strong>the</strong> error correction<br />

pathway. Thus, we are investigating whe<strong>the</strong>r inactivation <strong>of</strong> Cdk1 at <strong>the</strong> metaphase-toanaphase<br />

transition could play an important role in terminating mitotic checkpoint surveillance.<br />

Our preliminary data are consistent with <strong>the</strong> notion that Cdk1 is a master regulator <strong>of</strong> <strong>the</strong> mitotic<br />

checkpoint in human cells. In conclusion, toge<strong>the</strong>r our results suggest that Cdk1 inactivation<br />

prevents mitotic checkpoint re-engagement at anaphase onset, through <strong>the</strong> translocation <strong>of</strong><br />

CPC and o<strong>the</strong>r events that control Mad1/2 recruitment to kinetochores.<br />

2275<br />

A mechanism linking microtubule destabilisation to maturation <strong>of</strong> chromosomemicrotubule<br />

attachments.<br />

R. L. Shrestha 1 , N. Tamura 1 , I. Zulkipli 1 , V. M. Draviam 1 ; 1 Department <strong>of</strong> Genetics, University <strong>of</strong><br />

Cambridge, Cambridge, United Kingdom<br />

Selective destabilization <strong>of</strong> erroneous chromosome-microtubule attachments is critical to<br />

prevent <strong>the</strong> mis-segregation <strong>of</strong> chromosomes. Initially, chromosomes attach to lateral walls <strong>of</strong><br />

microtubules and <strong>the</strong>n convert this attachment to <strong>the</strong> ends <strong>of</strong> microtubules through a poorly<br />

understood maturation process. We demonstrate here a role for microtubule destabilization in


TUESDAY<br />

<strong>the</strong> maturation <strong>of</strong> attachments made to microtubule walls, and describe <strong>the</strong> consequence <strong>of</strong><br />

persistent immature attachments. TAO1 kinase mediates microtubule destabilisation during<br />

mitosis and cells depleted <strong>of</strong> TAO1 display persistent laterally attached kinetochores that fail to<br />

congress properly. By briefly exposing cells to mild microtubule stabilizing drugs, we<br />

demonstrate that suppressing microtubule dynamics is sufficient to deter <strong>the</strong> maturation <strong>of</strong><br />

lateral attachments. <strong>Late</strong>rally attached kinetochores, under microtubule stabilizing conditions,<br />

fail to retain Mad2 or MPS1 and compromise error-correction in cells released from monastrol<br />

treatment. Toge<strong>the</strong>r our findings provide first insight into <strong>the</strong> fate <strong>of</strong> unresolved lateral<br />

attachments, and highlight <strong>the</strong> importance <strong>of</strong> maturation in ensuring <strong>the</strong> accurate segregation <strong>of</strong><br />

chromosomes.<br />

Signal Transduction and Signaling Networks II<br />

2276/L50<br />

Regulation <strong>of</strong> Notch1 signaling by Jagged-1 intracellular domain.<br />

J-S. Ahn 1 , E-J. Ann 1 , J-H. Yoon 1 , H-S. Park 1 ; 1 School <strong>of</strong> Biological Sciences and Technology,<br />

Chonnam national university, Bukku, Gwangju, Korea<br />

Notch signaling involves <strong>the</strong> proteolytic cleavage <strong>of</strong> <strong>the</strong> transmembrane Notch receptor after<br />

binding to its transmembrane ligands. Jagged-1 also undergoes proteolytic cleavage by<br />

gamma-secretase and releases an intracellular fragment. In this study, we have demonstrated<br />

that <strong>the</strong> Jagged-1 intracellular domain (JICD) inhibits Notch1 signaling via a reduction in <strong>the</strong><br />

protein stability <strong>of</strong> <strong>the</strong> Notch1 intracellular domain (Notch1-IC). The formation <strong>of</strong> <strong>the</strong> Notch1-IC-<br />

RBP-Jk-Mastermind complex is prevented in <strong>the</strong> presence <strong>of</strong> JICD, via a physical interaction.<br />

Fur<strong>the</strong>rmore, JICD accelerates <strong>the</strong> protein degradation <strong>of</strong> Notch1-IC via Fbw7-dependent<br />

proteasomal pathway. These results indicate that JICD functions as a negative regulator in<br />

Notch1 signaling via <strong>the</strong> promotion <strong>of</strong> Notch1-IC degradation.<br />

2277<br />

Role <strong>of</strong> R-Ras GTPases in <strong>the</strong> Wnt-Planar Cell Polarity pathway.<br />

N. Hartig 1 , M. Muñoz-Alegre 1 , L. Young 1 , P. Rodriguez-Viciana 1 ; 1 UCL Cancer Institute, London,<br />

United Kingdom<br />

Ras genes are mutated in about 30% <strong>of</strong> human tumours and likely play a role by indirect<br />

activation (e.g. receptor tyrosine kinases) in an even larger number <strong>of</strong> cancers. The closely<br />

related R-Ras GTPases share many <strong>of</strong> <strong>the</strong> properties <strong>of</strong> Ras genes including <strong>the</strong> ability to<br />

behave as oncogenes. Despite sharing biochemical and biological properties, <strong>the</strong> R-Ras<br />

subgroup also have distinct functions. In order to identify novel R-Ras effector proteins<br />

potentially accounting for <strong>the</strong>se differences, we employed a Tandem Affinity Approach (TAP)<br />

using activated R-Ras as bait. This approach lead to <strong>the</strong> identification <strong>of</strong> Wnt-Planar Cell<br />

Polarity (PCP) protein Vangl1 as a novel R-Ras interacting protein.<br />

The Wnt pathway plays a key role in development and disease. In addition to <strong>the</strong> better studied<br />

ß-catenin dependent pathway, Wnt ligands can also activate <strong>the</strong> separate ‘non-canonical’ or<br />

PCP pathway, which coordinates complex cell migration during development. Perturbations in<br />

<strong>the</strong> PCP pathway contribute to <strong>the</strong> pathogenesis <strong>of</strong> a variety <strong>of</strong> diseases including cardiac and<br />

neuronal tube defects, cilia based disorders and to <strong>the</strong> invasiveness <strong>of</strong> cancer cells.<br />

We have shown that Vangl1 functions as a true R-Ras effector and mapped <strong>the</strong> interaction to<br />

<strong>the</strong> Vangl1 C-terminus. We have identified several Vangl1 interacting proteins that confirm it’s


TUESDAY<br />

conserved role in polarity and conducted biological assays that show that Vangl1 is critically<br />

required in migration. We have also shown that certain Frizzled receptors cooperate with <strong>the</strong><br />

Ror2 tyrosine kinase to selectively stimulate Vangl1 function and possibly activation <strong>of</strong> R-Ras<br />

GTPases.<br />

In conclusion, <strong>the</strong> identification <strong>of</strong> Vangl proteins as novel effectors <strong>of</strong> R-Ras GTPases provides<br />

an exciting new link between Ras signalling and <strong>the</strong> Wnt-PCP pathway.<br />

2278<br />

Loss <strong>of</strong> serine protease HtrA1 potentiates transforming growth factor-β signaling and<br />

bone formation.<br />

J. R. Graham 1 , A. Chamberland 1 , X. Li 1 , D. Dai 1 , Q. Lin 2 , M. S. Ryan 3 , W. Zeng 1 , T. Blanchet 1 ,<br />

M. A. Rivéra-Bermúdez 1 , C. R. Flannery 1 , Z. Yang 1 ; 1 Inflammation & Remodeling Research<br />

Unit, Pfizer Inc, Cambridge, MA, 2 Global Bio<strong>the</strong>rapeutics Technologies, Pfizer Inc, Cambridge,<br />

MA, 3 Immunology & Autoimmunity Research Unit, Pfizer Inc, Cambridge, MA<br />

HtrA1 is a member <strong>of</strong> <strong>the</strong> High Temperature Requirement (HtrA) family <strong>of</strong> serine proteases.<br />

Genetic and gene expression studies have implicated HtrA1 in <strong>the</strong> progression <strong>of</strong> several<br />

pathologies, most notably in conditions with aberrant deposition <strong>of</strong> extracellular matrix. The<br />

inhibitory activity <strong>of</strong> HtrA1 on <strong>the</strong> transforming growth factor (TGF)-β family pathway has been<br />

reported, partially explaining <strong>the</strong> function <strong>of</strong> HtrA1 in physiological or disease processes. This<br />

study characterized <strong>the</strong> molecular mechanism by which HtrA1 regulates TGF-β signaling and<br />

examined <strong>the</strong> effect <strong>of</strong> HtrA1 gene deletion on musculoskeletal tissues in mice.<br />

Overexpression <strong>of</strong> HtrA1 inhibited <strong>the</strong> TGF-β-mediated transcription <strong>of</strong> a Smad-responsive<br />

promoter. Incubation <strong>of</strong> HtrA1 with several candidate substrates in <strong>the</strong> TGF-β pathway revealed<br />

that this protease was capable <strong>of</strong> efficiently cleaving type II and III TGF-β receptors (TβRII and<br />

TβRIII), but not TβRI or TGF-β itself. Fur<strong>the</strong>rmore, cleavage increased with <strong>the</strong> addition <strong>of</strong> CPII,<br />

an HtrA1 agonist. Overexpression <strong>of</strong> HtrA1 decreased <strong>the</strong> cell surface levels <strong>of</strong> both TβRII and<br />

III, but not TβRI. Conversely, silencing HtrA1 expression resulted in significantly more TGF-β<br />

cell surface binding, increased Smad2 phosphorylation, and enhanced TGF-β-regulated gene<br />

expression. To better characterize <strong>the</strong> role <strong>of</strong> HtrA1, we generated mice with targeted gene<br />

deletion. MicroCT analysis showed that deletion <strong>of</strong> <strong>the</strong> HtrA1 gene resulted in a marked<br />

increase in bone volume in <strong>the</strong> distal femur and lumbar vertebrae (48% and 20%, respectively).<br />

Embryonic fibroblasts from HtrA1 knockout mice exhibited increased TGF-β-induced expression<br />

<strong>of</strong> three genes known to promote bone formation, Runx2, CTGF and PAI-1.<br />

Our results reveal that <strong>the</strong> cleavage <strong>of</strong> two TGF-β receptors (TβRII and III) is a novel<br />

mechanism for HtrA1 inhibition <strong>of</strong> TGF-β signaling. Deletion <strong>of</strong> HtrA1 in mice leads to a dramatic<br />

increase in bone formation, most likely due to enhanced signaling by TGF-β or TGF-β family<br />

proteins.


TUESDAY<br />

2279<br />

Cellular and Biochemical Activities <strong>of</strong> <strong>the</strong> Non-Canonical Wnt Receptors ROR1 and<br />

ROR2.<br />

T. W. Bainbridge 1 , V. De Almeida 2 , C. Chalouni 3 , R. Kelly 1,4 , J. Goldsmith 2 , A. Majeed 1,5 , B.<br />

Pan 6 , G. Quinones 1 , J. Lill 1 , W. Sandoval 1 , A. Izrael-Tomasevic 1 , D. Arnott 1 , M. Costa 2 , P.<br />

Polakis 2 , B. Rubinfeld 2 , J. A. Ernst 1,7 ; 1 Protein Chemistry, Genentech, South San Francisco, CA,<br />

2 Cancer Pathways, Genentech, South San Francisco, CA, 3 Center for Advanced Light<br />

Microscopy, Genentech, South San Francisco, CA, 4 University <strong>of</strong> California, Los Angeles, CA,<br />

5 Materials Science & Engineering, Stanford University,Palo Alto, CA, 6 Structural <strong>Biology</strong>,<br />

Genentech, South San Francisco, CA, 7 Early Discovery Biochemistry, Genentech, South San<br />

Francisco, CA<br />

The receptor tyrosine kinases, ROR1 and ROR2, activate non-canonical Wnt signaling and<br />

planar cell polarity pathways in response to Wnt5a. Various mutations within <strong>the</strong> intracellular<br />

domain (ICD) <strong>of</strong> ROR2 have been linked to Robinow syndrome and brachydactyly type B, while<br />

increased expression <strong>of</strong> ROR1 has been associated with acute lymphoblastic leukemia, chronic<br />

lymphocytic leukemia and mantle cell lymphoma. Previous studies have shown phosphorylation<br />

<strong>of</strong> ROR2 in response to Wnt5a stimulation, although autophosphorylation has not been<br />

demonstrated. The human ROR kinase domains lack conservation <strong>of</strong> <strong>the</strong> P-loop consensus<br />

sequence and are expected to be deficient in phosphotransferase activity, but studies with<br />

isolated ROR have provided conflicting results in kinase activity assays. CAM-1, <strong>the</strong> C. elegans<br />

ortholog <strong>of</strong> ROR, retains <strong>the</strong> P-loop consensus sequence, but <strong>the</strong> kinase activity has not been<br />

evaluated in vitro. In this work, we investigate <strong>the</strong> cellular role <strong>of</strong> <strong>the</strong> ROR ICD and compare <strong>the</strong><br />

in vitro kinase activities <strong>of</strong> <strong>the</strong> isolated ICDs <strong>of</strong> ROR1, ROR2, CAM-1, and <strong>the</strong> most closely<br />

related human kinase, MuSK.<br />

2280<br />

Serotonin Receptor Signaling in JC Virus Infection.<br />

M. Maginnis 1 , W. Atwood 1 ; 1 <strong>Molecular</strong> <strong>Biology</strong>, Cell <strong>Biology</strong> & Biochemistry, Brown University,<br />

Providence, RI<br />

The JC polyomavirus (JCV) is a prevalent human pathogen and <strong>the</strong> causative agent <strong>of</strong> <strong>the</strong> fatal,<br />

demyelinating disease Progressive Multifocal Leukoencephalopathy (PML). JCV infection is<br />

initiated by interactions with cell-surface alpha 2,6-linked sialic acid on lactoseries<br />

tetrasaccharide c (LSTc) and serotonin 5-hydroxytryptamine2A receptor (5-HT2AR). Engagement<br />

<strong>of</strong> viral receptors can activate intracellular signaling pathways that regulate key steps in <strong>the</strong><br />

virus life cycle. The objective <strong>of</strong> this study is to define whe<strong>the</strong>r cellular signaling events emanate<br />

from <strong>the</strong> G-protein coupled-receptor (GPCR) 5-HT2AR in JCV infection. 5-HT2AR activates <strong>the</strong><br />

Ca 2+ /calmodulin pathway leading to activation <strong>of</strong> <strong>the</strong> ERK/MAPK signaling pathway and<br />

phosphorylation <strong>of</strong> ERK. To determine whe<strong>the</strong>r ERK is phosphorylated following engagement <strong>of</strong><br />

5-HT2AR, cell lysates were harvested at early time points following JCV infection and analyzed<br />

by immunoblot analysis for ERK phosphorylation. ERK was phosphorylated as early as 5<br />

minutes post-infection consistent with receptor engagement. Fur<strong>the</strong>rmore, treatment <strong>of</strong> cells<br />

with inhibitors <strong>of</strong> MEK and ERK abolished JCV infection in a dose-dependent manner<br />

suggesting that activation <strong>of</strong> <strong>the</strong> ERK/MAPK signaling pathway is critical for JCV infection. To<br />

define whe<strong>the</strong>r Ca 2+ /calmodulin signaling is activated during JCV infection, cells were treated<br />

with inhibitors <strong>of</strong> calcium release and calmodulin kinase, both <strong>of</strong> which decreased JCV infection.<br />

5-HT2AR contains two calmodulin binding sites expressed in <strong>the</strong> second transmembrane loop<br />

and carboxyl terminus. Mutations in key hydrophobic residues in <strong>the</strong> calmodulin binding sites<br />

were generated in a 5-HT2AR construct and expressed in poorly permissive HEK cells.<br />

Expression <strong>of</strong> a wild-type 5-HT2AR rescues infection in HEK cells, but a mutation in <strong>the</strong>


TUESDAY<br />

calmodulin-binding site in <strong>the</strong> carboxyl terminus resulted in a decrease in JCV infection. These<br />

findings provide new information regarding <strong>the</strong> role 5-HT2AR in JCV infection, and suggest that<br />

signals initiated upon engagement <strong>of</strong> <strong>the</strong> 5-HT2AR are essential for a productive viral infection.<br />

2281<br />

A genetic approach to identify new regulators for Smoo<strong>the</strong>ned.<br />

H-T. Shih 1 , W-Y. Chen 1 , J-T. Wu 1 ; 1 Institute <strong>of</strong> <strong>Molecular</strong> Medicine, National Taiwan University,<br />

Taipei City, Taiwan<br />

G protein-coupled receptors (GPCRs) are seven-transmembrane receptors that initiate signaling<br />

processes <strong>of</strong> many physiological responses upon ligand stimulation. In Drosophila, Smoo<strong>the</strong>ned<br />

(Smo) belongs to GPCR family and transduces Hedgehog signaling that is critical in embryonic<br />

and larval development. Whereas <strong>the</strong> strength <strong>of</strong> GPCR-mediated signaling could be modulated<br />

at <strong>the</strong> level <strong>of</strong> GPCR by desensitization and internalization <strong>of</strong> <strong>the</strong> GPCRs, it is not clear whe<strong>the</strong>r<br />

<strong>the</strong> level <strong>of</strong> Smo is also subjective to any regulation that eventually changes <strong>the</strong> strength <strong>of</strong><br />

Hedgehog signaling. To test this possibility, we performed an in vivo RNAi screen in a smo loss<strong>of</strong>-function<br />

mutant background to identify regulators for Smo expression that also modulate <strong>the</strong><br />

strength <strong>of</strong> Hedgehog signaling. First, we used a MS1096-Gal4 driven USA-smo5A to generate<br />

a fused L2 and L3 wing vein, <strong>the</strong> characteristic smo deficient phenotype in adult wing. Next, we<br />

crossed MS1096>smo5A to RNAi or dominant negative stocks for small G proteins, Rac1,<br />

Cdc42, RhoA, and Rab, and GTPase-activating proteins (GAPs). Our preliminary data showed<br />

that ectopic expression <strong>of</strong> small G proteins caused a marked wing shrinkage or lethality. But<br />

dominant negative form <strong>of</strong> small G proteins enhanced Smo deficient phenotype. Interestingly,<br />

most GAPs had no obvious effect on <strong>the</strong> smo deficient phenotype, suggesting a functional<br />

redundancy among GAPs. Our long term goal is to understand <strong>the</strong> distinct regulation impinged<br />

on Smo as oppose to those on conventional GPCRs.<br />

2282<br />

Dihydrotestosterone activates Src to increase EGF-induced Erk activation in fetal lung<br />

fibroblasts.<br />

M. K. Lee 1 , S. M. Smith 1 , S. Murray 2 , L. D. Pham 2 , H. C. Nielsen 2 ; 1 Center for Crani<strong>of</strong>acial<br />

<strong>Molecular</strong> <strong>Biology</strong>, University <strong>of</strong> Sou<strong>the</strong>rn California, Los Angeles, CA, 2 Newborn Medicine,<br />

Tufts Medical Center, Boston, MA<br />

Neonatal respiratory distress syndrome has long been recognized as being more severe in<br />

premature male infants. This observation has been ascribed to differences in lung maturation,<br />

which is regulated by interactions between mesenchymal and epi<strong>the</strong>lial cell populations.<br />

Fibroblast-type II cell communications are, in turn, modulated by epidermal growth factor (EGF)mediated<br />

Erk1/2 activation. Because dihydrotestosterone (DHT) has been implicated in delayed<br />

lung maturation, we hypo<strong>the</strong>sized that androgens could affect tyrosine kinase signaling in fetal<br />

lung fibroblasts. Androgens act by both translation-dependent and independent mechanisms,<br />

and we fur<strong>the</strong>r hypo<strong>the</strong>sized that DHT regulates fetal lung fibroblast signaling by inducing<br />

cytosolic Src to phosphorylate EGF receptors, <strong>the</strong>reby enhancing <strong>the</strong>ir subsequent activation by<br />

EGF. To assess <strong>the</strong>se postulates, fetal lung fibroblasts were isolated from ei<strong>the</strong>r pregnant mice<br />

treated with subcutaneous timed-release dihydrotestosterone (DHT) pellets or from untreated<br />

timed-gestation mice at 16 days gestation. Cells were maintained in culture containing 100 nM<br />

DHT for up to 4 d prior to overnight serum starvation and stimulation with 100 ng/ml EGF for up<br />

to 30 m. Cells were lysed in RIPA buffer, resolved by SDS-PAGE, and transferred to<br />

nitrocellulose for Western analysis. Membranes were probed with antibodies against<br />

phosphoErk1/2, phosphoEGF receptor, and phosphoSrc. To attenuate Src activity, selected<br />

cells were transfected with siRNA against Src, treated with <strong>the</strong> pharmacological Src inhibitor


TUESDAY<br />

PP2, or infected with adenoviruses expressing a dominant negative Src. Both long-term (in<br />

utero plus in culture) and short-term (in culture only) DHT exposure increased peak Erk<br />

activation by EGF, and this increase was evident with as little as 60 min <strong>of</strong> DHT exposure. The<br />

phosphorylation <strong>of</strong> Src and EGF receptor was increased by DHT. EGF-induced Shc<br />

phosphorylation and Grb2 association with Shc were similarly increased by prior DHT exposure,<br />

confirming pathway activation. Finally, Src silencing, PP2 treatment, and dominant negative Src<br />

adenoviruses all prevented DHT from increasing EGF-mediated Erk1/2 phosphorylation. These<br />

results suggest that DHT activates Src to increase <strong>the</strong> activity <strong>of</strong> EGF receptors and, hence, <strong>the</strong><br />

EGF signaling pathway leading to Erk activation in primary fetal lung fibroblasts.<br />

2283<br />

Regulation <strong>of</strong> calcium-dependent calcineurin activity by inositol sphingolipids.<br />

S. A. Jesch 1 , S. A. Henry 1 ; 1 <strong>Molecular</strong> <strong>Biology</strong> and Genetics, Cornell University, Ithaca, NY<br />

Inositol-containing sphingolipids and phosphoinositides at <strong>the</strong> plasma membrane play important<br />

roles in regulating stress response pathways, which are critical for lipid homeostasis and cell<br />

viability. We previously showed that inhibiting inositol sphingolipid metabolism triggers protein<br />

kinase C signaling and subsequent activation <strong>of</strong> <strong>the</strong> MAP kinase, Slt2p/Mpk1p, by regulating <strong>the</strong><br />

Stt4p-dependent pool <strong>of</strong> phosphatidylinositol-4-phosphate (PI4P) at <strong>the</strong> plasma membrane. To<br />

identify additional lipid-mediated signaling networks that are activated by a similar mechanism,<br />

we carried out gene expression pr<strong>of</strong>iling following interruption <strong>of</strong> inositol sphingolipid syn<strong>the</strong>sis<br />

by starving cells for inositol. We found that calcineurin-dependent genes are highly up-regulated<br />

following inositol starvation. Previous work has demonstrated that calcineurin, a calciumdependent<br />

protein phosphatase, regulates <strong>the</strong> activities <strong>of</strong> <strong>the</strong> PH-domain proteins Slm1p and<br />

Slm2p, which are necessary for normal sphingolipid metabolism. Consistent with <strong>the</strong>se data,<br />

calcineurin, stt4, and slm mutants exhibit inositol auxotrophy. Moreover, cells lacking Csg2p, a<br />

calcium binding regulator <strong>of</strong> mannosyl-inositolphosphorylceramide syn<strong>the</strong>sis, constitutively<br />

activates calcium-dependent calcineurin signaling. This activation requires stt4, slt2/mpk1, and<br />

<strong>the</strong> plasma membrane-localized calcium channel Cch1p, which is a known target <strong>of</strong><br />

Slt2p/Mpk1p. Toge<strong>the</strong>r, <strong>the</strong>se results suggest a feedback mechanism for regulating sphingolipid<br />

mediated signaling by regulating plasma membrane pools <strong>of</strong> PI4P and intracellular calcium<br />

levels.<br />

2284<br />

Anti-Inflammatory Effect <strong>of</strong> Benzylideneacetophenone Derivative on Thyroid-Associated<br />

Ophthalmopathy through Suppression CXCL10/IP-10 Production.<br />

S. Lee 1 , Y-H. Choi 1 ; 1 Ewha Woman's University, Seoul, Korea<br />

Thyroid-associated ophthalmopathy (TAO) is an autoimmune inflammatory disorder <strong>of</strong> <strong>the</strong><br />

orbital fatty tissue/connective tissue and <strong>the</strong> extra ocular muscles that is closely associated with<br />

Graves’ hyperthyroidism. Recent experimental evidence has shown that CXCL10/IP-10 is<br />

elevated in patients with TAO and plays an important role in <strong>the</strong> initial phases <strong>of</strong> TAO.<br />

Yakuchinone B from Alpinia oxyphylla,that belongs to ginger family has been used in oriental<br />

herbal medicine. It has been reported as one <strong>of</strong> anti-inflammatory phytochemicals which<br />

regulate COX-2 expression. To search for new candidate <strong>of</strong> anti-inflammatory compound at <strong>the</strong><br />

inflammatory stage <strong>of</strong> TAO, benzylideneacetophenone compound JC3, (2E)-3-(4-hydroxy-3methoxyphenyl)phenylpro-2-en-l-one),<br />

was syn<strong>the</strong>sized from Yakuchinone B and <strong>the</strong>ir potential<br />

to suppress CXCL10/IP-10 production was evaluated. IFN-γ strongly increased <strong>the</strong> level <strong>of</strong><br />

CXCL10/IP-10 in orbital fibroblasts with TAO and normal. JC3 compound revealed <strong>the</strong> potent<br />

inhibitory effect on IFN-γ-induced CXCL10/IP-10 protein expression. JC3 compound effectively<br />

decreased mRNA expression level <strong>of</strong> CXCL10/IP-10 and exhibited no toxicity in orbital


TUESDAY<br />

fibroblasts at <strong>the</strong> 10μM concentration. Moreover, <strong>the</strong> result obtained from gel shift assay<br />

showed that JC3 suppressed IFN-γ-induced DNA binding <strong>of</strong> STAT-1. These results suggest that<br />

JC3 modulates high levels <strong>of</strong> CXCL10/IP-10 protein in response to IFN-γ, indicating that JC3<br />

may be an attractive <strong>the</strong>rapeutic agent to reduce <strong>the</strong> inflammatory stage <strong>of</strong> TAO.<br />

2285<br />

IL-15-induced IL-10 increases <strong>the</strong> cytolytic activity <strong>of</strong> human natural killer cells.<br />

T-D. Kim 1 , J. Park 1 , I. Choi 1 ; 1 Korea Research Institute <strong>of</strong> Bioscience and Biotechnology,<br />

Daejeon, Korea<br />

Interleukin 10 (IL-10) is a multifunctional cytokine that regulates diverse functions <strong>of</strong> immune<br />

cells. Natural killer (NK) cells express <strong>the</strong> IL-10 and IL-10 receptor, but little is known about <strong>the</strong><br />

function <strong>of</strong> IL-10 on NK cell activation. In this study, we show <strong>the</strong> expression and role <strong>of</strong> IL-10 in<br />

human NK cells. Among <strong>the</strong> cytokines tested, IL-15 was <strong>the</strong> most potent inducer <strong>of</strong> IL-10, with a<br />

maximal peak expression at 5 h after treatment. Fur<strong>the</strong>rmore, IL-10 receptor was shown to be<br />

expressed in NK cells. IL-10 alone had a significant effect on NK cytotoxicity which additively<br />

increased NK cell cytotoxicity in <strong>the</strong> presence <strong>of</strong> IL-15. Neutralizing IL-10 with anti-IL-10<br />

antibody suppressed <strong>the</strong> inductive effect <strong>of</strong> IL-10 on NK cell cytotoxicity; however, IL-10 had no<br />

effect on IFN-γ or TNF-α production or NK cell activatory receptor expression. STAT signals are<br />

implicated as a key mediator <strong>of</strong> IL-10/IL-15 cytotoxicity response. Thus, <strong>the</strong> effect <strong>of</strong> IL-10 on<br />

NK cells is particularly interesting with regard to <strong>the</strong> STAT3 signal that was enhanced by IL-10<br />

or IL-15.<br />

2286<br />

Anthrax Toxin Receptor 2 Carboxyl-terminal 12-residues are Important for Toxin Entry<br />

into Cells.<br />

R. Jimenez 1 , G. J. Chaudry 1 , S. Manam 1 ; 1 South Texas Center for Emerging Infectious Disease,<br />

Department <strong>of</strong> <strong>Biology</strong>, Univeristy <strong>of</strong> Texas at San Antonio, San Antonio, TX<br />

Anthrax toxin is secreted by <strong>the</strong> Gram-positive, spore-forming, rod-shaped bacterium Bacillus<br />

anthracis. The toxin is one <strong>of</strong> two virulence factors <strong>of</strong> <strong>the</strong> organism, <strong>the</strong> o<strong>the</strong>r being its<br />

antiphagocytic poly-D-glutamate capsule. Three secreted proteins make up anthrax toxin:<br />

protective antigen (PA), edema factor (EF), and lethal factor (LF). EF is an adenylyl cyclase that<br />

harms cells by producing excessive amounts <strong>of</strong> cAMP. LF is a Zn-dependent metalloprotease<br />

that cleaves MAP kinase kinases. PA delivers EF and LF to cytosol, but itself is without any<br />

toxic catalytic activity. Thus, active anthrax toxins are PA+EF and PA+LF. The toxins enter cells<br />

by receptor-mediated endocytosis. PA utilizes two receptors for entry, tumor endo<strong>the</strong>lial marker<br />

8 (TEM8; ANTXR1) and capillary morphogenesis gene 2 protein (CMG2, ANTXR2). The two<br />

membrane-bound forms <strong>of</strong> CMG2 that function as PA receptors comprise 488 and 489<br />

residues, and <strong>the</strong>y differ only in <strong>the</strong> last 12 residues. Our analysis shows that absence <strong>of</strong> <strong>the</strong>se<br />

residues results in a receptor that affords PA binding and its cleavage by furin, but does not<br />

support intoxication by <strong>the</strong> toxin. The block to intoxication appears at <strong>the</strong> level <strong>of</strong> <strong>the</strong> SDSresistant<br />

heptamer, a structure that forms upon exposure to acidic pH in endosomes. Functional<br />

analysis by alanine-scanning mutagenesis revealed that essentially all 12 residues are<br />

important for receptor function. Surprisingly, deletion <strong>of</strong> <strong>the</strong> entire cytoplasmic portion <strong>of</strong> CMG2<br />

left <strong>the</strong> truncated receptor fully functional for anthrax toxin. Thus, <strong>the</strong> 12-residue C-terminal<br />

segment must remain intact for <strong>the</strong> full-length receptor to function, but <strong>the</strong> segment is<br />

dispensable when <strong>the</strong> rest <strong>of</strong> <strong>the</strong> cytoplasmic domain is also absent. The results also suggest<br />

that different mechanisms underlie toxin entry via <strong>the</strong> full-length CMG2 and <strong>the</strong> receptor lacking<br />

<strong>the</strong> cytoplasmic domain.


