11.07.2015 Views

imcb symposium 2008 - Institute of Molecular and Cell Biology - A*Star

imcb symposium 2008 - Institute of Molecular and Cell Biology - A*Star

imcb symposium 2008 - Institute of Molecular and Cell Biology - A*Star

SHOW MORE
SHOW LESS

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

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

IMCBSYMPOSIUM<strong>2008</strong>Wednesday, August 13 th <strong>2008</strong>Breakthrough Theatrette,Matrix Building1


CONTENTSProgramme page 3Oral Presentations: Abstracts page 4Poster Presentations: List <strong>of</strong> Abstracts page 12Poster Presentations: Abstracts page 182


ProgrammeWednesday 13 th August, <strong>2008</strong>09:30Welcome Address by Pr<strong>of</strong>essor Neal Copel<strong>and</strong>;Keynote Address: “Unfinished Genomes” by Pr<strong>of</strong>essor SydneyBrenner10:20Wip1 phosphatase modulates dendritic morphology <strong>and</strong> learning<strong>and</strong> memory processes in hippocampus: Francesca Fern<strong>and</strong>ez,XZC lab, GDD Division.10:50 C<strong>of</strong>fee Break <strong>and</strong> 1st Poster Session11:4012:10MRCK-LRAP35a-Myosin 18A Complex Regulates LamellarActomyosin Flow Essential for <strong>Cell</strong> Migration: Ivan Tan, GSKlab, GDD Division.An intronic Runx1 enhancer marks hematopoietic stem cells:Cherry Ng, RUNX lab, GGD Division.12:40 Lunch14:0014:3015:00Pin1 plays a role in zebrafish neuromasts development bystabilizing NeuroD: Steven Fong, VK lab, CDCBD Division.c-Cbl’s polygamy explained: Jan P. Buschdorf, GG lab, GGDDivisionProbing the genetic networks that regulate the Golgi apparatus:Susanne Ng, FB lab, SBD Division15:30 C<strong>of</strong>fee Break <strong>and</strong> 2 nd Poster Session16:3017:0017:30Aph2, a palmitoyl acyltransferase for phospholamban, is anessential regulator <strong>of</strong> cardiac physiological function <strong>and</strong> theonset <strong>of</strong> heart failure: Tielin Zhou, LBJ lab, CDCBD DivisionRegulation <strong>of</strong> p120ctn in E-cadherin-mediated intercellularadhesion strengthening: Yeh-Shiu Chu, JPT lab, SBD DivisionHappy Hour (sponsored by Invitrogen), including Prize AwardCeremony19:00 Thank You for Coming!!3


ABSTRACTS – ORAL PRESENTATIONSWip1 phosphatase modulates dendritic morphology <strong>and</strong> learning <strong>and</strong> memoryprocesses in hippocampusF. Fern<strong>and</strong>ez, Q.H. Ma, S.H. Wei, L. Zeng, A.I. Soon, S.M. Wong, O. Demidov, G.Dawe, D. Bulavin , <strong>and</strong> Z.C. Xiao.Dendritic spines form the postsynaptic compartment <strong>of</strong> most excitatory synapses in thebrain, undergoing morphological changes in response to neuronal activity. Thiscorrelation between dendritic morphology <strong>and</strong> synaptic plasticity may contribute tolearning <strong>and</strong> memory, but the mechanisms underlying this association remain unclear.The polymerization <strong>and</strong>/or depolymerization <strong>of</strong> actin filament plays a key role in thespine motility, growth <strong>and</strong> shape. Several Protein Kinases (including mitogen-activatedprotein kinase (MAPK)) <strong>and</strong> protein phosphatases (PP) such as PP1 families, present inthe post synaptic compartment, are essential in the regulation <strong>of</strong> actin filament <strong>and</strong>/ordendritic morphology, <strong>and</strong> also in the synaptic plasticity. The PP wip1, encoded byoncogene ppm1d, participates to the regulation <strong>of</strong> the protein F-actin. In this study, weinvestigated the role <strong>of</strong> wip1 in dendritic morphology within the CA1 hippocampalregion <strong>and</strong> its effect on the synaptic plasticity <strong>and</strong> learning memory, via behaviouraltesting on mice deficient <strong>of</strong> wip1 gene. We report a colocation <strong>of</strong> wip1 <strong>and</strong> p38 MAPK,main effectors <strong>of</strong> wip1 protein, at neuronal dendrites. Deficiency <strong>of</strong> wip1 leads to adecrease <strong>of</strong> dendritic spine size <strong>and</strong> <strong>of</strong> the dendrite density in pyramidal neurons. Thesemorphological dendritic changes are accompanied by an impairment <strong>of</strong> memory in wip1deficient mice <strong>and</strong> by a reduction <strong>of</strong> the Long-Term Potentiation measured in the CA1hippocampal area. We also demonstrated that p38MAPK may contribute to modification<strong>of</strong> the synaptic activity observed in the CA1 hippocampal region <strong>of</strong> wip1 deficient mice,but not to the morphological dendritic changes.4


MRCK-LRAP35a-Myosin 18A Complex Regulates Lamellar Actomyosin FlowEssential for <strong>Cell</strong> MigrationIvan Tan, Jeffery Yong, Jing Ming Dong, Louis Lim <strong>and</strong> Thomas Leung(GSK-IMCB Group)<strong>Cell</strong> migration is important in both development <strong>and</strong> diseases. It’s a cyclical processinvolving membrane protrusion at leading edge, movement <strong>of</strong> cell body <strong>and</strong> retraction <strong>of</strong>cell rear. Actomyosin retrograde flow underlies the contraction essential for cell motility.Both lamellipodial <strong>and</strong> lamellar retrograde flows are essential for membrane protrusion<strong>and</strong> generation <strong>of</strong> contractile force by coupling to cell adhesion. We recently found thatRac/Cdc42-binding kinase MRCKa/b, a novel adaptor LRAP35a <strong>and</strong> myosin II-relatedMYO18A form a functional tripartite complex that is specifically colocalized with <strong>and</strong>regulates the lamellar retrograde flow required for persistent cell protrusion <strong>and</strong> asubnuclear actomyosin network. LRAP35a binds independently to MYO18A <strong>and</strong> MRCK;the binding leads to MRCK activation <strong>and</strong> its phosphorylation <strong>of</strong> MYO18A-associatedregulatory myosin light chain on Serine-19, independent <strong>of</strong> ROK <strong>and</strong> MLCK. Live cellimaging showed that MRCK-complex moves in concert with the retrograde actomyosinflow, allowing it to regulate the flow continuously. In addition, MRCK-complex wasfound to be functionally linked to specific MYO2A-network known to be responsible forlamellar actomyosin flow <strong>and</strong> force generation. The promotion <strong>of</strong> persistent protrusiveactivity <strong>and</strong> inhibition <strong>of</strong> cell motility by the respective expression <strong>of</strong> wild type <strong>and</strong>dominant negative mutant components <strong>of</strong> the MRCK-complex show it to be crucial tocell protrusion <strong>and</strong> migration. Taken together, our findings point to specific roles <strong>of</strong>MRCK-complex in actomyosin regulation, <strong>and</strong> they provide new insights on theregulatory mechanisms underlying the coordination <strong>of</strong> cellular actomyosin in cellmigration.5


An intronic Runx1 enhancer marks hematopoietic stem cellsCherry Ee Lin Ng, Namiko Yamashita, Giselle Nah, Bindya Jacob, Chelsia Qiuxia Wang,Hao Jin, Zilong Wen, Motomi Osato, Yoshiaki Ito<strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>, Singapore, 138673, Oncology Research <strong>Institute</strong>,National University <strong>of</strong> Singapore, Singapore 117597RUNX1 is a transcription factor critical for generation <strong>and</strong> maintenance <strong>of</strong> hematopoieticstem cells (HSC) <strong>and</strong> is frequently mutated in human leukemias. Germline RUNX1mutations cause familial leukemia, named FPD/AML. Despite such widespreadinvolvement in diseases, the precise transcriptional regulation <strong>of</strong> Runx1 remains largelyunknown due to the extremely large size (approximately 1.0 Mb) <strong>of</strong> its gene locus. Wefirst took a comparative genomics approach <strong>and</strong> identified 93 conserved non-codingelements (CNE). In addition, we utilised retroviral integration sites (RIS) information asRIS are potential indicators <strong>of</strong> active chromatin region. Using this novel approach wehighlighted 10 CNE. These CNE were individually fused with EGFP <strong>and</strong> injected intozebrafish embryos to assess their function in vivo. We identified a mouse CNE (+24.1mCNE) which drives GFP expression in the intermediate cell mass (ICM) region, whichis a known site <strong>of</strong> endogenous Runx1 expression <strong>and</strong> hematopoiesis in zebrafish.Quantitative chromatin-immunoprecipitation (qChIP), using antibodies against modifiedhistones which mark either open or close regions <strong>of</strong> chromatin, showed a strongcorrelation between state <strong>of</strong> chromatin structure at the +24.1 mCNE locus <strong>and</strong> expression<strong>of</strong> Runx1. To further characterize this element, +24.1 mCNE-GFP transgenic mouse lineswere generated. Analyses <strong>of</strong> mouse embryos revealed that this CNE is active in emergingHSC. In adult bone marrow, +24.1 mCNE-driven GFP+ cells were highly enriched forHSC. These results support the role <strong>of</strong> +24.1 mCNE as a HSC enhancer. +24.1 mCNE-GFP transgenic mice hold the promise to be a powerful tool for research on stem cell <strong>and</strong>cancer biology.Reference:Ng CEL, Osato M, Ito Y. Retroviral integration sites (RIS) mark cis-regulatoryelements. Critical Reviews in Hematology/Oncology, in press.6


Pin1 plays a role in zebrafish neuromasts development by stabilizing NeuroDSteven Fong 1 , Li-Qun Chao 2 , Vladimir Korzh 1 , Yi-Cherng Liou 21. <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>, Singapore.2. Department <strong>of</strong> Biological Sciences, National University <strong>of</strong> Singapore, Singapore.The vertebrate pSer/pThr-Pro specific peptidyl-prolyl-isomerase Pin1 has been shown toplay important roles in cell cycle regulation, apoptosis, oncogenesis, <strong>and</strong> neuronaldegeneration. However, its role in neuronal development is not clear. We clonedzebrafish Pin1 <strong>and</strong> investigated its interaction with bHLH factors involved in neuronalspecification. Pin1 is expressed maternally <strong>and</strong> in a ubiquitous manner early indevelopment, but by 48 hpf it is restricted to the brain <strong>and</strong> neuromasts. Coimmunoprecipitationassays (CoIP) in cell lines showed that Pin1 interacts with neuroDbut not ngn1 <strong>and</strong> atoh1a. This binding was reduced when Ser/Thr phosphorylation siteswere mutated. Antisense morpholino oligonucleotide (MO) knockdown <strong>of</strong> Pin1 led todelay in development. Accounting for the delay, neuroD expression was significantlydiminished in the hindbrain <strong>of</strong> morphants by 48 hpf equivalent. Morphants in thebackground <strong>of</strong> Tol2/GFP enhancer trap lines with specific expressions in hair cells (ET4)<strong>and</strong> mantle cells (ET20) also displayed defects in neuromasts formation. It has beenshown that specification <strong>of</strong> hair cells in neuromasts is neuroD dependent but ngn1independent. Using siRNA Pin1 knockdown 293T cells <strong>and</strong> Pin1 knockout MEF cells,we showed that neuroD protein is degraded more rapidly in the absence <strong>of</strong> Pin1. NeuroDstability was restored when Pin1 is over-expressed. This is the first study to demonstratethe functional regulation <strong>of</strong> a bHLH factor by post-phosphorylation cis-transisomerization in neuronal specification. In view <strong>of</strong> the role <strong>of</strong> Pin1 in neurodegenativediseases, our results may have important pharmaco-therapeutic applications.7


c-Cbl’s polygamy explainedCherlyn Ng, Rebecca A. Jackson, Jan P. Buschdorf, Qingxiang Sun, J. Sivaraman <strong>and</strong>Graeme R. GuyThe proto-oncogene c-Cbl inhibits several signaling proteins involved in cell growth <strong>and</strong>the immune response. c-Cbl keeps in check, via ubiquitination, a number <strong>of</strong> tyrosinekinases implicated in cancer, including the epidermal growth factor receptor, the c-Metreceptor <strong>and</strong> c-Src. This requires binding <strong>of</strong> c-Cbl’s ‘tyrosine kinase-binding domain’(TKB), essentially a derivative SH2 (src homology 2) domain, to specific sequencesaround a phosphotyrosine residue. c-Cbl is unique among the other generally motifspecificphosphotyrosine binding domains in recognizing three unrelated motifs; how thishappens at the molecular level has been enigmatic. To underst<strong>and</strong> the structural basis <strong>of</strong>this feat, we obtained X-ray crystallography structures <strong>of</strong> c-Cbl bound to five targetpeptide fragments. While the target phosphotyrosine forms several H-bonds with theTKB, only an H-bond from the phosphotyrosine to another residue within the target isconserved. The importance <strong>of</strong> this H-bond is demonstrated by mutagenesis <strong>and</strong> byconverting VEGFR1, a receptor not recognized by c-Cbl, into a c-Cbltarget. Surprisingly, one target, c-Met, binds to c-Cbl in the reverse orientation. Thestudy explains how c-Cbl manages to regulate such a diverse set <strong>of</strong> targets.8


Probing the genetic networks that regulate the Golgi apparatusSusanne San San Ng, Victor Racine <strong>and</strong> Frederic BardThe Golgi apparatus in mammalian cells is a dynamic <strong>and</strong> complex organelle regulatedby multiple fluxes <strong>of</strong> membranes <strong>and</strong> material. The molecular pathways that orchestratethis complex organization are still poorly understood. We have used a functionalgenomics approach, combining high-throughput RNA interference (RNAi) screeningwith automated microscopy to characterize the genes involved in regulation <strong>of</strong> the Golgistructure. Hela cells were stained for three different Golgi markers specific for the cis,medial <strong>and</strong> trans cisternae, <strong>and</strong> a nuclear dye. Automated image analysis extractsnumerical features from the images. To explore the range <strong>of</strong> phenotypes that could beobtained after RNAi, we tested a set <strong>of</strong> known regulators <strong>of</strong> membrane trafficking. Wefound that Golgi phenotypes can be used to reconstruct functional pathways. For exampletargeting genes implicated in ER to Golgi traffic yields a specific dispersion <strong>of</strong> the cisGolgi marker. By contrast, the trans Golgi marker is specifically affected when Golgi toplasma membrane traffic genes are targeted. A preliminary systematic screen coveringthe human kinome reveals several classes <strong>of</strong> Golgi regulating kinases. Some kinasephenotypes suggest a regulation <strong>of</strong> ER to Golgi traffic while other kinases appear to act atthe trans side <strong>of</strong> the Golgi. Overall, our preliminary results indicate that up to 70 kinasesare involved in regulating various aspects <strong>of</strong> Golgi organization. This figure is muchlarger than expected <strong>and</strong> suggest that a significant number <strong>of</strong> unknown regulatorynetworks impinge on the secretory pathway <strong>and</strong> on the Golgi apparatus.9


Aph2, a palmitoyl acyltransferase for phospholamban, is an essential regulator <strong>of</strong>cardiac physiological function <strong>and</strong> the onset <strong>of</strong> heart failureTielin Zhou (LBJ lab)Reversible protein palmitoylation regulates many aspects <strong>of</strong> cell function <strong>and</strong> is largelycarried out by zf-DHHC containing proteins. However, the in vivo physiological function<strong>of</strong> protein palmitoylation is largely unknown. To address this question, we generatedAph2-/-(zf-DHHC16) mice, revealing an essential role for Aph2 in postnatal survival <strong>and</strong>heart development, evidenced by histological/electrophysiological defects <strong>and</strong> neonatalonset <strong>of</strong> cardiomyopathy <strong>and</strong> heart failure. Aph2 is shown to be a palmitoyl acyltransferase for phospholamban (PLN), where palmitoylation on cysteine 36 promotesPLN nucleus envelope localization, its interaction with PKA, <strong>and</strong> subsequent serine 16phosphorylation. Aph2 deficiency led to PLN hypophosphorylation, defects in PLNpentamer formation <strong>and</strong> Ca2+ homeostasis. Deletion <strong>of</strong> PLN in Aph2-/- mice rescuedseveral <strong>of</strong> the heart phenotypes.These findings establish that Aph2 is a critical in vivo regulator <strong>of</strong> heart contractilefunction, partially via the modification <strong>of</strong> PLN, <strong>and</strong> that Aph2-/- mice represent acongenital heart disease model in which one member <strong>of</strong> PATs is proven to be involved incardiomyopathy formation, thereby identifying a novel therapeutic target forcardiomyopathy <strong>and</strong> heart failure.10


Regulation <strong>of</strong> p120ctn in E-cadherin-mediated intercellular adhesion strengtheningYeh-Shiu Chu, Pierre Nassoy, Alex<strong>and</strong>er Bershadsky, Sylvie Dufour, Jean-Paul Thieryp120ctn is a member <strong>of</strong> the catenin protein family that controls a wide range <strong>of</strong> cellularprocesses, including cell adhesion, intracellular signalling, <strong>and</strong> transcriptional regulation.p120ctn associates with the juxta-membrane domain <strong>of</strong> E-cadherin via armadillo repeats.The armadillo repeats constitute a core region which indirectly associates with Rho smallGTPases, such as Rac <strong>and</strong> Cdc42. Although the key roles <strong>of</strong> p120ctn as a regulator <strong>of</strong>cadherin transport from the Golgi complex to the plasma membrane <strong>and</strong> for cadherinstabilization at the plasma membrane are well-established, the contribution <strong>of</strong> p120ctn tocadherin-mediated adhesion remains to be investigated.We show here that the strengthening <strong>of</strong> E-cadherin-mediated cell adhesion is abrogatedby transient expression <strong>of</strong> an E-cadherin mutant lacking the p120ctn binding site insarcoma S180 cells. In addition, TIRF analysis <strong>of</strong> the spreading on EcadFC-coatedsurfaces <strong>of</strong> cells expressing GFP-tagged cadherins indicates that uncoupling <strong>of</strong> p120ctnfrom cadherin prevents cadherin adhesions <strong>and</strong> induces increasing lamellipodia formation.The treatment <strong>of</strong> cells with the Rac inhibitor NSC23766 partially rescues formation <strong>of</strong>cadherin adhesions. p120 ctn knockdown experiments shows that reducing p120 ctn incells also weakens E-cadherin-mediated adhesion strengthening. Our data highlight thefact that the association <strong>of</strong> p120ctn with E-cadherin is important to strengthen thedevelopment <strong>of</strong> adhesion, possibly by actin cytoskeletal reorganization11