TUESDAY<br />

2287<br />

Rho A signaling contributes to statin-induced osteogenesis in bone marrow<br />

mesenchymal stem cells.<br />

I-C. TAI 1,2 , M-L. Ho 2,3 , J-K. Chang 2,4 ; 1 Graduate Institute <strong>of</strong> Medicine, Kaohsiung Medical<br />

University, Kaohsiung, Taiwan, 2 Orthopaedic Research Center, Kaohsiung Medical University,<br />

Kaohsiung, Taiwan, 3 Department <strong>of</strong> Physiology, Kaohsiung Medical University, Taiwan,<br />

4 Department <strong>of</strong> Orthopedic Surgery, Kaohsiung Medical UniversityHospital, Taiwan<br />

Statins, 3-Hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, reduce<br />

cholesterol syn<strong>the</strong>sis and prevent cardiovascular disease. Previous in vitro and in vivo studies<br />

showed that statin stimulated bone formation. They also have been found to inhibit prenylation<br />

<strong>of</strong> Rho proteins in recent decade. Previous reports showed that statins inhibited protein<br />

prenylation and decreased <strong>the</strong> active form <strong>of</strong> Rho A in osteoclasts. O<strong>the</strong>rs reports indicated that<br />

statins inhibited protein prenylation but increased <strong>the</strong> active form <strong>of</strong> Rho A in human<br />

erythroleukemia cells. Therefore, <strong>the</strong> role <strong>of</strong> statins regulate Rho A activity remains unclear. Rho<br />

GTPases act as molecular switches to regulate mesenchymal stem cell differentiation. Previous<br />

study showed that transfected constitutively active-form <strong>of</strong> RhoA into human mensenchymal<br />

stem cells (hMSCs) which leaded differentiation <strong>of</strong> hMSCs into osteoblasts. On <strong>the</strong> o<strong>the</strong>r hand,<br />

dominant negative RhoA leaded differentiation <strong>of</strong> hMSCs into adipocytes. According to <strong>the</strong><br />

description above, we want to investigate whe<strong>the</strong>r Rho A signaling contributes to statin-induced<br />

osteogenesis in BMSCs. Pluripotent mesenchymal cells, D1, which were cloned from Balb/c<br />

mouse bone marrow cells and purchased from ATCC. For all experiments, cells were seeded at<br />

a density <strong>of</strong> 80% confluence and treated with or without simvastatin. The mRNA expression <strong>of</strong><br />

Rho A was detected and quantified by real time PCR. And <strong>the</strong> mineralization effect on rat bone<br />

mesenchymal stem cells (rBMSCs) was tested by Alizarin Red S Staining. The Alizrin red S<br />

staining assay showed that <strong>the</strong> simvastatin were potentially enhanced <strong>the</strong> cell mineralization on<br />

BMSCs. The data <strong>of</strong> mRNA expression <strong>of</strong> Rho A showed that simvastatin 1uM significantly<br />

increased Rho A gene expression on first day. However, <strong>the</strong>re were no significantly different<br />

between control and treatment group on third and fifth days. The protein level <strong>of</strong> Rho A<br />

increased with 0.5uM and 1uM but not 0.1uM simvastatin treatment on first day. From <strong>the</strong>se<br />

results, we suggest Rho A signaling may contribute to simvastatin-enhanced osteogenesis in rat<br />

bone mesenchymal stem cells (rBMSCs).<br />

2288<br />

Magnetic Manipulation <strong>of</strong> signaling “hotspots” inside living cells shows contextdependent<br />

amplification <strong>of</strong> <strong>the</strong> Rac pathway.<br />

F. Etoc 1 , M. Coppey 1 , D. Lisse 2 , Y. Bellaiche 3 , J. Piehler 2 , M. Dahan 1 ; 1 LKB-IBENS, Paris,<br />

France, 2 biophysics, Osnabruck University, Germany, 3 Curie Institute, France<br />

In vivo imaging has shown how <strong>the</strong> establishment and maintenance <strong>of</strong> a polarized cellular state<br />

relies on complex mechanisms by which signaling cascades become activated and regulated at<br />

sub-cellular levels. It has recently led to <strong>the</strong> concept <strong>of</strong> spatially-restricted signaling modules.<br />

Yet, it remains unclear which ingredients and processes specify a module. In this context, we<br />

used a new tool to locally perturb and probe signaling pathways associated to <strong>the</strong> small Rho-<br />

GTPases.<br />

First, magnetic nanoparticles (500nm), functionalized with constitutively active Rho-GTPases<br />

(Rac1 and cdc42) were microinjected inside living cells. We observed <strong>the</strong> induction <strong>of</strong><br />

downstream signaling at <strong>the</strong> particle surface by visualization <strong>of</strong> effectors binding (Pak and N-<br />

WASP) and <strong>the</strong> local polymerization <strong>of</strong> actin structures, <strong>the</strong>reby demonstrating that <strong>the</strong>se<br />

particles behave in <strong>the</strong> cytosol as signaling "hotspots". Using FRAP, we fur<strong>the</strong>r measured in


TUESDAY<br />

vivo dissociation rates between <strong>the</strong> immobilized GTPase and its direct effector. Next, <strong>the</strong><br />

nanoparticles were manipulated with a magnetic tip to position <strong>the</strong>ir signaling activity at different<br />

subcellular locations. With nanoparticles functionalized in situ with Tiam (a known Rac1<br />

activator), we were able to specifically activates Rac1 signaling by bringing <strong>the</strong> nanoparticles in<br />

contact with <strong>the</strong> plasma membrane. The Rac1 activation occurred regardless <strong>of</strong> <strong>the</strong> subcellular<br />

localization at <strong>the</strong> membrane. However, <strong>the</strong> signal was non-linearly amplified only when <strong>the</strong><br />

signaling activity was carried to protrusive areas <strong>of</strong> <strong>the</strong> cell. This result reinforces <strong>the</strong> concept <strong>of</strong><br />

subcellular signaling modules and shows that a module could be specified by its ability to<br />

amplify ra<strong>the</strong>r than just linearly transduce incoming signals.<br />

2289<br />

Implications <strong>of</strong> RhoA regulation in Hela cells exposed to stress radiation.<br />

J. H. Osaki 1 , G. E. Silva 1 , F. L. Forti 2 ; 1 Biochemistry, Institute <strong>of</strong> Chemistry, University <strong>of</strong> Sao<br />

Paulo, 2 Biochemistry, Institute <strong>of</strong> Chemistry, University <strong>of</strong> Sao Paulo, Sao Paulo, Brazil<br />

The protein RhoA GTPase belongs to <strong>the</strong> superfamily <strong>of</strong> small GTPases related to Ras. RhoA<br />

are enzymes acting as molecular switches to control a wide variety <strong>of</strong> signal transduction<br />

pathways in eukaryotic cells such as remodeling <strong>of</strong> actin cytoskeleton, cell cycle, and cell death.<br />

RhoA overexpression is found in aggressive breast cancer and cervix carcinomas, where <strong>the</strong><br />

enzyme is usually constitutively activated. Ionizing radiation (gamma and UV) causes single and<br />

double strand breaks in DNA, and also pyrimidine dimers lesions, damages usually repaired by<br />

homolog recombination, non-homologous end-joining and nucleotide excision repair<br />

mechanisms. In this study, using HeLa cells and mutants expressing dominant-negative RhoA<br />

(HeLa-RhoA-N19) and constitutively-active RhoA (HeLa-RhoA-V14), <strong>the</strong> RhoA protein was<br />

analyzed as a possible player in survival, senescence, migration, DNA damage response and<br />

repair after irradiation <strong>of</strong> cells with low doses <strong>of</strong> gamma and UV light. Results show that RhoA<br />

activity was downregulated by <strong>the</strong> highest energy ionizing. The HeLa-RhoA-V14 presents an<br />

increase in cell proliferation, survival and migration rate compared to HeLa-RhoA-N19.<br />

Fur<strong>the</strong>rmore, in RhoA-V14 clones ei<strong>the</strong>r <strong>the</strong> DNA damage is decreased or <strong>the</strong> repair<br />

mechanisms are increased resulting in attenuated cellular senescence after <strong>the</strong> different<br />

ionizing treatments. In conclusion, <strong>the</strong>se data confirm <strong>the</strong> essential role <strong>of</strong> RhoA protein in<br />

proliferation and migration and more intriguingly point to possible involvements <strong>of</strong> this enzyme in<br />

mechanisms <strong>of</strong> DNA repair and cell senescence after genotoxic stress induced by ionizing<br />

radiation.<br />

Supported by Fapesp: Project # 2008/58264-5.<br />

2290<br />

Rho small GTPases activates PI3K via a cooperative positive feedback loop.<br />

H. Lee 1 , H. Yang 1 , M-G. Shin 1 , S. Lee 1 , W. Park 1 , J-R. Kim 1 , K-H. Cho 1 , T. Meyer 2 , W. Heo 1 ;<br />

1 Korea Advanced Institute <strong>of</strong> Science and Technology, Daejeon, Korea, 2 Stanford University<br />

Phosphoinositide3-kinases (PI3Ks) and Ras and Rho family small GTPases are key regulators<br />

<strong>of</strong> processes such as cell polarization, motility and chemotaxis. These signaling components<br />

may control each o<strong>the</strong>r¡¯s activities by direct and indirect feedback processes that are only<br />

partially understood. Here, we used a live cell screen to evaluate <strong>the</strong> role <strong>of</strong> 100 human small<br />

GTPases in <strong>the</strong> regulation <strong>of</strong> PI3K and found that 21 small GTPase homologs activate PI3K.<br />

Using a protein-protein interaction assay that we recently developed, we found that K-Ras, H-<br />

Ras and 5 o<strong>the</strong>r homologs function upstream <strong>of</strong> PI3K and directly bind to <strong>the</strong> PI3K catalytic<br />

subunit, p110, while three Rac is<strong>of</strong>orms and 11 o<strong>the</strong>r small GTPase homologs activate PI3K by<br />

indirect feedback mechanisms. Fur<strong>the</strong>rmore, live cell experiments and in silico analysis<br />

revealed that significant level <strong>of</strong> PI3K activation can be generated by cooperation <strong>of</strong> multiple


TUESDAY<br />

activators in endogenous level. We demonstrate two modes <strong>of</strong> PI3K activation that is initiated by<br />

upstream activators and is fur<strong>the</strong>r enhanced by <strong>the</strong> cooperation <strong>of</strong> PI3K activators.<br />

2291<br />

Arl4A complexes with ELMO to promote actin cytoskeleton remodeling.<br />

T-C. Chiang 1 , M. Patel 2,3 , V. Tran 2 , F-J. Lee 1 , J-F. Cote 2,3 ; 1 Institute <strong>of</strong> <strong>Molecular</strong> Medicine,<br />

Taipei, Taiwan, 2 Institut de Recherches Cliniques de Montreal, 3 Departement de Medicine,<br />

Université de Montréal<br />

The prototypical DOCK protein, DOCK180, is an evolutionarily conserved Rac regulator and is<br />

indispensable during processes such as cell migration and myoblast fusion. The biological<br />

activity <strong>of</strong> DOCK180 is tightly linked to its binding partner ELMO. We previously reported that<br />

auto-inhibited ELMO proteins regulate signaling from this pathway. One mechanism to activate<br />

<strong>the</strong> ELMO/DOCK180 complex appears to be <strong>the</strong> recruitment <strong>of</strong> this complex to <strong>the</strong> membrane<br />

via <strong>the</strong> Ras-binding domain (RBD) <strong>of</strong> ELMO. In <strong>the</strong> present study, we aimed to identify novel<br />

ELMO-interacting proteins to fur<strong>the</strong>r define <strong>the</strong> molecular events capable <strong>of</strong> controling ELMO<br />

recruitment to <strong>the</strong> membrane. To do so, we performed two independent interaction screens: one<br />

specifically interrogated an active GTPase library while <strong>the</strong> o<strong>the</strong>r probed a brain cDNA library.<br />

Both methods converged on Arl4A, an Arf-related GTPase, as a specific ELMO interactor.<br />

Biochemically, Arl4A is constitutively GTP-loaded and our binding assays confirm that both wildtype<br />

and constitutively active forms <strong>of</strong> <strong>the</strong> GTPase associate with ELMO. Mechanistically, we<br />

report that Arl4A binds <strong>the</strong> ELMO RBD and acts as a membrane localization signal for ELMO. In<br />

addition, we report that membrane targeting <strong>of</strong> ELMO via Arl4A promotes cytoskeletal<br />

reorganization including membrane ruffling and stress fiber disassembly via an<br />

ELMO/DOCK180/Rac signaling pathway. We conclude that ELMO is capable <strong>of</strong> interacting with<br />

GTPases from Rho and Arf families, leading to <strong>the</strong> conclusion that ELMO contains a versatile<br />

RBD. Fur<strong>the</strong>rmore, via binding <strong>of</strong> an Arf family GTPase, <strong>the</strong> ELMO/DOCK180 is uniquely<br />

positioned at <strong>the</strong> membrane to activate Rac signaling and remodel <strong>the</strong> actin cytoskeleton.<br />

2292<br />

New biosensor for Cdc42-GDI complexation reveals dynamic regulation <strong>of</strong> GDI-Cdc42<br />

binding by Src-kinase.<br />

D. Spiering 1 , L. Hodgson 1 , M. Sabouri 2 , C. der Mardirossian 2 , G. Danuser 3 , K. Hahn 4 ; 1 Albert<br />

Einstein College <strong>of</strong> Medicine, Bronx, NY, 2 The Scripps Research Institute, La Jolla, CA,<br />

3 Harvard Medical School, Boston, MA, 4 University <strong>of</strong> North Carolina at Chapel Hill, Chapel Hill,<br />

NC<br />

Signaling by <strong>the</strong> p21 Rho family GTPases is critically important in a broad range <strong>of</strong> cell<br />

behaviors. They have been most extensively studied in motility and polarization, where <strong>the</strong>ir<br />

activation (GTP binding) is tightly coordinated in space and time to generate cell morphological<br />

changes. Guanosine Dissociation Inhibitors (GDI) sequester inactive (GDP bound) GTPases in<br />

<strong>the</strong> cytosol, and mediate localization <strong>of</strong> <strong>the</strong> activated GTPase in <strong>the</strong> plasma membrane. The<br />

role <strong>of</strong> GDI in controlling <strong>the</strong> precise placement and timing <strong>of</strong> GTPase activation remains poorly<br />

understood. To investigate this, we constructed fluorescent biosensors reporting <strong>the</strong> interaction<br />

<strong>of</strong> endogenous RhoGDI with ei<strong>the</strong>r Cdc42, Rac1 or RhoA. Using <strong>the</strong> Cdc42-GDI biosensor<br />

toge<strong>the</strong>r with <strong>the</strong> meroCBD biosensor detecting <strong>the</strong> activation <strong>of</strong> endogenous Cdc42 in MEFs,<br />

we imaged simultaneously <strong>the</strong> Cdc42–endogenous GDI complexation dynamics and <strong>the</strong><br />

endogenous Cdc42 activation dynamics in single living cells. Using <strong>the</strong> morphodynamics<br />

mapping and computational multiplexing technology, we show directly for <strong>the</strong> first time that <strong>the</strong><br />

Cdc42 uptake rate by <strong>the</strong> GDI is regulated through Src-kinase-mediated phosphorylation.


Proteostasis, Cell Stress, and Aging<br />

TUESDAY<br />

2293<br />

Is <strong>the</strong> ubiquitin ligase itch phosphorylation modulating its binding and ligase activity<br />

towards all substrates?<br />

R. Forget 1 , A. Angers 1 ; 1 Biological Sciences, University <strong>of</strong> Montreal, Montréal, Québec, Canada<br />

Itch is an ubiquitin ligase closely related to Nedd4. These enzymes bind to <strong>the</strong>ir substrate<br />

proteins with <strong>the</strong>ir central region and transfer ubiquitin through <strong>the</strong>ir C-terminal catalytic HECT<br />

domain. By promoting <strong>the</strong> proteasomal degradation <strong>of</strong> substrate proteins, ubiquitin ligases are<br />

involved in a variety <strong>of</strong> process like endocytosis, apoptosis, inflammation, cell signaling, etc.<br />

Recently, <strong>the</strong> regulation <strong>of</strong> <strong>the</strong> activity <strong>of</strong> ubiquitin ligases <strong>the</strong>mselves has received a lot <strong>of</strong><br />

attention. Itch is <strong>the</strong> target <strong>of</strong> <strong>the</strong> cJun N-terminal kinase (JNK) which phosphorylates three<br />

residues in a region overlapping with a proline-rich region <strong>of</strong> Itch that mediates recognition by<br />

SH3-domain proteins. Phosphorylation <strong>of</strong> Itch by JNK has been shown to regulate its activity<br />

towards itself and substrates cJun and JunB. An inhibitory intramolecular interaction abrogated<br />

by JNK-phosphorylation has been proposed to mediate this effect. Here, we evaluate if this<br />

model can be generalized to o<strong>the</strong>r Itch substrates, most specifically endophilin that has been<br />

shown to interact with Itch through its SH3 domain, distinct from most Itch known substrates.<br />

Using site-directed mutagenesis, we created mutants <strong>of</strong> Itch that could not be phosphorylated<br />

by JNK, and a phosphomimic mutant. These mutants were tested for <strong>the</strong>ir ability to interact with<br />

endophilin, and to mediate endophilin ubiquitylation and proteasomal degradation. We found<br />

that Itch phosphorylation by JNK has no effect on endophilin recognition by Itch, nor on <strong>the</strong><br />

ligase ability to promote endophilin ubiquitylation. Although Itch phosphorylation is not required<br />

for proteasomal degradation <strong>of</strong> endophilin, a small facilitating effect <strong>of</strong> Itch phosphorylation<br />

could be observed. In conclusion, if JNK- mediated phosphorylation <strong>of</strong> Itch can slightly<br />

potentiate its ubiquitin- ligase activity, it has no incidence on <strong>the</strong> interaction between Itch and<br />

SH3 proteins like endophilin. Therefore, <strong>the</strong> regulation <strong>of</strong> Itch activity seems to differ according<br />

to <strong>the</strong> selected substrate.<br />

2294<br />

Potentiation <strong>of</strong> ER stress-mediated apoptotic pathway by protein tyrosine kinase p56lck<br />

in proteasome inhibitor MG132-induced apoptosis human Jurkat T cells.<br />

C. Han 1 , H. Park 1 , D. Jun 1 , H. Woo 1 , J. Lee 1 , Y. Kim 1 ; 1 School <strong>of</strong> Life Science and<br />

Biotechnology, Kyungpook National University, Daegu, Korea<br />

Exposure <strong>of</strong> human Jurkat T cells to MG132 caused apoptosis along with upregulation <strong>of</strong><br />

Grp78/BiP and CHOP/GADD153, activation <strong>of</strong> JNK and p38MAPK, activation <strong>of</strong> Bak,<br />

mitochondrial membrane potential (¥Ä¥÷m) loss, cytochrome c release, activation <strong>of</strong> caspase-<br />

12, -9, -3, -7, and -8, cleavage <strong>of</strong> Bid and PARP, and DNA fragmentation. However, <strong>the</strong>se<br />

MG132-induced apoptotic events, with <strong>the</strong> exceptions <strong>of</strong> upregulation <strong>of</strong> Grp78/BiP and<br />

CHOP/GADD153 and activation <strong>of</strong> JNK and p38MAPK, were abrogated by Bcl-xL<br />

overexpression. Pretreatment with <strong>the</strong> pan-caspase inhibitor z-VAD-fmk prevented MG132induced<br />

apoptotic caspase cascade, but allowed upregulation <strong>of</strong> Grp78/BiP and<br />

CHOP/GADD153 levels, activation <strong>of</strong> JNK and p38MAPK, ¥Ä¥÷m loss, and cleavage <strong>of</strong><br />

procaspase-9 (47 kDa) to active form (35 kDa). Fur<strong>the</strong>r analysis using selective caspase<br />

inhibitors revealed that caspase-12 activation was required for activation <strong>of</strong> caspase-9 and -3 to<br />

<strong>the</strong> sufficient level for subsequent activation <strong>of</strong> caspase-7 and -8. MG132-induced cytotoxicity,<br />

apoptotic sub-G1 peak, Bak activation, and ¥Ä¥÷m loss were reduced in part by p38MAPK<br />

inhibitor, but not by JNK inhibitor. MG132-induced apoptotic changes, including upregulation <strong>of</strong><br />

Grp78/BiP and CHOP/GADD153 levels, activation <strong>of</strong> caspase-12, p38MAPK and Bak, and


TUESDAY<br />

mitochondria-dependent activation <strong>of</strong> caspase cascade were more significant in p56lck-stable<br />

transfectant JCaM1.6/lck than in p56lck-dificient JCaM1.6/vector. The cytotoxicity <strong>of</strong> MG132<br />

toward p56lck-positive Jurkat T cell clone was not affected by <strong>the</strong> Src-like kinase inhibitor PP2.<br />

These results demonstrated that MG132-induced apoptosis was caused by ER stress and<br />

subsequent activation <strong>of</strong> mitochondria-dependent caspase cascade, and that <strong>the</strong> presence <strong>of</strong><br />

p56lck enhances MG132-induced apoptosis by augmenting ER stress-mediated apoptotic<br />

events in Jurkat T cells.<br />

Key words: proteasome inhibitor, apoptosis, ER stress, mitochondria-dependent caspase<br />

cascade,<br />

p56lck<br />

2295<br />

Translation elongation factor eEF3 and translation termination factor eRF3 are novel<br />

constituents <strong>of</strong> heat-induced stress granules in Saccharomyces cerevisiae.<br />

T. Grousl 1 , P. Ivanov 2 , I. Malcova 1 , I. Frydlova 1 , P. Pompach 1 , R. Slaba 1 , P. Vasicova 1 , L.<br />

Novakova 1 , J. Hasek 1 ; 1 Inst. <strong>of</strong> Microbiology <strong>of</strong> ASCR, v.v.i., Prague 4, Czech Republic, 2 Faculty<br />

<strong>of</strong> <strong>Biology</strong>, Moscow State University, Moscow, Russia<br />

In response to severe environmental stresses eukaryotic cells shut down translation and form<br />

transient mRNA assemblies called stress granules (SGs). Stress granules are <strong>of</strong>ten referred to<br />

as dominant accumulations <strong>of</strong> stalled translation preinitiation complexes since <strong>the</strong>ir typical<br />

components are <strong>the</strong> translation initiation factor eIF3 and <strong>the</strong> small ribosomal subunit 40S. We<br />

have shown previously that in <strong>the</strong> budding yeast Saccharomyces cerevisiae <strong>the</strong> SGs containing<br />

<strong>the</strong> eIF3 complex and <strong>the</strong> 40S ribosomal subunit are formed upon <strong>the</strong> robust heat shock at 46°C<br />

only. The formation <strong>of</strong> <strong>the</strong>se SGs is independent <strong>of</strong> phosphorylation <strong>of</strong> eIF2alpha by Gcn2<br />

kinase and also <strong>of</strong> <strong>the</strong> scaffolding proteins <strong>of</strong> P bodies Edc3 and Lsm4. Here we present <strong>the</strong><br />

evidence that <strong>the</strong> robust heat shock-induced SGs <strong>of</strong> S. cerevisiae contain also <strong>the</strong> translation<br />

elongation factor eEF3 (Yef3). In addition, distinct accumulations <strong>of</strong> Yef3GFP were observed in<br />

<strong>the</strong> edc3lsm4C mutant even upon a mild heat shock at 42°C and <strong>the</strong>y pre-determined<br />

accumulations <strong>of</strong> eIF3a upon <strong>the</strong> robust heat shock at 46°C. Fur<strong>the</strong>rmore, some <strong>of</strong> <strong>the</strong> robust<br />

heat shock-induced stress granules co-localized with accumulations <strong>of</strong> Sup35 suggesting that<br />

also translation termination factor eRF3 is a constituent <strong>of</strong> SGs. While <strong>the</strong> N-terminal domain <strong>of</strong><br />

Sup35 is essential for [PSI + ] prion formation its absence did not affect assembly <strong>of</strong> SGs. Our<br />

data indicate that SGs containing translation preinitiation complexes and termination factors are<br />

successively assembled on accumulations <strong>of</strong> <strong>the</strong> elongation factor eEF3 (Yef3) that was<br />

released from translation during heat shock. This work was financed by grants CSF<br />

204/09/1924, LC545 and IRCAV0Z50200510.<br />

2296<br />

Pathogenic missense mutations located in distinct domains affect <strong>the</strong> folding and<br />

function <strong>of</strong> dystrophin.<br />

D. M. Strandjord 1 , D. M. Henderson 1 , B. Li 1 , J. M. Ervasti 1 ; 1 University <strong>of</strong> Minnesota,<br />

Minneapolis, MN<br />

Dystrophin is a 427-kDa protein encoded by <strong>the</strong> DMD gene on <strong>the</strong> X chromosome. This<br />

structural protein localizes to <strong>the</strong> plasma membrane <strong>of</strong> muscle cells, where it establishes a<br />

mechanical link between <strong>the</strong> cytoskeleton and <strong>the</strong> extracellular matrix. The N-terminus and a<br />

segment <strong>of</strong> <strong>the</strong> middle rod domain in dystrophin interact with cytoplasmic actin filaments while<br />

modules near <strong>the</strong> C-terminus interact with <strong>the</strong> transmembrane protein �-dystroglycan.<br />

Dystrophin and �-dystroglycan, along with several o<strong>the</strong>r proteins, function as a complex known


TUESDAY<br />

as <strong>the</strong> dystrophin-glycoprotein complex, or DGC, which is thought to stabilize <strong>the</strong> sarcolemma<br />

against mechanical stress.<br />

Males with a mutation in <strong>the</strong>ir only copy <strong>of</strong> <strong>the</strong> gene encoding dystrophin are diagnosed<br />

as having ei<strong>the</strong>r Duchene Muscular Dystrophy (DMD) or Becker Muscular Dystrophy (BMD).<br />

Many patients have mutations that cause a dystrophin-deficiency in cells and develop extreme<br />

muscle deterioration in <strong>the</strong>ir first decade <strong>of</strong> life. More recently, sequencing has revealed that<br />

some patients have mutations in <strong>the</strong> dystrophin gene that result in a single amino acid change<br />

in <strong>the</strong> protein. Less is known about <strong>the</strong> molecular pathomechanism <strong>of</strong> <strong>the</strong>se missense<br />

mutations. We recently demonstrated that missense mutations in <strong>the</strong> N-terminal actin-binding<br />

domain (ABD1) <strong>of</strong> dystrophin had minimal effect on actin binding activity, but caused <strong>the</strong>rmal<br />

instability and aggregation in vitro (D.M. Henderson et al., 2010 PNAS). Additional pathogenic<br />

missense mutations have been identified in <strong>the</strong> �-dystroglycan binding domain (DgBD) <strong>of</strong><br />

dystrophin. To determine <strong>the</strong> effect <strong>of</strong> <strong>the</strong>se pathogenic changes in a location distinct from<br />

ABD1, we engineered <strong>the</strong> disease-causing missense mutations D3187G, C3207R, F3228L,<br />

A3311P, C3313F, D3335Y into dystrophin. Both wild-type dystrophin and E3032K, a nonsynonomous<br />

Single Nucleotide Polymorphism (SNP) found in <strong>the</strong> population, served as<br />

controls. When expressed in insect cell culture, quantification <strong>of</strong> <strong>the</strong> protein in <strong>the</strong> soluble versus<br />

insoluble fractions <strong>of</strong> <strong>the</strong> cell lysate revealed that some missense mutations significantly<br />

decreased <strong>the</strong> solubility <strong>of</strong> dystrophin. Circular Dichroism melt curves <strong>of</strong> purified proteins<br />

revealed that missense mutations decreased <strong>the</strong> cooperative unfolding transition which is<br />

indicative <strong>of</strong> a misfolded, heterogeneous population <strong>of</strong> protein. Supported by <strong>the</strong> Torske<br />

Klubben Graduate Fellowship and a grant from <strong>the</strong> National Institutes <strong>of</strong> Health (AR042423).<br />

2297<br />

Two distinguishable aging states found in an isogenic population at a single<br />

chronological age.<br />

D. M. Eckley 1 , S. Rahimi 1 , N. V. Orlov 1 , S. Mantilla 1 , C. E. Coletta 1 , J. D. Delaney 1 , Y. Zhang 2 ,<br />

K. Becker 2 , W. Iser 3 , M. Wilson 3 , I. G. Goldberg 1 ; 1 IICBU, Laboratory <strong>of</strong> Genetics, National<br />

Institute on Aging (NIA), National Institutes <strong>of</strong> Health, Baltimore, MD, 2 Research Resources<br />

Branch, NIA-Intramural Research Program, 3 IMGU, Laboratory <strong>of</strong> Neuroscience, NIA<br />

We present <strong>the</strong> initial molecular characterization <strong>of</strong> a transition between two early aging states.<br />

An age score reflecting physiological age was developed using a machine classifier. Images <strong>of</strong><br />

C. elegans populations at fixed chronological ages throughout <strong>the</strong>ir lifespan were used for<br />

training. The distribution <strong>of</strong> age scores identified three stable post-developmental states and<br />

transitions. We examined <strong>the</strong> transition that occurs at day 5 post L4 molt, because a significant<br />

percentage <strong>of</strong> that population exists in both State I and State II. We measured <strong>the</strong> temperature<br />

dependence <strong>of</strong> <strong>the</strong> timing <strong>of</strong> <strong>the</strong> transition (Q10 ~ 1.17). This value is too low to be explained by<br />

a step-wise process with an enzymatic or chemical rate-limiting step, implicating a more<br />

complex timing mechanism. Individual animals at day 5 were sorted into State I and State II<br />

groups by <strong>the</strong> machine classifier and analyzed by microarray expression pr<strong>of</strong>iling. Despite being<br />

isogenic, grown for <strong>the</strong> same amount <strong>of</strong> time, and indistinguishable by eye, <strong>the</strong>se two<br />

morphological states were confirmed to be molecularly distinct by hierarchical clustering and<br />

principal component analysis. The distinction demonstrates that pharynx morphology must<br />

reflect <strong>the</strong> aging state <strong>of</strong> <strong>the</strong> whole organism. Our expression pr<strong>of</strong>iling also yielded a gene set<br />

that showed significant overlap with those from three previous age-related studies and identified<br />

several genes not previously implicated in aging. A highly represented group <strong>of</strong> genes unique to<br />

this study is involved in <strong>the</strong> ubiquitination pathway, including Skip1-related (SKR) as well as Fbox-containing<br />

adaptors which target specific proteins for degradation.


TUESDAY<br />

2298<br />

Glucose Stress Reduces Fertility and Impairs Mating in Caenorhabditis elegans.<br />

A. K. Engstrom 1 , M. R. Liggett 1 , M. A. Mondoux 1 ; 1 Department <strong>of</strong> <strong>Biology</strong>, College <strong>of</strong> <strong>the</strong> Holy<br />

Cross, Worcester, MA<br />

Diabetes is a chronic disease marked by insulin resistance and <strong>the</strong> inability <strong>of</strong> cells to correctly<br />

respond to high levels <strong>of</strong> blood glucose. Diabetes currently affects 25.8 million people in <strong>the</strong><br />

United States, and has been shown to lead to increased potential for birth defects, miscarriages,<br />

and infertility. Caenorhadiditis elegans has a conserved insulin-signaling pathway, making it an<br />

ideal model organism for studying <strong>the</strong> effects <strong>of</strong> excess glucose exposure. In previous studies,<br />

glucose stress in C. elegans was shown to lead to both a developmental delay and a reduction<br />

in hermaphrodite fertility; however <strong>the</strong> cause(s) <strong>of</strong> <strong>the</strong>se defects is unknown. By exposing<br />

worms to high glucose during ei<strong>the</strong>r development or adulthood, and <strong>the</strong>n studying <strong>the</strong>ir<br />

reproductive pr<strong>of</strong>ile and brood size, we found that <strong>the</strong> developmental delay is separable from<br />

<strong>the</strong> decrease in fertility. Feeding high glucose only during development led to a two-day<br />

reproductive delay but had no effect on fertility. Likewise, glucose stress during adulthood<br />

reduced fertility as much as lifetime exposure to high glucose. We assayed post-fertilization<br />

phenotypes under high glucose conditions and saw no change in external or internal hatching,<br />

suggesting that glucose stress causes defects in <strong>the</strong> adult germline. We also tested whe<strong>the</strong>r<br />

glucose stress affects male fertility and mating and find that glucose stress interferes with<br />

mating, leading to decreased brood sizes and decreased numbers <strong>of</strong> male <strong>of</strong>fspring. Preliminary<br />

evidence suggests that male fertility is also decreased by glucose stress. We are currently<br />

investigating <strong>the</strong> roles <strong>of</strong> <strong>the</strong> insulin-signaling pathway and apoptosis in <strong>the</strong> response to glucose<br />

stress in hermaphrodites and determining which components <strong>of</strong> <strong>the</strong> male fertility and mating<br />

pathways respond to glucose stress.<br />

2299<br />

Differences in neuropeptide Y and Tirosine Hydroxylase levels related to ageing and<br />

gender in Locus Coeruleus.<br />

K. L. Farizatto 1 , M. F. Ferrari 1 , D. R. Chadi 2 ; 1 Faculdade de Medicina, Fisiopatologia<br />

Experimental, Universidade de Sao Paulo, Sao Paulo, Brazil, 2 Universidade de Sao Paulo, Sao<br />

Paulo, Brazil<br />

Age is a important factor for emerging <strong>of</strong> some diseases, as neurodegeneration and<br />

hypertension. Locus Coeruleus (LC) is a brain area involved with autonomic functions and<br />

behavior wich play a role in differences <strong>of</strong> blood pressure between males and females during<br />

lifespan. The goal <strong>of</strong> this study was to analyze <strong>the</strong> NPY and TH levels in <strong>the</strong> LC <strong>of</strong><br />

spontaneously hypertensive (SHR) rats during aging in males and females. Methods: All <strong>the</strong><br />

procedures and protocols were performed in accordance with <strong>the</strong> Institutional and International<br />

Guidelines for Animal Experimentation. We have employed SHR rats <strong>of</strong>: 3 months, 1 and 2<br />

years (N=6) old. Animals were decapitated, <strong>the</strong> brains were frozen to achieve coronal sections<br />

from pons for <strong>the</strong> analysis <strong>of</strong> LC, through in situ hybridization and imunohistochemistry<br />

techniques. Afterwards, sections were analyzed by means <strong>of</strong> a semiquantitative<br />

microdensitometric procedure. Results are showed as mean ± standard error in arbitrary units.<br />

Two-way ANOVA was used for statistical analysis. Results: NPY mRNA expression in <strong>the</strong> LC is<br />

increased at 2 years females (0,056 ± 0,005) as compared to 1 year (0.036 ± 0.004) and<br />

decreased at 1 year (0.036 ± 0.004) as compared to 3 months old female rats (0.090 ± 0.010).<br />

In males group is decreased at 2 years (0.031 ± 0.002) as compared to 3 months old male rats<br />

(0.090 ± 0.016), (p


TUESDAY<br />

peptide is increased at 2 years females (80.5 ± 2.5) as compared to 1 year (68.0 ± 2.5) and 3<br />

months old female rats (60.0 ± 1.5). In males group is decreased at 2 years (54.5 ± 3.0) as<br />

compared to 3 months old male rats (65.5 ± 2.0), (p


TUESDAY<br />

2301<br />

Depletion <strong>of</strong> ARMS enhances reactive oxygen species-induced autophagy.<br />

Y-H. Liao 1,2 , P-H. Huang 3,4 ; 1 Department <strong>of</strong> Dermatology, National Taiwan University College <strong>of</strong><br />

Medicine, Taipei, Taiwan, 2 Department <strong>of</strong> Dermatology, National Taiwan University Hospital,<br />

Taipei, Taiwan, 3 Institute <strong>of</strong> Pathology, National Taiwan University College <strong>of</strong> Medicine, Taipei,<br />

Taiwan, 4 Department <strong>of</strong> Pathology, National Taiwan University Hospital, Taipei, Taiwan<br />

Increased resistance to oxidative stress in melanoma cells plays an important role for<br />

melanoma development and contributes to <strong>the</strong> ineffectiveness <strong>of</strong> treatment aimed at oxidative<br />

stress-induced cell death. Autophagy is a self-digestion process in response to stimuli including<br />

oxidative stress. We have previously found that depletion <strong>of</strong> ankyrin repeat-rich membrane<br />

spanning (ARMS) protein in melanoma cells caused significantly increased autophagosomes.<br />

To determine <strong>the</strong> role <strong>of</strong> ARMS in oxidative stress-induced autophagy, we established siRNAmediated<br />

ARMS-knockdown and control B16F0 stable melanoma cell lines. We found that<br />

depletion <strong>of</strong> ARMS significantly enhanced autophagosome formation, as evidenced by<br />

autophagic vacuolization examined by electron microscopy, punctate translocation <strong>of</strong> GFP-LC3<br />

(microtubule-associated protein light chain 3) by immun<strong>of</strong>luorescence, and increased LC3-II by<br />

Western blotting. Significantly increased intracellular reactive oxygen species production in<br />

ARMS-RNAi cells at baseline and in time course <strong>of</strong> hydrogen peroxide treatment was revealed<br />

by DCHF-DA flow cytometry. Moreover, <strong>the</strong> percentage <strong>of</strong> cells with GFP-LC3 puncta was<br />

suppressed by treatment with antioxidant N-acetyl-L-cysteine or tiron in ARMS-RNAi cells. In<br />

vivo knockdown <strong>of</strong> ARMS in Drosophila imaginal eye discs also showed robust autophagy. In<br />

conclusion, <strong>the</strong>se results suggested that ARMS silencing increased ROS production, which led<br />

to enhanced autophagy.<br />

2302<br />

Physiological role <strong>of</strong> <strong>the</strong> membrane contact site:Nucleus-Vacuole (NV) junctions.<br />

C-C. Lin 1 , D. S. Goldfarb 1 ; 1 <strong>Biology</strong>, University <strong>of</strong> Rochester, Rochester, NY<br />

Nucleus-Vacuole (NV) junctions are membrane contact sites (MCSs) where perinuclear ER<br />

(nuclear envelope) and vacuole membrane come into close apposition. NV junctions are formed<br />

by <strong>the</strong> direct interaction between outer nuclear membrane protein Nvj1 and <strong>the</strong> vacuole<br />

membrane protein Vac8. A unique autophagic process in yeast Saccharomyces cerevisiae<br />

called ‘Pieacemeal microautophagy <strong>of</strong> <strong>the</strong> nucleus (PMN)’ emanates from NV junctions. Osh1<br />

and Tsc13, which are involved in lipid homeostasis, are recruited to NV junctions and are<br />

essential for normal PMN biogenesis. Here, we report ano<strong>the</strong>r protein that is sequestered to NV<br />

junctions-Nvj2. Unlike Osh1 and Tsc13, Nvj2 is nei<strong>the</strong>r required for NV junctions formation nor<br />

PMN process. Nvj2 localizes on ER, and is enriched on NV junctions upon Nvj1 expression is<br />

up regulated. Nvj1-Vac8 complex and <strong>the</strong> Y321 <strong>of</strong> Nvj1 are essential but not sufficient for <strong>the</strong><br />

enrichment <strong>of</strong> Nvj2 on NV junctions. The absence <strong>of</strong> NVJ2 confers fungicide resistance, and<br />

overexpression <strong>of</strong> Nvj1, which sequesters Nvj2 to NV junctions, phenocopy <strong>the</strong> deletion <strong>of</strong><br />

NVJ2. These results suggest an alternative role <strong>of</strong> NV junctions for <strong>the</strong> fungicide resistance<br />

o<strong>the</strong>r than PMN process.<br />

2303<br />

Premature aging <strong>of</strong> yeast Saccharomyces cerevisiae RAS2 val19 mutant.<br />

A. Pichová 1 , K. Sigler 1 ; 1 Institute <strong>of</strong> Microbiology, Academy <strong>of</strong> Sciences <strong>of</strong> <strong>the</strong> Czech Republic,<br />

Praha 4, Czech Republic<br />

Aging and apoptosis, multifactorial processes involving a complex network <strong>of</strong> regulations, are<br />

conveniently studied on single cell models. Yeast, especially S. cerevisiae, is suitable for this


TUESDAY<br />

purpose because it exhibits mo<strong>the</strong>r cell specific aging, where <strong>the</strong> cell divides asymmetrically,<br />

giving rise to a next-generation large mo<strong>the</strong>r cell and a smaller daughter cell that resets <strong>the</strong><br />

clock to zero while mo<strong>the</strong>r cell accumulates all aging-connected changes. This asymmetry is<br />

disturbed in <strong>the</strong> oncogene mutant RAS2 val19 . This strain, known as a short-lived mutant, shows<br />

also a higher sensitivity to starvation and heat shocks. Micromanipulations revealed a high<br />

percentage <strong>of</strong> virgins unable to form micro- and macro-colonies. Young cells, mostly virgins,<br />

separated by centrifugal elutriation show markers <strong>of</strong> apoptosis, such as having a high load <strong>of</strong><br />

ROS (reactive oxygen species) detected by DHR (dihydrorhodamine); most cells appeared with<br />

DNA breaks stained by <strong>the</strong> TUNEL test and <strong>the</strong>ir nuclei also appeared to have aberrant<br />

morphology. Quantitative comparison <strong>of</strong> <strong>the</strong> amount <strong>of</strong> extrachromosomal DNA rings (ECR) in<br />

separated young and old cells <strong>of</strong> <strong>the</strong> wild type, RAS2 val19 mutant and a strain with delayed aging<br />

toge<strong>the</strong>r with o<strong>the</strong>r data point to <strong>the</strong> premature aging being connected with symmetric aging,<br />

where <strong>the</strong> daughter cell is already inheriting some features <strong>of</strong> aging.<br />

2304<br />

The effects <strong>of</strong> OYE2 and ZWF1 overexpression in furfural stressed Saccharomyces<br />

cerevisiae.<br />

N. Carlson 1 , R. McRae 1 , S. Raehtz 1 , S. W. Gorsich 1 ; 1 <strong>Biology</strong>, Central Michigan University,<br />

Mount Pleasant, MI<br />

A renewable fuel source is needed as economical, environmental, and security concerns<br />

continue to rise regarding <strong>the</strong> use <strong>of</strong> fossil fuels. Bio-ethanol is one potential solution. In <strong>the</strong> U.S.<br />

bio-ethanol is mainly produced by fermenting agricultural products such as corn starch.<br />

However, <strong>the</strong> use <strong>of</strong> corn starch is problematic due to concerns with quantity <strong>of</strong> <strong>of</strong> corn supply<br />

and <strong>the</strong> competing use <strong>of</strong> corn as a food source. Alternatively, biomass waste products such as<br />

lignocellulose (e.g. corn stover, wood chips) could be used. Currently, lignocellulose has not<br />

been optimized for fermentation. One major barrier to using lignocellulose is that during its<br />

hydrolysis many growth inhibitors are produced. Furfural is a major inhibitor generated. Furfural<br />

has been shown to inhibit growth and metabolic enzymes, reduce ethanol yield, and increase<br />

reactive oxygen species (ROS) that go onto damage internal membranes, actin filaments, and<br />

chromatin. Furfural can be removed using various chemical and physical methods, but at <strong>the</strong><br />

expense <strong>of</strong> increased cost and environmental waste production. Our method to reduce <strong>the</strong><br />

effect <strong>of</strong> furfural is to genetically engineer yeast that are robust enough to tolerate high<br />

concentrations <strong>of</strong> furfural while optimizing ethanol yield. For instance, overexpressing ei<strong>the</strong>r<br />

ZWF1 (Zwf1p produces NADPH from <strong>the</strong> pentose phosphate pathway) or OYE2 (Oye2p is an<br />

NADPH-dependent reductase linked to oxidative stress tolerance) individually improves<br />

tolerance to furfural and reduces ROS accumulation and damage to cellular structures. In <strong>the</strong><br />

present study we show how overexpressing both OYE2 (pRS425-MET25-OYE2) and ZWF1<br />

(pRS426-ZWF1) in <strong>the</strong> same yeast strain effects overall furfural tolerance as assayed by growth<br />

(absorbance A600), reactive oxygen species (ROS) accumulation (dihydroethidium stain),<br />

membrane damage (mitochondrial targeted-GFP and FM 4-64), and chromatin damage (DAPI<br />

stain). Our strategy <strong>of</strong> genetic engineering fermenting microorganisms is valuable in improving<br />

<strong>the</strong> technology needed to optimize bio-ethanol fermentation from lignocellulose.<br />

2305/L80<br />

Vitamin C recycling and glutamate effects in astrocytes.<br />

P. Cisternas 1 , K. Oyarce 1 , P. Llanos 1 , C. Silva-Alvarez 1 , F. Nualart 1 ; 1 Department <strong>of</strong> Cell<br />

<strong>Biology</strong>, University <strong>of</strong> Concepcion, Concepcion, Chile<br />

Specialized cells transport vitamin C in its reduced form using sodium-dependent cotransporters<br />

(SVCT1 and SVCT2). Additionally, different cells transport <strong>the</strong> oxidized form <strong>of</strong> vitamin C,


TUESDAY<br />

dehydroascorbic acid (DHA), through glucose transporters (GLUTs). Vitamin C recycling<br />

associated with neuron-astrocyte interactions needs specific AA uptake in neurons and DHA<br />

uptake in astrocytes. SVCT2 is expressed principally in neurons mediating temperature- and<br />

sodium- dependent AA uptake. GLUT1 is expressed in astrocytes mediating glucose-DHA<br />

uptake. It has been suggested that glutamate increases vitamin C recycling. However, <strong>the</strong><br />

specific mechanism has not yet been defined. In this work we analyzed <strong>the</strong> expression <strong>of</strong><br />

GLUT1 and GLUT3 primary cultures <strong>of</strong> astrocytes and neurons obtained from rat brain, and<br />

assessed <strong>the</strong>ir differential capacities to incorporate AA or DHA. Using HPLC and radioactive<br />

pulse tracing we defined <strong>the</strong> effect <strong>of</strong> glutamate in <strong>the</strong> regeneration <strong>of</strong> AA in neurons and<br />

astrocytes. Our results confirmed that, in vitro, astrocytes only express GLUT1 and neurons<br />

preferentially express GLUT3. Functional data indicate that astrocytes transport mainly<br />

dehydroascorbic acid, using <strong>the</strong> glucose transporter GLUT1. Additionally, we defined that<br />

glutamate stimulates intracellular reduction <strong>of</strong> DHA in astrocytes <strong>of</strong> 15 and 30 DIV. In parallel,<br />

we observed that neurons have a lower reductive capacity, which increases only in <strong>the</strong><br />

presence <strong>of</strong> high concentrations <strong>of</strong> glutamate. In this study we determined that glutamate is<br />

directly involved in <strong>the</strong> recycling <strong>of</strong> vitamin C in <strong>the</strong> CNS. This recycling model may work as an<br />

efficient system for <strong>the</strong> salvage <strong>of</strong> vitamin C by avoiding <strong>the</strong> hydrolisis <strong>of</strong> DHA produced by<br />

antioxidative protection.<br />

Grants Support by FONDECYT 1100396.<br />

2306<br />

ER Calcium Depletion and ER Stress by Palmitate in Mouse Podocyte.<br />

S. Xu 1 , J-H. Kim 1 , R. Das 1 , X. Quan 1 , S-K. Cha 1,2 , K-S. Park 1,2 ; 1 Physiology, Yonsei University<br />

Wonju College <strong>of</strong> Medicine, Wonju, Korea, 2 Institute <strong>of</strong> Lifestyle Medicine<br />

Podocytes play a major role in glomerular filtration barrier, but also participate in pathogenic<br />

process <strong>of</strong> diabetic nephropathy. Diabetic patients have elevated plasma levels <strong>of</strong> saturated free<br />

fatty acid (FFA) that induces ER stress and apoptosis in different cell types. In this study, we<br />

investigated <strong>the</strong> cytotoxic mechanism <strong>of</strong> FFA in immortalized mouse podocyte. Incubation with<br />

palmitate, a saturated FFA, elicited apoptotic cell death in a dose dependent manner<br />

(50~500�M). Palmitate elevated cytosolic or mitochondrial reactive oxygen species (ROS)<br />

production measured by fluorescence imaging system, which was blocked by N-acetyl-cystein<br />

(NAC). However, a mono-unsaturated FFA oleate itself or combination with palmitate did not<br />

increase <strong>the</strong> ROS level in podocyte. ER stress proteins such as GRP78/Bip, spliced Xbp1 and<br />

CHOP were strongly upregulated by palmitate, but not by oleate or oleate with palmitate.<br />

Palmitate decreased <strong>the</strong> luminal Ca 2+ level in ER measured with ER Ca 2+ sensing protein<br />

D1ER. In <strong>the</strong> absence <strong>of</strong> extracellular Ca 2+ , cytosolic Ca 2+ increase induced by blocking ER<br />

Ca 2+ uptake with cyclopiazonic acid (CPA) was markedly reduced in palmitate-treated cells,<br />

which could be recovered by NAC pretreatment. These data suggest that palmitate as <strong>the</strong><br />

predominant circulating saturated FFA leads to ER stress and apoptosis in podocyte, which<br />

might be mediated by ROS-triggered ER Ca 2+ depletion.<br />

2307<br />

Involvement <strong>of</strong> yeast homolog MOH1 <strong>of</strong> human YPEL5 gene in DNA damage-induced<br />

apoptosis in <strong>the</strong> budding yeast Saccharomyces cerevisiae.<br />

J. Lee 1 , D. Jun 1 , Y. Kim 1 ; 1 School <strong>of</strong> Life Science and Biotechnology, Kyungpook National<br />

University, Daegu, Korea<br />

The homolog <strong>of</strong> Drosophila yippee protein is known to be highly conserved among eukaryotes<br />

from yeast to human. For five members <strong>of</strong> <strong>the</strong> human YPEL family, YPEL5 is evolutionarily


TUESDAY<br />

distinct from o<strong>the</strong>r four members, YPEL1 through YPEL4, especially. However, <strong>the</strong>ir functional<br />

roles are still poorly understood. To understand <strong>the</strong> role <strong>of</strong> yippee protein, we have employed<br />

<strong>the</strong> budding yeast (Saccharomyces cerevisiae) mutant possessing disrupted yeast homolog<br />