ABSTRACTS CONTENTS – POSTER PRESENTATIONSPosterS/NPresenter Group Title1 Choong‐Tat Keng CAVR2 Ning Sheng Liu HWJ3 Honghui Huang PJR4 Sharon Ling SHW5 Portia G. Loh SHW6 William Go VK7 Igor Kondrychyn VK8 Kar Lai Poon VKGeneration <strong>of</strong> SARS coronavirus escapemutants by using monoclonal antibodiesthat block membrane fusionTom1L1 as a regulated adaptor for EGFstimulatedendocytosis <strong>of</strong> EGF receptorMypt1‐Mediated Spatial Positioning <strong>of</strong>Bmp2‐Producing <strong>Cell</strong>s Is Essential forLiver OrganogenesisCrystal structure <strong>of</strong> human Edc3 <strong>and</strong> itsfunctional implicationsStructural basis <strong>of</strong> translation inhibitionby PDCD4Plasma membrane Ca2+‐ATPase 1 isrequired for lateral line development inzebrafishDevelopmental analysis <strong>of</strong> the novelzebrafish gene, zic6.Analysis <strong>of</strong> zebrafish inducible nitricoxide synthase genes duringembryogenesis9 Terk Shin Teng CXM Cytoplasmic role <strong>of</strong> Stat3 in cell migration10 Alison P Lee FUGE/SB11 Samantha Quah ZQIdentification <strong>and</strong> characterization <strong>of</strong>conserved cis‐regulatory elementsassociated with transcription‐factorencoding genes in the human genomePRL‐3 Initiates Tumor Angiogenesis byRecruiting Endothelial <strong>Cell</strong>s In vitro <strong>and</strong>In vivo12 Khay Chun Ang BR Structure <strong>of</strong> Actin in the Nucleus13 Shalini Nag BR Nuances <strong>of</strong> Gelsolin Function12


14 Hai Yun See DPLDesigning a synthetic tumour suppressorprotein to validate eIF4E as a potentialanti‐cancer target.15Joanne Zhi HuiChiaFBSNARE genes expression regulation uponvariation <strong>of</strong> secretory cargo load16 Fa‐Xing Yu LY17 Mei Chin Lee LY18 Jun Yan LY19 Jenn Hui Khong US20 Keng Boon Wee USAdenosine‐containing Molecules AmplifyGlucose Signaling <strong>and</strong> Enhance TxnipExpressionDissecting a Drosophila melanogasterHistone Transcriptional RegulationPathwayNm23 <strong>and</strong> Histone H2B gene expressionin HeLa cellsA Mechanism that prevents prematurechromosome segregation when initiation<strong>of</strong> DNA replication failsRoles <strong>of</strong> intracellular p53 oscillations incell survival <strong>and</strong> death21 Kelly Hogue WB Mass Spectrometry in IMCB22 Jasmine Lee ZLH23 Chiaw‐Hwee Lim JYJ24 Ashish Maurya PWI25 Hoe Peng Liew SR26 Hao Zou WYNon‐ribosomal peptides in Pseudomonasaeruginosa quorum sensing regulationLoss <strong>of</strong> udu function causes activation <strong>of</strong>DNA damage checkpointEngrailed gene expression <strong>and</strong> its controlby hedgehog signalingSpecification <strong>of</strong> vertebrate slow‐twitchmuscle fiber fate by the transcriptionalregulator Blimp1G‐actin binds to the adenylyl cyclase Cyr1through Cap1 <strong>and</strong> regulates cAMPsynthesis in C. albicans hyphalmorphogenesis13


27 Yun‐Hua Zhu XZC28 Lihui Goh YXH29 Zhenxing Huang YXHWip1 regulates neural stemcell/progenitor proliferation through p53dependent G2 cell cycle controlMouse embryonic neural stem cells: Amodel for asymmetric cell divisionSearching For Novel Proteins Involved InAsymmetric <strong>Cell</strong> Division30Joan Siew ChingLeeYXHApim genetically interacts withInscuteable in the dividing Drosophilaneuroblast31 Valentina Migliori EG32 Cheng Chun Wang HWJ33 Cathleen Teh VK34 R.N.Mahalakshmi WH35 Shuo Shen CAVR36 Lei Li AGP37 Jenny Chau LBJIdentification <strong>of</strong> novel epigeneticregulatorsVAMP8 Is Required for <strong>Cell</strong> SurfaceDeployment <strong>of</strong> the Water ChannelAquaporin 2Wnt3 is Essential for Dorsal DiencephalicDifferentiationA regulatory role for Kir/Gem <strong>and</strong> thenovel muscle‐specific protein CCDC95 inmyogenesisComparing the antibody responsesagainst recombinant hemagglutininproteins <strong>of</strong> avian influenza A (H5N1)virus expressed in insect cells <strong>and</strong> bacteriaJNK‐dependent activation <strong>of</strong> p53 <strong>and</strong> c‐Jun is required for nitric oxide‐inducedapoptosis <strong>of</strong> tumor cellsSmad1 provides a link between BMP‐Smad signaling <strong>and</strong> DNA damageresponse <strong>and</strong> a target for cancer therapy38 Canhe Chen NNHarnessing piggyBac Transposons forcancer gene discovery in Zebrafish14


39 Bindya Jacob RUNX40 Shreeram Sathya BD41 San San Lee VY42 Permeen Yus<strong>of</strong>f GG43 Lu Hao CXMStem cell exhaustion due to Runx1deficiency is prevented by Evi5 activationin leukemogenesisCdc25A Serine 123 phosphorylationcouples centrosome duplication withDNA replication <strong>and</strong> regulatestumorigenesisTrim39 is a MOAP‐1 binding protein thatstabilizes MOAP‐1 through inhibition <strong>of</strong>its polyubiquitination processA complex affair between two tumoursuppressors <strong>and</strong> a famous regulatorGRIM‐19 Is Essential for Maintenance <strong>of</strong>Mitochondrial Membrane Potential44 Hsiang Ling Teo VT New Controls <strong>of</strong> the NFkB Pathway45 Xinde Zheng PRK46 Shuhei Kotoshiba PRK47 Haihe Wang ZQ48 Rashmi P Kulkarni FUGE/SB49 Chit Fang Cheok DPLCharacterization <strong>of</strong> Speedy/RINGO, anew family <strong>of</strong> cyclin‐like proteinsRb/Cdk2/Cdk4 triple mutant mice elicit anew mechanism for G1/S regulationPRL‐3 Down‐regulates PTEN Expression<strong>and</strong> Signals through PI3K to PromoteEpithelial‐Mesenchymal TransitionEvolutionary changes in the regulatoryregion <strong>of</strong> vertebrate erythropoietin genelocus<strong>Molecular</strong> therapy : Targeting the p53pathway using small molecules50Christopher JBrownDPLCrystallographic analysis <strong>of</strong> cap free <strong>and</strong>cap bound eIF4E with N7 alkylated capderivatives51BalakrishnanKannanBRSpatio‐temporal Mapping <strong>of</strong> ActinPolymerisation Regulated by Actin‐Binding Proteins15


52 Marten E Larsson BRDirected Evolution <strong>of</strong> Actin by In VitroCompartmentalization53JayanthaGunaratneWBTracking cellular events using stableisotopelabeling by amino acids in cellculture (SILAC)54 Ya‐Wen He ZLH55 Dimitri Moreau FB56 Xiao Xing Wang LDX57 Tao Zhang US58 Xin Gang Wang PWI59 Shang‐Wei Chong JYJ60 Xuehui Qiu JYJSynergistic regulation <strong>of</strong> Xanthomonascampestris virulence <strong>and</strong> adaptation byDSF signaling system <strong>and</strong> a novel twocomponentsignal transduction systemRavS/RavRProbing the retrograde transport pathwayby RNA interference screeningInhibition <strong>of</strong> PKR Activation <strong>and</strong> Upregulation<strong>of</strong> GADD34 Expression play asynergistic role in the maintenance <strong>of</strong> denovo Protein Synthesis in Coronavirus‐Infected <strong>Cell</strong>sDNA damage checkpoint maintains Cdh1in quasi‐active state to prevent spindleelongation <strong>and</strong> the completion <strong>of</strong>AnaphaseSearching for novel factors involved inneutrophil chemotaxis by chemicalgeneticsZebrafish nicastrin is required formidbrain <strong>and</strong> hindbrain developmentTemporal Notch activation throughNotch1a <strong>and</strong> Notch3 is required formaintaining zebrafish rhombomereboundaries61 Ka Fai William Tse JYJ The Characterization <strong>of</strong> Novel mib Alleles62 Semil P. Choksi SRIdentifying gene regulatory networks thatcontrol zebrafish regeneration16


63 Xianwen Yu SR64 Yijun Yin YXH65 Chang Run Li WYThe Forkhead domain containingtranscription factor Foxj1 is the masterregulator <strong>of</strong> the motile ciliogenic programA mouse embryonic stem/progenitor cellsas a model to investigate asymmetric celldivisionCDK regulates cytokinesis byphosphorylation <strong>of</strong> an IQGAP familyprotein in fungal pathogen C<strong>and</strong>idaalbicans66YohendranBaskaranGSKRegulation <strong>of</strong> PAK4, a non‐conventionalPAK17


ABSTRACTS – POSTER PRESENTATIONS(1) Generation <strong>of</strong> SARS coronavirus escape mutants by using monoclonal antibodiesthat block membrane fusionKeng Choong-Tat (CAVR lab)We have previously shown that a bacterially expressed, denatured spike (S) proteinfragment <strong>of</strong> the severe acute respiratory syndrome (SARS) coronavirus, containingresidues 1029 to 1192, which include the heptad repeat 2 (HR2) domain, could induceneutralizing polyclonal antibodies (Keng et. al., 2005. J. Virol. 79:3289-3296). We havealso produced monoclonal antibodies (MAbs) against this fragment to identify the linearneutralizing epitopes in this functional domain (Lip et al., 2006. J. Virol. 80:941-950).Eighteen hybridomas secreting the S protein-specific MAbs were obtained. Binding sites<strong>of</strong> these MAbs were mapped to 4 linear epitopes. Two <strong>of</strong> them were located within theHR2 region <strong>and</strong> two immediately upstream <strong>of</strong> the HR2 domain. MAbs targeting to theseepitopes show in vitro neutralizing activities <strong>and</strong> can inhibit cell-cell membrane fusion.These results provide evidence <strong>of</strong> novel neutralizing epitopes that are located in the HR2domain <strong>and</strong> the spacer region immediately upstream <strong>of</strong> it. Using these MAbs, wegenerated SARS coronavirus escape mutants by growing the virus at an antibodyconcentration that reduces the virus concentration by 4 logs. After several passages, wemanaged to isolate viruses that are no longer neutralized by the MAbs. The spike protein<strong>of</strong> the escaped viruses was sequenced to identify crucial mutations that enable the virus toescape the inhibition by the MAbs. How such mutations affect the entry ability <strong>of</strong> theSARS coronavirus will be further investigated using a pseudotyped virus system. Thedynamics <strong>of</strong> the growth <strong>and</strong> viability <strong>of</strong> the escape viruses will also be studied later.18


(2) Tom1L1 as a regulated adaptor for EGF-stimulated endocytosis <strong>of</strong> EGF receptorNing Sheng Liu (HWJ lab)The molecular mechanism governing lig<strong>and</strong>-stimulated endocytosis <strong>of</strong> receptor tyrosinekinases remains elusive. I show here that EGF stimulates transient tyrosinephosphorylation<strong>of</strong> Tom1L1(Tom-Like 1) by the Src family kinases, resulting in itstransient interaction with the activated EGF receptor (EGFR) bridged by the receptorboundGrb2. Cytosolic Tom1L1 is recruited onto the plasma membrane <strong>and</strong> subsequentlyredistributes with EGFR into the early endosome. Mutant forms <strong>of</strong> Tom1L1 defective intyrosine-phosphorylation or interaction with Grb2 is incapable <strong>of</strong> interaction with EGFR<strong>and</strong> inhibits endocytosis <strong>of</strong> EGFR. In addition, siRNA-mediated knockdown <strong>of</strong> Tom1L1inhibits endocytosis <strong>of</strong> EGFR. The C-terminal tail <strong>of</strong> Tom1L1 contains a novel clathrininteractingmotif, which is important for exogenous Tom1L1 to rescue endocytosis <strong>of</strong>EGFR in Tom1L1 knocked-down cells. These results suggest that EGF triggers atransient association <strong>of</strong> EGFR with Tom1L1 to engage the endocytic machinery forendocytosis <strong>of</strong> the lig<strong>and</strong>-receptor complex. Moreover, Tom1L1 interacts with ubiquitin<strong>and</strong> ESCRT family proteins, such as: Hrs, TSG101, STAM1/2, <strong>and</strong> it is recruited toendosome upon over-expression HA-Hrs. These results suggest that Tom1L1 couldparticipate in the machinery for EGFR sorting <strong>and</strong> degradation. In addition, Tom1L1negatively regulates Ras activation upon EGF stimulation <strong>and</strong> A431 colony formation,which indicate that it may play a negative role in Src kinase signaling.19


(3) Mypt1-Mediated Spatial Positioning <strong>of</strong> Bmp2-Producing <strong>Cell</strong>s Is Essential forLiver OrganogenesisHonghui Huang 1,5 , Hua Ruan 1,5,7 , Meng Yuan Aw 1,7 , Alamgir Hussain 1,7 , Lin Guo 1,5,7 ,Chuan Gao 5 , Feng Qian 1 , Changqing Chang 1 , Thomas Leung 2 , Haiwei Song 3 , DavidKimelman 6 , Zilong Wen 4* <strong>and</strong> Jinrong Peng 1*1 Laboratory <strong>of</strong> Functional Genomics, 2 Laboratory <strong>of</strong> Neural Differentiation <strong>and</strong>Degeneration, 3 Laboratory <strong>of</strong> Translation Termination <strong>and</strong> Messenger RNA Decay, 4Laboratory <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> Developmental Immunology, <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong><strong>Cell</strong> <strong>Biology</strong>, 61 Biopolis Drive, Proteos, Singapore 138673. 5 Department <strong>of</strong> BiologicalSciences, National University <strong>of</strong> Singapore, Singapore 117543. 6 Department <strong>of</strong>Biochemistry, University <strong>of</strong> Washington, Seattle, WA, 98195-7350, USA7 These authors contributed equally to this work.*To whom correspondence should be addressed. E-mail: pengjr@<strong>imcb</strong>.a-star.edu.sg;zilong@ust.hk;Mesodermal tissues produce various inductive signals essential for morphogenesis <strong>of</strong>endodermal organs. However, little is known about how the spatial relationship betweenthe mesodermal signal-producing cells <strong>and</strong> their target endodermal organs is establishedduring morphogenesis. Here we report that a mutation in the zebrafish myosinphosphatase targeting subunit 1 (mypt1) causes abnormal bundling <strong>of</strong> actin filaments <strong>and</strong>disorganization <strong>of</strong> lateral plate mesoderm (LPM) cells. As a result, the coordinationbetween mesoderm <strong>and</strong> endoderm cell movements is disrupted. Consequently, the twostripes <strong>of</strong> Bmp2a-expressing cells in the LPM fail to align in a V-shaped pockets<strong>and</strong>wiching the liver primordium. Mispositioning Bmp2a producing cells with respect tothe liver primordium leads to a reduction <strong>of</strong> hepatoblast proliferation <strong>and</strong> final abortion <strong>of</strong>hepatoblasts by apoptosis that causes the liverless phenotype. Our results demonstratethat Mypt1 mediates coordination between mesoderm <strong>and</strong> endoderm cell movements inorder to carefully position the liver primordium such that it receives a Bmp signal that isessential for liver formation in zebrafish.20


(4) Crystal structure <strong>of</strong> human Edc3 <strong>and</strong> its functional implicationsSharon Ling (SHW lab)Edc3p is an enhancer <strong>of</strong> decapping <strong>and</strong> serves as a scaffold that aggregates mRNAribonucleoproteins (mRNPs) together for P-body formation. It has a modular domainarchitecture consisting <strong>of</strong> an N-terminal Lsm domain, a central FDF domain <strong>and</strong> a C-terminal YjeF-N domain, which forms a network <strong>of</strong> interactions with the components <strong>of</strong>the mRNA decapping machinery. We have determined the crystal structure <strong>of</strong> the N-terminally truncated human Edc3 at a resolution <strong>of</strong> 2.2Å. The structure reveals that theYjeF-N domain <strong>of</strong> Edc3 possesses a divergent Rossmann fold topology that forms adimer, which is supported by sedimentation velocity <strong>and</strong> sedimentation equilibriumanalysis in solution. The dimerization interface <strong>of</strong> Edc3 is highly conserved in eukaryotesdespite the overall low sequence homology across species. Structure based site-directedmutagenesis revealed dimerization is required for efficient RNA binding, P-bodyformation <strong>and</strong> likely for regulating the yeast Rps28B mRNA as well, suggesting that thedimeric form <strong>of</strong> Edc3 is a structural <strong>and</strong> functional unit in mRNA degradation.21


(5) STRUCTURAL BASIS OF TRANSLATION INHIBITION BY PDCD4Portia G. Loh 1,2 , <strong>and</strong> Haiwei Song 1,21 <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>, 61 Biopolis Drive, Proteos, Singapore 138673,2 Department <strong>of</strong> Biological Sciences, National University <strong>of</strong> Singapore, Lower Kent RidgeRoad, Singapore 117543Pdcd4 is a tumor suppressor protein thought to be an inhibitor <strong>of</strong> tumor progression <strong>and</strong>promoter <strong>of</strong> apoptosis. The mechanism by which it does so involves interaction with theeukaryotic translation initiation factor eIF4A that functions to unwind secondary structurein mRNA in an ATP-dependent fashion. Loss <strong>of</strong> Pdcd4 is thought to contribute to celltransformation. Decreased expression <strong>of</strong> Pdcd4 has been observed in lung, breast,prostate, liver tumors <strong>and</strong> neuroendocrine carcinomas. To better underst<strong>and</strong> theregulation <strong>of</strong> eIF4A by Pdcd4, we present here the crystal structures <strong>of</strong> the MA3 domains<strong>of</strong> Pdcd4 at 2.8Å resolution <strong>and</strong> that <strong>of</strong> Pdcd4 in complex with eIF4A at 3.5Å resolution.To confirm the Pdcd4 <strong>and</strong> eIF4A binding interface as seen in the structure, we haveidentified a conserved surface on Pdcd4 extending across both t<strong>and</strong>em MA3 domains aswell as specific residues on eIF4A for mutagenesis. We analyzed the binding <strong>of</strong> eIF4A toa series <strong>of</strong> Pdcd4 mutants in vitro, <strong>and</strong> vice versa. Mutational analyses have also revealedthat inhibition <strong>of</strong> ATP hydrolysis induced by interaction with Pdcd4 correlates withconcomitant reduction <strong>of</strong> RNA unwinding, <strong>and</strong> that these mutants reduce the ability <strong>of</strong>Pdcd4 to inhibit the enzymatic activities <strong>of</strong> eIF4A. These same mutants also reduce theinhibitory effect <strong>of</strong> Pdcd4 on AP-1 transactivation. Based on our structures <strong>and</strong> availablestructures <strong>of</strong> other RNA helicases, we will present a model <strong>of</strong> the mechanism <strong>of</strong>translation inhibition by Pdcd4.22


(6) Plasma membrane Ca2+-ATPase 1 is required for lateral line development inzebrafishWilliam Go 1 , Dmitri Bessarab 2 , Vladimir Korzh 11 <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>, Singapore2 <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>Biology</strong>, SingaporeThe plasma membrane calcium ATPase (PMCA) transports Ca2+ ions out <strong>of</strong> the cells byATP hydrolysis (Ca2+ pump). It is essential in the control <strong>of</strong> Ca2+ concentration in thecytosol to maintain a low cytosolic free Ca2+ concentration for the correct functioning <strong>of</strong>cells. In mammals, there are four different genes that generate four is<strong>of</strong>orms (PMCA1,PMCA2, PMCA3, <strong>and</strong> PMCA4) by alternative splicing. The efforts so far were focusedmainly on is<strong>of</strong>orms’ physiological significance. Expression <strong>of</strong> some is<strong>of</strong>orms started veryearly during embryogenesis but PMCA role in development has not been demonstrated(Sepúlveda et al., 2007).To facilitate developmental studies, enhanced green fluorescent protein (EGFP) geneplaced under control <strong>of</strong> the keratin8 promoter was used as a reporter in the Tol2transposon based enhancer trap (ET) designed in our laboratory (Parinov et al., 2004). Inline ET4, Tol2 element insertion occurred 100kb upstream <strong>of</strong> the gene coding forPMCA1. Expression <strong>of</strong> EGFP faithfully recapitulates the expression <strong>of</strong> PMCA1 in brain,muscle, inner ear <strong>and</strong> lateral line neuromasts. During posterior lateral line development,pmca1 transcripts were first detected in the presumptive hair cell precursors locatedwithin the migrating primordium.We used morpholino approach to address the role <strong>of</strong> PMCA1 in early zebrafishdevelopment <strong>and</strong> found that abnormalities <strong>of</strong> development were detected in all organs ortissues expressing pmca1. We focused on the role <strong>of</strong> PMCA1 in the lateral linedevelopment <strong>and</strong> our results suggest that PMCA1 is required during development <strong>of</strong>lateral line from proneuromast deposition to maturation <strong>of</strong> functional neuromast.References:1. Sepúlveda MR, Hidalgo-Sánchez M, Marcos D, Mata AM. (2007), Developmental distribution <strong>of</strong> plasmamembrane Ca2+-ATPase is<strong>of</strong>orms in chick cerebellum, Dev Dyn.;236(5):1227-36.2. Parinov S, Kondrichin I, Korzh V, Emelyanov A. (2004), Tol2 transposon-mediated enhancer trap toidentify developmentally regulated zebrafish genes in vivo. Dev Dyn.;231(2):449-59.23


(7) Developmental analysis <strong>of</strong> the novel zebrafish gene, zic6.Kondrychyn, Igor <strong>and</strong> Korzh, VladimirThe zic (zinc finger protein for cerebellum) gene family encoding transcription factors invertebrates is comprised <strong>of</strong> five members, which play a role in formation <strong>of</strong> the dorsalneural tube. Developmental analysis <strong>of</strong> zic genes showed that during the early stage <strong>of</strong>neurogenesis they act as pre-pattern genes downstream <strong>of</strong> BMPs <strong>and</strong> upstream <strong>of</strong> Wnts.We isolated <strong>and</strong> characterized the novel member <strong>of</strong> the zebrafish zic gene family, zic6,which 3’-untranslated region was tagged by insertion <strong>of</strong> Tol2 transposon in the ET33enhancer trap transgenic zebrafish line (Parinov et al., 2004). Genomic analysis showedthat in contrast to all other zic’s this gene is present only in teleost phyla. Probably, it waslost during later evolution. During development zic6 mRNA was first detected by RT-PCR at an early blastula stage. By whole mount in situ hybridization zic6 expression wasdetected soon after bud stage at the prospective midbrain-hindbrain boundary <strong>and</strong> dorsalmidbrain followed by dorsal hindbrain <strong>and</strong> pretectum. At the later stages, zic6 continuesto be expressed in the dorsal neural tube, in particular, the ro<strong>of</strong> plate. The inhibition <strong>of</strong>zic6 translation by antisense morpholino oligonucleotide knockdown revealed that zic6plays a role during morphogenesis <strong>of</strong> the ro<strong>of</strong> plate cells.24


(8) Analysis <strong>of</strong> Zebrafish Inducible Nitric Oxide Synthase Genes DuringEmbryogenesisKar Lai Poon (VK lab)Inducible nitric oxide synthase (NOS2) catalyzes the production <strong>of</strong> nitric oxide (NO), <strong>and</strong>is one <strong>of</strong> the players establishing innate immunity. In zebrafish, nos2 is represented bynos2a <strong>and</strong> nos2b. Here, we report the cloning <strong>and</strong> expression pattern <strong>of</strong> the zebrafishnos2b genes, which does not participate in immune defence. nos2b was mapped tozebrafish linkage group 15. The spatial <strong>and</strong> temporal expression pattern <strong>of</strong> nos2b inembryonic zebrafish was analyzed by whole mount in situ hybridization. Duringembryogenesis nos2b is expressed constitutively in a highly restricted manner.Expression <strong>of</strong> zebrafish nos2b appeared at 14 hpf in the forebrain. Later on it was presentin the hypothalamus, where initially it was expressed in the ventral-most cell layer.During late embryogenesis, expression <strong>of</strong> nos2b was detected immediately posterior tothe adenohypophysis <strong>and</strong> later on a low level was present in the midline ventral to MHBthat move rostrally. By 72 hpf, the only expression domain <strong>of</strong> nos2b is associated withthe middle lower jaw.25


(9) Cytoplasmic role <strong>of</strong> Stat3 in cell migrationTerk Shin Teng (CXM lab)Stat3 is a member <strong>of</strong> the signal transducer <strong>and</strong> activator <strong>of</strong> transcription protein family,which is important for cytokine signaling as well as a number <strong>of</strong> cellular processesincluding cell proliferation, anti-apoptosis <strong>and</strong> immune responses. In recent years, thereis emerging evidence that Stat3 also participates in cell invasion <strong>and</strong> motility. However,how Stat3 regulates these processes still remains poorly understood. In our current study,we use Stat3-deficient <strong>and</strong> wild-type Stat3-expressing mouse embryonic fibroblasts(MEFs) as a model to investigate the role <strong>of</strong> Stat3 in cell migration. We find that loss <strong>of</strong>Stat3 expression leads to an elevation <strong>of</strong> Rac1 activity, which promotes a r<strong>and</strong>om mode<strong>of</strong> migration by reducing directional persistence. By performing rescued experiments, wedemonstrate that Stat3 can regulate the activation <strong>of</strong> Rac1 to mediate persisted directionalmigration <strong>and</strong> this function is not dependent on Stat3 transcriptional activity. We findthat Stat3 binds to -Pix <strong>and</strong> this interaction could represent a mechanism, whichcytoplasmic Stat3 regulates Rac1 activity to modulate the organization <strong>of</strong> actincytoskeleton <strong>and</strong> directional migration.26


(10) Identification <strong>and</strong> characterization <strong>of</strong> conserved cis-regulatory elementsassociated with transcription-factor encoding genes in the human genomeAlison P Lee (FUGE/SB lab)<strong>Molecular</strong> Genetics Laboratory, <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>Comparative genomics is a powerful strategy for identifying conserved gene regulatoryelements in the human genome. We have used this approach for identifying <strong>and</strong>characterizing conserved cis-regulatory elements associated with genes that encodetranscription factors (TFs) in the human genome. TFs represent key nodes in theregulatory networks <strong>of</strong> important biological processes, <strong>and</strong> underst<strong>and</strong>ing the molecularbasis <strong>of</strong> regulation <strong>of</strong> TF-encoding genes is an important step towards decipheringregulatory networks. Human genome is estimated to contain about 2,000 TF-encodinggenes. We created a database <strong>of</strong> human, mouse <strong>and</strong> fugu TF-encoding genes <strong>and</strong>identified orthologous genes in the three genomes. The genomic sequences <strong>of</strong> theorthologous loci in the three genomes were aligned using a global alignment program <strong>and</strong>conserved noncoding elements (CNEs) were predicted using VISTA. These CNEsrepresent regulatory elements under evolutionary selection. A total <strong>of</strong> 2,843 CNEsassociated with 389 human TF-encoding genes were identified. TF-encoding genes thatare expressed in the nervous system <strong>and</strong> involved in development contain a significantlyhigher number <strong>of</strong> CNEs. We predicted four 8-bp motifs that are enriched in CNEs <strong>of</strong>central nervous system genes. These motifs could be the binding sites <strong>of</strong> transcriptionfactors important for gene expression in the central nervous system. The functions <strong>of</strong> aselected set <strong>of</strong> CNEs are being assayed in transgenic mice. The database <strong>of</strong> vertebratetranscription factors <strong>and</strong> the CNEs associated with them, known as TFCONES, is madeavailable in the public domain (http://tfcones.fugu-sg.org/).27


(11) PRL-3 Initiates Tumor Angiogenesis by Recruiting Endothelial <strong>Cell</strong>s In vitro<strong>and</strong> In vivoSamantha Quah (ZQ lab)Metastasis is the terminal stage <strong>of</strong> cancer progression. It involves the transformation <strong>of</strong>primary tumors to malignant secondary tumors, <strong>and</strong> is a multi-step process. Themembers <strong>of</strong> the PRL (Phosphatase <strong>of</strong> Regenerating Liver) family belong to a subclass <strong>of</strong>protein tyrosine phosphatases. PRL-3 has been linked to promote cell migration,invasion <strong>and</strong> metastases. PRL-3 is proposed to be an indicator <strong>of</strong> poor prognosticoutcome. The molecular mechanisms by which PRL-3 operates in metastasis are onlyjust beginning to be understood. We have investigated the possible functions <strong>of</strong> PRL-3 inboth physiological <strong>and</strong> tumor angiogenesis, which is an important step in metastasis.We investigated PRL-3 expressing cells’ ability to crosstalk to human umbilical vascularendothelial cells (HUVEC) using an in vitro co-culture system. HUVEC were grown withfibroblasts, <strong>and</strong> later overlaid with PRL-3-expressing cells. PRL-3-expressing CHO cellsredirected the migration <strong>of</strong> HUVEC towards them; while PRL-3-expressing DLD-1 cellsenhanced HUVEC vascular formation. In vivo injection <strong>of</strong> Matrigel plugs containingPRL-3-expressing CHO cells into nude mice to form local tumors resulted in therecruitment <strong>of</strong> host endothelial cells (ECs) to the vicinity <strong>of</strong> the tumor sites <strong>and</strong> initiation<strong>of</strong> angiogenesis. Our results suggest that PRL-3 may play a role in initiatingestablishment <strong>of</strong> microvasculature. We further show that PRL-3 expressing cells reducedIL-4 expression levels, attenuating IL-4 inhibitory effects on the HUVEC vasculature.Our findings provide direct evidence that PRL-3 may be involved in triggeringangiogenesis <strong>and</strong> may serve as an attractive therapeutic target with respect to bothangiogenesis <strong>and</strong> cancer metastasis.28


(12) Structure <strong>of</strong> Actin in the NucleusKhay Chun Ang (BR lab)The existence <strong>of</strong> actin in the nucleus has been controversial. Traditional cystoplasmicmarkers for actin, such as antibodies <strong>and</strong> phalloidin, have indicated that conventionalactin filaments do not generally exist in the nucleus. However, over the last few yearsactin has been identified as an integral subunit <strong>of</strong> the transcription apparatus, DNA <strong>and</strong>RNA modifying complexes on top <strong>of</strong> other nuclear binding partners. Hence, actin mostdefinitely is present in the nucleus, however, nothing is known <strong>of</strong> its conformational state.Actin, as well as actin related proteins (Arps), are found to be integral subunits <strong>of</strong>multisubunit chromatin remodeling complexes (CRC), complexes intimately tasked toactivate genes in eukaryotes by relieving topological constraints exerted by nucleosomes.The role <strong>of</strong> actin within these complexes is unclear <strong>and</strong> therefore we believe that a 3-Dstructure approach will not only be vital in clarifying the paradigm but also provideinvaluable structural correlation <strong>of</strong> actin with existing biochemical <strong>and</strong> biophysicalobservations.Experimental approaches include purifying targeted nuclear actin binding proteins tohomogeneity, with the final intention <strong>of</strong> co-crystallization with actin. In addition, at<strong>and</strong>em affinity purification (TAP)-tag approach is being implemented to harvest native,multisubunit actin complexes <strong>and</strong>/or Arps containing nuclear complexes forcrystallographic studies. From these structures we expect to clarify the conformationalstate <strong>of</strong> actin <strong>and</strong> gain insight into its nuclear roles.29


(13) Nuances <strong>of</strong> Gelsolin FunctionShalini Nag (BR lab)The gelsolin superfamily <strong>of</strong> calcium-activated, actin binding proteins regulates actindynamics by severing, capping, nucleating or bundling actin filaments. Expressed in awide range <strong>of</strong> eukaryotes, they regulate cell motility <strong>and</strong> phagocytosis, transduce signalsinto dynamic cytoskeletal rearrangements, stimulate apoptosis in some vertebrate cells,<strong>and</strong> regulate transcription. Due to their varied functions the misregulation <strong>of</strong> theseproteins results in pathological conditions like inflammation <strong>and</strong> diseases such as cancer<strong>and</strong> amyloidosis. Proteins <strong>of</strong> this family are composed <strong>of</strong> 100 – 125 residue homologousmodules that are folded into compact domains. Movement between these domains isresponsible for the diverse activities <strong>of</strong> these proteins. Gelsolin, the founding member <strong>of</strong>this family, has 6 such domains connected by linkers <strong>of</strong> various lengths. Other membersdiffer in the number <strong>of</strong> domains, or have residue substitutions that tailor their expression,localization <strong>and</strong> regulation to the appropriate regions <strong>of</strong> motile cells. Although 3 decades<strong>of</strong> studies have provided numerous insights, the holy grail <strong>of</strong> the gelsolin field, amolecular movie <strong>of</strong> this dynamic molecule as it undergoes large conformational changesduring calcium activation <strong>and</strong> function, has been elusive because <strong>of</strong> the speed <strong>of</strong> the actinsevering reaction, multiplicity <strong>of</strong> steps in the reaction, small size <strong>of</strong> gelsolin, <strong>and</strong> theinherently dynamic nature <strong>of</strong> actin. In an effort to contribute to this molecular movie, Ipresent the structure <strong>of</strong> human gelsolin domains G1 – G3 bound to actin, along withinsights about gelsolin obtained from various biophysical, biochemical <strong>and</strong> molecularbiology methods.30


(14) Designing a synthetic tumour suppressor protein to validate eIF4E as apotential anti-cancer target.See Hai Yun, Christopher J.Brown, David Coomber, David P.LaneUpregulation <strong>of</strong> translation initiation factor eIF4E has been found to selectively increasethe synthesis <strong>of</strong> oncoproteins, inducing tumourigenesis. Our study aims to design <strong>and</strong>construct a synthetic protein that will bind to <strong>and</strong> inhibit the tumourigenic activity <strong>of</strong>eIF4E.12 amino acids derived from eIF4G, one <strong>of</strong> eIF4E’s endogenous binding partners, wereinserted between Gly33 <strong>and</strong> Pro34 <strong>of</strong> an Escherichia coli protein: thioredoxin (Trx). Thisinsertion disrupts the active site loop <strong>of</strong> the protein <strong>and</strong> allows the binding peptide to bepresented at the surface <strong>of</strong> the aptamer (Trx-eIF4G). Critical contact residues in thebinding amino acid sequence were mutated to alanines to act as the control aptamer (TrxeIF4G-ala).Immunoprecipitations demonstrated the ability <strong>of</strong> the Trx-eIF4G aptamer to bind toeIF4E, while there was negligible binding <strong>of</strong> the control aptamer Trx-eIF4G-ala. Theinsertion <strong>of</strong> additional residues in front <strong>of</strong> the peptide sequence to allow for increasedflexibility enhanced the binding affinity <strong>of</strong> the aptamer (Trx-SG-eIF4G). Fluorescentpolarization (FP) experiments using Trx constructs to compete with peptides for bindingfurther confirmed the interaction between the aptamer <strong>and</strong> its target protein eIF4E. TheFP assays corroborated with the pulldown assays demonstrating enhanced binding <strong>of</strong> theTrx-SG-eIF4G aptamer to eIF4E as compared to the original Trx-eIF4G aptamer.In vivo transformation assays that involve growing transfected cells on s<strong>of</strong>t agar to testfor independence <strong>of</strong> contact inhibition (characteristic <strong>of</strong> tumourigenicity <strong>and</strong> malignancy)demonstrates the ability <strong>of</strong> the aptamer to inhibit the tumourigenic effects <strong>of</strong> eIF4E. Thecapacity <strong>of</strong> the aptamer to inhibit malignant cell growth in transformation assays wouldprovide strong evidence <strong>and</strong> support for further research to exploit the therapeuticpotential <strong>of</strong> eIF4E as an anti-cancer target.31


(15) SNARE genes expression regulation upon variation <strong>of</strong> secretory cargo loadChia Zhi Hui, Joanne <strong>and</strong> Frederic BardThe secretory apparatus in higher eukaryotes must be able to adapt to variation in theamount <strong>of</strong> secretory cargo. For example, terminal B cell differentiation requires anincrease biosynthetic capacity to synthesize massive amounts <strong>of</strong> antibodies. It remainspoorly understood how the secretory apparatus copes with increase in cargo load.In this study we have used an inducible system in Drosophila S2 cells wherebyexpression <strong>of</strong> secreted horseradish peroxidase (HRP) is induced in the presence <strong>of</strong> Cu2+ions. Using quantitative imaging to probe the intracellular HRP (HRPi) levels, we foundthat HRPi increases <strong>and</strong> decreases over time after Cu2+ induction. This suggests that thesecretory apparatus adapts to the increase in cargo load. To test if genes important forsecretion are being upregulated, we turned to the SNARE family <strong>of</strong> proteins. The exactSNAREs that regulate Golgi to plasma membrane traffic in Drosophila are currentlyunknown. A systematic survey <strong>of</strong> SNAREs by RNAi indicated that none <strong>of</strong> the SNAREc<strong>and</strong>idates is actually essential for secretion. However, using a co-RNAi screen <strong>of</strong> allSNAREs in the drosophila genome, we have identified Syntaxin1A, Syntaxin4, VAMP3<strong>and</strong> possibly VAMP2 as key SNAREs participating in the late secretory pathway.Following activation <strong>of</strong> secreted HRP production, we found that expression <strong>of</strong> bothVAMP3 <strong>and</strong> VAMP2 genes is upregulated 3 to 4 folds. Our results have revealedextensive SNARE functional redundancy in the Golgi to plasma membrane traffic. Theyalso indicate that the secretory apparatus is able to sense an increase <strong>of</strong> cargo load <strong>and</strong> toup regulate the expression <strong>of</strong> secretory genes in response.32