(MOH1; YBL049W) <strong>of</strong> yippee protein gene, and compared its physiological characteristics with<br />

those <strong>of</strong> <strong>the</strong> wild-type strain. The mutant for <strong>the</strong> MOH1 gene exhibited an enhanced resistance<br />

to UVC (254 nm) irradiation and genotoxic agents as compared to <strong>the</strong> wild-type. This<br />

phenomenon was abrogated when <strong>the</strong> disrupted MOH1 gene in <strong>the</strong> mutant was restored by<br />

transformation <strong>of</strong> <strong>the</strong> functional gene. The level <strong>of</strong> MOH1 mRNA in <strong>the</strong> wild-type was<br />

upregulated in response to UVC irradiation and genotoxic agents. And exposed<br />

phosphatidylserine was detected by reaction with annexin V-FITC and PI staining after UVC<br />

treated. Our findings provide evidence for functional conservation <strong>of</strong> YPEL5 protein between<br />

lower and higher eukaryotes, which can confer apoptotic cell death to DNA damage in S.<br />

cerevisieae.<br />

Key word: MOH1, YBL049W, Saccharomyces cerevisiae, complementation test, YPEL5, UVC<br />

irradiation, genotoxic agent<br />

Development and Morphogenesis II<br />

2308<br />

Two periods <strong>of</strong> testicular regression, not directly linked to apoptosis, are peculiar events<br />

<strong>of</strong> <strong>the</strong> annual reproductive cycle <strong>of</strong> Myotis nigricans (Chiroptera: Vespertilionidae).<br />

M. R. Beguelini 1 , C. C. Puga 2 , F. F. Martins 2 , S. R. Taboga 1 , E. Morielle-Versute 2 ; 1 <strong>Biology</strong>,<br />

UNESP - University <strong>of</strong> Estadual Paulista, São José do Rio Preto, Brazil, 2 Zoology and Botany,<br />

UNESP - University <strong>of</strong> Estadual Paulista, São José do Rio Preto, Brazil<br />

The reproductive data available for Myotis nigricans, an endemic Neotropical species <strong>of</strong><br />

vespertilionid bat, are few and controversial; and indicate that its reproductive cycle is<br />

geographically variable, presenting an active pattern throughout <strong>the</strong> year in some regions and a<br />

hibernating pattern in o<strong>the</strong>rs. Thus, this study aimed to evaluate <strong>the</strong> seasonal modifications in<br />

testicular morphology in a tropical environment. Twenty-eight mature specimens, collected in<br />

São Paulo State, Brazil (49W22'45" 20S49'11"), between September 2008 and August 2009,<br />

were submitted to morphometric and immunohistochemical (cell proliferation-PCNA; apoptosis-<br />

TUNEL; androgen receptor-AR) analysis. Morphological observations revealed that <strong>the</strong> testes<br />

present two periods <strong>of</strong> regression (TR) during <strong>the</strong> year: in late spring (November) and in early<br />

winter (June); and peaks <strong>of</strong> spermatogenic activity in early spring (September-October) and<br />

middle autumn (April). The TR in spring begins in October, with a sharply decrease in testicular<br />

epi<strong>the</strong>lium and in <strong>the</strong> amount <strong>of</strong> PCNA+ cells; however, <strong>the</strong> amount <strong>of</strong> TUNEL+ and AR+ cells<br />

were basal in this period, indicating that <strong>the</strong> regression did not occur by apoptosis. In November<br />

<strong>the</strong> testes were totally regressed but, <strong>the</strong>y already presented patterns <strong>of</strong> reactivation, as <strong>the</strong><br />

increase in expression <strong>of</strong> AR and PCNA. In December was noted <strong>the</strong> greatest expression <strong>of</strong><br />

PCNA and a great expression <strong>of</strong> AR, indicating an accelerated development, which can lead to<br />

errors that are eliminated by apoptosis, thus explaining <strong>the</strong> great amount <strong>of</strong> TUNEL+ cells. The<br />

recrudescence process extended from December to <strong>the</strong> production <strong>of</strong> <strong>the</strong> first spermatozoa only<br />

in March, with this period presenting a gradually decrease in <strong>the</strong> AR, PCNA and TUNEL<br />

expressions, culminating in <strong>the</strong> second period <strong>of</strong> TR in June. The TR in June was less<br />

accentuated than <strong>the</strong> first (November), presenting a more rapidly reactivation <strong>of</strong> <strong>the</strong><br />

spermatogenesis (July). This reactivation was induced by <strong>the</strong> greatest AR expression and<br />

culminated in <strong>the</strong> greatest occurrence <strong>of</strong> apoptotic cells. All data demonstrated <strong>the</strong> presence <strong>of</strong><br />

two periods <strong>of</strong> TR in <strong>the</strong> reproductive cycle <strong>of</strong> M. nigricans, a pattern never previously observed


TUESDAY<br />

in bats; and that <strong>the</strong> apoptosis was not linked to <strong>the</strong> TR, but with <strong>the</strong> elimination <strong>of</strong> errors<br />

caused by <strong>the</strong> rapidly reactivation <strong>of</strong> <strong>the</strong> spermatogenesis.<br />

2309<br />

Reactive oxygen species cause reduced spore development and spore fitness in <strong>the</strong><br />

budding yeast, Saccharomyces cerevisiae.<br />

M. M. Postema 1 , T. Stokes 1 , M. Scott 1 , S. W. Gorsich 1 ; 1 <strong>Biology</strong>, Central Michigan University,<br />

Mount Pleasant, MI<br />

The generation <strong>of</strong> spores in yeast is essential for sexual reproduction and survival <strong>of</strong> <strong>the</strong><br />

organism. When diploid yeast cells undergo meiotic division it is important that four optimally fit<br />

spores are produced. During this specialized division <strong>the</strong> four resulting spores not only have a<br />

haploid copy <strong>of</strong> nuclear chromosomes, but also a complete complement <strong>of</strong> organelles, such as<br />

mitochondria. Mitochondria undergo temporally regulated fusion and fission events to assure<br />

that mitochondria are represented equally in each <strong>of</strong> <strong>the</strong> resulting spores. When this<br />

mitochondrial network is not maintained, due to mutations in mitochondrial fission genes (eg.<br />

dnm1/dnm1), it has been shown that fewer spores survive and <strong>the</strong> ones that do survive are<br />

more likely to have reduced respiratory fitness. We hypo<strong>the</strong>sized that environmental factors<br />

could also influence spore development in wild-type diploid yeast. In <strong>the</strong> present study, we<br />

demonstrated that hydrogen peroxide phenocopied spore development phenotypes associated<br />

with mitochondrial fission mutants. Wild-type yeast exposed to hydrogen peroxide have<br />

mitochondrial morphology and distribution defects during spore development, reduced spore<br />

viability, and decreased respiratory competency just as seen in dnm1/dnm1 fission mutants.<br />

Next we demonstrated that reactive oxygen species (ROS) sensitive mutants have increased<br />

ROS accumulation during spore development. In addition spore development defects<br />

associated with <strong>the</strong>se mutants and wild-type cells exposed to hydrogen peroxide were reduced<br />

when treated with various antioxidant strategies. These findings suggest that <strong>the</strong> ability <strong>of</strong> yeast<br />

to produce optimal numbers <strong>of</strong> fit spores is heavily influenced by <strong>the</strong>ir ability to protect<br />

<strong>the</strong>mselves from exogenous or endogenous ROS.<br />

2310<br />

A Role <strong>of</strong> Xenopus Zygote Arrest 2 (Xzar2) in Translational Regulation <strong>of</strong> Key Cell Cycle<br />

mRNAs.<br />

J. M. Cook 1 , T. M. Yamamoto 1 , A. Charlesworth 1 ; 1 Department <strong>of</strong> Integrative <strong>Biology</strong>, University<br />

<strong>of</strong> Colorado Denver, Denver, CO<br />

Zygote arrest proteins, Zar1 and Zar2 (aka Zar1-like), have been implicated in <strong>the</strong> oocyte to<br />

embryo transition, zygotic genome activation, pre-implantation development and<br />

epidermalization. However, <strong>the</strong> mechanism <strong>of</strong> action <strong>of</strong> Zar proteins is unknown: transcriptional<br />

regulation, chromatin remodeling and RNA metabolism have all been suggested. Early Xenopus<br />

development is coordinated through <strong>the</strong> translational regulation <strong>of</strong> maternal mRNAs and is<br />

mediated by a variety <strong>of</strong> cis-elements in <strong>the</strong> 3’ untranslated region (3’UTR) <strong>of</strong> <strong>the</strong> mRNA. The<br />

recently described translational control sequence (TCS) is one such cis-element found in<br />

mRNAs, such as Wee1 and Pcm1. Here, we show that Xenopus Zar2 (Xzar2) binds <strong>the</strong> TCS <strong>of</strong><br />

maternal mRNAs and can regulate translation in a progesterone responsive manner.<br />

In an electrophoretic mobility shift assay (EMSA), <strong>the</strong> C-terminal portion <strong>of</strong> XZar2 (CXzar2)<br />

bound a fluorescent labeled wild-type Wee1 3’UTR probe, resulting in a shifted protein-RNA<br />

complex. This band was super-shifted with a Zar2 antibody, verifying <strong>the</strong> presence <strong>of</strong> Zar2<br />

protein in <strong>the</strong> band. Complex formation was inhibited by <strong>the</strong> addition <strong>of</strong> unlabeled Wee1 3’UTR<br />

or Pcm1 3’UTR probe, but was not inhibited if <strong>the</strong> TCSs were mutated, indicating <strong>the</strong> TCS is<br />

required for RNA binding. In a dual luciferase reporter (DLR) assay, <strong>the</strong> N-terminal domain <strong>of</strong>


TUESDAY<br />

Xzar2 fused to an MS2 te<strong>the</strong>ring protein (NXzar2-MS2) was injected into oocytes and allowed to<br />

incubate with or without <strong>the</strong> addition <strong>of</strong> progesterone. NXzar2-MS2 repressed translation <strong>of</strong> <strong>the</strong><br />

reporter mRNA in immature oocytes in a dose responsive manner. Fur<strong>the</strong>rmore, that repression<br />

was relieved during progesterone-induced meiotic maturation. Endogenous Zar2 levels were<br />

evaluated during maturation via Western blot, and it was found that levels drop dramatically<br />

during oocyte maturation.<br />

These data suggest that Xzar2 protein binds developmentally important mRNAs in a sequence<br />

specific manner and exerts translational control over those mRNAs differentially during meiotic<br />

maturation. We propose that one role <strong>of</strong> Xzar2 in early development may be to modulate <strong>the</strong><br />

syn<strong>the</strong>sis <strong>of</strong> maternal cell cycle regulatory proteins at <strong>the</strong> translational level in <strong>the</strong> maturing<br />

oocyte.<br />

2311<br />

Zygote Arrest Proteins Bind Maternal mRNAs.<br />

K. D. Silva 1 , T. Khat 1 , T. M. Yamamoto 1 , A. Charlesworth 1 ; 1 Department <strong>of</strong> Integrative <strong>Biology</strong>,<br />

University <strong>of</strong> Colorado Denver, Denver, CO<br />

Zygote arrest proteins, Zar1 and Zar2 (aka Zar1-like), are important in early development. Zar1<br />

has been implicated in <strong>the</strong> activation <strong>of</strong> <strong>the</strong> zygotic genome and progression past <strong>the</strong> two-cell<br />

stage. However, <strong>the</strong> mechanism <strong>of</strong> Zar protein action has yet to be characterized. The product<br />

<strong>of</strong> <strong>the</strong> maternal Wee 1 mRNA has also been identified as a regulator <strong>of</strong> early development.<br />

Xenopus laevis Wee 1 mRNA contains a newly characterized cis-element in its 3’untranslated<br />

region (UTR) called <strong>the</strong> Translation Control Sequence (TCS), which regulates mRNA translation<br />

during early development. The protein that binds to <strong>the</strong> TCS has yet to be identified. The<br />

objective <strong>of</strong> this study is to show that Zar proteins bind to Wee 1 mRNA.<br />

Zar1 and Zar2 proteins possess a highly conserved C-terminal region containing five pairs <strong>of</strong><br />

invariable cysteine residues, four <strong>of</strong> which are thought to make up a putative atypical<br />

FYVE/plant homeodomain (PHD) zinc finger motif. Our data shows that Zar proteins use <strong>the</strong><br />

conserved C-terminal region to bind <strong>the</strong> TCS <strong>of</strong> Wee1 maternal mRNA. To characterize binding<br />

to mRNA, Xenopus laevis Zar protein binding mutants, with point mutations at specified cysteine<br />

residues, were designed and used to perform electrophoretic motility shift assays (EMSA) with a<br />

Cy5 labeled Wee 1 3’ UTR probe. We show that cysteine to alanine mutation at any <strong>of</strong> <strong>the</strong><br />

invariable cysteines disrupted Zar binding to Wee 1 mRNA. To test if zinc was a requirement for<br />

binding interactions, Zar protein was purified and an EMSA was performed in a zinc free buffer.<br />

This EMSA showed that without zinc, Zar proteins did not bind to Wee 1 mRNA.<br />

This data suggests that Zygote arrest proteins contain a new type <strong>of</strong> zinc finger motif that<br />

constitutes an mRNA binding domain, providing support to <strong>the</strong> hypo<strong>the</strong>sis that Zar proteins play<br />

an important role in translational regulation <strong>of</strong> maternal mRNAs.<br />

2312<br />

Three-dimensional cell cultures <strong>of</strong> human uro<strong>the</strong>lium by using a collagen gel scaffold.<br />

C. Wasén 1 , M. Levin 2,3 , D. Giglio 1,3 ; 1 Pharmacology, Neuroscience and Physiology, Göteborg,<br />

Sweden, 2 Wallenberg Laboratory, Sahlgrenska Center for Cardiovascular and Metabolic<br />

Research, Göteborg, Sweden, 3 The Sahlgrenska University hospital, Jubileumskliniken,<br />

Göteborg, Sweden<br />

In vivo, uro<strong>the</strong>lial cells create a 3-6 cell thick epi<strong>the</strong>lial layer in <strong>the</strong> urinary bladder. In vitro,<br />

uro<strong>the</strong>lial cells are most <strong>of</strong>ten grown two-dimensionally (2D) forming a one-cell layer. Three<br />

dimensional cell culture (3D) is an alternative to classic 2D culture. In 3D, epi<strong>the</strong>lial cells form in<br />

vivo-like structures. In <strong>the</strong> present study, we investigated if uro<strong>the</strong>lial cells may be grown threedimensionally<br />

in a collagen gel scaffold. We used <strong>the</strong> UROtsa cell line, which is an immortalized


TUESDAY<br />

normal human bladder uro<strong>the</strong>lial cell line (Petzoldt et. al., 1995). UROtsa cells grown threedimensionally<br />

in a collagen I gel formed spherical multicellular formations after 7 days. The cells<br />

expressed occludin and laminin indicating presence <strong>of</strong> tight junctions and basal membrane,<br />

respectively. The present findings reveal a new and promising way to study uro<strong>the</strong>lial function in<br />

vitro by growing uro<strong>the</strong>lial cells three-dimensionally in a collagen gel scaffold.<br />

2313<br />

The 3Ds - development, differentiation and death, <strong>of</strong> yeast colony.<br />

L-C. Liou 1 , Q. Ren 1 , Z. Zhang 1 ; 1 Zoology and Physiology, University <strong>of</strong> Wyoming, Laramie, WY<br />

The budding yeast Saccharomyces cerevisiae has long been used as a model system to study<br />

all aspects <strong>of</strong> biology. Majority <strong>of</strong> <strong>the</strong> studies, however, are conducted in liquid culture. Yeast<br />

cells, when growing on solid medium, form organized colony structure. Cells inside <strong>the</strong> colony<br />

develop and differentiate into a mature structure under normal conditions. Colony cells may die<br />

when <strong>the</strong> colony encounters severe stress (internal, such as mutation or external, like drought).<br />

The objective <strong>of</strong> this study is to understand <strong>the</strong> mechanism <strong>of</strong> <strong>the</strong> development, differentiation<br />

and death <strong>of</strong> yeast colony. Using transmission electron microscopy (TEM), we showed that for a<br />

4 day-colony, <strong>the</strong> dividing cells are mostly located at <strong>the</strong> periphery and <strong>the</strong> bottom <strong>of</strong> <strong>the</strong> colony,<br />

where cells are in direct contact with <strong>the</strong> nutrient. Dividing cells are also observed at <strong>the</strong> top<br />

surface <strong>of</strong> <strong>the</strong> colony, where cells are far away from <strong>the</strong> nutrient. The non-dividing cells are<br />

mostly located at <strong>the</strong> middle portion <strong>of</strong> <strong>the</strong> colony. TEM analyses <strong>of</strong> mutants that are defective<br />

<strong>of</strong> colony growth and development, including chc1 and sro7/sro77 double deletion, revealed that<br />

cell death occurs mostly in <strong>the</strong> middle portion <strong>of</strong> <strong>the</strong> colony. Our results suggest <strong>the</strong> cells in <strong>the</strong><br />

middle portion <strong>of</strong> <strong>the</strong> colony play an important, supporting role for <strong>the</strong> yeast colony development<br />

and differentiation. They likely cease to divide, or even die, to support <strong>the</strong> proliferation <strong>of</strong> o<strong>the</strong>r<br />

cells inside <strong>the</strong> colony.<br />

2314<br />

An immunoglobulin domain-containing protein, IGP, is involved in trafficking <strong>of</strong> FGFR1.<br />

S. Kim 1 , J. Liu 1 , G. Yagnik 1 , A. Stoner 1 , C. Stevens 1 , E. Cherkez 1 , J. kim 1 , S. Boyadjiev 1 ;<br />

1 UCDMC MIND Inst, Sacramento, CA<br />

Fibroblast growth factor receptors (FGFRs) are critical for cell growth, differentiation, and animal<br />

development. Mutations in this receptor family are associated with crani<strong>of</strong>acial defects and<br />

cancer. In this study, we characterize an immunoglobulin domain-containing protein (IGP) that<br />

influences stability and function <strong>of</strong> FGFR1. When IGP and FGFR1 are coexpressed, <strong>the</strong>ir levels<br />

are increased compared to individual expression. In addition, IGP enhances conversion <strong>of</strong><br />

FGFR1 from ER forms to Golgi forms. Fur<strong>the</strong>rmore, IGP enhances phosphorylation <strong>of</strong> PLCgamma,<br />

but does not affect o<strong>the</strong>r FGFR1 signaling components such as AKT and ERK1/2.<br />

Interestingly, IGP predominantly exists as ER forms. These results indicate that IGP facilitates<br />

ER export <strong>of</strong> FGFR1 and activates <strong>the</strong> FGFR1-mediated PLC-gamma signaling pathway.<br />

Because aberrant activation <strong>of</strong> <strong>the</strong> FGFR signaling pathways is implicated in craniosynostosis<br />

(CS, premature fusion <strong>of</strong> cranial sutures), we have screened IGP for rare mutations from a<br />

cohort <strong>of</strong> nonsyndromic CS patients (431 individuals) who do not carry mutations in <strong>the</strong><br />

previously known CS-causing genes. We have identified three rare missense mutations from<br />

this screening. Strikingly, one <strong>of</strong> <strong>the</strong>se mutant IGPs fur<strong>the</strong>r enhances phosphorylation <strong>of</strong> PLCgamma.<br />

In summary, IGP plays a critical role in ER export/maturation/signaling <strong>of</strong> FGFR1.<br />

Fur<strong>the</strong>rmore, our results fur<strong>the</strong>r suggest that IGP mutations may be a risk factor for CS.


TUESDAY<br />

2315<br />

Sonic Hedge Hog Pathway Expression in Normal and Megabladder Mice During<br />

Development.<br />

K. R. DeSouza 1 , M. Saha 1 , A. R. Carpenter 1 , K. M. McHugh 1 ; 1 Center for <strong>Molecular</strong> & Human<br />

Genetics, The Research Institute at Nationwide Children's Hospital, Columbus, OH<br />

In this study, we present <strong>the</strong> first comprehensive assessment <strong>of</strong> <strong>the</strong> expression pattern <strong>of</strong> <strong>the</strong><br />

Sonic Hedge Hog (shh) signaling pathway in <strong>the</strong> bladders <strong>of</strong> normal mice at consecutive stages<br />

during development. Additionally, we compared <strong>the</strong> expression pattern <strong>of</strong> <strong>the</strong> shh signaling<br />

pathway in megabladder (mgb-/-) mice to wild type mice to determine if this critical signaling<br />

pathway is altered in <strong>the</strong> mutant bladders <strong>of</strong> <strong>the</strong>se animals.<br />

We characterized <strong>the</strong> expression pattern <strong>of</strong> <strong>the</strong> shh pathway’s ligand (Shh), receptor (Ptch), and<br />

transcription factor (Gli) and <strong>the</strong> key smooth muscle regulatory gene, Myocardin (Myocd), in <strong>the</strong><br />

bladders <strong>of</strong> male and female wild type and mgb-/- mutant mice at embryonic days 13.5, 14.5,<br />

15.5 and 16.5 (E13.5-E16.5), by in situ hybridization analysis.<br />

This study provides evidence that <strong>the</strong> shh pathway exhibits a canonical pattern <strong>of</strong> expression in<br />

wild type bladders, with each molecule displaying a specific spatial localization within <strong>the</strong><br />

bladder. Shh expresses exclusively in <strong>the</strong> uro<strong>the</strong>lium <strong>of</strong> <strong>the</strong> developing bladder. Ptch expresses<br />

in <strong>the</strong> proximal mesenchyme immediately adjacent to <strong>the</strong> uro<strong>the</strong>lium, while Gli expresses in <strong>the</strong><br />

distal mesenchyme, where smooth muscle cells eventually develop. Myocd, a key smooth<br />

muscle regulatory gene, expresses in <strong>the</strong> distal mesenchyme, where <strong>the</strong> presumptive smooth<br />

muscle develops in <strong>the</strong> bladder. Our study shows <strong>the</strong> shh pathway is functional in mgb-/- mutant<br />

bladders but shows a potential developmental delay with poorly restricted signal boundaries. In<br />

contrast, our results indicate that Myocd is minimally expressed in mgb-/- bladders when<br />

compared to controls.<br />

This study represents <strong>the</strong> first comprehensive analysis <strong>of</strong> <strong>the</strong> Shh signaling pathway during<br />

normal bladder development in mice. In addition, we show <strong>the</strong> shh pathway is functional in mgb-<br />

/- bladders but <strong>the</strong> key smooth muscle regulator, Myocd, is drastically reduced or absent. This<br />

finding suggests that <strong>the</strong> lack <strong>of</strong> detrusor smooth muscle development in mgb-/- bladders is not<br />

due absent short axis patterning but appears to result from a dramatic reduction in <strong>the</strong><br />

expression <strong>of</strong> Myocd.<br />

2316<br />

Imbalance on cell proliferation and basal cell frequency <strong>of</strong> senile gerbil prostate:<br />

exposure effects <strong>of</strong> ethinylestradiol during <strong>the</strong> puberty.<br />

A. P. Perez 1 , M. F. Biancardi 1 , B. F. Gonçalves 1 , L. R. Falleiros Junior 2 , F. C. Santos 3 , S. R.<br />

Taboga 2 ; 1 <strong>Biology</strong>, Unicamp, Campinas, Brazil, 2 <strong>Biology</strong>, IBILCE/UNESP, São Joé do Rio Preto,<br />

Brazil, 3 Morphology, Federal University <strong>of</strong> Goias, Goiânia, Brazil<br />

Endocrine disruptor chemicals (EDCs) are exogenous compounds with <strong>the</strong> potential to disturb<br />

hormonal regulation. Male and female rodent exposed to EDCs during critical time points <strong>of</strong><br />

development, such as prenatal and neonatal as well as puberty, affect <strong>the</strong> reproductive tract <strong>of</strong><br />

both sexes. Some studies show that <strong>the</strong> prostate is a highly sensitive gland to <strong>the</strong> EDCs action<br />

that might increase <strong>the</strong> risk <strong>of</strong> emergence <strong>of</strong> prostatic pathologies with aging. Thus, this study<br />

examined <strong>the</strong> ethinylestradiol effects during puberal development on histological differentiation,<br />

cell proliferation and p63-positive cells frequency <strong>of</strong> senile gerbil male ventral prostate (VP) and<br />

female prostate. For this, male and female gerbil (Meriones unguiculatus) with 42 days old<br />

received by gavage 15µg/kg/day <strong>of</strong> 17α–ethinylestradiol (EE- component oral contraceptive)<br />

diluted in mineral oil during one week (Pub group) and control group (males and females) were<br />

remained untreated. When males and females <strong>of</strong> control and Pub groups completed 12 months<br />

<strong>of</strong> age, <strong>the</strong>y were sacrificed and male VP and female prostate were retired, processed and


TUESDAY<br />

histological sections for each experimental group were subjected to immunohistochemistry for<br />

proliferating (PCNA) and basal (p63) cells. Puberty exposure to ethinylestradiol promoted<br />

different alterations in <strong>the</strong> senile gerbil male VP and female prostate. The Pub group male VP<br />

presented a significant increase in percentage cell proliferation (25.65±3.19) compared to<br />

control group (10.57±1.37) mainly in regions with invasive lesions. While this increase was not<br />

observed in <strong>the</strong> Pub group female prostate. Regarding <strong>the</strong> prostatic basal cells frequency, was<br />

observed a significant increment in female Pub group (23.24±1.18) compared to control group<br />

(12.82±0.91), indicating difference in proportion <strong>of</strong> main cell populations in acinar epi<strong>the</strong>lium.<br />

The Pub group male VP did not presented significant difference in this frequency. In conclusion<br />

<strong>the</strong> syn<strong>the</strong>tic estrogen exposure during puberty causes aberrant proliferation in prostatic cells <strong>of</strong><br />

senile male, while in female prostate was observed basal cells stratification, mainly in regions<br />

with epi<strong>the</strong>lial hyperplasia. Although prostatic epi<strong>the</strong>lium proliferation is regulated by androgen,<br />

<strong>the</strong> estrogen is also able to cause this event. Thus <strong>the</strong> syn<strong>the</strong>tic estrogen exposure during<br />

prostatic morphogenesis can favor <strong>the</strong> emergence <strong>of</strong> invasive lesions mainly in senile male<br />

prostate.<br />

2317<br />

F3 ENU genetic screen – a quest for endo<strong>the</strong>lial cells directional movement cues.<br />

S. Krispin 1 , J. Yu 2 , T. Mulligan 2 , A. Davis 2 , D. Castranova 2 , B. M. Weinstein 2 ; 1 Program in<br />

Genomics <strong>of</strong> Differentiation, National Institute <strong>of</strong> Child Health and Human Development, NIH<br />

Be<strong>the</strong>sda, MD, 2 National Institute <strong>of</strong> Child Health and Human Development, NIH Be<strong>the</strong>sda, MD<br />

Blood vessels are essential for tissue homeostasis within <strong>the</strong> complex bodies <strong>of</strong> vertebrates.<br />

Studies in developing vertebrates have uncovered many genes crucial for embryonic endo<strong>the</strong>lial<br />

specification and for blood vessel differentiation and growth, but we still know little about what<br />

guides <strong>the</strong> patterning <strong>of</strong> developing blood vessels and determines <strong>the</strong> anatomical architecture <strong>of</strong><br />

<strong>the</strong> vascular system.<br />

The metameric arrangement and relative anatomical simplicity <strong>of</strong> <strong>the</strong> zebrafish trunk primary<br />

angiogenic network makes it ideally suited to study <strong>the</strong> cues and mechanisms leading to <strong>the</strong><br />

formation <strong>of</strong> vascular network. Of particular interest are <strong>the</strong> intersegmental vessels (ISVs),<br />

which emerge from <strong>the</strong> dorsal aorta (DA) and migrate dorsally along somites boundaries<br />

through angiogenesis. The cues that control this dorsal migration are still not understood. We<br />

have been using genetic screens to identify novel genes involved in endo<strong>the</strong>lial cell (EC)<br />

directional movement.<br />

We used ENU (N-ethyl-N-nitrosourea) to induce random, single-base-pair mutations in <strong>the</strong><br />

zebrafish genome, followed by a large-scale F3 genetic screen to uncover <strong>the</strong> homozygous<br />

vascular phenotypes <strong>of</strong> induced mutations. We facilitated <strong>the</strong> screening process by visualizing<br />

alterations in <strong>the</strong> vasculature primary network formation using a stable germline transgenic line,<br />

fli1-EGFP, which expresses enhanced green fluorescent protein in endo<strong>the</strong>lial cells in vivo<br />

under <strong>the</strong> control <strong>of</strong> <strong>the</strong> fli1 promoter. We identified a new mutant, y210, in which trunk ISVs fail<br />

to migrate dorsally properly. We are in <strong>the</strong> process <strong>of</strong> mapping and positionally cloning <strong>the</strong><br />

affected gene from <strong>the</strong> mutant, and <strong>the</strong>n study its functional role in EC directional movements.<br />

Addressing this question will provide insight into one <strong>of</strong> <strong>the</strong> basic enigmas in<br />

developmental/vascular biology: what cues guide vessel positioning relative to o<strong>the</strong>r tissues and<br />

organs.