(16) Adenosine-containing Molecules Amplify Glucose Signaling <strong>and</strong> EnhanceTxnip ExpressionFa-Xing YU <strong>and</strong> Yan LUOGlucose uptake <strong>and</strong> utilization are tightly regulated <strong>and</strong> fundamental for cellularfunctions. Eukaryotic cells have developed different mechanisms that sense the glucose<strong>and</strong> regulate the expression <strong>of</strong> certain genes involved in metabolism. The thioredoxininteractionprotein (Txnip) has been identified as a glucose sensor, for the expression <strong>of</strong>which is tightly correlated with extracellular glucose concentrations. Here, we presentdata showing that, at a transcriptional level, the Txnip expression is induced by a novelclass <strong>of</strong> adenosine-containing molecules such as NAD(H), NADP(H), FAD, ATP, ADP,AMP <strong>and</strong> adenosine, <strong>and</strong> that an intact adenosine moiety is necessary <strong>and</strong> sufficient forthis induction. The induction is abolished in presence <strong>of</strong> glucose transporter inhibitors orupon glucose withdrawal, thus is critically dependent on glucose. This is consistent withthat the Txnip promoter contains a cis-regulatory element, carbohydrate response element(ChoRE), which mediates the induction. MondoA <strong>and</strong> MLX have been recentlydocumented to be ChoRE-binding transcription factor partners involved in the glucosestimulatedTxnip expression, <strong>and</strong> are shown here to be required to transmit stimulatorysignals from extracellular adenosine-containing molecules to the Txnip promoter. Ourstudies suggest that adenosine-containing molecules can amplify the glucose-mediatedsignaling <strong>and</strong> further stimulate the Txnip expression, <strong>and</strong> that the revealed function <strong>of</strong>these molecules might set the stage for developing novel pharmacological interventionsor modulations <strong>of</strong> certain cellular processes in which glucose plays fundamental roles.33


(17) Dissecting a Drosophila melanogaster Histone Transcriptional RegulationPathwayMei Chin LEE, Ling Ling TOH <strong>and</strong> Yan LUOMammalian OCA-S acts as an Oct-1 Co-Activator in S-phase that stimulates thetranscription <strong>of</strong> the histone 2B (H2B) gene. Oct-1 binds the essential octamer element inthe H2B promoter, whilst OCA-S occupies the promoter via an interaction with Oct-1.As a multi-component protein complex, OCA-S comprises <strong>of</strong> p38/GAPDH, p36/LDH<strong>and</strong> other components that are essential for OCA-S function. This exemplifiesmoonlighting nuclear gene-switching activities <strong>of</strong> certain metabolic enzymes, thusexp<strong>and</strong>s the one-protein-multi-function paradigm.A similar regulatory pattern may be conserved in Drosophila melanogaster (dm). Wethus studied expression patterns <strong>of</strong> the dmH2B gene. The octamer elements in thedmH2B promoter are bound by Pdm-1, a counterpart <strong>of</strong> mammalian Oct-1; this binding isessential for dmH2B expression. We have developed an effective RNAi approach toanalyze functions <strong>of</strong> Pdm-1 <strong>and</strong> the components <strong>of</strong> the fly OCA-S (dmOCA-S) in the dmSchneider-2 cell line. Each dmOCA-S component is essential for H2B expression, <strong>and</strong>the H2B promoter occupancy by dmOCA-S is critically dependent upon Pdm-1. This isin line with an OCA-S-Oct-1 interaction in mammalian cells, <strong>and</strong> suggests a conservedhistone expression program in higher eukaryotes. We are testing the functions <strong>of</strong> Pdm-1<strong>and</strong> dmOCA-S at an organism level--in embryos--<strong>and</strong> are using a genome-wide RNAiapproach to search for other potential transcription (co)-activators involved in regulatingthe expression <strong>of</strong> the dmH2B gene as well as other dm histone genes.34


(18) Nm23 <strong>and</strong> Histone H2B gene expression in HeLa cellsJun Yan (LY lab)OCA-S is a key coactivator that enables transcription factor Oct 1 to regulate histoneH2B transcription. OCA-S is composed <strong>of</strong> seven components including nm23 H1 <strong>and</strong>nm23 H2, two subunits <strong>of</strong> nucleotide diphosphate (NDP) kinase. To investigate the role<strong>of</strong> nm23 H1/H2 in OCA-S coactivation <strong>of</strong> H2B transcription, we knocked down the H1<strong>and</strong>/or H2 transiently with siRNA; <strong>and</strong> created 3 types <strong>of</strong> stable knockdown cell lines forlong term effects by using shRNA. We found that the combination <strong>of</strong> siRNA against bothH1 <strong>and</strong> H2 decreased H2B expression by 30% at 72 hours post-transfection. In stableknockdown cells, H1 <strong>and</strong> double knockdown lines grew at a faster rate than parentalHeLa cells whereas H2 knockdown lines grew at a slower rate. Compared with HeLacells by real time PCR, all stable knockdown lines didn’t produce significant differencein overall H2B mRNA level. However, double knockdown lines showed a reduced H2Btranscription initiation by 70%-80% using luciferase reporter assay, suggesting theexistence <strong>of</strong> compensatory mechanisms at posttranscriptional level. Here we report themRNA stability increase by 50% in double knockdown lines at late S phase (6 hours afterrelease from thymidine); <strong>and</strong> stable knockdown lines had lower NDP kinase activities.These findings indicate that nm 23 H1 <strong>and</strong> H2 positively contributed to OCA-S mediatedH2B expression at transcription initiation <strong>and</strong> we further suggest that nm23 mightfunction through its NDP kinase activity.35


(19) A Mechanism that prevents premature chromosome segregation wheninitiation <strong>of</strong> DNA replication failsJenn Hui KhongUS lab, Systems <strong>Biology</strong> Division, <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>Cdc6, an evolutionarily conserved protein, is required for the initiation <strong>of</strong> DNAreplication <strong>and</strong> is essential for cell viability. Yeast cells deficient in Cdc6 fail to initiateDNA replication but proceed to elongate their spindles <strong>and</strong> segregate the un-replicatedchromosomes leading to a “reductional” anaphase. Therefore, Cdc6 has been defined asan important component <strong>of</strong> the G1-M checkpoint that prevents untimely onset <strong>of</strong> mitosiswhen cells fail to initiate DNA replication. Our results suggest that untimelychromosome segregation in the absence <strong>of</strong> Cdc6 function is not due to premature entryinto mitosis but is a result <strong>of</strong> the deregulation <strong>of</strong> spindle dynamics. This dramaticderegulation is a result <strong>of</strong> the interplay <strong>of</strong> three cellular events as cells proceed to DNAreplication: activation <strong>of</strong> the E3 ubiquitin ligase SCF, destruction <strong>of</strong> Cdk inhibitorSic1<strong>and</strong> inactivation <strong>of</strong> another ubiquitin ligase APCCdh1 (Anapahase PromotingComplex activated by Cdh1). Surprisingly, we find that premature chromosomesegregation is a not a property specifically associated with the loss <strong>of</strong> Cdc6 function but itis a common characteristic <strong>of</strong> cells (such as cdc7 or cdc45 mutants) that execute the threeevents but fail to initiate DNA replication. These results suggest that initiation <strong>of</strong> DNAreplication saves the cells from potential chromosome-segregation catastrophe which theconvergence <strong>of</strong> the three regulatory elements APC, SCF <strong>and</strong> Sic1 can cause.36


(20) Roles <strong>of</strong> intracellular p53 oscillations in cell survival <strong>and</strong> deathKeng Boon WeeUS lab, Systems <strong>Biology</strong> Division, <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>,NUS Graduate School for Integrative Sciences & Engineering, National University <strong>of</strong>SingaporeIntracellular levels <strong>of</strong> p53 <strong>and</strong> MDM2 proteins have been shown to oscillate in responseto ionizing radiation (IR). The p53-MDM2 negative feedback loop – the putative cause<strong>of</strong> the oscillations – is embedded in a larger network involving a mutual antagonismbetween p53 <strong>and</strong> AKT. We have shown earlier that this p53-AKT network predicts anall-or-none switching behavior from a pro-survival cellular state (low p53 <strong>and</strong> high AKTsteady state) to a pro-apoptotic state (high p53 <strong>and</strong> low AKT steady state). Here, weshow that upon IR, this p53-AKT network model can also reproduce experimentallyobserved oscillations, <strong>and</strong> that these oscillations are possible only at pro-survival states.Notably, p53 oscillations markedly decrease the IR intensity threshold at which switchingto a pro-apoptotic state occurs. Several biological advantages conferred by suchoscillations are demonstrated, including the increased ability <strong>of</strong> p53 as a transcriptionfactor to induce expression <strong>of</strong> DNA repair <strong>and</strong> pro-apoptotic target genes at higher levels,though no advantage <strong>of</strong> p53 oscillations in non-proliferating cell-types is deduced.Taking the analyses <strong>of</strong> p53-AKT regulation <strong>of</strong> the BCL2 family proteins <strong>and</strong>experimental evidence together, p53 oscillations appear to confer proliferative cells withthe ability to strike the critical balance between rapid responses to DNA damage <strong>and</strong>timely apoptosis after irreparable damage but yet prevent premature cell death.37


(21) Mass Spectrometry in IMCBKelly Hogue <strong>and</strong> Walter BlackstockIn his 2007 essay in <strong>Cell</strong> “Is Proteomics the New Genomics?” Mann proposed that “theexpression levels <strong>of</strong> all proteins would arguably provide the most relevant single data setcharacterizing a biological system.” Realizing this goal is the aim <strong>of</strong> quantitativeproteomics. Of late there have been major advances in mass spectrometry, mostnoticeably in the manipulation <strong>and</strong> storage <strong>of</strong> ions <strong>and</strong> image-current detection in thefrequency domain. With stable isotope labeling this makes a potent combination forlarge-scale protein quantification, the protein analogue <strong>of</strong> the micro-array. Thetechnology is expensive <strong>and</strong> at the moment its use far from routine. Mass spectrometerskills are scarce as there is little opportunity to learn mass spectrometry in graduateschools. There is also a need to improve underst<strong>and</strong>ing <strong>of</strong> what is possible <strong>and</strong> ensure thatinstrument time is productively used.To improve mass spectrometry in IMCB specifically <strong>and</strong> <strong>A*Star</strong> in general we arebuilding a small team <strong>of</strong> biologists, mass spectroscopists, <strong>and</strong> s<strong>of</strong>tware engineers workingcollaboratively in the area <strong>of</strong> quantitative biology using stable isotope labeling Weemploy two state-<strong>of</strong>-the art Orbitrap mass spectrometers, an investment <strong>of</strong> over S$ 3.5million, in work-flows optimized for the analysis <strong>of</strong> protein digests. Data analysis is aprocess <strong>of</strong> inference as the protein information has to be recovered from the peptideinformation. This can be computationally intensive particularly for post-translationalmodifications <strong>and</strong> is being actively developed.38


(22) Non-ribosomal peptides in Pseudomonas aeruginosa quorum sensingregulationJasmine Lee (ZLH lab)Quorum Sensing (QS) bacteria sense <strong>and</strong> communicate with one another through therelease <strong>of</strong> chemical signals known as autoinducers. These signals are recognized by theircognate receptors, which then act as transcriptional regulators to activate the expression<strong>of</strong> downstream target genes. There are currently three known QS systems inPseudomonas aeruginosa, i.e., the LasI/R, RhlI/R, <strong>and</strong> Pseudomonas quinolone signal(PQS) systems. R<strong>and</strong>om mutagenesis revealed an operon, consisting <strong>of</strong> 4 genesdesignated as qrpABCD, implicated in regulation <strong>of</strong> PQS biosynthesis. They arepredicted to be a non-ribosomal peptide synthetase gene cluster. When individuallydeleted from the genome, a dramatic reduction in transcriptional expression <strong>of</strong> PQSbiosynthesis genes is observed. This is translated into an actual physiological reduction inPQS level, as well as corresponding decrease in production <strong>of</strong> PQS-controlled virulencefactors, including pyocyanin <strong>and</strong> elastase. In trans expression <strong>of</strong> wild type qrp genes incorresponding mutants reversed the trends <strong>and</strong> caused increased production <strong>of</strong> PQS <strong>and</strong>its related virulence factors. These evidences attest to the role <strong>of</strong> QrpABCD in positiveregulation <strong>of</strong> the PQS quorum sensing system in P. aeruginosa. Work has started onunderst<strong>and</strong>ing the mechanism behind the control. Recent data suggests that theqrpABCD-dependent products are secreted into the extracellular environment <strong>and</strong> can bechemically extracted from the bacterial culture supernatant. When the extracts are addedto the qrpABCD-null mutants, PQS production is restored to wild type level. Furtherexperiments are underway to purify the peptide for structural analysis in a bid to identifyhow PQS is regulated.39


(23) Loss <strong>of</strong> udu function causes activation <strong>of</strong> DNA damage checkpointChiaw-Hwee Lim, Shang-Wei Chong <strong>and</strong> Yun-Jin JiangLaboratory <strong>of</strong> Developmental Signalling <strong>and</strong> Patterning, Genes <strong>and</strong> DevelopmentDivision, <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>, A*STAR (Agency for Science,Technology <strong>and</strong> Research), 61 Biopolis Drive, Singapore 138673Although Udu has been showed to play an essential role during blood cell development,its roles in other cellular processes remain largely unexplored. We used a zebrafishmutant; ugly duckling (udutu24) isolated in the 1996 Tübingen genetic screen <strong>and</strong>showed that its extensive p53-dependent apoptosis is a consequence <strong>of</strong> activation <strong>of</strong> theAtm-Chk2 pathway. The DNA damage response pathway is a cellular surveillancesystem that senses the presence <strong>of</strong> damaged DNA <strong>and</strong> elicits an appropriate response tothe damage. Our FACS analysis showed that the loss <strong>of</strong> udu function resulted in defectivecell cycle progression <strong>and</strong> comet assay indicated the presence <strong>of</strong> increased DNA damagein udutu24 mutants. Immun<strong>of</strong>luoresence staining <strong>of</strong> transfected COS7 cells demonstratesa dynamic association between PAH-L (Paired Amphipathic α-Helix like), SANT-L(SW13, ADA2, N-Cor <strong>and</strong> TFIIIB like) domains <strong>and</strong> replicating pericentromericheterochromatin, which is known to serve functions from gene regulation to protection <strong>of</strong>the integrity <strong>of</strong> chromosomes. Taken together, these data suggest a possible role <strong>of</strong> Uduin protecting the integrity <strong>of</strong> the genome <strong>and</strong> the loss <strong>of</strong> Udu function, particularly PAH-L <strong>and</strong> SANT-L domains can contribute to DNA damage, leading to the activation <strong>of</strong>DNA damage pathway <strong>and</strong> subsequently, cell cycle arrest <strong>and</strong> apoptosis.40


(24) Engrailed gene expression <strong>and</strong> its control by hedgehog signalingAshish Maurya (PWI lab)In the zebrafish myotome several muscle cell types are specified by varying levels <strong>and</strong>timing <strong>of</strong> hedgehog signaling. Muscle pioneer cells that are specified by the highestamount <strong>of</strong> hedgehog signaling express high levels <strong>of</strong> engrailed homeeobox genes. In thepresent study, I am studying regulation <strong>of</strong> engrailed genes in the muscle pioneers <strong>and</strong>how it is controlled by hedgehog signaling?To isolate engrailed cis-elements I have tested various conserved elements around theengrailed genes for activity in an enhancer assay in transgenic zebrafish. I have alsotested various lengths <strong>of</strong> the En2a promoter <strong>and</strong> mapped a muscle pioneer enhancerelement within 4 to 8kb region upstream <strong>of</strong> the gene. Currently I am trying to identify theminimal region necessary for this muscle pioneer activity. Within this 4kb region no Glibinding sites (Gli transcription factors mediate hedgehog responsive gene expression) arepredicted. This suggests that hedgehog signaling is indirectly regulating engrailed. Inagreement with this the -4 to -8kb region does contain binding sites for transcriptionfactors that are positively <strong>and</strong> strongly regulated by hedgehog signaling like MyoD <strong>and</strong>Prdm1.To identify the trans-acting factors that bind to this element I plan to screen a medakacDNA library in a cell culture based assay. In this assay a reporter gene will be driven bythe engrailed regulatory elements <strong>and</strong> the cDNA clones will be individually tested fortheir ability to either enhance or suppress the reporter.41


(25) Specification <strong>of</strong> vertebrate slow-twitch muscle fiber fate by the transcriptionalregulator Blimp1Hoe Peng Liew, Semil P. Choksi, Kangli Noel Wong, Sudipto Roy<strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>Vertebrate skeletal muscles are typically composed <strong>of</strong> slow <strong>and</strong> fast-twitch fibers thatdiffer in their morphology, gene expression pr<strong>of</strong>iles, contraction speeds, metabolicproperties <strong>and</strong> patterns <strong>of</strong> innervation. During myogenesis, how muscle precursors areinduced to mature into distinct slow or fast-twitch fiber-types remains inadequatelyunderstood. We have previously shown that within the somites <strong>of</strong> the zebrafish embryo,activity <strong>of</strong> the zinc finger <strong>and</strong> SET domain containing transcriptional regulator Blimp1 isessential for the specification <strong>of</strong> slow muscle fibers. Here, we have investigated themechanism by which Blimp1 programs myoblasts to adopt the slow-twitch fiber fate. Inslow myoblasts, expression <strong>of</strong> the Blimp1 protein is transient, <strong>and</strong> precedes the onset <strong>of</strong>expression <strong>of</strong> slow muscle-specific differentiation genes. We demonstrate that thecompetence <strong>of</strong> somitic myoblasts to commit to the slow lineage in response to Blimp1changes as a function <strong>of</strong> developmental time. Furthermore, we provide evidence thatmammalian Blimp1 can fully recapitulate the slow myogenic program in the zebrafish,suggesting that zebrafish Blimp1 can bind to the same consensus DNA sequence that hasbeen ascribed to the mammalian protein. In line with this, zebrafish Blimp1 can target therepression <strong>of</strong> fast muscle-specific myosin light chain through direct binding near thepromoter <strong>of</strong> this gene, indicating that an important aspect <strong>of</strong> the transcriptional activity <strong>of</strong>Blimp1 in slow muscle development is the suppression <strong>of</strong> fast-muscle-specific geneexpression. Taken together, all <strong>of</strong> these findings provide new insights into the molecularbasis <strong>of</strong> vertebrate muscle fiber-type specification, <strong>and</strong> underscore Blimp1 as the centraldeterminant <strong>of</strong> this process.42


(26) G-actin binds to the adenylyl cyclase Cyr1 through Cap1 <strong>and</strong> regulates cAMPsynthesis in C. albicans hyphal morphogenesisHao Zou <strong>and</strong> Yue WangC<strong>and</strong>ida albicans <strong>Molecular</strong> & <strong>Cell</strong> <strong>Biology</strong> Laboratory, Genes <strong>and</strong> DevelopmentDivision, <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>The yeast-to-hyphal growth switch is a key virulence trait <strong>of</strong> C. albicans. It is induced bycertain environmental signals <strong>and</strong> primarily mediated by the cAMP/PKA pathway. Actincytoskeleton plays a central role in the hyphal growth. However, the mechanisms <strong>of</strong>interaction between the cAMP/PKA pathway <strong>and</strong> the actin cytoskeleton remain unclear.The aim <strong>of</strong> this study is to test the hypothesis that the Cyclase Associated Protein, Cap1<strong>of</strong> C. albicans, which has the capacity to interact with both actin <strong>and</strong> the adenylyl cyclaseCyr1, may play an important role in linking the actin cytoskeleton <strong>and</strong> the cAMP/PKApathway. We have found that Cyr1, Cap1 <strong>and</strong> actin can indeed form a ternary complex inwhich Cap1 acts as a bridge, <strong>and</strong> this complex is sufficient for producing cAMP in vitrounder hyphal-inducing conditions. Since the C-terminus <strong>of</strong> Cap1 contains the G-actinbinding site <strong>and</strong> is required for the formation <strong>of</strong> the ternary complex, it is reasonable tospeculate that G-actin might be able to influence Cyr1 activity through its interactionwith Cap1. Consistent with this view, cells expressing a Cap1 mutant lacking the G-actinbinding site exhibited delayed <strong>and</strong> reduced cAMP production <strong>and</strong> defects in hyphalmorphogenesis. Consistently, while wild-type cells exhibited suppressed adenylyl cyclaseactivity when treated with actin depolymerization drugs, the mutant cells were resistant.These results for the first time demonstrate that G-actin plays an active role in directlyregulating cAMP synthesis by forming a ternary complex with Cyr1 <strong>and</strong> Cap1, revealinga mechanistic link for regulation <strong>of</strong> the cAMP/PKA pathway by cellular actin status.43


(27) Wip1 regulates neural stem cell/progenitor proliferation through p53dependent G2 cell cycle controlYun-Hua. Zhu, Lu Li, Dmitry V. Bulavin <strong>and</strong> Zhi-Cheng XiaoContinuing new neuron formation in adult mammalian brain has been realized as animportant process. However, the intrinsic regulation <strong>of</strong> Neural Stem/progenitor <strong>Cell</strong>s(NSC) remains to be elucidated. Here we report an essential function <strong>of</strong> theserine/thronine phosphatase wip1 in NSC proliferation <strong>and</strong> new neuron formation. wip1knockout (ko) resulted in a 90% decrease in new neuron formation in the Olfactory Bulb(OB), without obvious defects in migration <strong>and</strong> apoptosis. wip1 ko mice show a over tw<strong>of</strong>old decrease in proliferating NSC number in the Sub-Ventricular Zone (SVZ). Similar toin vivo data, in vitro findings indicated the defect <strong>of</strong> wip1 ko NSC in proliferation, selfrenewal <strong>and</strong> new neuron formation. <strong>Cell</strong> cycle analysis by CSFE pulse labeling proved adirect evidence <strong>of</strong> prolonged cell cycle in wip1 ko NSC, <strong>and</strong> that could be a result fromrepressed G2/M transition. <strong>Molecular</strong> analysis revealed that wip1 ko NSC have increasedp21 <strong>and</strong> reprimo levels in a p53 dependent manner, which might be responsible for therepression in G2 progression. Most interestingly, wip1 ko could not trigger G2/Maccumulation without p53. Finally, we confirmed that NSC regulation by wip1 is p53dependent in vitro <strong>and</strong> in vivo by the finding that ko <strong>of</strong> p53, but not other targets <strong>of</strong> wip1including atm or p38, diminishes the effect <strong>of</strong> wip1 ko on NSC proliferation. Thus, ourfinding established wip1 as an essential regulator in maintaining NSC proliferation <strong>and</strong>new cell formation during through p53 dependent cell cycle control at G2/M transition.44


(28) Mouse embryonic neural stem cells: A model for asymmetric cell divisionGoh Lihui (YXH lab)A stem cell typically divides asymmetrically to give two different daughter cells, one <strong>of</strong>which remains as an undifferentiated stem cell, the other which would differentiate intospecialized cells. The mechanism <strong>of</strong> asymmetric cell division is well studied inDrosophila Melanogaster neuroblasts <strong>and</strong> sensory organ precursor cells. The mechanismdirecting asymmetric cell division in mammals, however, is largely unknown.Polarity genes like Partioning Defective (Par) 3, Par 6, Lethal Giant Larvae (Lgl), whichare known to be involved in asymmetric cell division in Caenorhabditis elegans <strong>and</strong>Drosophila Melanogaster, are highly conserved in mammals. Studies have found thatthese genes <strong>and</strong> their gene product are required for the establishment <strong>of</strong> polarity inmammalian epithelial cells. This brings us to wonder if they also play a role in theprocess <strong>of</strong> asymmetric cell division in mammals.Using embryonic neural stem cells (NSC) from mouse embryonic cerebral cortex as amodel, we aim to investigate if these different polarity genes are involved in asymmetriccell division. By knocking down these genes in NSC using lentiviral or retroviralparticles carrying the gene specific shRNA constructs, we seek to determine the functions<strong>of</strong> the genes in asymmetric cell division using various real time cell markers like Hes-1promoter <strong>and</strong> Notch Receptors.45


(29) Searching For Novel Proteins Involved In Asymmetric <strong>Cell</strong> DivisionZhenxing Huang, Siu Kwan Sze <strong>and</strong> Xiaohang YangAsymmetric cell division is a fundamental mechanism <strong>of</strong> generating cell diversity duringdevelopment. Drosophila Neuroblast is a model system for studying cell polarity,asymmetric cell division as well as stem cell self-renewal. The apical proteins, such asPar6, aPKC <strong>and</strong> Pins, control <strong>and</strong> regulate various aspects <strong>of</strong> neuroblast asymmetricdivision, such as basal cell fate determinant localization, spindle orientation as well assibling cell size. Despite the fact that large numbers <strong>of</strong> proteins have been identified asplayers in this mechanism in the recent decade, the complicated network <strong>of</strong> proteininteractions during asymmetric cell division is still poorly understood. This suggests thatthere are still a large number <strong>of</strong> proteins contributing in this event awaiting to bediscovered. In this study, we generated transgenic flies expressing TAP (t<strong>and</strong>em affinitypurification)-tagged Par6 fusion protein specifically in neuronal cells. The fusion proteintogether with its interacting partners was co-pulled down from fly embryos extracts. Thecomponents <strong>of</strong> the protein complex were subsequently separated <strong>and</strong> identified byLC/MS/MS. Some known interacting partners, such as aPKC <strong>and</strong> Lgl, together withdozens <strong>of</strong> novel interacting proteins have been identified. Base on the expression patterns,we chose six c<strong>and</strong>idate genes for further investigation.46


(30) Apim genetically interacts with Inscuteable in the dividing DrosophilaneuroblastJoan Siew Ching Lee (YXH lab)Asymmetric cell division is an evolutionary conserved mechanism used by progenitorcells to generate cellular diversity during development. Apim (Apical Mir<strong>and</strong>a), a coiledcoilprotein, was isolated from a genetic screen. Embryos lacking Apim <strong>and</strong> Inscuteable(Insc) exhibit delayed development. Mislocalization <strong>of</strong> proteins normally segregated tothe basal cortex <strong>of</strong> dividing neuroblasts such as Mir<strong>and</strong>a <strong>and</strong> Prospero (Pros) is observedin double mutant embryos. This mislocalization occurs progressively from the cortex <strong>of</strong>the dividing neuroblast to the mitotic spindle. Neuroblasts in double mutant embryos areeventually stalled in metaphase. Our findings show that in the absence <strong>of</strong> Inscuteable,Apim is involved in embryonic development, regulates the basal localization <strong>of</strong> the keycell fate determinant Pros, <strong>and</strong> plays a role in cell cycle progression in Drosophilaembryos.47


(31) Identification <strong>of</strong> Novel Epigenetic RegulatorsMigliori, V 1 , Casadio F 2 , Bassi C 2 , Amati B 2 , Gozani O 3 <strong>and</strong> Guccione E 1 .1IMCB, Singapore 2 Ifom-Ieo-Campus, Dpt <strong>of</strong> Experimental Oncology, Milan Italy 3 Stanford University,Dept. <strong>of</strong> Biological Sciences, USAIn recent years several groups have generated genome wide epigenetic maps <strong>of</strong> differentmouse <strong>and</strong> human cell types. One <strong>of</strong> the next challenging questions in biological researchis to underst<strong>and</strong> how this epigenetic information is then transduced by chromatinassociatedproteins to downstream effects.The project aims at identifying the network <strong>of</strong> interaction between protein-motifs that arecommonly found among chromatin-associated proteins <strong>and</strong> methyl-histone modifications(HMs).The experimental strategy involved the cloning <strong>of</strong> a library <strong>of</strong> protein domains into avector, which allows in vitro transcription <strong>and</strong> translation. The library includes the entireset <strong>of</strong> human protein domains potentially associated with modified histone tails: Chromo-,Tudor-, MBT-, Plant Agenet-, PWWP-, all belonging to the Tudor domain "Royalfamily", as well as BRK domains <strong>and</strong> selected PHD fingers. The second step <strong>of</strong> theproject consisted in hybridizing the library onto an array <strong>of</strong> Histone H3 <strong>and</strong> H4 modifiedpeptides.This has resulted in the identification <strong>of</strong> new histone binders, like the chromodomain <strong>of</strong>CDYL1 <strong>and</strong> CDYL2 that recognize the trimethylation <strong>of</strong> lysine 9 <strong>of</strong> histone H3(H3K9me3), <strong>and</strong> the WD40 domain <strong>of</strong> WDR5 that binds to the symmetric dimethylation<strong>of</strong> histone H3 (H3R2me2s).We have further validated the newly discovered interactions by alternative methods suchas st<strong>and</strong>ard peptide immunoprecipitations from bacterial lysates, immun<strong>of</strong>luorescence,BIAcore <strong>and</strong> crystalization. The project, which is still ongoing, as allowed so far toincrease the information available on how proteins "read" the histone epigeneticinformation <strong>and</strong> to further investigate the function <strong>of</strong> some <strong>of</strong> these interactions indownstream signaling events.48


(32) VAMP8 Is Required for <strong>Cell</strong> Surface Deployment <strong>of</strong> the Water ChannelAquaporin 2Wang Cheng Chun (HWJ lab)Water is the major constituent <strong>of</strong> living cells <strong>and</strong> water homeostasis is fundamentallyimportant for cell metabolism. AQP2 is a vasopressin-regulated water channel that playsa critical role in maintaining body water balance. Despite the major progress indeciphering the structure <strong>and</strong> function <strong>of</strong> AQP2, the SNARE proteins mediating AQP2exocytosis remain elusive. Here we show that VAMP8 is required for regulated surfaceexpression <strong>of</strong> AQP2. Ablation <strong>of</strong> VAMP8 in mice resulted in hydronephrosis associatedwith a three to five folds up-regulation <strong>of</strong> AQP2. Forskolin <strong>and</strong> [desamino-Cys1, D-Arg8]-vasopressin (DDAVP)-induced AQP2 exocytosis was impaired in VAMP8-nullcollecting duct cells. VAMP8 was revealed to co-localize with AQP2 on intracellularvesicles <strong>and</strong> to interact with the plasma membrane t-SNARE proteins syntaxin4 <strong>and</strong>syntaxin3, suggesting that VAMP8 may mediate regulated fusion <strong>of</strong> AQP2-positivevesicles with the plasma membrane.49


(33) Wnt3 is Essential for Dorsal Diencephalic DifferentiationCathleen Teh, Michael Richardson, Yin-Ming Lim, Lee-Thean Chu, Igor Kondrychyn<strong>and</strong> Vladimir KorzhThe Wnt signaling pathway has been implicated in dorsal diencephalic differentiation.However, the Wnt involved has not been defined. In zebrafish, wnt3 is persistentlyexpressed in the dorsal diencephalon, specifically, the zona limitans intrathalamica (ZLI),ventral epithalamus <strong>and</strong> thalamus. We developed a zebrafish stable transgenic line,Tg(wnt3:GFP) whose GFP expression during development faithfully recapitulates theexpression pattern <strong>of</strong> endogenous wnt3 at the mRNA level. We showed that during neuraldevelopment Wnt3 acts within the context <strong>of</strong> the canonical Wnt signaling pathway.Morpholino-mediated Wnt3 knockdown results in morphants characterized by atruncated ZLI <strong>and</strong> lack <strong>of</strong> neurogenesis in the ventral epithalamus <strong>and</strong> thalamus, whichparallels the deficient GFP expression in Tg(wnt3:GFP) embryos. In vivo electroporation<strong>of</strong> either human or zebrafish Wnt3 expression constructs into morphant backgroundrescues these neural developmental defects. Taken together, our results demonstrate thatWnt3 plays an evolutionarily conserved role during cell differentiation in the dorsaldiencephalon.50


(34) A regulatory role for Kir/Gem <strong>and</strong> the novel muscle-specific protein CCDC95in myogenesisR.N.Mahalakshmi (WH lab)Kir/Gem, a member <strong>of</strong> the RGK family <strong>of</strong> Ras related small G proteins, is implicated inregulation <strong>of</strong> voltage gate calcium channel activity <strong>and</strong> cytoskeleton remodeling. Thefunctions <strong>of</strong> Kir/Gem are modulated by its subcellular localization, which in turn is underregulation by calmodulin, 14.3.3, importins <strong>and</strong> phosphorylation.In our present study, we identify CCDC95, a novel heart <strong>and</strong> muscle specific protein, asan interacting partner <strong>of</strong> Kir/Gem. CCDC95 modulates the subcellular localization <strong>of</strong>Kir/Gem by effectively mediating its nuclear translocation, independent <strong>of</strong> the importinα5 pathway previously reported by us. It thereby affects the role <strong>of</strong> Kir/Gem incytoskeleton remodeling <strong>and</strong> cell shape changes. In an invitro model <strong>of</strong> muscledifferentiation, Kir/Gem relocalizes from nucleus to cytosol in the initial stages <strong>of</strong>differentiation. Followed by the induction <strong>of</strong> CCDC95 expression in the late stages,Kir/Gem <strong>and</strong> CCDC95 are found to be present in apoptotic nuclei <strong>of</strong> myotubes.The process <strong>of</strong> muscle development is a series <strong>of</strong> well orchestrated events- withdrawal <strong>of</strong>myoblasts from cell cycle, expression <strong>of</strong> myogenic regulatory factors (MRFs) <strong>and</strong>formation <strong>of</strong> multinucleated mytoubes by fusion. Interestingly, overexpression <strong>of</strong>Kir/Gem interferes with the differentiation <strong>of</strong> myoblasts to myotubes. This effect <strong>of</strong>Kir/Gem on myotube formation is overcome by co-expression <strong>of</strong> CCDC95 <strong>and</strong> Kir/Gem.Thus our study unravels a novel function <strong>of</strong> Kir/Gem, in co-ordination with CCDC95, inmuscle development or myogenesis.51


(35) Comparing the antibody responses against recombinant hemagglutinin proteins<strong>of</strong> avian influenza A (H5N1) virus expressed in insect cells <strong>and</strong> bacteriaShen Shuo (CAVR lab)The hemagglutinin (HA) <strong>of</strong> influenza A virus plays an essential role in mediating theentry <strong>of</strong> the virus into host cells. Here, recombinant full-length HA5 protein from a H5N1isolate (A/chicken/hatay/2004(H5N1)) was expressed <strong>and</strong> purified from the baculovirusinsectcell system. As expected, full-length HA5 elicits strong neutralizing antibodies, asevaluated in micro-neutralization tests using HA5 pseudotyped lentiviral particles. Inaddition, we have expressed two fragments <strong>of</strong> HA5 in bacteria <strong>and</strong> found that the N-terminal fragment, covering the ectodomain before the HA1/HA2 polybasic cleavage site,can also elicit neutralizing antibodies. But the C-terminal fragment, which covers theremaining portion <strong>of</strong> the ectodomain, did not. Neutralizing titer <strong>of</strong> the anti-serum againstthe N-terminal fragment is only four times lower than the anti-serum against the fulllengthHA5 protein. Using a novel membrane fusion assay, we further showed that theabilities <strong>of</strong> these antibodies to block membrane fusion correlate well with theneutralization activities.52


(36) JNK-dependent activation <strong>of</strong> p53 <strong>and</strong> c-Jun is required for nitric oxide-inducedapoptosis <strong>of</strong> tumor cellsLei Li, S.H. Lim, G. Aw <strong>and</strong> Alan G Porter<strong>Cell</strong> Death & Human Disease Group, Cancer & Developmental <strong>Biology</strong> Division,<strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>, #07-03B, 61 Biopolis Drive, Proteos,Singapore, e-mail: mcblil@<strong>imcb</strong>.a-star.edu.sgExcessive nitric oxide (NO) contributes to cell death in trauma, strokes <strong>and</strong>neurodegenerative diseases. Here we investigate the mechanisms <strong>of</strong> NO-inducedapoptosis in human neuronal <strong>and</strong> other cell lines. We show that NO-induced, JNKdependentactivation <strong>of</strong> c-Jun by phosphorylation on Ser-63 promotes caspase-3-dependent apoptosis in human SH-Sy5y cells; <strong>and</strong> that p53 is also an essential mediator<strong>of</strong> NO-induced apoptosis in these cells. With published evidence that JNK canphosphorylate various amino acids on p53, we addressed if NO-activated JNK is requiredfor p53 Ser-15 phosphorylation <strong>and</strong> p53 activation. Time course <strong>and</strong> dose responseexperiments in neuroblastoma <strong>and</strong> colon carcinoma cells treated with two different NOdonors demonstrated activation <strong>of</strong> JNK concomitant with or prior to phosphorylation <strong>of</strong>p53 on Ser-15 <strong>and</strong> p53 up-regulation. A JNK chemical inhibitor substantially decreasedJNK activation, p53 Ser-15 phosphorylation, p53 up-regulation, apoptosis, <strong>and</strong>expression <strong>of</strong> the p53 transcriptional targets <strong>of</strong> NO we recently identified - viz. maspin<strong>and</strong> plasminogen activator inhibitor-1 (PAI-1). Neuroblastoma cells expressingdominant-negative JNK2 were relatively resistant to NO-induced apoptosis, exhibitedreduced p53 activation, <strong>and</strong> showed decreases in maspin <strong>and</strong> PAI-1 at both thetranscriptional <strong>and</strong> translational levels. Conversely, in p53-deficient cells JNK was fullyactivated by NO even though the cells were resistant to apoptosis. Altogether our resultssuggest that JNK-dependent activation <strong>of</strong> c-Jun <strong>and</strong> p53 leads to NO-induced apoptosis indifferent tumor cell types.53