TUESDAY<br />

2318<br />

Novel antibodies to track neurogenesis in planarians.<br />

K. G. Ross 1 , M. Taylor 1 , R. K. Munday 1 , A. Hubert 1 , R. M. Zayas 1 ; 1 <strong>Biology</strong>, San Diego State<br />

University, San Diego, CA<br />

Neural stem cells proliferate and integrate into <strong>the</strong> adult central nervous system (CNS) <strong>of</strong><br />

diverse organisms, however <strong>the</strong>re are gaps in our knowledge <strong>of</strong> <strong>the</strong> precise molecular<br />

mechanisms controlling <strong>the</strong>se events. Planarian flatworms are amongst <strong>the</strong> simplest organisms<br />

to possess a CNS. Their nervous system is surprisingly complex at <strong>the</strong> molecular level,<br />

containing numerous diverse cell types, many <strong>of</strong> which demonstrate functional similarity with<br />

those found in vertebrates. Planarians maintain a population <strong>of</strong> adult pluripotent stem cells<br />

(neoblasts) that proliferate and differentiate in response to injury or during normal tissue<br />

homeostasis, enabling <strong>the</strong>m to regenerate all <strong>of</strong> <strong>the</strong>ir tissues, including a complete nervous<br />

system. These features make planarians excellent models for in vivo studies <strong>of</strong> stem cell based<br />

CNS regeneration. However, many neurons in planarians have not been fully characterized and<br />

little is known about <strong>the</strong> spatial distribution <strong>of</strong> differentiating cells and temporal sequence <strong>of</strong> cell<br />

replacement during <strong>the</strong>ir regeneration. To improve our understanding <strong>of</strong> cell turnover and<br />

regeneration in <strong>the</strong> planarian CNS, we have performed a monoclonal antibody screen for<br />

neuronal markers in <strong>the</strong> planarian Schmidtea mediterranea. We have produced nine<br />

monoclonal antibodies thus far, each <strong>of</strong> which is specific to different planarian tissues, including<br />

discrete neuronal populations. We have characterized <strong>the</strong> staining patterns <strong>of</strong> <strong>the</strong>se antibodies<br />

in intact and regenerating animals and are using <strong>the</strong>m in combination with bromodeoxyuridine<br />

pulse-chase experiments to track neural differentiation in regenerating planarians. Our work has<br />

yielded new markers to help visualize CNS architecture and quantify differentiation <strong>of</strong> neuronal<br />

subpopulations. These analyses will help us understand how <strong>the</strong>se animals maintain and<br />

reconstruct <strong>the</strong>ir nervous system and provide useful tools for phenotypic screening <strong>of</strong> gene<br />

knockdown experiments in planarians. This work was supported by CIRM Grant RN2-00940-1<br />

to RMZ.<br />

2319<br />

Imaging Epi<strong>the</strong>lial Morphogenesis in <strong>the</strong> Developing Hair Placode.<br />

L. Ahtiainen 1 , S. Lefebvre 1 , P. Lindfors 1 , I. Thesleff 1 , M. L. Mikkola 1 ; 1 Institute <strong>of</strong> Biotechnology,<br />

University <strong>of</strong> Helsinki, Finland<br />

Development <strong>of</strong> different ectodermal appendages (hair follicle, mammary gland, tooth) shares<br />

similarities at <strong>the</strong> morphological and molecular level. All epidermal appendages begin as a<br />

thickening <strong>of</strong> <strong>the</strong> epi<strong>the</strong>lium, a placode. The key signaling pathways regulating placode<br />

formation are ra<strong>the</strong>r well known, but <strong>the</strong> cellular mechanisms involved are still poorly<br />

understood. Two hypo<strong>the</strong>ses have been proposed: local cellular proliferation or cell migration.<br />

Only very recently light microscopy imaging techniques have advanced to enable tracking <strong>of</strong> <strong>the</strong><br />

fate <strong>of</strong> single cells in live tissue. This study aims at establishing and developing 3D and 4D live<br />

tissue imaging methodology for studying cellular processes regulating epi<strong>the</strong>lial morphogenesis,<br />

in particular <strong>the</strong> role <strong>of</strong> <strong>the</strong> Ectodysplasin (Eda)/Eda receptor(Edar)/NF-κB signaling pathway in<br />

skin appendage development and disease. We have established laser scanning confocal<br />

imaging methodology that allows live visualization <strong>of</strong> E13.5-E14.5 mouse embryonic skin whole<br />

mount explants for up to 24h during <strong>the</strong> first wave <strong>of</strong> hair placode formation. As a tool we use<br />

K17-GFP embryonic mouse skin, where GFP expression driven by <strong>the</strong> K17 promoter marks<br />

emerging placode cells. We provide evidence indicating that epi<strong>the</strong>lial cell migration is<br />

associated with hair placode formation and that inhibition <strong>of</strong> migration and actin remodeling<br />

suppresses placode formation. Imaging studies in <strong>the</strong> Fluorescent Cell Cycle Indicator (Fucci)


TUESDAY<br />

mouse suggest that proliferation has a less prominent role in placode formation. Accordingly,<br />

inhibition <strong>of</strong> proliferation had no gross effect on timing or morphology <strong>of</strong> forming placodes.<br />

2320<br />

Multiparametric image analysis <strong>of</strong> lung branching morphogenesis.<br />

C. Schnatwinkel 1 , L. Niswander 1 ; 1 Pediatrics, Universtiy <strong>of</strong> Colorado, Denver/HHMI, Aurora, CO<br />

Morphogenesis remains a fascinating problem in biology and proper morphogenesis is required<br />

for <strong>the</strong> shape and function <strong>of</strong> all organs and tissues. However, <strong>the</strong> morphogenetic mechanisms<br />

by which <strong>the</strong> genome moulds developing tissue masses into specific shapes remain poorly<br />

understood.<br />

To provide <strong>the</strong> first insights into <strong>the</strong> cellular dynamics <strong>of</strong> lung branching, we developed a life<br />

imaging set up to study cellular dynamics during branching morphogenesis. This approach<br />

helped in many ways to understand <strong>the</strong> interpretation <strong>of</strong> various phenotypes we have observed<br />

in mutant lungs. In this study, we performed a systematic time-lapse analysis <strong>of</strong> two <strong>of</strong> <strong>the</strong><br />

recently postulated branching subroutines domain branching and planar bifurcation <strong>of</strong> epi<strong>the</strong>lial<br />

cells in both lung explant- and organotypic three-dimensional (3D) culture. Our analysis<br />

revealed morphological and molecular distinct stages during branching morphogenesis that<br />

occurred in normal cultured lungs. We have classified a single bifurcation event into four<br />

morphological and molecularly distinct stages. Throughout <strong>the</strong> stages, we noticed a proximal to<br />

distal increase in proliferation, as well as a consecutive reduction in bud size before undergoing<br />

ano<strong>the</strong>r round <strong>of</strong> bifurcation. This phenomenon may be a potential reason why lungs in culture<br />

only have a limited branching capacity. The stages are <strong>the</strong> following: (1) The bud stage: <strong>the</strong> size<br />

<strong>of</strong> <strong>the</strong> initial epi<strong>the</strong>lial bud is increased due to proliferation, which appears to occur throughout<br />

<strong>the</strong> epi<strong>the</strong>lium. (2) The flattening stage: Initially <strong>the</strong> distal tip cells apply mechanical tension<br />

along <strong>the</strong> basal membrane, whereas <strong>the</strong> surrounding mesenchyme acts partially as a stabilizing<br />

scaffold. This step may be a checkpoint stage and is required to precisely position and<br />

determine <strong>the</strong> cleft region for <strong>the</strong> subsequent step. (3) The bifurcation stage: epi<strong>the</strong>lial cells on<br />

<strong>the</strong> two sides <strong>of</strong> <strong>the</strong> bud move slightly outward and <strong>the</strong>n towards <strong>the</strong> distal end <strong>of</strong> <strong>the</strong> lung<br />

explants. This movement is partially provided by proliferation along <strong>the</strong> epi<strong>the</strong>lial axis, which<br />

leads to <strong>the</strong> extension <strong>of</strong> <strong>the</strong> monolayered epi<strong>the</strong>lium. Cells in <strong>the</strong> cleft region seem not to<br />

respond to migrational cues and remain in place. (4) The outgrowing stage: The cells at <strong>the</strong> two<br />

sides <strong>of</strong> <strong>the</strong> bifurcating bud continue <strong>the</strong>ir directional growth. Previously lateral cells become tip<br />

cells and cleft cells stay behind and localize to what becomes <strong>the</strong> stalk.<br />

To address <strong>the</strong> role <strong>of</strong> distinct cellular behaviors, we applied pharmacological inhibitors. Lung<br />

explants treated with an inhibitor <strong>of</strong> cell proliferation and apoptosis, prevented bud growth but<br />

maintained <strong>the</strong> capability to branch. On <strong>the</strong> o<strong>the</strong>r hand, treatment <strong>of</strong> explants with inhibitors<br />

against MLCK to break force generating processes in <strong>the</strong> cell allowed <strong>the</strong> bud to continue<br />

growing but failed to bifurcate if <strong>the</strong> inhibitor was applied before <strong>the</strong> flattening stage. However,<br />

new formation <strong>of</strong> a bud from <strong>the</strong> stalk region (domain branch) was still present. In fact, blocking<br />

<strong>the</strong> ATPase activity <strong>of</strong> myosin by blebbistatin increased <strong>the</strong> number and location <strong>of</strong> domain<br />

branches. Among o<strong>the</strong>r findings, this suggests that proliferation is a key step in driving domain<br />

branch formation, whereas local changes in force generating mechanisms mold and divide <strong>the</strong><br />

distal bud into two.<br />

Our finding defined a new set <strong>of</strong> parameters that can be applied to future studies with various<br />

types <strong>of</strong> mutant lungs as well as indicated that force generating mechanisms play an important<br />

role in <strong>the</strong> progression through <strong>the</strong> stages <strong>of</strong> branching morphogenesis.


TUESDAY<br />

2321<br />

In vitro assessement <strong>of</strong> bioactivities <strong>of</strong> rmGDF-9 and rhBMP-15.<br />

L. Niu 1 , V. Hernandez 1 , J. Li 1 , V. Kalabokis 1 , G. Wu 1 , J. Humphrey 1 , R. Hao 1 ; 1 R&D Systems,<br />

Minneapolis, MN<br />

Growth differentiation factor -9 (GDF-9) and bone morphogenetic protein -15 (BMP-15) are<br />

oocyte-derived growth factors that are well known to play important roles in regulating<br />

folliculogenesis and female fertility. However, recent publications implicating <strong>the</strong>ir biological<br />

functions in human cancers raised <strong>the</strong> questions <strong>of</strong> whe<strong>the</strong>r GDF-9 and BMP-15 exert diverse<br />

biological functions in non-ovarian tissues. We have purified recombinant mouse GDF-9 and<br />

human BMP-15 from Chinese Hamster Ovary cells and evaluated <strong>the</strong>ir bioactivities using an<br />

array <strong>of</strong> bioassays. As expected, both mouse GDF-9 and human BMP-15 bind to <strong>the</strong><br />

recombinant human/mouse BMP-RII/Fc extracellular domain with Kd values <strong>of</strong> 1.4 nM and 2 nM<br />

respectively, in a functional ELISA. In addition, <strong>the</strong> two proteins activate Smad 2/3 in a mouse<br />

embryonic teratocarcinoma P19 cells. Fur<strong>the</strong>rmore, we discovered that <strong>the</strong> recombinant mouse<br />

GDF-9, similar to o<strong>the</strong>r members <strong>of</strong> <strong>the</strong> transforming growth factor -βfamily, is able to induce<br />

apoptosis <strong>of</strong> mink lung epi<strong>the</strong>lia MV1Lu and prostate carcinoma DU145 cells. In contrast, <strong>the</strong><br />

recombinant human BMP-15, like o<strong>the</strong>r BMP family members, is osteogenic, promoting<br />

differentiation <strong>of</strong> MC3T3-E1 cells to osteoblasts. We also found that human BMP-15 is very<br />

potent in supporting <strong>the</strong> survival and proliferation <strong>of</strong> NIH3T3 cells in nutrient deprived conditions.<br />

These in vitro differential functions <strong>of</strong> GDF-9 and BMP-15 provide <strong>the</strong> stepping stones for new<br />

research initiatives and also open a whole new avenue to explore <strong>the</strong>ir functions in cancer<br />

biology, in addition to <strong>the</strong> area <strong>of</strong> reproductive physiology. These studies in turn may eventually<br />

lead to an expanded role in <strong>the</strong>ir <strong>the</strong>rapeutic potential.<br />

2322<br />

Reg1 and Reg3β expression in <strong>the</strong> pancreas <strong>of</strong> adult diabetic Goto-Kakizaki (GK) rats.<br />

J-C. Irminger 1 , M-H. Giroix 2 , S. Calderari 3 , J. Ehses 4 , J. Coulaud 2 , K. Rickenbach 1 , P.<br />

Serradas 3 , F. Homo-Delarche 2 ; 1 University <strong>of</strong> Geneva, Geneva, Switzerland, 2 University <strong>of</strong><br />

Paris-Diderot, Paris, 3 University Pierre et Marie Curie, Paris, 4 University <strong>of</strong> British Columbia,<br />

Vancouver, BC, Canada<br />

Reg protein expression is associated with islet development, β-cell damage, diabetes and<br />

pancreatitis. We previously reported that islets <strong>of</strong> 4-month-old (4-mo) GK rats, a spontaneous<br />

model <strong>of</strong> type 2 diabetes, overexpress Reg1, 3α, 3β and 3γ vs age-matched Wistar control<br />

islets. Reg1 and 3β are involved in cell growth/survival control and inflammation, respectively.<br />

Diabetic GK islets also exhibit progressive inflammation reaction, consisting <strong>of</strong> high CCL2<br />

(MCP-1), CCL3 (MIP-1α), CXCL-1 (murine IL-8 analog) and IL-6 expression/release and mostly<br />

peri-islet macrophage infiltration. Importantly, Reg1 gene promoters contain IL-6-responsive<br />

elements. Here we analyzed in greater detail <strong>the</strong> pancreatic expression <strong>of</strong> Reg1 and Reg3β in<br />

diabetic GK rats.<br />

Isolated pancreatic islets and acinar tissue from male Wistar and GK rats were used for<br />

quantitative RT-PCR analysis. Islet IL-6, CCL2, CCL3 and CXCL1 release was measured by<br />

LuminexTM technology after a 48h islet culture on collagen. Pancreatic Reg<br />

immunohistochemistry (IHC) was performed on paraffin sections with a rabbit anti-human<br />

polyclonal antibody (Ab) and a mouse anti-rat monoclonal anti-Reg1 Ab. Macrophage infiltration<br />

was detected on cryostat sections using CD68 and MHC class II antibodies. Islet macrophage+<br />

area was quantified and expressed as % <strong>of</strong> corresponding islet area. Statistical analyses used<br />

<strong>the</strong> Student’s t-test for unpaired data.<br />

The exocrine/endocrine ratio <strong>of</strong> Reg mRNA expression in 4-mo normoglycemic Wistar and<br />

diabetic GK rats was: 1) Reg1: 41.3±2.4 and 5.0±1.5, respectively (n=3 different


TUESDAY<br />

isolations/group, p


TUESDAY<br />

formation we are using <strong>the</strong> zebrafish gut as model system. The zebrafish gut tube forms from a<br />

solid rod <strong>of</strong> endodermal cells that differentiate and start to polarize and assemble tight junctions<br />

which give rise to multiple small lumens. These small lumens subsequently enlarge though <strong>the</strong><br />

accumulation <strong>of</strong> fluid and eventually coalesce to form a single lumen. However, o<strong>the</strong>r cellular<br />

processes such as <strong>the</strong> remodeling <strong>of</strong> cell contacts must also be involved to facilitate<br />

coalescence into a single lumen. We are interested in <strong>the</strong>se additional cellular processes that<br />

regulate <strong>the</strong> transition into a single lumen. Analysis <strong>of</strong> lumen formation in wild-type (WT)<br />

embryos revealed that <strong>the</strong> coalescence <strong>of</strong> multiple small lumens into one involves <strong>the</strong> formation<br />

<strong>of</strong> a double lumen intermediate. Interestingly, we found that smoo<strong>the</strong>ned (smo) mutants <strong>of</strong>ten<br />

fail to properly form a single lumen and instead display a double lumen phenotype. In smo<br />

mutants, cells are frequently found in between <strong>the</strong> two lumens and display mislocalization <strong>of</strong><br />

adhesion proteins which suggests a defect in <strong>the</strong> remodeling <strong>of</strong> adhesive contacts. We analyzed<br />

<strong>the</strong> transcriptome <strong>of</strong> smo mutants and found that several genes associated with intracellular<br />

trafficking were down-regulated. In light <strong>of</strong> this finding, we generated transgenic zebrafish lines<br />

to determine <strong>the</strong> function <strong>of</strong> trafficking and recycling in <strong>the</strong> process <strong>of</strong> single lumen formation.<br />

Our analyses suggest that that intracellular membrane recycling is required for single lumen<br />

formation. Toge<strong>the</strong>r, our data suggest that smo regulates a transitional step in single lumen<br />

formation that facilitates intercellular contact remodeling through recycling.<br />

Cancer Cell <strong>Biology</strong> II<br />

2325<br />

Rapamycin treatment increases mTORC2 activity and selectively activates <strong>the</strong> AKT1<br />

is<strong>of</strong>orm in epidermis by inhibition <strong>of</strong> <strong>the</strong> mTORC1-dependent feedback loop to IRS-1.<br />

K. Sully 1 , O. A. Akinduro 1 , M. P. Philpott 1 , C. A. Harwood 1 , R. F. O'Shaughnessy 2 , C. Byrne 1 ;<br />

1 Cutaneous Research, Queen Mary University <strong>of</strong> London, London, United Kingdom,<br />

2 Immunobiology, UCL Institute <strong>of</strong> Child Health, London, United Kingdom<br />

Epidermal squamous cell carcinoma (SCC) is dramatically increased in patients undergoing<br />

immunosuppression following organ transplantation. The mammalian target <strong>of</strong> rapamycin<br />

complex 1 (mTORC1) inhibitor, rapamycin, when used as a post-transplantation<br />

immunosuppressive, significantly reduces SCCs, though <strong>the</strong> mechanism is not understood.<br />

Epidermal SCC is associated with changes in AKT signaling. AKT2, associated with less<br />

differentiated keratinocytes, is upregulated in SCC and in response to ultraviolet (UV) radiation,<br />

suggesting a tumour-promoting role. In contrast, AKT1, associated with differentiated<br />

keratinocytes, is downregulated in SCC and after UV radiation, suggesting a tumour-suppressor<br />

role. In some tumour types rapamycin activates AKT signaling via inhibition <strong>of</strong> mTORC1dependent<br />

negative feedback loops acting on <strong>the</strong> insulin receptor substrate-1 (IRS-1). It is<br />

unclear whe<strong>the</strong>r this feedback loop is active in epidermis. Upregulation <strong>of</strong> epidermal AKT<br />

signaling by rapamycin would have different effects on tumourigenesis depending on which AKT<br />

is<strong>of</strong>orm is upregulated. The objective was to establish whe<strong>the</strong>r <strong>the</strong> mTORC1-dependent<br />

negative feedback loop to IRS-1 is active in epidermis and find which epidermal AKT is<strong>of</strong>orm is<br />

affected.<br />

We show that rapamycin increases keratinocyte AKT phosphorylation at both S473 and T308,<br />

<strong>the</strong> two sites required for full activation, with a single treatment <strong>of</strong> rapamycin maintaining<br />

increased AKT phosphorylation for over 24 hours. Both acute and prolonged rapamycin<br />

treatment also increases epidermal mTORC2 activity, shown by upregulation <strong>of</strong> downstream<br />

targets. We demonstrate that <strong>the</strong> mTORC1-dependent negative feedback loop to IRS-1 is active<br />

in keratinocytes and responsible for rapamycin-induced mTORC2 activity and AKT<br />

phosphorylation. Rapamycin inhibits mTORC1-dependent phosphorylation <strong>of</strong> IRS-1 at


TUESDAY<br />

Ser636/639 and increases IRS-1 tyrosine phosphorylation, protein stability and downstream<br />

signaling to AKT. Epidermal AKT1 is<strong>of</strong>orm, but not AKT2, is selectively upregulated via<br />

rapamycin’s effects on epidermal mTORC2 and inhibition <strong>of</strong> <strong>the</strong> mTORC1-dependent feedback<br />

loop to IRS-1.<br />

These findings suggest a mechanism for rapamycin’s anti-tumour activity in skin. We propose<br />

that rapamycin’s selective activation <strong>of</strong> epidermal AKT1 promotes AKT1’s tumour-suppressing<br />

role in epidermis and protects against epidermal tumourigenesis.<br />

2326<br />

Gab2 regulates <strong>the</strong> migratory behaviors and E-cadherin expression via activation <strong>of</strong> <strong>the</strong><br />

PI3K pathway in ovarian cancer cells.<br />

Y. Wang 1 , Q. Sheng 2 , M. A. Spillman 3 , K. Behbakht 3 , H. Gu 1 ; 1 Department <strong>of</strong> Pathology,<br />

University <strong>of</strong> Colorado Denver Anschutz Med Campus, Aurora, CO, 2 Department <strong>of</strong> Cancer<br />

<strong>Biology</strong>, Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA, 3 Department<br />

<strong>of</strong> Obstetrics and Gynecology, University <strong>of</strong> Colorado Denver Anschutz Med Campus, Aurora,<br />

CO<br />

Ovarian cancer, <strong>the</strong> most deadly gynecologic malignancy, is <strong>of</strong>ten diagnosed late and at <strong>the</strong><br />

advanced stage when <strong>the</strong> cancer cells have already migrated and invaded into o<strong>the</strong>r tissues<br />

and organs. Better understanding <strong>of</strong> <strong>the</strong> mechanism <strong>of</strong> metastasis in ovarian cancer cells is<br />

essential to <strong>the</strong> design <strong>of</strong> effective <strong>the</strong>rapy. In this study, we investigated <strong>the</strong> function <strong>of</strong><br />

scaffolding adaptor protein Gab2 in ovarian cancer cells. Gab2 is found to be overexpressed in<br />

a subset <strong>of</strong> ovarian tumors and cancer cell lines. Gab2 expression mainly regulates <strong>the</strong><br />

migratory behaviors <strong>of</strong> ovarian cancer cells. Overexpression <strong>of</strong> Gab2 promotes <strong>the</strong> migration<br />

and invasion, and down-regulates E-cadherin expression in ovarian cancer cells with low-Gab2<br />

expression. Conversely, knockdown <strong>of</strong> Gab2 expression inhibits <strong>the</strong> migration and invasion, and<br />

promotes E-cadherin expression in ovarian cancer cells with high-Gab2 expression. By<br />

expressing Gab2 wild type and Gab2 mutants that are defective in activation <strong>the</strong> PI3K and<br />

Shp2-Erk pathways, we find that Gab2 inhibits E-cadherin expression and enhances <strong>the</strong><br />

expression <strong>of</strong> Zeb1, a transcription factor involved in epi<strong>the</strong>lial-to-mesenchymal transition<br />

(EMT), and cell migration and invasion through <strong>the</strong> activation <strong>of</strong> <strong>the</strong> PI3K pathway. Knockdown<br />

<strong>of</strong> Zeb1 expression blocks Gab2-induced suppression <strong>of</strong> E-cadherin expression and increase in<br />

cell invasion. LY294002 and GDC-0941, inhibitors <strong>of</strong> PI3K, or Rapamycin, an inhibitor <strong>of</strong> PI3K<br />

downstream target mTOR, can reverse <strong>the</strong> effects <strong>of</strong> Gab2 on migration and invasion. Overall,<br />

our studies reveal that Gab2 overexpression, via activation <strong>of</strong> <strong>the</strong> PI3K-Zeb1 pathway, promotes<br />

characteristics <strong>of</strong> EMT in ovarian cancer cells. Ovarian cancer cells with high Gab2 expression<br />

are potentially more metastatic in vivo and drugs that target <strong>the</strong> PI3K pathway may be used to<br />

treat a subset <strong>of</strong> ovarian cancer with high Gab2 expression.<br />

2327<br />

LRIG2 promotes PDGF induced experimental glioma.<br />

C. Holmlund 1 , T. Karlsson 1 , M. Faraz 1 , O. Casar Borota 2,3 , J. Miller 4 , B. Wang 1 , C. Sweeney 4 , R.<br />

Henriksson 1 , H. Hedman 1 ; 1 Radiation Sciences, Oncology, Umeå, Sweden, 2 Medical<br />

Biosciences, Pathology, Umeå, Sweden, 3 Laboratory Medicine, Pathology/Cytology, Umeå,<br />

Sweden, 4 University <strong>of</strong> California, UC Davis Medical Center, Sacramento, CA<br />

Leucine-rich and immunoglobulin-like domains protein 2 (LRIG2) is a transmembrane protein,<br />

whose expression is associated with poor survival <strong>of</strong> oligodendroglioma patients. This is in<br />

contrast with findings for <strong>the</strong> homologous protein LRIG1, a negative regulator <strong>of</strong> growth factor<br />

signaling, whose expression is associated with good survival in several different malignancies.<br />

Here, we address <strong>the</strong> role <strong>of</strong> LRIG2 in an experimental glioma model and its possible regulation


TUESDAY<br />

<strong>of</strong> platelet-derived growth factor (PDGF) receptor (PDGFR). Gliomas were induced in Ntv-a<br />

mice by intracranial injection <strong>of</strong> PDGFB-expressing RCAS viruses. All injected Lrig2-wild-type<br />

mice developed oligodendroglioma-like brain tumors <strong>of</strong> grade II/III (82%) or glioblastoma-like<br />

tumors <strong>of</strong> grade IV (18%), whereas Lrig2-deficient mice only developed grade II/III tumors (77%)<br />

or no detectable tumors (23%). In transfected cells, LRIG2 and PDGFRα co-localized in<br />

intracellular vesicle-like structures. Intriguingly, LRIG2, in contrast to LRIG1, did not negatively<br />

regulate <strong>the</strong> protein levels <strong>of</strong> PDGFRα or PDGFRβ. Instead, LRIG2 up-regulated <strong>the</strong> protein<br />

levels <strong>of</strong> PDGFRα. In summary, we show that LRIG2 promoted <strong>the</strong> genesis and/or progression<br />