(37) Smad1 provides a link between BMP-Smad signaling <strong>and</strong> DNA damageresponse <strong>and</strong> a target for cancer therapyJenny Chau, Yuanyu Hu, Jianhe Wang, <strong>and</strong> Baojie LiThe <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>A*STAR (Agency for Science, Technology <strong>and</strong> Research)SingaporeGenome instability is a major driving force <strong>of</strong> tumorigenesis <strong>and</strong> DNA damage activatedAtm-p53 pathway acts as an anti-cancer barrier by limiting cell growth. Here we reportthat the BMP-Smad pathway acts downstream <strong>of</strong> Atm in DNA damage response, as a p53alternative pathway. Atm phosphorylates Smad1 on Ser240 in the linker region, whichinhibits Smad1 dephosphorylation by PPM1A, leading to Smad1 activation, nuclearretention <strong>and</strong> stabilization, accompanied by transactivation <strong>of</strong> some BMP target genes.Furthermore, when signaling from Atm/Atr to p53 is disrupted in p53 deficient cells,Smad1 is up-regulated, as a backup mechanism. Elevation <strong>of</strong> Smad1 enhances DNAdamage induced apoptosis <strong>and</strong> diminishes cell transformation <strong>and</strong> tumorigenesis, whiledeficiency <strong>of</strong> Smad1 shows opposite effects. Moreover, up-regulation <strong>of</strong> Smad1 is amechanism by which many <strong>of</strong> the non-genotoxic chemotherapeutic drugs kill tumor cells.These results suggest that the BMP-Smad1 pathway is an integral part <strong>of</strong> DNA damageresponse that suppresses oncogenesis, <strong>and</strong> that Smad1 is a target for cancer therapy.54


(38) Harnessing piggyBac Transposons for cancer gene discovery in ZebrafishCanhe Chen 1 , Jolene Y.Z. Ooi 1 , Lifang Li 1 , Sook Yee Wong 1 , Sergey Parinov 2 ,Alex<strong>and</strong>er Emelyanov 2 , Nancy A. Jenkins 1 <strong>and</strong> Neal G. Copel<strong>and</strong> 11 <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>, Singapore 138673; 2 Temasek Life Sciences<strong>Institute</strong>, Singapore 117604Transposon-based insertional mutagenesis <strong>of</strong>fers enormous potential for systematicallyidentifying the genes that cause cancer, which is a prerequisite for developing new targetbasedtherapeutics for treating cancer. While such screens have recently been developedin mice, their cost is a major limitation for most labs. The development <strong>of</strong> such screensin zebrafish would dramatically reduce their cost <strong>and</strong> make it possible to performsaturation screens for new cancer genes in fish. In my experiments, I am making use <strong>of</strong>piggyBac (PB), a novel class II transposon, which can accept large inserts <strong>and</strong> transposein both dividing <strong>and</strong> non-dividing cells. I have shown that PB can transpose in zebrafishat very high frequencies where it preferentially inserts into the coding-region <strong>of</strong> genes,similar to what has been reported for mammalian cells. I have also constructedmutagenic versions <strong>of</strong> the SB transposon that are capable <strong>of</strong> insertionally activatingoncogenes as well as insertionally inactivating tumor suppressor genes <strong>and</strong> generatedzebrafish lines that harbor multiple copies <strong>of</strong> these mutagenic transposons. In addition, Ihave generated a number <strong>of</strong> zebrafish transgenic lines that carry a codon-optimizedversion <strong>of</strong> the PB transposase under the control <strong>of</strong> a chemically inducible promoter.Since the site <strong>of</strong> integration affects PB transposase expression, it has been possible toselect zebrafish transposase lines that vary in their intensity <strong>and</strong> pattern <strong>of</strong> transposaseexpression. I have also made polyclonal <strong>and</strong> monoclonal PB transposase antibodies <strong>and</strong>used them to characterize these zebrafish transposase lines. Finally, I have generatedlarge numbers <strong>of</strong> zebrafish that carry the mutagenic transposase <strong>and</strong> a chemically inducedubiquitously expressed transposase <strong>and</strong> I am currently monitoring these zebrafish fortumor development. If my experiments are successful they have the potential todramatically accelerate cancer gene discovery in zebrafish <strong>and</strong> open up a whole new area<strong>of</strong> zebrafish genetics.55


(39) Stem cell exhaustion due to Runx1 deficiency is prevented by Evi5 activation inleukemogenesisBindya Jacob *1 , Motomi Osato 1, 2 , Namiko Yamashita 1 , Ichiro Taniuchi 3 , Dan Littman 4 ,Norio Asou 5 , Yoshiaki Ito 1, 2*Postdoctoral fellow, Runx lab1 <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>, Singapore, 2 Oncology Research <strong>Institute</strong>,National University <strong>of</strong> Singapore, Singapore 117456, 3 RIKEN, Research Center forAllergy <strong>and</strong> Immunology, Yokohama, Japan, 4 New York University, New York, U.S.A,5 Department <strong>of</strong> Hematology, Kumamoto University School <strong>of</strong> Medicine, Kumamoto,Japan.The balance between the self-renewal <strong>and</strong> differentiation <strong>of</strong> hematopoietic stem cells(HSC) is perturbed in acute leukemia. Quiescence is an alternative state for HSC, evendominant in adult. However, its involvement in leukemogenesis remains poorlyunderstood. We show that conditional deletion <strong>of</strong> Runx1, the most frequently targetedgene in human leukemia, results in an increase <strong>of</strong> HSC due to downregulation <strong>of</strong> CXCR4,a key factor for niche interaction. The weakened niche interaction <strong>of</strong> Runx1-/- HSC failedto sustain quiescence <strong>and</strong> instead facilitated cell cycle entry. The decreased dormancy inturn progressively resulted in stem cell loss, called HSC exhaustion. In the leukemiadevelopment, the stem cell exhaustion was rescued by additional genetic changes, such asoverexpression <strong>of</strong> Evi5. These results reiterate the importance <strong>of</strong> interaction <strong>of</strong> leukemiainitiating cells with niche in leukemogenesis. Our results also emphasize the usefulness<strong>of</strong> retroviral insertional mutagenesis to precisely dissect the multiple steps <strong>of</strong>leukemogenesis: subsequent genetic hits compensate negative aspects given by precedinggenetic alterations. Elucidation <strong>of</strong> these combinatorial mechanisms, in particular stem cellquiescence related mechanism, may provide novel direction for therapeutic applications.56


(40) Cdc25A Serine 123 phosphorylation couples centrosome duplication with DNAreplication <strong>and</strong> regulates tumorigenesisShreeram Sathya (BD lab)The cell division cycle 25A (Cdc25A) phosphatase is a critical regulator <strong>of</strong> cell cycleprogression under normal conditions <strong>and</strong> after stress. Stress-induced degradation <strong>of</strong>Cdc25A has been proposed as a major way <strong>of</strong> delaying cell cycle progression. In vitrostudies pointed towards serine 123 as a key site in regulation <strong>of</strong> Cdc25A stability afterexposure to ionizing radiation (IR). To address the role <strong>of</strong> this phosphorylation site invivo, we generated a knock-in mouse in which serine 123 was substituted for alanine.The Cdc25 Ser123A knock-in mice appeared normal,<strong>and</strong>, unexpectedly, cells derived from them exhibited unperturbed cell cycle <strong>and</strong> DNAdamage responses. In turn, we found that Cdc25A was present in centrosomes <strong>and</strong> thatCdc25A levels were not reduced after IR in knock-in cells. This resulted in centrosomeamplification due to lack <strong>of</strong> induction <strong>of</strong> Cdk2 inhibitory phosphorylation after IRspecifically in centrosomes. Further, Cdc25A knock-in animals appeared sensitive to IRinducedcarcinogenesis. Our findings indicate that Cdc25A Ser123 phosphorylation iscrucial for coupling centrosome duplication to DNA replication cycles after DNAdamage, <strong>and</strong> therefore is likely to play a role in the regulation <strong>of</strong> tumorigenesis.57


(41) TRIM39 is a MOAP-1 Binding Protein That Stabilizes MOAP-1 ThroughInhibition <strong>of</strong> its Polyubiquitination ProcessSan San Lee, Nai Yang Fu, Sunil K. Sukumaran, Kah Fei Wan, Qian Wan, <strong>and</strong> Victor YuBax, a multi-domain pro-apoptotic member <strong>of</strong> the Bcl-2 family, is a key regulator for therelease <strong>of</strong> apoptogenic factors from mitochondria. MOAP-1, which was first isolatedfrom a screen for Bax-associating proteins, interacts with Bax upon apoptotic induction.Gain- <strong>and</strong> loss-<strong>of</strong>-function analyses suggest that MOAP-1 may play an effector role infacilitating the function <strong>of</strong> Bax in mitochondria. MOAP-1 is a short-lived protein that isconstitutively degraded by the ubiquitin-proteasome system. Apoptotic stimuli upregulateMOAP-1 rapidly through inhibition <strong>of</strong> its poly-ubiquitination process. However,cellular factors that play a role in regulating the stability <strong>of</strong> MOAP-1 have not yet beenidentified. In this study, we report the identification <strong>of</strong> TRIM39 as a MOAP-1-associatingprotein. TRIM39 belongs to a family <strong>of</strong> proteins characterized by a Tripartite Motif(TRIM), consisting <strong>of</strong> a RING domain, a B-box <strong>and</strong> a coiled-coil domain. Severalmembers <strong>of</strong> the TRIM family are known to demonstrate E3 ubiquitin ligase activity.Surprisingly, TRIM39 significantly extends the half-life <strong>of</strong> MOAP-1 by inhibiting itspoly-ubiquitination process. TRIM39 localizes predominantly in the cytosol but a portion<strong>of</strong> TRIM39 co-localizes with MOAP-1 in the mitochondria. In agreement with its effecton enhancing MOAP-1 stability, TRIM39 sensitizes cells to etoposide-induced apoptosis.Furthermore, TRIM39 elevates the level <strong>of</strong> MOAP-1 in mitochondria <strong>and</strong> promotescytochrome c release from isolated mitochondria stimulated by recombinant Bax. Takentogether, these data suggest that TRIM39 is a regulator <strong>of</strong> MOAP-1 stability that canpromote Bax-dependent apoptotic function in mitochondria.58


(42) A complex affair between two tumour suppressors <strong>and</strong> a famous regulatorPermeen Yus<strong>of</strong>f, Yim GR Daniel <strong>and</strong> Graeme R. GuySprouty (Spry) proteins are inhibitors <strong>of</strong> the central, cancer-implicated Ras/ERKsignaling pathway <strong>and</strong> have been implicated as likely tumour suppressors. Spry proteinsshare a common C-terminal, cysteine-rich domain <strong>and</strong> two short sequences in the N-terminus The function <strong>of</strong> Spry2, the most pr<strong>of</strong>ound inhibitor <strong>of</strong> the Ras/ERK pathway,appears to be governed by a controlled level <strong>of</strong> phosphorylation on key tyrosine <strong>and</strong>serine residues. Tyr 55 is situated within a conserved, canonical c-Cbl TKB domainbinding site <strong>and</strong> upon receptor tyrosine kinase (RTK) activation c-Cbl binds with highaffinity to this sequence. c-Cbl is a well-characterised docker protein that also hasubiquitin E3 ligase activity <strong>and</strong> appears to be central in the downregulation <strong>of</strong> manyRTKs <strong>and</strong> non-RTKs <strong>of</strong> the Src family. The second conserved sequence consists mainly<strong>of</strong> Ser <strong>and</strong> Thr residues, some or all <strong>of</strong> which are dephosphorylated upon RTK activation,despite the whole protein showing an increased phosphorylation. Evidence indicates thatthe phosphatase responsible for the specific RTK-induced dephosphorylation is theubiquitous PP2A, which also plays an important role in controlling the activity <strong>of</strong> variousproteins within <strong>and</strong> those controlling the Ras/ERK pathway. PP2A, consisting <strong>of</strong> threevariable scaffold, catalytic <strong>and</strong> activator subunits, has also been implicated as playing arole as a tumour suppressor <strong>and</strong> is a major target <strong>of</strong> tumourigenic viruses. PP2A-A, thescaffold subunit binds directly to Spry2, coincidently at the same location as c-Cbl. Thecellular pools <strong>of</strong> Spry2 binding c-Cbl <strong>and</strong> to PP2A are mutually exclusive <strong>and</strong> we arecurrently working on the physiological implication <strong>of</strong> this complex relationship.59


(43) GRIM-19 Is Essential for Maintenance <strong>of</strong> Mitochondrial Membrane PotentialLu Hao (CXM lab)GRIM-19 was found to copurify with complex I <strong>of</strong> mitochondrial respiratory chain <strong>and</strong>subsequently was demonstrated to be involved in complex I assembly <strong>and</strong> activity. T<strong>of</strong>urther underst<strong>and</strong> its function in complex I, we dissected its functional domains bygenerating a number <strong>of</strong> deletion, truncation, <strong>and</strong> point mutants. The mitochondriallocalization sequences were located at the N-terminus. Strikingly, deletion <strong>of</strong> residues70–80, 90–100, or the whole C-terminal region (70–144) led to a loss <strong>of</strong> mitochondrialtransmembrane potential (m). However, similar deletions <strong>of</strong> another two complex Isubunits, NDUFA9 <strong>and</strong> NDUFS3, did not show such effect. We also found that deletion<strong>of</strong> the last 10 residues affected GRIM-19's ability to be assembled to complex I. Weconstructed a dominant-negative mutant containing the N-terminal 60 <strong>and</strong> the last C-terminal 10 residues, which could be assembled into complex I, but failed to maintainnormal m. <strong>Cell</strong>s overexpressing this mutant did not spontaneously undergo cell death, butwere sensitized to apoptosis induced by cell death agents. Our results demonstrate thatGRIM-19 is required for electron transfer activity <strong>of</strong> complex I, <strong>and</strong> disruption <strong>of</strong> m byGRIM-19 mutants enhances the cells' sensitivity to apoptotic stimuli.60


(44) New Controls <strong>of</strong> the NFkB PathwayHsiang Ling Teo (VT lab)The transcription factor NFkB is critical for several cellular <strong>and</strong> developmental events inmetazoans. Given that over 200 physiological stimuli activate NFkB, which in turnregulates an equally large number <strong>of</strong> genes, underst<strong>and</strong>ing how specificity is generated insuch a pleiotropic pathway is also a major challenge. Using a large-scale functionalgenomics approach, parallel interrogation <strong>of</strong> approximately 20,000 sequence-annotatedgenes was carried out <strong>and</strong> several novel modifiers <strong>of</strong> NFkB activity were identified. I willdescribe the characterization <strong>of</strong> one <strong>of</strong> these modulators <strong>and</strong> the mechanisms by which itregulates NFkB signalling.61


(45) Characterization <strong>of</strong> Speedy/RINGO, a new family <strong>of</strong> cyclin-like proteinsXinde Zheng <strong>and</strong> Philipp KaldisCyclin-dependent kinases (Cdk) are master regulators <strong>of</strong> cell cycle progression. Thefunction <strong>of</strong> Cdk is largely decided by the bound cyclins, which determine the substratespecificity. Recently, a new family <strong>of</strong> cyclin-like proteins, Speedy/RINGO, has beenidentified. Similar to classical cyclins, Speedy/RINGO family members can bind <strong>and</strong>activate Cdk1, Cdk2, <strong>and</strong> Cdk3. Through phylogenetic analysis, Speedys can only befound in the species <strong>of</strong> Deuterostome, including sea squirt, bird, fish, mouse, <strong>and</strong> primate.Mouse <strong>and</strong> human have four Speedy genes. Several cell culture studies indicated thatSpeedys may have functions in cell cycle <strong>and</strong> DNA damage control. In Xenopus, Speedycan promote the meiotic maturation. However, their roles on meiosis <strong>and</strong> somatic cellcycle have not been firmly established. We use mouse models to explore their in viv<strong>of</strong>unctions. Conditional knockout mice <strong>of</strong> Speedy A, B1, B2 <strong>and</strong> B3 are being made. Todifferentiate these highly homologous speedy genes, monoclonal antibody <strong>and</strong> peptidepolyclonal antibody are being developed. Testis-specific expression <strong>of</strong> Speedy A, B1 <strong>and</strong>B2 suggest that mammalian Speedys may be key players in meiosis. Ubiquitousexpression <strong>of</strong> Speedy B3 indicates that Speedy may also have functions in the somaticcell cycle. The potential binding partners found in yeast two hybrid screens suggest thatSpeedys have close relation with Cdks, <strong>and</strong> may contribute to the unique chromosomebehavior during meiosis. Interestingly, the only phenotype <strong>of</strong> Cdk2-/- male mice isspermatogenesis block. We hypothesize that at least one <strong>of</strong> the Speedys may be themeiotic partner <strong>of</strong> Cdk2.62


(46) Rb/Cdk2/Cdk4 triple mutant mice elicit a new mechanism for G1/S regulationShuhei Kotoshiba 1 , Weimin Li 2 , <strong>and</strong> Philipp Kaldis 11 <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong> (IMCB), <strong>Cell</strong> Division <strong>and</strong> CancerLaboratory (PRK), 2 University <strong>of</strong> Wisconsin-Madison, Department <strong>of</strong> PharmacologyThe G1/S transition is a well-toned switch in the mammalian mitotic cell cycle. The cellcycle kinases, Cdk2 <strong>and</strong> Cdk4, <strong>and</strong> the rate-limiting tumor suppressor Retinoblastomaprotein (Rb) have been studied in separate animal models leaving an unattendedinteraction between the kinases <strong>and</strong> Rb in vivo. To further dissect the regulation <strong>of</strong> theG1 to S phase progression, we generated Cdk2-/-Cdk4-/-Rb-/- (TKO) mutant mice. Themice died in mid-gestation with major defects in the circulatory systems <strong>and</strong> displayedcombined phenotypes <strong>of</strong> Rb-/- <strong>and</strong> Cdk2-/-Cdk4-/- mutants. However, the TKO MEFswere not only resistant to senescence <strong>and</strong> became immortal but displayed enhanced Sphase entry as well as proliferation rates similar to wild type. The effects were moreremarkable in hypoxia compared to normoxia. Interestingly, depletion <strong>of</strong> the pocketproteins by HPV-E7 in the TKO background elicited a second mechanism for the G1/Sregulation, where p27Kip1 bound to Cdk1 <strong>and</strong> cyclin E complex due to disrupted Skp2-p27Kip1 interactions. Our work indicates that the G1/S transition can be controlled indifferent ways depending on the situation, resembling a regulatory network63