<strong>of</strong> PDGF-induced oligodendroglioma and positively regulated PDGFRα protein levels. This<br />

fur<strong>the</strong>r supports <strong>the</strong> notion that LRIG proteins play important roles in cancer biology and<br />

suggests that LRIG1 and LRIG2 may have opposing functions in <strong>the</strong> regulation <strong>of</strong> PDGF<br />

signaling.<br />

2328<br />

The Study <strong>of</strong> Androgen Effects on LPA-induced VEGF-C Expression in Prostate Cancer<br />

Cell Lines.<br />

Y-C. Lin 1 , H. Lee 2 ; 1 Institute <strong>of</strong> Zoology, National Taiwan University, Taipei, Taiwan,<br />

2 Department <strong>of</strong> Life Science, National Taiwan University, Taipei, Taiwan<br />

Lysophosphatidic acid (LPA) is a bioactive lysophospholipid that binds to a family <strong>of</strong> G proteincoupled<br />

receptors (GPCRs), termed LPA1~LPA5. It has been reported that vascular endo<strong>the</strong>lial<br />

growth factor-C (VEGF-C) was related to <strong>the</strong> lymph node metastasis in human prostate<br />

cancer. Our previous study has indicated that LPA induces <strong>the</strong> expression <strong>of</strong> VEGF-C in human<br />

prostate cancer PC-3 cell line. Moreover, a well-known effector, androgen, modulating prostate<br />

cancer cell progression was also shown to negatively regulate VEGF-C expression in prostate<br />

cancer. Therefore, we attempt to investigate whe<strong>the</strong>r LPA-induced VEGF-C expression is<br />

affected by androgen in different prostate cancer cells. By real-time PCR analysis, we proved<br />

that LPA could trigger VEGF-C expression in dosage and time-dependent manners in different<br />

prostate cancer cell lines. In addition, our results fur<strong>the</strong>r showed that androgen agonist<br />

decreased LPA-induced VEGF-C expression. On <strong>the</strong> contrary, androgen antagonist enhanced<br />

LPA-induced VEGF-C expression. In summary, our results demonstrated that androgen was<br />

involved in LPA-induced VEGF-C expression in human prostate cancer cells.<br />

2329<br />

Mitochondrial Genome Instability and ROS Enhance Intestinal Tumorigenesis in<br />

APCMin/+ mice.<br />

D. Woo 1 , P. Green 2 , J. Santos 2 , A. D’Souza 3 , Z. Wal<strong>the</strong>r 3 , D. Martin 4 , B. Christian 3 , N. Chandel 5 ,<br />

G. Shadel 3 ; 1 Gyeongsang National University, Jinju, Korea, 2 University <strong>of</strong> Medicine and<br />

Dentistry, New Jersey-New Jersey Medical School, Newark, NJ, 3 Yale University School <strong>of</strong><br />

Medicine, New Haven, CT, 4 Emory University School <strong>of</strong> Medicine, Atlanta, GA, 5 Northwestern<br />

Medical School, Chicago, IL<br />

Alterations in mitochondrial oxidative phosphorylation have long been documented in tumors.<br />

O<strong>the</strong>r types <strong>of</strong> mitochondrial dysfunction, including altered reactive oxygen species (ROS)<br />

production and apoptosis, can also contribute to tumorigenesis and cancer phenotypes.<br />

Fur<strong>the</strong>rmore, mitochondria contain mtDNA, mutation and altered amounts <strong>of</strong> which have been<br />

observed in cancer cells. However, how mtDNA instability per se contributes to cancer remains<br />

largely undetermined. Tfam is a mitochondrial transcription factor and mtDNA-packaging protein<br />

required for expression and maintenance <strong>of</strong> mtDNA. Tfam heterozygous knock-out (Tfam +/- )<br />

mice exhibit mild mtDNA depletion, but have no overt phenotypes. Here, we show that Tfam +/-<br />

mouse cells and tissues not only have less mtDNA, but also increased oxidative mtDNA


TUESDAY<br />

damage. Crossing Tfam +/- mice to <strong>the</strong> APC Min/+ mouse cancer model revealed that mtDNA<br />

instability increases tumor number and growth in <strong>the</strong> small intestine. This was not due to<br />

enhancement <strong>of</strong> Wnt/ -catenin signaling, but ra<strong>the</strong>r appears to involve a propensity for<br />

increased mitochondrial ROS production. Direct involvement <strong>of</strong> mitochondrial ROS in intestinal<br />

tumorigenesis was demonstrated by crossing APC Min/+ mice to those that have catalase targeted<br />

to mitochondria (MCAT), which resulted in a significant reduction in tumorigenesis in <strong>the</strong> colon.<br />

Altoge<strong>the</strong>r, <strong>the</strong>se results show that mitochondrial genome instability and ROS can enhance<br />

tumorigenesis, and elucidate Tfam +/- mice as relevant model to address <strong>the</strong> role <strong>of</strong> mtDNA<br />

instability in disease states where mitochondrial dysfunction is implicated, such as cancer,<br />

neurodegeneration and aging.<br />

2330<br />

Upregulation <strong>of</strong> Plk1 in castration-resistant prostate cancer cells.<br />

A. Deeraksa 1 , J. Pan 1 , Y. Sha 1 , X. Liu 1 , A. C. Ferrari 2 , N. T. Eissa 1 , S-H. Lin 3 , L-Y. Yu-Lee 1 ;<br />

1 Medicine, Baylor College <strong>of</strong> Medicine, Houston, TX, 2 Medicine, New York University School <strong>of</strong><br />

Medicine, New York, NY, 3 <strong>Molecular</strong> Pathology, The University <strong>of</strong> Texas M. D. Anderson<br />

Cancer Center, Houston, TX<br />

Castration-resistant progression <strong>of</strong> prostate cancer (PCa) is a challenge in PCa treatment. A<br />

recent study found that PCa cells cultured in charcoal-stripped media (androgen-independent AI<br />

condition) have undergone a genetic reprogramming that selectively upregulated <strong>the</strong> expression<br />

<strong>of</strong> M-phase cell cycle genes. We found that <strong>the</strong> mitotic kinase Plk1 is upregulated during <strong>the</strong><br />

reprogramming process in AI PCa cells. We employed a pair <strong>of</strong> PCa cells, LNCaP (androgendependent<br />

PCa cells derived from lymph node metastasis) and LNCaP-AI cells (AI PCa cells<br />

derived from LNCaP), to determine <strong>the</strong> role <strong>of</strong> Plk1 in castration-resistant progression <strong>of</strong> PCa.<br />

We found that LNCaP-AI cells express 3 – 5 fold higher levels <strong>of</strong> Plk1 relative to LNCaP cells.<br />

Cell cycle proteins along <strong>the</strong> Plk1-regulated mitotic pathway, including Cdc25C (mitotic entry),<br />

Sgo2 (chromosome cohesion), PICH (chromosome architecture), BubR1 (spindle checkpoint),<br />

and NudC (cytokinesis) are all elevated in LNCaP-AI cells. Both LNCaP and LNCaP-AI cells<br />

were highly sensitive to a 5-day treatment with a small molecule inhibitor <strong>of</strong> Plk1, BI2536, with<br />

IC50 <strong>of</strong> 0.6 nM and 0.2 nM, respectively, compared to many o<strong>the</strong>r cell types with IC50 between<br />

10 – 50 nM. Under charcoal-stripped culture conditions, LNCaP did not grow or respond to<br />

BI2536 treatment. In contrast, LNCaP-AI cells remained highly sensitive to BI2536 inhibition,<br />

suggesting that <strong>the</strong> upregulated Plk1 pathway is inhibited by BI2536 in LNCaP-AI cells.<br />

We fur<strong>the</strong>r examined whe<strong>the</strong>r growth inhibition by BI2536 leads to cell death. In full medium<br />

culture conditions, LNCaP cells responded to BI2536 with caspase 3 cleavage and membrane<br />

blebbing, characteristics <strong>of</strong> apoptosis. In contrast, LNCaP-AI cells did not show caspase 3<br />

cleavage but exhibited PARP-1 cleavage, a common end point for cell death.<br />

Immun<strong>of</strong>luorescence microscopy showed unusual nuclear morphologies in BI2536-treated cells.<br />

LNCaP cells contained a single enlarged/lobular nucleus, a phenomenon previously observed<br />

with Plk1 inhibition in o<strong>the</strong>r cell types. In contrast, LNCaP-AI cells formed giant cells that<br />

contained up to 30 small and large independent nuclei within a single cell unit, likely due to<br />

nuclear fragmentation and cytokinesis failure. Live cell imaging indicate that <strong>the</strong>se giant cells<br />

underwent membrane disintegration and cell death characteristic <strong>of</strong> necroptosis. These results<br />

suggest that LNCaP-AI cells utilize a caspase 3-independent cell death modality in response to<br />

BI2536 inhibition. These observations suggest that Plk1 has a novel role in cell growth/death<br />

regulation in LNCaP-AI cells and Plk1 may be targeted for <strong>the</strong>rapy <strong>of</strong> castration-resistant PCa.


2331<br />

HPV E6 oncoproteins from high and low risk types associate with <strong>the</strong> proteasome<br />

independently <strong>of</strong> E6AP.<br />

L. M. Banks 1 , V. Tomaic 1 ; 1 ICGEB, Trieste, Italy<br />

TUESDAY<br />

Human papillomavirus (HPV) E6 oncoproteins associate with components <strong>of</strong> <strong>the</strong> ubiquitin<br />

proteasome pathway, including <strong>the</strong> cellular ubiqutin ligases E6AP, EDD and HERC2. E6AP was<br />

shown to play a major role in E6’s degradatory activities, while more recent studies suggest that<br />

its presence is also critical for E6 protein stability. In addition, it has been suggested that E6’s<br />

association with EDD could have a pr<strong>of</strong>ound effect upon <strong>the</strong> ability <strong>of</strong> E6 to target various<br />

substrates for proteolytic degradation and <strong>the</strong>reby directly influence <strong>the</strong> development <strong>of</strong> HPVinduced<br />

malignancy. To fur<strong>the</strong>r investigate E6 function, we performed a mass spectrometric<br />

analysis on HPV-18 E6 and E6AP protein complexes. Besides identifying several known binding<br />

partners <strong>of</strong> <strong>the</strong>se two proteins, we also identified several proteasome regulatory subunits as<br />

being associated with E6 and E6AP. We also show that both HPV-18 E6 and E6AP,<br />

independently <strong>of</strong> each o<strong>the</strong>r, interact with proteasome regulatory subunits in vitro. HPV-18 E6<br />

preferentially interacts with S2 and S4 subunits, while E6AP interacts with S2, S4, S5a, S7 and<br />

S8 subunits, an interaction that is independent <strong>of</strong> E6AP’s catalytic activity. Fur<strong>the</strong>rmore, HPV-16<br />

E6 and HPV-11 E6 also preferentially interact with <strong>the</strong> S2 and S4 subunits, although <strong>the</strong> HPV-<br />

16 E6 interaction is significantly stronger than that with HPV-11 E6. In presence <strong>of</strong> endogenous<br />

E6AP HPV-18 E6 also binds strongly to <strong>the</strong> subunit S5a, while binding to S2 and S4 subunits is<br />

reduced. These results demonstrate that E6AP can recruit E6 to <strong>the</strong> S5a subunit. Ongoing<br />

studies will now aim to elucidate <strong>the</strong>se aspects fur<strong>the</strong>r and clarify how E6AP mediates E6s’<br />

association with <strong>the</strong> proteasome.<br />

2332<br />

Cyclin B1 and B2 have oncogenic properties but drive tumorigenesis through distinct<br />

mechanisms.<br />

H-J. Nam 1 , J. van Deursen 1,2 ; 1 Pediaric and Adolescent Medicine, Mayo Clinic, Rochester,<br />

Rochester, MN, 2 Biochemistry and <strong>Molecular</strong> <strong>Biology</strong>, Mayo Clinic, Rochester, Rochester, MN<br />

High cyclin B1 and B2 levels are observed in a large proportion <strong>of</strong> human tumors and correlate<br />

with poor clinical outcome. However, <strong>the</strong> molecular and cellular defects caused by increased<br />

expression <strong>of</strong> <strong>the</strong>se mitotic cyclins and <strong>the</strong>ir roles in tumorigenesis are unknown. To address<br />

<strong>the</strong>se questions, we generated transgenic mice that overexpress cyclin B1 or B2. These mice<br />

show high aneuploidy rates, with cyclin B1 overexpression inducing chromatin bridges and<br />

cyclin B2 overexpression lagging chromosomes. Fur<strong>the</strong>rmore, cyclin B1 overexpression leads<br />

to persistent inhibition <strong>of</strong> separase, while cyclin B2 overexpression does not. Conversely, cyclin<br />

B2 overexpression induces supernumerary centrosomes, whereas cyclin B1 does not. These<br />

results demonstrate that both cyclin B1 and B2 induce chromosome missegregation, although<br />

through distinct mechanisms. Fur<strong>the</strong>rmore, preliminary results indicate that both cyclin B1 and<br />

B2 transgenic mice are prone to spontaneous tumors, although <strong>the</strong> tumor spectra <strong>of</strong> <strong>the</strong><br />

transgenes are remarkably different. In addition, it seems that cyclin B2 overexpression, but not<br />

cyclin B1 overexpression, predisposes mice to DMBA-induced cancers. Taken toge<strong>the</strong>r, <strong>the</strong><br />

data suggest that both cyclin B1 and cyclin B2 drive chromosomal instability and tumorigenesis,<br />

but through markedly different mechanisms and in distinct tissues.


TUESDAY<br />

2333<br />

Reactive oxygen species regulates CDK2 activity differently in normal and cancer cells to<br />

induce senescence and proliferation.<br />

C. Hwang 1 , S-M. Lee 1 , K-S. Kwon 1 ; 1 Aging Research Center, KRIBB, Daejeon, Korea<br />

Reactive oxygen species (ROS) were previously shown to trigger cellular senescence through<br />

several pathways such as Ras, p38 MAPK , p66shc, Rb, p53, and p21 Cip1 . However, <strong>the</strong> detailed<br />

mechanisms remain to be elucidated. Previously we demonstrated that p21 Cip1 was degraded at<br />

an early phase after low dose H2O2 treatment in a variety <strong>of</strong> cell types. Here, we observed that<br />

low dose <strong>of</strong> H2O2 induced cellular senescence in HDFs and IMR90 cells but induced cellular<br />

proliferation in cancer cells, even though p21 Cip1 was degraded in both cases. Interestingly,<br />

normal cells showed sustained inhibition <strong>of</strong> CDK2 activity upon H2O2 stimulation, which is a<br />

likely mechanism in cell cycle arrest and subsequent senescence. However, H2O2 stimulation<br />

resulted in temporary CDK2 activation in cancer cells, which might cause H2O2-induced<br />

proliferation. Still unclear are how ROS control CDK2 activity differently in <strong>the</strong> normal and<br />

cancer cells.<br />

Key words: H2O2, CDK2, p21 Cip1 , senescence, proliferation, cell fate<br />

2334<br />

The nuclear translocation <strong>of</strong> hTERT is mediated by <strong>the</strong> classical importin alpha/betadependent<br />

import pathway.<br />

S. Jeong 1 , J. Lee 1 , I. Chung 1 ; 1 Department <strong>of</strong> <strong>Biology</strong> and Integrated Omics for Biomedical<br />

Science, WCU Program <strong>of</strong> Graduate School, Yonsei university, Seoul, Korea<br />

The maintenance <strong>of</strong> eukaryotic telomeres requires telomerase, which is minimally comprised <strong>of</strong><br />

a telomerase reverse transcriptase (TERT) and an associated RNA component (TERC).<br />

Telomerase activity is tightly regulated by expression <strong>of</strong> hTERT at both <strong>the</strong> transcriptional and<br />

posttranslational levels. In our previous study, we identified <strong>the</strong> region <strong>of</strong> bipartite nuclear<br />

localization signal (NLS) <strong>of</strong> hTERT. However, <strong>the</strong> molecular mechanism <strong>of</strong> nuclear translocation<br />

<strong>of</strong> hTERT remains unclear. To investigate clear nuclear translocation mechanism, we minimized<br />

<strong>the</strong> NLS region <strong>of</strong> hTERT, and <strong>the</strong>n this NLS region was conjugated to Glutathionine Stransferase<br />

(GST). Clearly, GST protein was diffused at both cytoplasm and nucleus. In contrast<br />

to GST protein, GST-conjugated wild type NLS protein was predominantly localized in <strong>the</strong><br />

nucleus and GST-conjugated mutant (7A+S227A) NLS protein was not. To prove <strong>the</strong> molecular<br />

mechanism <strong>of</strong> nuclear translocation, we co-immunoprecipitated GST-conjugated wild-type NLS<br />

or endogenous hTERT with importin α/β1. And we also measured telomerase activity after<br />

importin alpha, beta and Ran overexpression by TRAP assay.<br />

We next investigated cells selectively depleted <strong>of</strong> importin alpha and beta1 by transfection with<br />

siRNAs. We observed <strong>the</strong> effect <strong>of</strong> importin siRNAs on endogenous hTERT by<br />

immun<strong>of</strong>luoresence analysis, subcellular fractionation assay and TRAP assay. These findings<br />

demonstrate that <strong>the</strong> nuclear translocation <strong>of</strong> hTERT is mediated by <strong>the</strong> classical importin α/βdependent<br />

import pathway.<br />

2335<br />

P300-mediated Acetylation <strong>of</strong> TRF2 is required for Telomere Maintenance.<br />

Y. Her 1 , I. Chung 1 ; 1 Department <strong>of</strong> <strong>Biology</strong> and Integrated Omics for Biomedical Science, WCU<br />

Program <strong>of</strong> Graduate School, Yonsei University, Seoul, Korea<br />

Human telomeres are protected from DNA damage by a nucleoprotein complex that includes<br />

<strong>the</strong> repeat-binding factor TRF2. The Abundance <strong>of</strong> TRF2 protein at telomeres is tightly


TUESDAY<br />

regulated by post-translational modifications, protein-protein interactions and protein<br />

stabilization. Here we report that TRF2 is physically interacts with p300 and its lysine 293 is<br />

acetylated by p300 in vitro and in vivo. Whereas overexpression <strong>of</strong> p300 increases <strong>the</strong> level <strong>of</strong><br />

telomere-bound TRF2, depletion <strong>of</strong> endogenous p300 expression decreases <strong>the</strong> level <strong>of</strong><br />

telomere-bound TRF2. We show that loss <strong>of</strong> TRF2 from telomeres results in ubiquitination and<br />

degradation <strong>of</strong> TRF2 by <strong>the</strong> proteasome. In addition, we found that p300-mediated TRF2<br />

stabilization is dependent <strong>of</strong> p300 catalytic activity, using ei<strong>the</strong>r <strong>the</strong> inactive form <strong>of</strong> p300 or <strong>the</strong><br />

acetylation mutant TRF2-K293R. Depletion <strong>of</strong> p300 in cancer cells induced DNA-damage<br />

response foci at internal genome as well as telomeres as evidenced by telomere dysfunctioninduced<br />

foci, indistinguishable from TRF2-K293R stably expressing cell lines. Some <strong>of</strong> <strong>the</strong><br />

metaphase chromosomes showed no telomeric signal at chromatid ends, suggesting an<br />

aberrant telomere structure. Fur<strong>the</strong>rmore, we found that stable overexpression <strong>of</strong> K293R<br />

induces growth arrest in human cancer cells, suggesting that K283R may control cell<br />

proliferation. Taken toge<strong>the</strong>r, <strong>the</strong>se results suggest p300-mediated acetylation <strong>of</strong> TRF2 is<br />

required for <strong>the</strong> efficient telomere binding <strong>of</strong> TRF2 as well as telomere protection and represents<br />

a new pathway for telomere maintenance by modulating <strong>the</strong> level <strong>of</strong> TRF2 at telomeres.<br />

2336<br />

Human chromosomal passenger Borealin co-exists with active telomerase and regulates<br />

telomerase activity.<br />

P. Khadka 1 , J. Lee 1 , S. Beak 1 , I. Chung 1 ; 1 Department <strong>of</strong> <strong>Biology</strong> and Integrated Omics for<br />

biomedical science, WCU program <strong>of</strong> Graduate school, Yonsei University, Seoul, Korea<br />

From different studies it has shown that <strong>the</strong> conventional DNA metabolism machineries directly<br />

or indirectly regulate telomerase activity and telomere, still it is not clearly known how DNA<br />

replication proteins regulate telomerase. Here, we report <strong>the</strong> role <strong>of</strong> borealin, member <strong>of</strong><br />

chromosomal passenger, which binds with hTERT and regulates telomerase activity. Borealin is<br />

a member <strong>of</strong> <strong>the</strong> chromosomal passenger complex that plays important role as a regulator <strong>of</strong><br />

DNA replication, chromosome alignment, and spindle assembly checkpoint. Here, we show that<br />

borealin co-exists with active telomerase in vivo via telomeric DNA in telomerase positive cells<br />

and telomerase-immortalized human somatic cells but not in human ALT cells. In <strong>the</strong> present<br />

study, we demonstrate that hTERT interacts with borealin both in vitro and in vivo. Here, we<br />

show that <strong>the</strong> association <strong>of</strong> borealin with hTERT is independent <strong>of</strong> RNA but requires DNA and<br />

also we show that N-terminal <strong>of</strong> hTERT binds to borealin. Fur<strong>the</strong>rmore, we show that borealin<br />

depletion by RNA interference reduces <strong>the</strong> telomerase activity inducing <strong>the</strong> cell growth arrest in<br />

MCF7 cells. Borealin has until now been described as a component <strong>of</strong> chromosomal passenger,<br />

mainly involved in DNA replication, here, we confirmed that about 30%-40% <strong>of</strong> borealin can bind<br />

to telomeric end. This is, to our knowledge, <strong>the</strong> first evidence that Borealin coexist with active<br />

telomerase and regulates telomerase activity, revealing a not yet recognized link <strong>of</strong> this CPC<br />

protein to telomere.<br />

2337<br />

PIAS1 and RNF4 regulate <strong>the</strong> homeostasis <strong>of</strong> SUMO-conjugated TRF2.<br />

J. Her 1 , I. Chung 1 ; 1 Department <strong>of</strong> <strong>Biology</strong> and Integrated Omics for biomedical science, WCU<br />

program <strong>of</strong> Graduate school, Yonsei University, Seoul, Korea<br />

Eukaryotic chromosome end is composed <strong>of</strong> DNA and proteins, called telomere and forms T<br />

loop to protect chromosome ends. TRF2 is a major component <strong>of</strong> sheltrin complex and function<br />

to prevent loss <strong>of</strong> genetic materials and shelter chromosome end to avoid DNA damage<br />

machinery. Without TRF2, DNA damage signal occurs and cells go to apoptosis. To avoid it,<br />

TRF2 mostly binds to telomere and is very stable. So we think that <strong>the</strong>re should be complex


TUESDAY<br />

mechanism to degrade. Sheltrin complex proteins are regulated by several post translational<br />

modifications. Among <strong>the</strong>se modifications, SUMOylation is one <strong>of</strong> essential modifications in ALT<br />

cells. It is known that TRF1 and TRF2 are sumoylated by MMS21 and <strong>the</strong>ir SUMOylation is<br />

required for APB formation in ALT cells but it is not clear what <strong>the</strong> function <strong>of</strong> TRF1 and TRF2<br />

SUMOylation is in telomerase positive cancer cells. Here, we found that PIAS1 is ano<strong>the</strong>r<br />

SUMO E3 ligase for TRF2 and SUMOylated TRF2 is regulated by proteosomal degradation.<br />

SUMO-specific E3 ubiquitin ligase, RNF4, is also involved in SUMOylated TRF2 degradtion and<br />

control <strong>the</strong> levels <strong>of</strong> SUMOylated TRF2.<br />

2338<br />

Disruption <strong>of</strong> CTCF at <strong>the</strong> miR-125b1 locus in gynecological cancers.<br />

E. Soto-Reyes 1 , R. Gonzalez 1 , F. Cisneros-Soberanis 1 , C. Castro 2 , D. Cantú 1 , F. Recillas-<br />

Targa 3 , L. A. Herrera 4,5 ; 1 Dirección de Investigación, Instituto Nacional de Cancerología,<br />

Mexico, Mexico, 2 Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas<br />

UNAM, Mexico, 3 Biología <strong>Molecular</strong>, Instituto de Fisiología Celular UNAM, 4 Instituto Nacional de<br />

Cancerología, Mexico, 5 Instituto de Investigaciones Biomédicas UNAM, Mexico<br />

In cancer cells, transcriptional gene silencing has been associated with genetic and epigenetic<br />

defects. The disruption <strong>of</strong> DNA methylation patterns and covalent histone marks has been<br />

associated with cancer development. Until recently, microRNA (miRNA) gene silencing was not<br />

well understood. In particular, miR-125b1 has been suggested to be an miRNA with tumor<br />

suppressor activity, and it has been shown to be deregulated in various human cancers. Herein,<br />

we characterized <strong>the</strong> CCCTC-binding factor (CTCF) at <strong>the</strong> miR-125b1 locus in normal and<br />

breast cancer cells. The disruption <strong>of</strong> CTCF in breast cancer cells correlated with <strong>the</strong><br />

incorporation <strong>of</strong> repressive histone marks such H3K9me3 and H3K27me3 as well as with<br />

aberrant DNA methylation patterns. To determine <strong>the</strong> effect <strong>of</strong> DNA methylation at <strong>the</strong> CpG<br />

island <strong>of</strong> miR-125b1 on <strong>the</strong> expression <strong>of</strong> this gene, we performed a qRT-PCR assay. We<br />

observed a significant reduction on <strong>the</strong> expression <strong>of</strong> miR-125b1 in cancer cells in comparison<br />

with controls, suggesting that DNA methylation at <strong>the</strong> CpG island might reduce miR-125b1<br />

expression. These effects were observed in o<strong>the</strong>r gynecological cancers, including ovarian and<br />

cervical tumors. Our data strongly suggest that <strong>the</strong> loss <strong>of</strong> CTCF may contribute to <strong>the</strong><br />

destabilization <strong>of</strong> chromatin permissive for transcription and <strong>the</strong> establishment <strong>of</strong> an aberrant<br />

repressive chromatin configuration, including DNA hypermethylation, that induce <strong>the</strong> epigenetic<br />

silencing <strong>of</strong> <strong>the</strong> miR-125b1 locus.<br />

This work was supported by <strong>the</strong> Consejo Nacional de Ciencia y Tecnología (CONACyT: 83959)<br />

and Programa de Apoyo a Proyectos de Investigación e Innovación Tecnológica <strong>of</strong> <strong>the</strong><br />

Universidad Nacional Autónoma de México (PAPIIT, IN213311).<br />

2339<br />

Cancer cell proliferation regulated by DOT1L histone methyltransferase.<br />

W. KIM 1 , R. KIM 1 , G. PARK 1 , J. KIM 1 ; 1 Kyung Hee University, Seoul, Korea<br />

DOT1L, a histone methyltransferase for histone H3 K79, participates in <strong>the</strong> regulation <strong>of</strong><br />

transcription, development, erythropoiesis, differentiation and proliferation <strong>of</strong> normal cells. With<br />

respect to cancer, DOT1L-mediated methylation contributes to development <strong>of</strong> MLL-rearranged<br />

leukemias. However, <strong>the</strong> role <strong>of</strong> DOT1L in cancer cell proliferation has not been fully elucidated.<br />

DOT1L siRNA-transfected A549 and NCI-H1299 lung cancer cells displayed a non-proliferating<br />

multinucleated phenotype. DOT1L-deficient cells also showed abnormal mitotic spindle<br />

formation and centrosome number, suggesting that DOT1L deficiency leads to chromosomal<br />

missegregation. This chromosomal instability led to cell cycle arrest at <strong>the</strong> G1 phase and


TUESDAY<br />

induced senescence determined by enhanced activity <strong>of</strong> senescence-associated (galactosidase<br />

activity. Meanwhile, overexpression <strong>of</strong> siRNA-resistant DOT1L, not a catalytically<br />

inactive mutant, substantially restores DOT1L siRNA-induced phenotypes. Overall, <strong>the</strong>se data<br />

imply that downregulation <strong>of</strong> DOT1L-mediated histone H3 Lys79 methylation inhibits<br />

proliferation <strong>of</strong> cancer cells. Therefore, <strong>the</strong> inhibition <strong>of</strong> DOT1L activity might act as a barrier to<br />

aggressive growth <strong>of</strong> cancer cells.<br />

2340<br />

Class 3 semaphorins and <strong>the</strong>ir receptor complexes- neuropilin/plexins regulated by E2F1<br />

and SOX4 in pancreatic ductal adenocarcinoma.<br />

H-Y. Huang 1,2 , Y-Y. Cheng 1 , P-H. Huang 1,2 ; 1 Department <strong>of</strong> Pathology, National Taiwan<br />

University Hospital, Taipei, Taiwan, 2 Institute <strong>of</strong> Pathology, National Taiwan University, College<br />

<strong>of</strong> Medicine, Taipei, Taiwan<br />

Ligand/receptor co-expression can allow cells to respond to growth and survival demand<br />

through signal transduction in a certain circumstance, like Eph/Ephrin co- expression modulates<br />

axon guidance in <strong>the</strong> developing central nervous system<br />

Individual members <strong>of</strong> class 3 semaphorins (SEMA3), secreted cues for axonal growth cone<br />

guidance molecules, are frequently over-expressed with <strong>the</strong>ir receptor complexes<br />

plexins/neuropilins (PLXNs/NRPs) in tumors such as breast cancer, hepatocellular carcinoma,<br />

pancreatic cancer, etc. We thoroughly examined expression <strong>of</strong> SEMA3 and <strong>the</strong>ir receptors in<br />

pancreatic ductal adenocarcinoma (PDAC) and found that, unlike o<strong>the</strong>r epi<strong>the</strong>lial malignancies<br />

that <strong>of</strong>ten over-express one or few specific members <strong>of</strong> SEMA3/PLXN family, combined<br />

expression <strong>of</strong> more than 5 members <strong>of</strong> SEMA3/Plexin family are noted in most human PDAC<br />

cases. Analysis <strong>of</strong> promoter regions <strong>of</strong> SEMA3, plexins and NRPs revealed that putative binding<br />

sites <strong>of</strong> E2F1 and SOX4 are almost always present in all SEMA3 and plexin promoters. With<br />

chromatin-immunoprecipitation, luciferase promoter activity assay, and electrophoresis mobility<br />

shift assay, E2F1 and SOX4 were demonstrated to directly bind <strong>the</strong> consensus sites at <strong>the</strong><br />

promoter <strong>of</strong> each SEMA3 and plexin gene we examined to enhance <strong>the</strong> transcriptional activity.<br />

Statistical analysis fur<strong>the</strong>r showed correlation between SOX4 expression and poor survival in<br />

human PDAC. Conversely, RNAi-knockdown <strong>of</strong> SOX4 resulted in concomitant decrease in<br />

expression level <strong>of</strong> SEMA3/plexin family members and accentuated tumor growth by affecting<br />

cell proliferation.<br />

2341<br />

microRNAs regulating matrix metalloproteinase 2 activities.<br />

S-Y. Choi 1 , J. Kim 1 , B. Shin 1 ; 1 Chonnam National University Medical School, Gwangju, Korea<br />

MMP-2 plays an important role in tumor cell invasion and metastasis. Because microRNAs<br />

(miRNAs) have recently been reported to be implicated in <strong>the</strong>se processes, we set out to<br />

determine a role for miRNAs regulating tumor cell invasion and metastasis. We screened 215<br />

human miRNAs for upregulating MMP2 activities in human fibrosarcoma HT1080 cells.<br />

Transfection <strong>of</strong> HT1080 cells with miRNAs followed by zelatin zymography identified 14 miRNAs<br />

raising MMP2 activities. Of <strong>the</strong> 14 miRNAs, fur<strong>the</strong>r analyses with Western blotting, luciferase<br />

assay and Matrigel invasion assay pointed to miR-105 and miR-128 as final candidates. We will<br />

perform in vivo pulmonary metastasis assay in mice under gain-<strong>of</strong>-function and loss-<strong>of</strong>-function<br />

contexts to validate <strong>the</strong> effect <strong>of</strong> miR-105 and miR-128 on tumor cell invasion and metastasis.


TUESDAY<br />

2342<br />

Pr<strong>of</strong>iling <strong>of</strong> linker histone variants in ovarian cancer.<br />

M. Medrzycki 1 , Y. Zhang 1 , J. McDonald 1 , Y. Fan 1 ; 1 Georgia Institute <strong>of</strong> Technology, Atlanta, GA<br />

H1 linker histones play a key role in facilitating higher order chromatin folding. There are 11 H1<br />

variants in mammals that are differentially regulated during development and cellular<br />

differentiation. Emerging evidence suggests that H1 and its specific H1 variants are important<br />

epigenetic factors in modulating chromatin function and gene expression. Ovarian cancer is a<br />

devastating disease, ranking <strong>the</strong> fifth leading cause <strong>of</strong> all women cancer death due to its poor<br />

prognosis and difficulty in early diagnosis. Although epigenetic alterations in ovarian cancers are<br />

being appreciated in general, <strong>the</strong> role <strong>of</strong> H1 in ovarian cancers has not been explored. Here,<br />

using quantitative RT-PCR assays, we systematically examined <strong>the</strong> expression levels <strong>of</strong> 7 H1<br />

genes in 33 human epi<strong>the</strong>lial ovarian tumors. We found that <strong>the</strong> expression <strong>of</strong> H1.3 is markedly<br />

increased, whereas <strong>the</strong> expression levels <strong>of</strong> H10, H1.1, H1.4 and H1x are significantly reduced<br />

in <strong>the</strong> malignant adenocarcinomas compared to benign adenomas. Strikingly, ovarian<br />

adenocarcinomas and adenomas exhibit characteristic expression patterns, and expression<br />

pr<strong>of</strong>iling <strong>of</strong> <strong>the</strong>se 7 H1 genes in tumor samples can discriminate adenocarcinomas vs.<br />

adenomas. We identified 6 H1 genes as <strong>the</strong> minimum gene set required for correct<br />

discrimination <strong>of</strong> ovarian adenomas and adenocarcinomas with high accuracy. These findings<br />

indicate that <strong>the</strong> expression <strong>of</strong> H1 variants is exquisitely regulated and may serve as potential<br />

epigenetic biomarkers for ovarian cancer.<br />

2343<br />

Investigating <strong>the</strong> contribution <strong>of</strong> centrosome amplification in tumorigenesis.<br />

B. D. Vitre 1 , A. J. Holland 1 , Y. Wang 1 , D. W. Cleveland 1,2 ; 1 Ludwig Institute for Cancer Research,<br />

La Jolla, CA, 2 Department <strong>of</strong> Cellular and <strong>Molecular</strong> Medicine, University <strong>of</strong> California San<br />

Diego, La Jolla, CA<br />

As <strong>the</strong> major microtubule organizing centers, centrosomes play a central role in facilitating <strong>the</strong><br />

formation <strong>of</strong> a bipolar mitotic spindle. Defects in centrosome biogenesis can induce an abnormal<br />

centrosome number and may lead to chromosome missegregation and subsequent aneuploidy.<br />

Although aneuploidy is an extremely common feature <strong>of</strong> tumor cells, its status as a cause or a<br />

consequence <strong>of</strong> cancer is highly controversial. In vertebrates and invertebrates, <strong>the</strong> conserved<br />

protein kinase Polo-like kinase 4 (Plk4) plays a key role in initiating centriole duplication and<br />

overexpression <strong>of</strong> Plk4 promotes <strong>the</strong> formation <strong>of</strong> extra centrosomes. Here we will describe <strong>the</strong><br />

construction <strong>of</strong> two mouse models in which centrosome amplification can be induced through<br />

conditional overexpression <strong>of</strong> Plk4. We have made use <strong>of</strong> both a doxycycline-inducible promoter<br />

and <strong>the</strong> Cre-LoxP system to allow reversible and non-reversible expression <strong>of</strong> Plk4 in a tissue<br />

specific manner. These mice will be used to study <strong>the</strong> contribution <strong>of</strong> centrosome amplification<br />

in tumorigenesis.<br />

2344<br />

LEKTI re-expression up-regulates p53 and p21 in OSC-19, a metastatic squamous cell<br />

carcinoma.<br />

A. Iarrobino 1,2 , T. Shellenberger 3 , S. Klemann 2,3 ; 1 School <strong>of</strong> Medicine, University <strong>of</strong> Central<br />

Florida, Orlando, FL, 2 Rollins College, Winter Park, FL, 3 MD Anderson Cancer Center - Orlando,<br />

Orlando, FL<br />

Lymphoepi<strong>the</strong>lial Kazal Type Inhibitor (LEKTI) is a 15-domain serine proteinase inhibitor with<br />

constitutive expression in squamous mucosa and with reduced or absent expression in <strong>the</strong><br />

majority <strong>of</strong> head and neck squamous cell carcinomas (HNSCC) and cell lines. The loss <strong>of</strong>


TUESDAY<br />

expression may alter dynamics in <strong>the</strong> extracellular matrix <strong>of</strong> <strong>the</strong> tumor environment to favor local<br />

invasion and metastasis. Re-expression <strong>of</strong> LEKTI in HNSCC lines alters cell behavior in vitro<br />

and reduces local invasion in vivo. p53 is a well-characterized tumor suppressor gene with lost<br />

expression in many cancers, including HNSCC. In <strong>the</strong> context <strong>of</strong> examining LEKTI and matrix<br />

metalloproteinase expression in <strong>the</strong> HNSCC line OSC-19, we found <strong>the</strong> expression <strong>of</strong> p53 was<br />

restored upon stable transfection <strong>of</strong> OSC-19 with LEKTI to induce re-expression. Using OSC-19<br />

parental, vector, and clones re-expressing LEKTI (LEKTI-17 and LEKTI-25), semi-quantitative<br />

and quantitative analysis demonstrate restored expression <strong>of</strong> p53 in OSC-19 cells after<br />

transfection with LEKTI and cultivation both in vitro and in vivo (murine flank and orthotopic<br />

tongue tumor models). With re-expression <strong>of</strong> LEKTI and <strong>the</strong> subsequent restoration <strong>of</strong> p53, <strong>the</strong><br />

expression <strong>of</strong> p21 was in turn up regulated. Differential expression <strong>of</strong> LEKTI was noted for<br />

parental cells cultivated in vitro and in vivo. Whereas LEKTI expression did not take place in<br />

vitro, its expression took place in both tumor types. We found p53 and p21 expression in<br />

xenografts derived from parental cells and both OSC-19 clones transfected to express LEKTI.<br />

Whenever LEKTI was expressed, re-expression <strong>of</strong> p53 and p21 was observed. Lastly, in vitro<br />

treatment <strong>of</strong> parental and vector cell lines with conditioned medium containing secretory factors<br />

<strong>of</strong> LEKTI-25 cells resulted in <strong>the</strong> re-expression <strong>of</strong> p53 and p21. Our results demonstrate <strong>the</strong><br />

integrity <strong>of</strong> signaling pathways through which extracellular factors act and highlight <strong>the</strong><br />

importance <strong>of</strong> <strong>the</strong> tumor microenvironment in regulating downstream mediators involved in <strong>the</strong><br />

biologic behavior <strong>of</strong> OSC-19. [The James and Es<strong>the</strong>r King New Investigator Award <strong>of</strong> <strong>the</strong><br />

Florida Biomedical Research Program (TS) and <strong>the</strong> Critchfield Fund <strong>of</strong> Rollins College (SK)<br />

funded this work.]<br />

Normal and Diseased Organs and Therapeutics<br />

2345<br />

Therapeutic Multiple Exon-skipping Using Cell-penetrating Morpholinos for Dystrophic<br />

Dogs.<br />

T. Yokota 1 , T. Nagata 2 , A. Nakamura 3 , N. Urasawa 4 , T. Saito 2 , R. Kole 5 , P. Sazani 5 , T.<br />

Partridge 6 , E. H<strong>of</strong>fman 6 , S. Takeda 2 ; 1 University <strong>of</strong> Alberta, Edmonton, AB, Canada, 2 National<br />

Center <strong>of</strong> Neurology and Psychiatry, Japan, 3 Shinshu University, Japan, 4 Nagano Red Cross<br />

Hospital, Japan, 5 AVI Biopharma, 6 Children's National Medical Center, Washington, DC<br />

Duchenne muscular dystrophy (DMD), <strong>the</strong> most common and fatal X-linked myopathy, and its<br />

milder form, Becker muscular dystrophy (BMD), are caused by mutations in <strong>the</strong> dystrophin<br />

(DMD) gene. Antisense-mediated exon skipping <strong>the</strong>rapy is currently one <strong>of</strong> <strong>the</strong> most promising<br />

molecular <strong>the</strong>rapies for DMD. The exon skipping leads to <strong>the</strong> production <strong>of</strong> internally deleted inframe<br />

mRNA transcripts but <strong>the</strong> truncated quasi-dystrophin retains some functions like BMD.<br />

Previously we demonstrated <strong>the</strong> first successful exon-skipping treatment in body-wide skeletal<br />

muscles in Canine X-linked muscular dystrophy (CXMD) using a cocktail <strong>of</strong> phosphorodiamidate<br />

morpholino oligomers (PMOs, morpholinos) targeting exon 6 and exon 8 <strong>of</strong> dystrophin mRNA.<br />

However, unmodified (bare) morpholino injections led to inefficient delivery to <strong>the</strong> heart, and<br />

dystrophin induction was barely detectable in <strong>the</strong> cardiac muscle. Here, we sought to recover<br />

<strong>the</strong> dystrophin expression in cardiac muscles in dystrophic dogs using morpholinos conjugated<br />

with negatively charged arginine-rich cell-penetrating peptides (PPMOs). We demonstrated that<br />

<strong>the</strong> delivery moieties significantly improved dystrophin production in both skeletal and cardiac<br />

muscles. Intravenous injections with PPMOs restored dystrophin expression in cardiac muscles<br />

accompanied by ameliorated histology. No obvious toxicity was detected by blood tests and<br />

histology. Our results show <strong>the</strong> potential <strong>of</strong> PMO conjugates as <strong>the</strong>rapeutic agents for DMD.


TUESDAY<br />

2346<br />

The Regenerative Response <strong>of</strong> Zebrafish Hearts to Long-Term Induced Exercise Stress.<br />

B. Sch<strong>of</strong>fstall 1 , P. deVerteuil 1 , M. Jean 1 , N. Lopez 1 , J. Tapia 1 ; 1 <strong>Biology</strong>, Barry University, Miami<br />

Shores, FL<br />

While adult human cardiomyocytes are capable <strong>of</strong> low level cell division, this hyperplastic<br />

response is not sufficient to repair damaged cardiac tissue. Danio rerio (zebrafish)<br />

cardiomyocytes have recently been shown to be capable <strong>of</strong> proliferating at high levels to<br />

completely repair hearts damaged by apex amputation or cryoinjury; thus, <strong>the</strong>se fish have<br />

become a popular model to study heart regeneration. We hypo<strong>the</strong>sized that exposure <strong>of</strong><br />

zebrafish to long-term cardiac overload stress would elicit a similar response without invasive<br />

surgery, to provide us with a simple cardiac regeneration research model. In addition, this model<br />

may have useful applications to human stress-induced cardiac responses, which can lead to<br />

pathological cardiac hypertrophy and ultimately death. We stressed zebrafish cardiac systems<br />

using a 10-week forced swimming exercise regimen, <strong>the</strong>n compared fully remodeled hearts to<br />

non-exercised controls for physiological function and histological evidence <strong>of</strong> cell proliferation.<br />

We sought to determine if <strong>the</strong> zebrafish heart response to cardiac overload stress results in<br />

heart enlargement, and whe<strong>the</strong>r hypertrophy or hyperplasia is involved. We also used a<br />

zebrafish full-genome expression microarray to screen for genes that may be key initiators <strong>of</strong> a<br />

regenerative response. Adult zebrafish were forced to swim twice daily against a strong water<br />

current. After a period <strong>of</strong> 10 weeks, individual fish were measured for overall size; hearts were<br />

filmed to determine heart rate, ventricular surface area, and percent shortening fraction; and<br />

hearts were extracted for histological tissue sectioning, cDNA preparation, and analysis <strong>of</strong> total<br />

protein. Our results indicate that fully remodeled exercised zebrafish heart ventricles are ~32%<br />

larger than non-exercised (p


TUESDAY<br />

1) Malformation <strong>of</strong> great toes present at birth. 2) Progressive heterotropic ossification which<br />

starts at <strong>the</strong> first decade <strong>of</strong> life. Ossification is manifested by recurrent and painful swelling flare<br />

ups <strong>of</strong> s<strong>of</strong>t tissues that involve fascias, ligaments, aponeuroses, tendons and skeletal muscles.<br />

A Mexican family with one member affected <strong>of</strong> FOP was studied. The patient with <strong>the</strong> illness<br />

was a 19 years old female, who started with this disease at <strong>the</strong> age <strong>of</strong> 8 years old; she<br />

developed spontaneous and painful swelling <strong>of</strong> <strong>the</strong> right scapular area accompanied <strong>of</strong><br />

functional limitation for movement, among clinical laboratory abnormalities it was found alkaline<br />

phosphatase activity increased. By radiography <strong>the</strong>re was abnormal calcification and "pseudoexostosis<br />

dependent <strong>of</strong> ligament ossification at <strong>the</strong> site <strong>of</strong> attachment to <strong>the</strong> long bones.<br />

Mutation analysis was carried out using genomic DNA from peripheral blood lymphocytes;<br />

samples were assessed by PCR amplification using exon flanking primers (Shore et al, Nature<br />

Genetics 38; 525), PCR products were digested with respective restriction enzymes 1h at 37°C.<br />

Main results were obtained with <strong>the</strong> exon 4 flanking primers and Cac81 restriction enzyme, this<br />

gave a 253 bp product that carries <strong>the</strong> ACVR1 617→A mutation which causes aminoacid<br />

substitution <strong>of</strong> arginine by histidine in codon 206. ACVR1 gene encodes a type I bone<br />

morphogenic protein (BMP) transmembrane receptor, and mutation <strong>of</strong> this gene results in<br />

dysregulation <strong>of</strong> BMP signaling. This rare case is a novo mutation since her relatives were<br />

clinically healty and molecular analysis did not show ACVR1 617→A mutation.<br />

2348<br />

TGF-β and BAFF derived from CD4+CD25+Foxp3+ T cells mediate mouse IgA isotype<br />

switching.<br />

G-Y. Seo 1 , P-H. Kim 1 ; 1 Department <strong>of</strong> <strong>Molecular</strong> Bioscience, Kangwon National University,<br />

Chuncheon, Korea<br />

TGF-β is generally accepted as <strong>the</strong> physiological IgA isotype switch factor. Never<strong>the</strong>less, it is<br />

virtually unknown what kinds <strong>of</strong> cells in mucus-associated lymphoid tissue (MALT) mainly<br />

provide this cytokine to B cells. Regulatory T cells (Tregs) has immune suppressive activity by<br />

secretion <strong>of</strong> inhibitory cytokine such as TGF-β and IL-10. Thus, it is plausible that Tregs may be<br />

involved in IgA class switching recombination (CSR) in MALT. We explored, in <strong>the</strong> present<br />

study, <strong>the</strong> possibility that CD4 + CD25 + T cells facilitate IgA CSR in mouse B cells.<br />

CD4 + CD25 + Foxp3 + T cells were more stimulatory in IgA production by <strong>the</strong> cocultured splenic B<br />

cells than CD4 + CD25 - Foxp3 - T cells, and this effect was markedly abrogated by anti-TGFβAb.<br />

This was paralleled by an increase <strong>of</strong> germ line transcriptα (GLTα), an indicative <strong>of</strong> IgA CSR. In<br />

contrast, CD4 + CD25 - Foxp3 - T cells were more potent in induction <strong>of</strong> GLTγ1 and GLTε by <strong>the</strong><br />

cocultured splenic B cells. Consistent to <strong>the</strong>se results, phenotypic analyses revealed that TGF-β<br />

and IL-4 were predominantly expressed by CD4 + CD25 + Foxp3 + T cells and CD4 + CD25 - Foxp3 - T<br />

cells, respectively. Fur<strong>the</strong>r, we found that CD4 + CD25 + T cells strongly express BAFF leading to<br />

AID expression in B cells. Taken toge<strong>the</strong>r, our results suggest that CD4 + CD25 + T cells may<br />

have an important effect on IgA isotype commitment through TGF-β and BAFF in MALT.


2349<br />

Anti-platelet activity <strong>of</strong> yuzu and its components is mediated by inhibition <strong>of</strong> TXA2<br />

production.<br />

H. Yu 1,2 , S. Park 3 , I. Chung 4 , Y-S. Jung 1,5 ; 1 College <strong>of</strong> Pharmacy, Ajou University School,<br />

Suwon, Korea, 2 Brain Korea 21 for Medical Sciences, Ajou University, Suwon, Korea,<br />

3 <strong>Molecular</strong> Biotechnology, Konkuk University, Seoul, Korea, 4 Applied Life Science, Konkuk<br />

University, Seoul, Korea, 5 Brain Korea 21 for <strong>Molecular</strong> Science and Technology, Ajou<br />

University, Suwon, Korea<br />

TUESDAY<br />

The platelet activation and subsequent platelet aggregation play an essential role in <strong>the</strong><br />

pathogenesis <strong>of</strong> a<strong>the</strong>rothrombotic disease such as cardiovascular, cerebrovascular, and<br />

peripheral vascular diseases. Food can be one <strong>of</strong> <strong>the</strong> most important factors that influence risks<br />

for a<strong>the</strong>rothrombotic diseases. Yuzu included in citrus is one candidate that may benefit <strong>the</strong><br />

cardiovascular system. In this study, we investigated whe<strong>the</strong>r <strong>the</strong> methanolic extract <strong>of</strong> yuzu<br />

(yuzu ME) and its components (hesperidin and naringin) have anti-platelet activities. Yuzu ME<br />

and hesperidin inhibited collagen-, arachidonic acid (AA)-, ADP- and thrombin-induced rat<br />

platelet aggregation in vitro and ex vivo. Naringin also inhibited platelet aggregation induced by<br />

collagen, AA, or thrombin, but not aggregation induced by ADP. The oral administration <strong>of</strong> yuzu<br />

ME or hesperidin prolonged mouse tail vein bleeding time in a dose-dependent manner in vivo.<br />

In addition, yuzu ME including hesperidin and naringin inhibited collagen and thrombin-induced<br />

platelet aggregation by interfering with TXA2 production as established by <strong>the</strong> measurement <strong>of</strong><br />

TXB2. These results suggest that yuzu ME and its component such as hesperidin and naringin<br />

have anti-platelet activity, and that intake <strong>of</strong> yuzu, which includes various flavonoids such as<br />

hesperidin, may be beneficial for individuals at high risk <strong>of</strong> cardiovascular diseases. This<br />

research was supported by <strong>the</strong> Technology Development Program (#308013-3) for ′Food′<br />

Ministry for Food, Agriculture, Forestry and Fisheries, and <strong>the</strong> Cooperative Research Program<br />

for Agriculture Science & Technology Development (Project No. PJ006986) Rural Development<br />

Administration, Republic <strong>of</strong> Korea.<br />

2350<br />

Dehydroepiandrosterone inhibits <strong>the</strong> activation and dysfunction <strong>of</strong> endo<strong>the</strong>lial cells<br />

induced by high glucose concentration.<br />

E. Huerta García 1 , J. L. Ventura Gallegos 2,3 , M. E. Crescencio Victoriano 1 , A. Montiel Dávalos 1,4 ,<br />

G. Tinoco Jaramillo 5 , R. López-Marure 6 ; 1 Biología Celular, Instituto Nacional de Cardiología<br />

“Ignacio Chávez”, México, 2 Departamento de Bioquímica, Instituto Nacional de Ciencias<br />

Médicas y Nutrición "Salvador Zubirán", México, 3 Departamento de Medicina Genómica y<br />

Toxicología Ambiental, Instituto de Investigaciones Biomédicas, UNAM, Mexico, 4 Subdirección<br />

de Investigación Básica, Instituto Nacional de Cancerología, México, 5 Jefatura de Ginecología,<br />

Hospital General de Zona 2A “Troncoso”, IMSS, México, 6 Biología Celular, Instituto Nacional de<br />

Cardiología “Ignacio Chávez”, México City, Mexico<br />

Dehydroepiandrosterone (DHEA), an adrenal steroid, has a protective role against diabetes;<br />

however, its mechanisms <strong>of</strong> action are unknown. Here, we focus on <strong>the</strong> effect <strong>of</strong> DHEA on <strong>the</strong><br />

activation <strong>of</strong> endo<strong>the</strong>lial cells induced by a high concentration <strong>of</strong> glucose. The adhesion on<br />

U937 cells, <strong>the</strong> expression <strong>of</strong> adhesion molecules, <strong>the</strong> production <strong>of</strong> ROS and NO, and <strong>the</strong><br />

translocation <strong>of</strong> NF-[kappa]B were evaluated in human umbilical vein endo<strong>the</strong>lial cells (HUVEC)<br />

treated with high concentrations <strong>of</strong> glucose, DHEA, and both. High concentrations <strong>of</strong> glucose (><br />

20 mM) induced an increase in adhesion, an increase in mainly E-selectin and PECAM-1<br />

expression, ROS and NO production, translocation <strong>of</strong> NF-[kappa]B, and degradation <strong>of</strong> its<br />

inhibitor I[kappa]B-[alpha]. DHEA abolished adhesion and <strong>the</strong> increase <strong>of</strong> E-selectin, ICAM-1,<br />

VCAM-1, and PECAM-1 induced by glucose. In addition, DHEA completely blocked oxidative


TUESDAY<br />

stress and decreased translocation <strong>of</strong> NF-[kappa]B and <strong>the</strong> degradation <strong>of</strong> I[kappa]B-[alpha]<br />

induced by glucose. These results suggest that DHEA protects against <strong>the</strong> activation <strong>of</strong><br />

endo<strong>the</strong>lial cells induced by high concentrations <strong>of</strong> glucose, indicating that DHEA could be<br />

useful in <strong>the</strong> treatment <strong>of</strong> hyperglycemia and diabetes.<br />