(47) PRL-3 Down-regulates PTEN Expression <strong>and</strong> Signals through PI3K toPromote Epithelial-Mesenchymal TransitionHaihe Wang, Samantha Yiling Quah, Jing Ming Dong, Edward Manser, Jing Ping Tang,<strong>and</strong> Qi ZengPRL-3, a metastasis-associated phosphatase has been demonstrated to increases cellmotility <strong>and</strong> invasiveness with its overexpression. These phenotypic changes arereminiscent <strong>of</strong> the epithelial-mesenchymal transition (EMT) that occurs duringembryonic development <strong>and</strong> oncogenesis <strong>and</strong> is a complex process that converts epitheliainto migratory mesenchymal cells. We here attempt to unravel the underlyingmechanistic basis <strong>of</strong> these phenomena. HeLa cells transiently expressing EGFP-PRL-3(HeLa-PRL-3) exhibit reduced levels <strong>of</strong> paxillin. Similarly, CHO cells stably expressingmyc-PRL-3 (CHO-PRL-3) also show marked reductions in paxillin, phospho-paxillin-Tyr31, <strong>and</strong> vinculin at focal adhesion complexes, <strong>and</strong> notable reductions in the levels <strong>of</strong>RhoA-GTP, Rac1-GTP, <strong>and</strong> F-actin filaments. DLD-1 human colorectal cancer cellsengineered to express EGFP-PRL-3 (DLD-1-PRL-3) underwent changes consistent withEMT. In these cells, PRL-3 activates Akt <strong>and</strong> inactivates GSK-3β as assessed byphosphospesific antibodies. PRL-3 upregulates mesenchymal markers: Fibronectin <strong>and</strong>Snail, <strong>and</strong> downregulates epithelial markers: E-cadherin, γ-catenin (plakoglobin), <strong>and</strong>Integrin β3, which are major effectors in the EMT pathway. The changes in these EMTcharacteristics brought about by PRL-3 can be abrogated by the phosphoinositide 3kinase (PI3K) inhibitor LY294002, implying that PRL-3 acts upstream <strong>of</strong> PI3K <strong>and</strong> couldplay an initiating role to trigger the EMT switch during cancer metastasis. In addition,PRL-3 can downregulate PTEN which is an important antagonist <strong>of</strong> PI3K, furtherreinforcing PI3K/Akt function in PRL-3-triggered EMT. Catalytically inactive PRL-3(C104S) was impaired in the above PRL-3-mediated events, indicating that theseproperties require phosphatase activity. Targeting PRL-3 may thus be a useful strategy toimpede cancer cell invasion <strong>and</strong> metastasis.64


(48) Evolutionary changes in the regulatory region <strong>of</strong> vertebrate erythropoietin genelocusRashmi P Kulkarni (FUGE/SB lab)Erythropoietin (Epo) is the principal regulator <strong>of</strong> red blood cell production. Decreasedoxygen levels induce a dramatic increase in the expression levels <strong>of</strong> Epo gene. Inmammals, Epo is expressed mainly in the fetal liver <strong>and</strong> adult kidney. A highly induciblehypoxia-response element (HRE) has been identified in the 3’-flanking sequences <strong>of</strong>mammalian Epo genes. Interestingly, cloning <strong>of</strong> Epo gene from a teleost fish (fugu)showed that in fishes, the major site <strong>of</strong> Epo expression is the adult heart. Furthermore, 3’-flanking region <strong>of</strong> fugu Epo does not contain a HRE. Thus, the regulatory regions <strong>of</strong> Epolocus seem to have undergone significant changes during the evolution <strong>of</strong> vertebrates. Totrace these evolutionary changes <strong>and</strong> to underst<strong>and</strong> better the regulation <strong>of</strong> Epo gene, wehave characterized tissue-specific enhancers <strong>and</strong> HRE in the fugu Epo locus. In vitrostudies in human Hep3B cells demonstrated that fugu Epo locus contains a HRE 2.3 kbupstream <strong>of</strong> the transcription initiation site, on the opposite str<strong>and</strong> <strong>of</strong> DNA. Using azebrafish reporter-assay system we were able to identify the heart-specific enhancerlocated 1.8 kb upstream <strong>of</strong> the transcription initiation site. To elucidate the molecularchanges that have led to the replacement <strong>of</strong> this heart-specific enhancer by a kidneyspecificenhancer in mammals, we are testing the fugu heart-specific enhancer intransgenic mice. These studies should provide insights into the architecture <strong>and</strong> evolution<strong>of</strong> tissue-specific enhancers in vertebrates.65


(49) <strong>Molecular</strong> therapy : Targeting the p53 pathway using small moleculesChris Brown, Chit Fang Cheok, David P LaneThe tumor suppressor p53 is frequently inactivated in a wide variety <strong>of</strong> cancers. At least50% <strong>of</strong> all tumours carry either point mutations or deletions <strong>of</strong> the p53 gene <strong>and</strong> amongmany <strong>of</strong> the remaining tumours, the signaling pathways <strong>of</strong> p53 is/are defective. One <strong>of</strong>the common lesions in tumours carrying inactivated p53 is the amplification <strong>of</strong> murinedouble minute 2 (MDM2) status, a negative regulator <strong>of</strong> p53. Disruption <strong>of</strong> the p53-MDM2 complex therefore provides an attractive strategy for reactivating p53 functions.Detailed analysis <strong>of</strong> the p53-MDM2 interaction interface provides clues to better designin small molecules targeting this interaction. We use peptide aptamer molecules as leadstructures for drug design to develop more potent <strong>and</strong> specific small molecule inhibitors.Fluorescence polarization-based assays are used to screen <strong>and</strong> validate potential lig<strong>and</strong>sthat bind to the target used for selection.As pro<strong>of</strong>-<strong>of</strong>-concept that inhibitors <strong>of</strong> p53-MDM2 complex have therapeutic value, weWe demonstrate the correlation between p53 activation <strong>and</strong> the induction <strong>of</strong> apoptosis,using an inhibitor <strong>of</strong> p53-MDM2 complex (nutlin-3a). Given that p53 activity is tightlycontrolled by multiple levels <strong>of</strong> regulation, simultaneously targeting these multi-pathwaysmay provide new therapeutic regimes. We explored the concept <strong>of</strong> drug synergism in theactivation <strong>of</strong> p53 responses. The antitumor effect <strong>of</strong> low doses <strong>of</strong> CDK inhibitors issynergistically enhanced by nutlin-3a, resulting in p53-induced apoptosis in varioustumor-derived cell lines. Importantly, the combination is nongenotoxic <strong>and</strong> may be usefulas a therapeutic strategy to circumvent the problems <strong>of</strong> using genotoxic drugs inchemotherapy.66


(50) Crystallographic analysis <strong>of</strong> cap free <strong>and</strong> cap bound eIF4E with N7 alkylatedcap derivativesChristopher J Brown (DPL lab)We present here the first reported crystal structure <strong>of</strong> cap-free human eIF4E. Thisstructure reveals that in the absence <strong>of</strong> m7GTP the cap-binding site undergoes significantstructural changes with the loop containing residue W56 becoming disordered <strong>and</strong> theside-chain <strong>of</strong> W102 rotating out <strong>of</strong> the cap-binding site. The eIF4E complex crystalstructures with Bn7GMP <strong>and</strong> FBn7GMP show that the eIF4E cap-binding pocket isformed by an induced fit mechanism, with the W102 side-chain rotating into analternative position compared to the m7GTP bound structure. The phosphate binding siteremains invariant between the cap-free <strong>and</strong> cap-analogue bound structures which implieselectrostatic steering <strong>of</strong> the cap-analogue into the cap-binding site followed by ordering<strong>of</strong> the W56 loop <strong>and</strong> rotation <strong>of</strong> W102 into the cap binding site. Mass spectrometry wasalso used to determine apparent gas-phase Kd constants <strong>of</strong> 0.15 M, 13.6 M, <strong>and</strong> 55.7M for eIF4E with m7GTP, GTP, <strong>and</strong> GMP, respectively. These measurements showthat for tight <strong>and</strong> specific binding to eIF4E there seems to be a clear requirement forguanosine derivatives to possess both the delocalised positive charge <strong>of</strong> the N7-methylated guanine system <strong>and</strong> at least one phosphate group. We also showed that theN7benzylated monophosphates Bn7GMP <strong>and</strong> FBn7GMP bind eIF4E substantially moretightly than non-N7-alkylated guanosine derivatives (Kd values <strong>of</strong> 7.0 M <strong>and</strong> 2.0 M,respectively). The eIF4E complex crystal structures with Bn7GMP <strong>and</strong> FBn7GMP showthat additional favourable contacts <strong>of</strong> the benzyl groups with eIF4E <strong>and</strong> the localstructured water network contribute binding energy that compensates for the loss <strong>of</strong> thebeta <strong>and</strong> gamma-phosphates. The structural observations made here will be useful in thedesign <strong>and</strong> discovery <strong>of</strong> new families <strong>of</strong> eIF4E inhibitors, which may have potentialtherapeutic applications in cancer. The cap free structure also <strong>of</strong>fers a new pose forvirtual drug docking studies67


(51) Spatio-temporal Mapping <strong>of</strong> Actin Polymerisation Regulated by Actin-BindingProteinsKannan Balakrishnan (BR lab)Actin filaments not only provide mechanical support to the cell but also participate in cellmovement. Assembly <strong>and</strong> disassembly <strong>of</strong> actin filaments are regulated by a variety <strong>of</strong>proteins called the actin-binding proteins. To underst<strong>and</strong> the mechanism <strong>of</strong> cellmovement in health <strong>and</strong> disease, a detailed underst<strong>and</strong>ing <strong>of</strong> the self-assembly <strong>of</strong> actinmonomers into filaments <strong>and</strong> its regulation by the actin-binding proteins is required.Conventional bulk measurements yield ensemble averaged data which does not allow anunderst<strong>and</strong>ing <strong>of</strong> the process at the single molecule level. Total internal reflectionfluorescence (TIRF) microscopy coupled with fluorescence anisotropy <strong>and</strong> spectroscopymeasurements provide a means to follow the actin dynamics with single moleculesensitivity. In the present work, in vitro real-time TIRF assays <strong>of</strong> actin polymerization inthe presence <strong>and</strong> absence <strong>of</strong> actin-binding proteins in combination with anisotropy <strong>and</strong>spectroscopy analyses will be presented.68


(52) Directed Evolution <strong>of</strong> Actin by In Vitro CompartmentalizationMarten E Larsson (BR lab)Actin is a protein involved in many important cellular functions, including musclecontraction, cell motility, cytokinesis <strong>and</strong> is a central protein <strong>of</strong> the cytoskeleton. Actinmonomers (G-actin) rapidly polymerize into heterogeneous filaments (F-actin) in thepresence <strong>of</strong> even low concentration <strong>of</strong> salt. This feature has been a major sumbling blockin elucidating the structure <strong>of</strong> F-actin.Expression <strong>of</strong> soluble, functional actin in E. coli has been difficult partly due to the lack<strong>of</strong> chaperonins needed for the correct folding, <strong>and</strong> perhaps, by the lack <strong>of</strong>posttranslational modifications. Overexpression in other systems like yeast,Dictyostelium discoideum or baculovirus/insect cells have either been lethal, yielded verylow expression or insoluble aggregates. Expression <strong>of</strong> actin with the option <strong>of</strong> addingdifferent purification tags, or functional mutations would be highly desirable forstructural studies <strong>of</strong> F-actin <strong>and</strong> its complexes with actin-binding proteins.Using in vitro transcription/translation <strong>of</strong> r<strong>and</strong>omly mutated biotin-labelled actin cDNAscoupled to streptavidin beads located within the aqueous compartment <strong>of</strong> an oil-in-wateremulsion (in vitro compartmentalization) we are trying to evolve a soluble yet functionalactin. Correctly folded actin will polymerize within the compartment <strong>and</strong> the producedpolymer can be captured by biotin-labelled phalloidin coupled to the beads. Selection <strong>of</strong>highly polymerizable actin is done by breaking open the emulsion, staining withfluorescein-labelled phalloidin <strong>and</strong> subsequent FACS sorting. cDNAs encodingpolymerizable actin can be amplified by PCR <strong>and</strong> used for further rounds <strong>of</strong> selection orfor expression in E. coli <strong>and</strong> functional analyzes.69


(53) Tracking cellular events using stable-isotope labeling by amino acids in cellculture (SILAC)Jayantha Gunaratne, PhD (WB lab)Recent progress in mass spectrometry (MS) technology <strong>of</strong>fers exciting new strategies foradvances in virtually all areas <strong>of</strong> biology. Among the MS strategies that focus onquantitative proteomics, stable (non-radio active) isotope labeling by amino acids in cellculture (SILAC) has emerged as a simple <strong>and</strong> robust approach to solve a wide range <strong>of</strong>biological problems. SILAC eliminates false positives in protein-interaction studies, <strong>and</strong>reveals large-scale dynamics <strong>of</strong> proteomes <strong>and</strong> protein turnover kinetics by adding a timedimension to proteomics. It has become an important assay for measuring changes in thephospho-proteome, ranging from the yeast pheromone pathway to stem-celldifferentiation. As a major research focus in our laboratory we initially established thistechnology by successfully labeling whole proteomes <strong>of</strong> commonly used cancer cell linesfor the study <strong>of</strong> p53, PTEN <strong>and</strong> Evi1. Currently we are examining SILAC’s capability toquantify both the proteome <strong>and</strong> its modifications in response to stimuli <strong>and</strong> perturbationsreflecting the research interests <strong>of</strong> collaborators. In a study to measure gene translation <strong>of</strong>p53 hot-spot mutant R175H transfected into p53 null cells (H1299) we have quantifiedmore than 6000 proteins in the whole proteome using SILAC under basic fractionationconditions. SILAC-based interactomic study <strong>of</strong> tagged PTEN has revealed newinteracting partners with competent removal <strong>of</strong> false positives. In this presentation I willdiscuss the principles, practical stratagems, applications, limitations <strong>and</strong> improvedformats <strong>of</strong> SILAC.70


(54) Synergistic regulation <strong>of</strong> Xanthomonas campestris virulence <strong>and</strong> adaptationby DSF signaling system <strong>and</strong> a novel two-component signal transduction systemRavS/RavR.Ya-Wen HE, Calvin Boon, Lian Zhou, <strong>and</strong> Lian-Hui ZhangThe ability <strong>of</strong> Xanthomonas campestris pv. campestris (Xcc) to incite disease in crucifersdepends upon a number <strong>of</strong> virulence factors including extracellular enzymes <strong>and</strong>extracellular polysaccharides (EPS). Previous work shows that production <strong>of</strong> thesevirulence factors is positively regulated by a DSF quorum sensing system, which acts bymodulating the intercellular content <strong>of</strong> a novel second messenger cyclic di-GMP (c-di-GMP). In this study, the roles <strong>of</strong> 18 genes encoding proteins with HD or EAL domains,which are implicated in cyclic di-GMP degradation, were examined in EPS production<strong>and</strong> extracellular enzyme activity in Xcc. We found that deletion <strong>of</strong> Xcc1958 (ravR),which encodes a response regulator with GGDEF-EAL domains, led to decreased EPSproduction <strong>and</strong> extracellular enzyme activity. Transcriptional analysis showed that ravRis co-transcribed with Xcc1960 (ravS), which encodes a sensor kinase with PAS-PACdomains. Deletion analysis confirmed that the null mutant <strong>of</strong> ravS displayed similarphenotypes as the ravR mutant, suggesting that RavS <strong>and</strong> RavR constitute a twocomponentregulatory system. Microarray analysis revealed that RavS/RavR systemregulates diverse functions. Bioinformatic <strong>and</strong> genetic evidence suggest that RavR mightfunction as a c-di-GMP phosphodiesterase. Further studies suggest that RavS/RavRsystem sense low oxygen conditions <strong>and</strong> regulated EPS production <strong>and</strong> extracellularenzyme activity through the Clp transcriptional regulatory network. Furthermore, genetic<strong>and</strong> biochemical analysis revealed that disruption <strong>of</strong> RavS <strong>and</strong> RavR system did notaffect the functionality <strong>of</strong> DSF signaling system <strong>and</strong> both systems synergisticallyregulated virulence factor production <strong>and</strong> virulence. These results outline a dualsignaling system that may allow Xcc to integrate population density <strong>and</strong> environmentalcues to modulate virulence factor production <strong>and</strong> adaptation.71


(55) Probing the retrograde transport pathway by RNA interference screeningDimitri Moreau, Shin Yi Chew <strong>and</strong> Frederic BardThe cellular secretory route involves an anterograde membrane flow that is compensatedby retrograde transport <strong>of</strong> proteins <strong>and</strong> lipids to maintain organelle homeostasis <strong>and</strong> torecycle components <strong>of</strong> the transport machineries. The retrograde pathway is activebetween the endosomes, the Golgi apparatus <strong>and</strong> the endoplasmic reticulum. Today, thisendogenous process appears to be <strong>of</strong> special importance for the targeted delivery <strong>of</strong> drugsto the cytosol <strong>of</strong> cells. Indeed, the retrograde traffic pathway can provide a route todeliver drugs bypassing the hydrolytic environment <strong>of</strong> the lysosome. However, relativelyfew molecular players in this pathway have been identified. Pseudomonas Exotoxin A(PE) <strong>and</strong> Ricin are two toxins able to travel from the cell surface through the Golgiapparatus <strong>and</strong> the endoplasmic reticulum before translocation to the cytosol where theyblock protein synthesis. Therefore, we have developed a biological assay based on theinhibition <strong>of</strong> protein synthesis induced by the PE <strong>and</strong> Ricin toxins. We used this assay toperform a pilot RNAi screen with the goal to identify missing key genes. We foundseveral genes already known to be involved in the retrograde pathway, like the clathrinheavy chain, the KDEL receptor <strong>and</strong> Rab6A. We also found several potential new genesin this pathway, including MAPK8IP1 (JIP1), which has been involved in neuronalvesicular trafficking. JIP1 is able to link vesicular cargo with the molecular motor kinesin.Various signal transduction pathways regulate JIP1 through phosphorylation, raising theintriguing possibility that retrograde traffic could also be a target <strong>of</strong> regulation byextracellular signals.72


(56) Inhibition <strong>of</strong> PKR Activation <strong>and</strong> Up-regulation <strong>of</strong> GADD34 Expression play asynergistic role in the maintenance <strong>of</strong> de novo Protein Synthesis in Coronavirus-Infected <strong>Cell</strong>sXiaoxingWang, Ying Liao, Kim J Png, Pei Ling Yap, <strong>and</strong> D.X. LiuA diversity <strong>of</strong> strategies is evolved by RNA viruses to manipulate the host translationmachinery in order to create an optimal environment for viral replication <strong>and</strong> progenyproduction. One <strong>of</strong> the common targets <strong>of</strong> the translation machinery is the eukaryoticinitiation factor 2a (eIF2a). In this report, we showed that phosphorylation <strong>of</strong> eIF2α wasseverely suppressed during IBV infection. To underst<strong>and</strong> if this suppression is throughinhibition <strong>of</strong> PKR, the double-str<strong>and</strong>ed RNA dependent kinase that is responsible forphosphorylation <strong>of</strong> eIF2α, cells infected with IBV were analyzed by Western blot. Theresults showed that phosphorylated PKR was greatly reduced in IBV-infected cells.Over-expression <strong>of</strong> a number <strong>of</strong> IBV-encoded RNA-binding proteins results in theinhibition <strong>of</strong> PKR activation, suggesting the involvement <strong>of</strong> these proteins in viralevasion <strong>of</strong> host-antiviral response. Furthermore, GADD34, a component <strong>of</strong> the proteinphosphatase 1 complex that dephosphorylates eIF2α, was significantly induced in IBVinfectedcells. Over-expression <strong>of</strong> GADD34 showed significant enhancement <strong>of</strong> IBVinfection in a dose-dependent manner. Taken together, these results confirm that IBVmay have exploited one combination <strong>of</strong> two mechanisms, i.e. by blocking PKR activation<strong>and</strong> inducing GADD34, to maintain a minimal level <strong>of</strong> de novo protein synthesis duringthe infection cycles.73


(57) DNA damage checkpoint maintains Cdh1 in quasi-active state to preventspindle elongation <strong>and</strong> the completion <strong>of</strong> AnaphaseTao ZhangUS lab, Systems <strong>Biology</strong> Division, <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>DNA damage checkpoint imposes cell cycle arrest to prevent segregation <strong>of</strong> damagedchromosomes <strong>and</strong> allows sufficient time for the damaged chromosomes to be repaired.In yeast, Mec1, Chk1 <strong>and</strong> Rad53 (homologous to human ATM/ATR, Chk1 <strong>and</strong> Chk2kinase, respectively) are among the main effectors <strong>of</strong> this surveillance pathway. Whileactivated Chk1 phosphorylates securin to prevent cohesin cleavage, thus inhibitingchromosome segregation, Rad53 branch inhibits the mitotic exit network (MEN),preventing cells from exiting mitosis. Rad53 also inhibits Cdc5 polo kinase, an essentialcomponents <strong>of</strong> MEN. Restraining cohesin cleavage by stabilizing Pds1 (securin) isconsidered to be the predominant mechanism by which DNA damage checkpointprevents segregation <strong>of</strong> damaged chromosomes. We have uncovered a new regulatorynetwork that prevents segregation <strong>of</strong> damaged chromosomes by suppressing spindleelongation, even after sister-chromatid cohesion is lost. This control circuit involvesRad53, polo kinase, anaphase promoting complex (APC) activator Cdh1 <strong>and</strong> the bimCkinesin family proteins Cin8 (homologous to human Eg5) <strong>and</strong> Kip1. Hence, mitoticspindle emerges as a novel target <strong>of</strong> the DNA damage checkpoint.74


(58) Searching for novel factors involved in neutrophil chemotaxis by chemicalgeneticsXingang Wang <strong>and</strong> Philip W. InghamDevelopmental <strong>and</strong> Biomedical Genetics, IMCBNeutrophil chemotaxis, directed cell movement in response to a gradient <strong>of</strong>chemoattractant, plays an important role in wound healing <strong>and</strong> immune response. Thoughsome factors involved in cell migration have been identified in vitro using primaryneutrophils, <strong>and</strong> HL-60 cells as well as through studies in Dictyostelium discoideum,there is a need to verify <strong>and</strong> extend these finding in an in vivo context. In this study,mpo:GFP transgenic zebrafish - that heave neutrophils labeled with green fluorescenceprotein – have been used as a model to screen for compounds that can affect theirbehavior in vivo. Two compounds have been found that completely inhibit the migration<strong>of</strong> neutrophils towards an injury site after screening a drug library containing about onethous<strong>and</strong> natural extracts isolated from different organisms. Time-lapse imaging analysisshowed that in animals treated with these compounds neutrophils failed to polarize, wereunable to form pseudopod or formed pseudopods in r<strong>and</strong>om directions. Further analysisis under way to determine whether these compounds affect the subcellular localization <strong>of</strong>proteins such as PI3K, Rac <strong>and</strong> Rho which are important in establishing the cell polarity<strong>and</strong> controlling the F-actin polymerization <strong>and</strong> myosin contraction. These compounds arealso being subfractioned <strong>and</strong> further analyzed to determine their molecular structure. Theprotein targets <strong>of</strong> these molecules are to be determined in the future.75


(59) Zebrafish nicastrin is required for midbrain <strong>and</strong> hindbrain developmentShang-Wei Chong 1 , Adam Amsterdam 2 , Haw Siang Ang 3 , He Yang 3 , Nancy Hopkins 2<strong>and</strong> Yun-Jin Jiang 11 Laboratory <strong>of</strong> Developmental Signalling <strong>and</strong> Patterning, <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong><strong>Biology</strong>, 61 Biopolis Dr., Proteos, 138673, Singapore. 2 Center for Cancer Research <strong>and</strong>Department <strong>of</strong> <strong>Biology</strong>, Massachusetts <strong>Institute</strong> <strong>of</strong> Technology, 77 Massachusetts AvenueE17-340, Cambridge, MA 02139, USA. 3 Genome <strong>and</strong> Gene Expression Data AnalysisDivision, Bioinformatics <strong>Institute</strong>, 30 Biopolis Street, Matrix, 138671, SingaporeWe carried out a zebrafish mind bomb (mib) genetic interaction screen <strong>and</strong> discoveredthat one such cross between heterozygous mibta52b X hi1384 produced a low percentage<strong>of</strong> phenotype (


(60) Temporal Notch activation through Notch1a <strong>and</strong> Notch3 is required formaintaining zebrafish rhombomere boundariesXuehui Qiu, Chiaw-Hwee Lim, Kian-Hong Lee <strong>and</strong> Yun-Jin JiangLaboratory <strong>of</strong> Developmental Signalling <strong>and</strong> Patterning, Genes <strong>and</strong> DevelopmentDivision, <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>, A*STAR (Agency for Science,Technology <strong>and</strong> Research), 61 Biopolis Drive, Singapore 138673In vertebrates, hindbrain is subdivided into seven segments termed rhombomeres <strong>and</strong> theinterface between each rhombomere forms the boundary. Similar to the D/V boundaryformation in Drosophila, Notch activation has been shown to regulate the segregation <strong>of</strong>rhombomere boundary cells. Here we further explored the function <strong>of</strong> Notch signaling inthe formation <strong>of</strong> rhombomere boundaries. By using bodipy-ceramide cell labelingtechnique, we found that the hindbrain boundary is formed initially in mib mutants butlost after 24 hpf. This phenotype was more severe in mibta52b allele than in mibtfi91allele. Similarly, injection <strong>of</strong> su(h)-MO led to boundary defects in a dosage-dependentmanner. Though cell proliferation was decreased in mibta52b mutants, inhibition <strong>of</strong> cellproliferation by aphidicolin did not lead to the loss <strong>of</strong> boundary cells. Boundary cellsdifferentiated into neurons at later stages <strong>and</strong> were recovered in mibta52b mutants afterinhibition <strong>of</strong> neurogenesis by injecting hdac1-MO. Furthermore, boundary cells lostsensitivity to reduced Notch activation from 15 somite onwards. We also showed thatknockdown <strong>of</strong> notch3 function in notch1a mutants leads to the loss <strong>of</strong> rhombomereboundary cells <strong>and</strong> causes neuronal hyperplasia, indicating Notch1a <strong>and</strong> Notch3 play aredundant role in the maintenance <strong>of</strong> rhombomere boundary.77


(61) The Characterization <strong>of</strong> Novel mib AllelesKa Fai William TSE*, Qimei NG <strong>and</strong> Yun-Jin JIANGLaboratory <strong>of</strong> Developmental Signalling <strong>and</strong> Patterning, Genes <strong>and</strong> DevelopmentDivision, <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>, A*STAR (Agency for Science,Technology <strong>and</strong> Research), 61 Biopolis Drive, Singapore 138673The Notch signaling pathway is an evolutionarily conserved intercellular signaling pathway thatplays essential roles in a wide range <strong>of</strong> developmental processes, such as pattern formation <strong>and</strong>cell fate determination. Three E3 ubiquitin ligases, Neuralized (Neur), Mind bomb (Mib), <strong>and</strong>Mind bomb 2 (Mib2) are essential in Notch activation by targeting Notch lig<strong>and</strong>, Delta <strong>and</strong>/orSerrate/Jagged for endocytosis. Mib protein contains mib/her2 domain, zz zinc finger domain,<strong>and</strong> Mib repeat in the N-terminus, ankyrin repeats in the middle, <strong>and</strong> three Ring Fingers (RFs) inthe C-terminus. Different mib alleles (mibta52b, mibm132, <strong>and</strong> mibtfi91) in Danio renio had beenidentified <strong>and</strong> characterized in our lab. In brief, the mib mutants display curly-up tail <strong>and</strong>different severity <strong>of</strong> delayed somite development <strong>and</strong> pigmentation. In addition, decreased her4expression <strong>and</strong> increased huC expression were found with different degree. Recently, three newmib mutant alleles, mibSJI-1006, mibSJB-1017, <strong>and</strong> mibSJT-1207 were discovered in our lab bynon-complementation screen. Both mibSJI-1006 <strong>and</strong> mibSJB-1017, showed typical mibphenotypes. Point mutation in mibSJI-1006 generates a premature stop codon (C898stop) in RF2,resulting in a truncated protein without RF3 that is required for Mib E3 ligase activity. FormibSJB-1017, preliminary analysis suggested a point mutation in intron between exon 8 <strong>and</strong> 9generating a splicing variant, which leads to a truncated protein <strong>of</strong> 968 amino acids that deletesRF3. Interestingly, mibSJT-1207 was found to have different morphological phenotypes whencompared to the typical mib alleles, though it did not complement with the existing mib alleles.The mibSJT-1207 embryos, besides its delayed somite development, started early cell death(apoptosis) at 18s stage. In addition, huC expression level was higher. Its stronger neurogenicphenotype <strong>and</strong> the lethal development are different from other mib alleles, which raised aquestion <strong>of</strong> its mutation nature: a new mib allele or a mutation in another gene that can interactwith Mib <strong>and</strong>/or its homolog. By mapping, using the SSLP in LG2, we have found that there is ahigh potential linkage between the mibSJT-1207 <strong>and</strong> a SSLP marker Z13620 (3.32cm away frommib). This result suggested that the new mibSJT-1207 mutant is another mib allele. We are nowworking on the mutation nature <strong>of</strong> this new mib allele.78


(62) Identifying gene regulatory networks that control zebrafish regenerationSemil P. Choksi, Leong Wan Ying, Sudipto Roy<strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>, SingaporeOne <strong>of</strong> the major obstacles in regenerative medicine is directing progenitor cells torestore damaged or diseased tissues. In order to overcome this hurdle, we can look tonature: endogenous regeneration occurs in several complex tissues <strong>of</strong> the zebrafish,Danio rerio. To underst<strong>and</strong> the molecular mechanisms underlying this process, we haveset out to identify networks <strong>of</strong> genes that regulate tissue regeneration in the adultzebrafish.We have performed a c<strong>and</strong>idate-based quantitative reverse transcription <strong>and</strong> PCR screento find transcriptional regulators that are induced during both fin <strong>and</strong> heart regeneration.Using RNA in situ hybridization, we have tracked the expression domains <strong>of</strong> several <strong>of</strong>these genes within the regenerating tissue. Through this analysis we have identified fourgenes which are induced during both fin regeneration <strong>and</strong> heart regeneration.One <strong>of</strong> these genes, LIM domain only 4a (lmo4a), is highly expressed in the underlyingmesenchyme <strong>of</strong> the regenerating fin. Morpholino-based gene knockdown indicates thatlmo4a is critical for fin regeneration. Currently we are performing overexpression <strong>and</strong>genetic interaction studies to further dissect the function <strong>of</strong> this gene. Finally, we will usechromatin immunopreciptiation to identify the targets <strong>of</strong> Lmo4a, placing it into aregeneration-specific transcriptional network.79


(63) The Forkhead domain containing transcription factor Foxj1 is the masterregulator <strong>of</strong> the motile ciliogenic programXianwen Yu, Chee Peng Ng, Hermann Habacher, Sudipto Roy<strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>, SingaporeIn many tissues, motile cilia induce fluid movement through their rhythmic beatingactivity. Dysfunctional motile cilia are the hallmark <strong>of</strong> human genetic abnormalities likePrimary Ciliary Dyskinesia or Kartagener syndrome. In mice, the transcription factorFoxj1 has been implicated in motile cilia formation. However, the regulation <strong>of</strong> Foxj1expression in ciliated tissues <strong>and</strong> its precise role in ciliogenesis have remained unclear.Here, we show that in zebrafish embryos, foxj1 is a novel target <strong>of</strong> Hedgehog signaling inthe floor-plate. Loss <strong>of</strong> Foxj1 severely compromised assembly <strong>of</strong> motile cilia whichdecorate floor-plate cells. Besides the floor-plate, foxj1 is expressed in the embryonicnode <strong>and</strong> pronephric ducts, <strong>and</strong> its function is also necessary for ciliary differentiation inthese tissues. We provide evidence that Foxj1 activates expression <strong>of</strong> a constellation <strong>of</strong>genes essential for motile cilia formation <strong>and</strong> function, <strong>and</strong> that its activity is sufficientfor motile cilia formation. These findings identify a pivotal master regulatory role forFoxj1 in the transcriptional program that controls the production <strong>of</strong> motile cilia.80


(64) A mouse embryonic stem/progenitor cells as a model to investigate asymmetriccell divisionYin Yijun <strong>and</strong> Yang Xiaohang<strong>Cell</strong>ular diversity in every organism is generated from asymmetric cell divisions. Ourunderst<strong>and</strong>ing <strong>of</strong> the molecular mechanism <strong>of</strong> asymmetric cell divisions in animaldevelopment comes mainly from studies in C. elegans <strong>and</strong> Drosophila. However,knowledge <strong>of</strong> asymmetric division in mammal is less studied. We use in vitro culturedmouse embryonic neural stem/progenitor cells as a model to investigate the asymmetriccell division in mammals. Time-lapse imaging demonstrated that 64% cell divisions (55division in 26 cells) were asymmetric based on several parameters in control, but thefrequency <strong>of</strong> asymmetric division decreased to 48% (148 division in 62 cells) whenmouse Inscuteable was knockdown (mInsc-kd). Data from pair-assay using Ki67 as amarker to label the proliferating cells further showed that in control, the percentages <strong>of</strong> p-n (p=positive, n=negative) were 24% (16 pairs), p-p 32% (22 pairs), <strong>and</strong> n-n 44% (30pairs), as compared with p-n 18% (12 pairs), p-p 58% (32 pairs), <strong>and</strong> n-n 31% (20 pairs)in mInsc-kd, respectively. The immunocytochemical staining <strong>of</strong> numb (cell fatedeterminant) at anaphase further showed that the angle between numb crescent staining<strong>and</strong> spindle axis was changed. From these data we conclude that asymmetric divisionoccurs in cultured mouse embryonic neural stem/progenitor cells, <strong>and</strong> there is a shift fromasymmetric to symmetric division in mInsc-kd cells.81


(65) CDK regulates cytokinesis by phosphorylation <strong>of</strong> an IQGAP family protein infungal pathogen C<strong>and</strong>ida albicansChang Run Li, Yan Ming Wang <strong>and</strong> Yue WangC<strong>and</strong>ida albicans <strong>Molecular</strong> & <strong>Cell</strong> <strong>Biology</strong> Laboratory, Genes <strong>and</strong> DevelopmentDivision, <strong>Institute</strong> <strong>of</strong> <strong>Molecular</strong> <strong>and</strong> <strong>Cell</strong> <strong>Biology</strong>Cyclin-dependent kinases (CDKs) drive <strong>and</strong> coordinate multiple cell-cycle events,including construction <strong>and</strong> contraction <strong>of</strong> the actomyosin ring during cytokinesis.However, it remains unclear whether CDKs regulate cytokinesis by directly targetingcomponents <strong>of</strong> the ring. Here we show that Iqg1, an IQGAP family protein, is directlyphosphorylated by CDK1 in vivo <strong>and</strong> in vitro. This phosphorylation event mediates theassembly <strong>and</strong> disassembly <strong>of</strong> the actomyosin ring, in part through the regulation <strong>of</strong> theinteraction between Iqg1 <strong>and</strong> actin-nucleating formins. Furthermore, the phosphorylation<strong>of</strong> Iqg1 regulates the stability <strong>of</strong> Iqg1. These results suggest that CDK1 regulatescytokinesis by directly targeting the actomyosin ring component Iqg1 in C<strong>and</strong>ida albicans.82


(66) Regulation <strong>of</strong> PAK4, a non-conventional PAKYohendran Baskaran (GSK lab)The p21-activated kinase (PAK) family <strong>of</strong> proteins are critical regulators <strong>of</strong> cell signalingdownstream <strong>of</strong> the Cdc42 <strong>and</strong> Rac subfamilies <strong>of</strong> Rho GTPases. The group II <strong>of</strong> PAKs(mammalian PAK4, 5 <strong>and</strong> 6) is less understood both in terms <strong>of</strong> their targets <strong>and</strong> kinaseregulation. Here we define novel mechanisms <strong>of</strong> regulation, not seen in the group IPAKs.The ubiquitous PAK4 can affect the actin cytoskeleton <strong>and</strong> is over-expressed in a number<strong>of</strong> tumours, however the basis for its role in adhesion-independent growth <strong>and</strong> survival isnot understood. We have mapped <strong>and</strong> functionally characterized all the conservedregulatory domains in PAK4 that determine both localization <strong>and</strong> activity <strong>of</strong> the kinase.This includes 14-3-3 binding sites that have pr<strong>of</strong>ound effects on the function <strong>of</strong> the kinase.Using zebrafish as a model system to study kinase function, we show that PAK4operates via kinase-independent mechanisms. This includes PAK4 nuclear-cytoplasmicshuttling which assists in relocalization <strong>of</strong> important PAK4-associated proteins (PAPs).83

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!