2351<br />

Activin A stimulates mouse antigen presenting cells to express APRIL and BAFF.<br />

G. Seo 1 , P-H. Kim 1 ; 1 Department <strong>of</strong> <strong>Molecular</strong> Bioscience, Kangwon National University,<br />

Chuncheon, Korea<br />

The TNF family ligands APRIL and BAFF are primarily expressed by macrophages and<br />

dendritic cells and stimulate <strong>the</strong> differentiation and survival <strong>of</strong> B cells and <strong>the</strong>ir Ig production. In<br />

this study, we investigated <strong>the</strong> role <strong>of</strong> activin A in APRIL and BAFF expression by mouse<br />

antigen presenting cells and <strong>the</strong> signaling mechanism involved. Activin A markedly enhanced<br />

APRIL expression in mouse macrophages at both <strong>the</strong> transcriptional and protein levels.<br />

Overexpression <strong>of</strong> DN-Smad3 and SB431542 abrogated activin-induced APRIL transcription.<br />

Fur<strong>the</strong>rmore, activin A induced Smad3 phosphorylation. These results indicate that activin A<br />

enhances APRIL expression through both ALK4 and Smad3. In a subsequent analysis <strong>of</strong> activin<br />

A signaling, it was found that PD98059, an ERK inhibitor, eliminated activin A-induced APRIL<br />

expression. On <strong>the</strong> o<strong>the</strong>r hand, overexpression <strong>of</strong> CREB, a molecule downstream <strong>of</strong> ERK,<br />

augmented activin A-induced APRIL expression, and this effect could be abolished by<br />

PD98059. This finding that activin A induces ERK and CREB phosphorylation suggests that<br />

ERK and CREB act as intermediates in APRIL expression. Therefore, <strong>the</strong>se results<br />

demonstrate that activin A can enhance APRIL expression through two different pathways,<br />

Smad3 and ERK/CREB. On <strong>the</strong> o<strong>the</strong>r hand, activin A also enhanced BAFF expression in mouse<br />

macrophages and dendritic cells at both <strong>the</strong> transcriptional and protein levels. Overexpression<br />

<strong>of</strong> DN-Smad3 and SB431542 abrogated activin-induced APRIL transcription. These results<br />

demonstrate that activin A can enhance BAFF expression through ALK4-Smad3 pathway.<br />

Taken toge<strong>the</strong>r, <strong>the</strong>se results suggest that activin A can modulate mouse APCs to express<br />

APRIL and BAFF.<br />

2352<br />

High-fat diet during sexual maturation influences <strong>the</strong> epi<strong>the</strong>lium cell turnover kinetics in<br />

<strong>the</strong> adult rat ventral prostate.<br />

E. Z. Pytlowanciv 1 , D. L. Ribeiro 2 , R. M. Góes 3 ; 1 <strong>Biology</strong> Institute, State University <strong>of</strong> Campinas<br />

(Unicamp), Campinas, Brazil, 2 Histology Sector, Institute <strong>of</strong> Biomedical Sciences, Federal<br />

University <strong>of</strong> Uberlandia (UFU), Uberlândia, Brazil, 3 Department <strong>of</strong> <strong>Biology</strong>, University <strong>of</strong><br />

Estadual Paulista (Unesp), São José do Rio Preto, Brazil<br />

The obesity epidemic has been recognized as one <strong>of</strong> <strong>the</strong> major global health problems and is<br />

associated to several diseases including reproductive problems. Various consequences <strong>of</strong><br />

obesity in <strong>the</strong> male genital tract have been related to changes in sex steroid hormones.<br />

Although this could be applied to prostate, <strong>the</strong> influence <strong>of</strong> obesity and high-fat ingestion to <strong>the</strong><br />

prostate histophysiology is unknown. The aim <strong>of</strong> this study was to evaluate if a high-fat diet (HF)<br />

interferes on rat ventral prostate pubertal maturation and harm <strong>the</strong> histology <strong>of</strong> adult gland. Male<br />

Wistar rats (4w old) were fed for 3, 6 or 9 weeks (HF3, 6 and 9) with high-fat diet (20%fat,<br />

4.9kcal/g) whereas control animals (C3, 6 and 9) received balanced chow (4%fat, 3.2Kcal/g).<br />

Ventral prostate was analyzed using stereological, immunocytochemical and western blotting<br />

methods. The relative proportion <strong>of</strong> tissue gland components were maintained in HF, except for<br />

an 1.1-fold increase in acinar epi<strong>the</strong>lium volume at HF6. Morphological analyses indicated that<br />

acinar epi<strong>the</strong>lium was significantly higher in HF groups, more pleated and exhibited mild diffuse


TUESDAY<br />

epi<strong>the</strong>lial hyperplasia. The high cellularity observed in acinar epi<strong>the</strong>lium <strong>of</strong> all HF-treated groups<br />

is evidenced by a significant rise in cell densities. HF during sexual maturation promoted a 4fold<br />

increment in cell proliferation as well as a 4-fold reduction in apoptosis rate and also<br />

elevated circulating testosterone serum levels in HF9 compared to C9 group.<br />

Immunocytochemistry indicated an increase in cells expressing AR under all HF conditions.<br />

However, <strong>the</strong> western blotting analyses surprisingly pointed to a tendency <strong>of</strong> 110kDa-AR band<br />

down expression and 45kDa band over expression in prostate crude protein extracts. This may<br />

be due to primary polyclonal antibody non-specific cross reaction and <strong>the</strong> small molecular<br />

weight bands found on prostate extracts possibly correspond to AR is<strong>of</strong>orms. Interestingly, <strong>the</strong>re<br />

was an inverse correlation between 110kDa AR and smaller band expression in HF3 and HF9,<br />

which may suggest a fat dietary interference on differential expression <strong>of</strong> AR is<strong>of</strong>orms. The<br />

circulating androgen elevation, could explain <strong>the</strong> cell proliferation increment, apoptosis reduction<br />

and high prostate growth observed during HF treatment, since <strong>the</strong> androgens are <strong>the</strong> main<br />

factors responsible for maintaining <strong>the</strong> homeostasis <strong>of</strong> <strong>the</strong> prostate, through <strong>the</strong> activation <strong>of</strong> its<br />

receptor (AR). Finally, <strong>the</strong>se experiments indicate that HF diet-treatment during puberty<br />

influences prostatic maturation causing hiperplastic alterations and higher gland volume at<br />

adulthood. In addition, <strong>the</strong> quantity <strong>of</strong> satured fat in diet interferes in <strong>the</strong> cell epi<strong>the</strong>lium turnover<br />

kinetics in <strong>the</strong> adult prostate.<br />

2353<br />

Effect <strong>of</strong> peripheral serotonin on glucose uptake in murine brown adipocyte.<br />

H. Watanabe 1 , K. Saito 1 , R. Saito 1 , T. Nakano 1 , N. Okada 1 , K. Sumiyoshi 1 , S. Ohwada 1 , K.<br />

Watanabe 1 , H. Aso 1 ; 1 Tohoku University, Miyagiken Sendaishi, Japan<br />

Serotonin is a neurotransmitter syn<strong>the</strong>sized in <strong>the</strong> raphe nuclei <strong>of</strong> <strong>the</strong> brain stem and involved in<br />

<strong>the</strong> central control <strong>of</strong> food intake, sleep and mood. Serotonin is also a peripheral hormone<br />

produced by enterochromaffin cells in <strong>the</strong> intestine and involved in vasoconstriction,<br />

haemostasis and immune system. Serotonin is syn<strong>the</strong>sized by two distinct tryptophan<br />

hydroxylase (TPH) rate-limiting enzyme in brain (TPH2) and in peripheral (TPH1). As serotonin<br />

is inability to cross <strong>the</strong> blood-brain barrier, <strong>the</strong>re are two serotonin systems in brain and<br />

periphery with independent functions. We revealed that serotonin increased plasma glucose<br />

concentration through inhibiting glucose uptake from blood to tissue. However, <strong>the</strong> functions <strong>of</strong><br />

serotonin in periphery have not yet been fully elucidated.<br />

The major <strong>the</strong>rmogenic center is <strong>the</strong> brown adipose tissue (BAT), which is composed <strong>of</strong><br />

multilocular lipid droplets and a large number <strong>of</strong> mitochondria. Thus, BAT is known as an<br />

important tissue in <strong>the</strong> regulation <strong>of</strong> body weight and energy metabolism. In this study, we have<br />

established a clonal murine brown preadipocyte (MBP) line and investigated <strong>the</strong> function <strong>of</strong><br />

serotonin on glucose metabolism in MBP cells. After fasted mice were intraperitoneally injected<br />

with 1 mg serotonin, <strong>the</strong> glucose uptake in BAT was elevated. Next, we tried to reveal <strong>the</strong> effect<br />

<strong>of</strong> serotonin on glucose uptake in BAT using established MBP cells. MBP cells showed <strong>the</strong><br />

characteristic <strong>of</strong> BAT followed induction <strong>of</strong> differentiation, such as multilocular lipid droplets and<br />

<strong>the</strong> expressions <strong>of</strong> BAT related genes including uncoupling protein 1 (UCP-1). Additionally, <strong>the</strong><br />

stimulation <strong>of</strong> serotonin induced <strong>the</strong> increase <strong>of</strong> glucose uptake in MBP cells in a dose<br />

dependent manner. To determine what kind <strong>of</strong> serotonin receptors were related to <strong>the</strong> glucose<br />

uptake, we confirmed <strong>the</strong> expression <strong>of</strong> several serotonin receptors and measured <strong>the</strong> glucose<br />

uptake in MBP cells using three kinds <strong>of</strong> serotonin receptor antagonist: SB-204070 (5HT4), Ro-<br />

04-6790 (5HT6) and methysergide (5HT1, 5HT2 and 5HT7). The pretreatment <strong>of</strong> serotonin<br />

antagonist SB-204070 inhibited <strong>the</strong> glucose uptake in MBP cells after serotonin treatment.<br />

These data indicate that serotonin regulates a glucose metabolism in BAT through serotonin 4<br />

receptor.


TUESDAY<br />

2354<br />

Topical application <strong>of</strong> capsaicin induces adipokine secretion in visceral adipose tissues.<br />

G-R. Lee 1 , D-J. Yoon 2 , I-S. Han 2 ; 1 Medical Sciences, University <strong>of</strong> Ulsan, Ulsan, Korea,<br />

2 Biological Sciences, University <strong>of</strong> Ulsan, Ulsan, Korea<br />

We attempted to determine whe<strong>the</strong>r topical application <strong>of</strong> capsaicin can protect fat accumulation<br />

in visceral adipose tissues. We first observed that <strong>the</strong> topical application <strong>of</strong> 0.075% capsaicin to<br />

male obese mice fed a high-fat diet significantly reduced weight gain and visceral fat compared<br />

to those <strong>of</strong> untreated obese control mice (p < 0.001). Fat cells were markedly reduced in<br />

mesenteric and epididymal adipose tissues <strong>of</strong> <strong>the</strong> mice treated with capsaicin cream. Capsaicin<br />

treatment lowered serum levels <strong>of</strong> fasting glucose, total cholesterol, and triglycerides.<br />

Immunoblot analysis and RT-PCR revealed increased adiponectin expression in capsaicintreated<br />

mice. In addition, application <strong>of</strong> capsaicin to obese mice downregulated mRNA levels <strong>of</strong><br />

TNF-Ą and IL-6 but upregulated o<strong>the</strong>r adipokines including PPARs,visfatin, and adipsin. These<br />

results indicate that topical application <strong>of</strong> capsaicin to obese mice limited fat accumulation in<br />

adipose tissues and may contribute to insulin sensitivity and anti-inflammation.<br />

2355<br />

Interleukin-6 / Stat3 axis mediates S100a9 expression in <strong>the</strong> colonic epi<strong>the</strong>lial cells in a<br />

mouse model <strong>of</strong> ulcerative colitis.<br />

J-K. Lee 1 , M-J. Lee 1 , J. Choi 1 , C-H. Cho 1 , S-K. Ye 1 ; 1 Department <strong>of</strong> Pharmacology, Seoul<br />

National University College <strong>of</strong> Medicine, Seoul, Korea<br />

In <strong>the</strong> intestinal epi<strong>the</strong>lium lining, <strong>the</strong> mucosal surface is a key player in homeostasis. However,<br />

when <strong>the</strong> balance is disturbed, it might lead to intestinal inflammation, including ulcerative colitis<br />

(UC). Interleukin-6 (IL-6) and <strong>the</strong> signal transducer and activator <strong>of</strong> transcription 3 (STAT3)<br />

phosphorylation have been reported to be highly expressed in <strong>the</strong> colonic tissues in UC. Also,<br />

S100A9, as a fecal biomarker, is correlated with <strong>the</strong> severity <strong>of</strong> UC. However, <strong>the</strong> expression<br />

mechanism <strong>of</strong> S100A9 in colonic epi<strong>the</strong>lial cells (CECs) remains elusive. Here, a mouse model<br />

<strong>of</strong> UC was developed to investigate <strong>the</strong> linkage <strong>of</strong> IL-6 and S100a9 in CECs. We injected<br />

sgp130Fc or siSTAT3/CH-NP into dextran sodium sulfate (DSS)–treated mice and observed a<br />

markedly decreased S100a9 expression in CECs compared to those treated with PBS or<br />

siNegative/CH-NP. Fur<strong>the</strong>rmore, <strong>the</strong> administration <strong>of</strong> JAK2 or STAT3 inhibitors post IL-6<br />

stimulation into Caco-2 indicated that IL-6-mediated S100A9 expression occurs through STAT3<br />

activation. Also, we found that STAT3 binds directly to <strong>the</strong> S100A9 promoter and enhances its<br />

activity. We provide insights on <strong>the</strong> role <strong>of</strong> S100a9, which may recruit granulocytes into inflamed<br />

colon tissues. Thus, we conclude that elevated S100a9 expression mediated by IL-6/STAT3 in<br />

CECs plays an important role in <strong>the</strong> manifestation <strong>of</strong> UC.<br />

2356<br />

Immune cell monitoring using a handheld cell counter.<br />

A. Cappione 1 , N. Thirumalapura 2 , E. Crossley 2 , D. Hoover 1 ; 1 EMD Millipore, Danvers, MA,<br />

2 University <strong>of</strong> Texas Medical Branch, Galveston, TX<br />

Biological samples, such as primary isolates or cultured cells, are <strong>of</strong>ten comprised <strong>of</strong> a phenotypically heterogeneous<br />

population. The ability to discriminate subsets and determine <strong>the</strong>ir frequencies (and concentrations) is critical to many aspects<br />

<strong>of</strong> research. Cellular complexity is typically resolved by flow cytometry using panels <strong>of</strong> protein-specific fluorescent antibodies.<br />

Notably, many cell states are also uniquely distinguishable on <strong>the</strong> basis <strong>of</strong> size alone.<br />

Cell counting devices, operating via <strong>the</strong> principle <strong>of</strong> Coulter-based particle detection, permit <strong>the</strong> determination cell diameter<br />

measurements at <strong>the</strong> submicron level, as well as providing accurate concentrations. We describe three examples where a<br />

handheld cell counter provided qualitative assessment <strong>of</strong> individual population frequencies in complex cell mixtures. These


TUESDAY<br />

include: (1) Determination <strong>of</strong> lymphocyte and monocyte concentrations and <strong>the</strong>ir relative frequency<br />

in PBMC isolates, (2) Assessment <strong>of</strong> T-cell activation as a function <strong>of</strong> cell size shift in PBMC cultures, and (3)<br />

Identification <strong>of</strong> changes in splenocyte population dynamics in a murine model <strong>of</strong> Human Monocytotropic Ehrlichiosis (HME).<br />

Following cell harvest, single cell suspensions were prepared prior to acquisition. All samples<br />

were also stained with cell-specific fluorescent antibodies and analyzed by flow cytometry. In<br />

PBMC, <strong>the</strong> cell counter identified <strong>the</strong> lymphocyte and monocyte fractions as distinctly sized<br />

peaks (7.2 and 10.0µm, respectively). Across <strong>the</strong> 9 samples tested, Counter-derived frequency<br />

and concentration values for each subset were consistent with flow cytometry. In PBMC cultures<br />

co-stimulated with anti-CD3/CD28 antibodies, <strong>the</strong> counter confirmed <strong>the</strong> presence <strong>of</strong> a larger<br />

activated T-cell fraction (7.3 and 12.4 µm, respectively); <strong>the</strong> increase in cell size was coincident<br />

with expression <strong>of</strong> CD25. In <strong>the</strong> murine HME model, a change in <strong>the</strong> relative frequency <strong>of</strong><br />

splenic erythroid (4.1µm) to lymphocyte (6.2µm) fractions was shown to be due to an increase<br />

in <strong>the</strong> Ter119+ erythroid fraction.<br />

The availability <strong>of</strong> simplified methods for monitoring changes in culture dynamics is very useful<br />

for accelerating research in <strong>the</strong> areas <strong>of</strong> diagnostics and drug design. While flow cytometry<br />

provides a more sophisticated and quantitative cell analysis platform, our findings indicate that<br />

cell counting devices can fill this role providing in-line insight into cellular responses.<br />

2357<br />

Effect <strong>of</strong> IL-12 on Negative Selection in <strong>the</strong> TNC microenvironment.<br />

S-A. Miller 1 , S. C. Henley 1 , F. R. Davis 1 , R. H. Lewis 1 , G. C. Bernard 1 , S. Samuels 1 , M. T.<br />

Martinez 1 ; 1 <strong>Biology</strong>, Tuskegee University, Tuskegee, AL<br />

Thymic nurse cells (TNCs) are cortical epi<strong>the</strong>lial cells <strong>of</strong> <strong>the</strong> thymus that appear to be significant<br />

participants in <strong>the</strong> negative selection <strong>of</strong> thymocytes. TNCs express both class I and class II<br />

major histocompatibility (MHC) proteins on <strong>the</strong>ir cell surfaces. TNCs internalize thymocytes that<br />

are CD4 + CD8 + TCR lo (triple positive) into cytoplasmic vacuoles. These thymocytes are at <strong>the</strong><br />

developmental stage where <strong>the</strong>y undergo MHC restriction. In addition to thymocytes, TNC<br />

vacuoles also contain macrophages that interact intimately with <strong>the</strong> thymocyte subset. Greater<br />

than 95% <strong>of</strong> <strong>the</strong> TNC-interactive thymocyte population has been reported to undergo apoptosis<br />

within TNC vacuoles suggesting a strong role for TNCs in <strong>the</strong> negative selection <strong>of</strong> thymocytes.<br />

Triple positive thymocytes are reported to be non-responsive to a variety <strong>of</strong> cytokines including<br />

IL-6, IL-7, IL-10, IL-15 and IFN-γ. However, <strong>the</strong>se thymocytes were found to be responsive to<br />

IL-12. Although IL-12 promotes pro-inflammatory responses in <strong>the</strong> periphery it was shown to<br />

significantly influence <strong>the</strong> deletion <strong>of</strong> <strong>the</strong> triple positive thymocyte subset in <strong>the</strong> thymus. We<br />

<strong>the</strong>refore hypo<strong>the</strong>sized that within <strong>the</strong> TNC microenvironment IL-12 can safely facilitate negative<br />

selection. We used cOVA-TCR transgenic (Tg), D011.10 mice that recognize <strong>the</strong> cOVA 323-339<br />

peptide to analyze negative selection within <strong>the</strong> TNC microenvironment. The Tg mice were<br />

injected with different combinations <strong>of</strong> anti-IL-12 antibody or rat IgG2a antibody and/or cOVA<br />

peptide over a four day period. TNCs and thymocyte populations were harvested and analyzed<br />

for apoptotis using Annexin V with flow cytometry and TUNEL with fluorescent microscopy.<br />

Unexpectedly, <strong>the</strong>re were no observable differences in thymocyte apoptosis within <strong>the</strong> TNCs <strong>of</strong><br />

mice injected with rat-IgG2a, anti-IL-12, or those that were not injected. These data suggest that<br />

IL-12 does not play a role in negative selection within <strong>the</strong> TNC microenvironment.


TUESDAY<br />

2358<br />

Overexpression <strong>of</strong> human alpha-1 antitrypsin (AAT) in PiZZ liver reduces <strong>the</strong><br />

polymerization and facilitates secretion in vitro and in vivo.<br />

K. Xiao 1,2 , L. Wang 1 , Y. Lu 3 , R. Oshins 1 , R. L. Bridges 1 , E. J. McAndrew 1 , A. Huegel 1 , S. Song 3 ,<br />

A. D. Fu 4 , C. Liu 4 , F. Rouhani 1 , M. L. Brantly 1 ; 1 Department <strong>of</strong> Medicine, University <strong>of</strong> Florida,<br />

Gainesville, FL, 2 Genetics & Genomics Program, University <strong>of</strong> Florida, Gainesville, FL,<br />

3 Departments <strong>of</strong> Pharmaceutics, University <strong>of</strong> Florida, Gainsville, FL, 4 Dept <strong>of</strong> Pathology,<br />

University <strong>of</strong> Florida, Gainsville, FL<br />

The Alpha-1 Antitrypsin Deficiency (AATD) is one <strong>of</strong> <strong>the</strong> most prevalent inherited lung diseases<br />

next to <strong>the</strong> cystic fibrosis and also most common metabolic-genetic indication for pediatric liver<br />

transplantation. However, in heterozygote PiMZ patient, <strong>the</strong> liver damage is mild. To evaluate<br />

<strong>the</strong> effect <strong>of</strong> human wild (M) type AAT (MAAT) as a possible chaperone on <strong>the</strong> trafficking <strong>of</strong><br />

secretion-incompetent PiZZ AAT protein (ZAAT), we established a PiZZ cell model from<br />

pediatric patient. Over-expression <strong>of</strong> human MAAT in this cell model prevents process <strong>of</strong><br />

polymerization <strong>of</strong> ZAAT in <strong>the</strong> cytoplasma. The result was confirmed by AAT Western blot,<br />

immunostaining and electron microscope (EM). We fur<strong>the</strong>r test this treatment on PiZ transgenic<br />

mouse through portal vein injection <strong>of</strong> 1 X 10 11 vg <strong>of</strong> AAV8 that expressing MAAT. The<br />

characteristic accumulation <strong>of</strong> ZAAT as polymer in hepatocyte has decrease sharply by polymer<br />

specific Ab staining and ELISA. Fur<strong>the</strong>rmore, <strong>the</strong> secretion <strong>of</strong> ZAAT in <strong>the</strong> serum increases<br />

about 5-fold after 12 weeks <strong>of</strong> treatment. The secretion <strong>of</strong> ZAAT is dosage dependent on <strong>the</strong><br />

MAAT secretion level. Secretion <strong>of</strong> one molecular <strong>of</strong> MAAT brings out approximately one<br />

molecular <strong>of</strong> ZAAT. The purified MAAT can also successfully block <strong>the</strong> formation <strong>of</strong> polymer <strong>of</strong><br />

ZAAT in vitro which explains <strong>the</strong> mechanism <strong>of</strong> this treatment. The average serum SGOT level,<br />

which reflects liver function, decreased about 21% after <strong>the</strong> treatment. Our result may be fur<strong>the</strong>r<br />

tested on clinical trial in patients with AAT liver diseases.<br />

2359<br />

Identification <strong>of</strong> Serum miRNAs as Potential Biomarkers for Acute Myocardial Infarction<br />

P-H. Chu 1 , H-C. Chen 2 , S-J. Chen 2 ; 1 Chang Gung Memorial Hospital, Taipei, Taiwan, 2 Chang<br />

Gung Univeristy, Gueishan, Taoyuan<br />

It is important to find specific biomarkers to understand <strong>the</strong> pathophysiology and identify<br />

patients with ST-segment elevation myocardial infarction (STEMI). Recent studies have<br />

revealed <strong>the</strong> role <strong>of</strong> microRNAs (miRNAs) in a variety <strong>of</strong> biological and pathological processes<br />

including acute myocardial infarction (AMI). We hypo<strong>the</strong>sized that circulating serum miRNAs<br />

may be useful in <strong>the</strong> diagnosis <strong>of</strong> STEMI. The aim <strong>of</strong> this study was to establish <strong>the</strong> ability to<br />

detect <strong>the</strong> miRNAs, and <strong>the</strong>n to elucidate <strong>the</strong> relationship between miRNAs and STEMI. We<br />

initially pr<strong>of</strong>iled <strong>the</strong> differential expressions <strong>of</strong> 270 serum miRNAs, and identified potential serum<br />

microRNAs from 8 STEMI patients and matched healthy controls. We <strong>the</strong>n used <strong>the</strong> candidate<br />

miRNAs to evaluate 62 patients, including 31 STEMI and 31 normal patients. The t-test and<br />

receiver operating characteristic (ROC) curves were established to discriminate AMI patients<br />

from <strong>the</strong> normal patients. P values less than 0.05 were defined as statistically significant. We<br />

initially identified 12 up-regulated miRNAs and 13 down-regulated miRNAs showing at least a<br />

two-fold change. Serum miR-486-3p, miR-150*, miR-126, miR-26a and miR-191 were validated.<br />

ROC analysis using <strong>the</strong> expression ratio <strong>of</strong> miR-486-3p and miR-191 showed an area under <strong>the</strong><br />

curve <strong>of</strong> 0.863. Our findings implicate that serum miRNAs may be used as potential diagnostic<br />

biomarkers for STEMI.


TUESDAY<br />

2360<br />

Therapeutic Potential <strong>of</strong> TGF-β2 -Treated Mesenchymal Stem Cells from Wharton′s Jelly<br />

in a Rat Myocardial Infarction Model.<br />

H-S. Wang 1 , P-C. Chu 1 ; 1 Anatomy, Natl Yang-Ming University, Taipei, Taiwan<br />

Myocardial infarction (MI) is one <strong>of</strong> <strong>the</strong> leading causes <strong>of</strong> morbidity and mortality worldwide. The<br />

development <strong>of</strong> stem cell <strong>the</strong>rapy for treating MI patients is a contemporary challenge in <strong>the</strong> field<br />

<strong>of</strong> cardiovascular medicine. In this study, left anterior descending artery ligation rats were used<br />

as a myocardial infarction model. The induction <strong>of</strong> left anterior descending artery ligation was<br />

confirmed by electrocardiogram, echocardiography, and <strong>the</strong> presence <strong>of</strong> c-troponin I in <strong>the</strong><br />

serum. To investigate <strong>the</strong> improved myocardial function after transplantation <strong>of</strong> undifferentiated<br />

and TGF-β2-treated cells, electrocardiogram, echocardiogram, Masson′s Trichrome staining,<br />

and immunohistochemical staining <strong>of</strong> heart tissues at different time points were assessed in <strong>the</strong><br />

infracted rat hearts. Our results indicated that transplantation <strong>of</strong> undifferentiated or TGF-β2treated<br />

human umbilical cord mesenchymal stem cells from Wharton′s jelly both improve left<br />

ventricular systolic function, reduce <strong>the</strong> fibrotic area <strong>of</strong> heart, and increase <strong>the</strong> survival rate <strong>of</strong> MI<br />

rat model. These results suggested that deliver stem cells from human umbilical cord<br />

mesenchymal stem cells to sites <strong>of</strong> injury are able to ameliorate <strong>the</strong> dysfunctional heart failure.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!