12.02.2013 Views

New Modes of GPCR Signalling

New Modes of GPCR Signalling

New Modes of GPCR Signalling

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.

INVITED LIST OF INVITED SPEAKER’S SESSION SPEAKER’S SPEAKER’S ABSTRACT<br />

ABSTRACT<br />

ABSTRACT<br />

<strong>New</strong> <strong>Modes</strong> <strong>of</strong> <strong>GPCR</strong> <strong>Signalling</strong> ……………………………………………………… 20<br />

Joel Bockaert<br />

Regulation <strong>of</strong> Cellular Metabolism by Protein Lysine<br />

Acetylation…………………………23<br />

Kun-Liang Guan<br />

Sensing mGlu Receptor Activation…………………………………………………….......26<br />

Jean-Philippe Pin<br />

mGlu Receptors : Complex Allosteric Machines to Tune Up Synaptic<br />

Transmission………...29<br />

Philippe Rondard<br />

Manipulation <strong>of</strong> Rac Activity and Cell Motility in vivo Using<br />

Light……………………...32<br />

Denise J. Montell<br />

Activation Mechanism <strong>of</strong> GABAB Receptor………………………………………………35<br />

Jianfeng Liu<br />

From Bench to Bedside: BACE1, the Beta-Secretase Enzyme, in Basic Science and<br />

Clinical<br />

Investigation……………………………………………………………………………..38<br />

Yong Shen<br />

TRPA1: a Sentinel for Noxious Chemicals and<br />

Temperatures……………………………...41<br />

Craig Montell<br />

Novel Genes and Pathways that Regulate Alzheimer’s Disease<br />

Pathogenesis……………….44<br />

Huaxi Xu<br />

Structural Feature and Function <strong>of</strong> hβ-subunits <strong>of</strong> BK Channel…………………………43<br />

Jiuping Ding<br />

The Synaptic Mechanisms <strong>of</strong> Glycogen Synathese Kinase-3 in Regulating Leaning and<br />

Memory………………………………………………………………………………….50<br />

Jianzhi Wang<br />

Sensory Transduction in C. elegans: What Can't a Worm<br />

Sense?................................................53<br />

Shawn X. Xu<br />

Spatial Control <strong>of</strong> Synaptic Transmission at a Cholinergic Synapse in C.<br />

elegans………..56<br />

Jean-Louis Bessereau<br />

The Function <strong>of</strong> Phosphatidylinositol-Linked Novel D1 Dopamine Receptor in Central<br />

Nervous System…………………………………………………………………………..59<br />

Jian-Guo Chen


INVITED SESSION ABSTRACT<br />

Mechanisms <strong>of</strong> Wnt Signaling and Regulation…………………………………………...62<br />

Xi He<br />

Acid Sensing Ion Channels: A Novel Therapeutic Target for Anxiety and<br />

Depression?............65<br />

John Wemmie<br />

Sequential Activation <strong>of</strong> Orai1 Subunits by Different Numbers <strong>of</strong> STIM1 Leads to<br />

Multistate Opening <strong>of</strong> the CRAC Channel………………………………………………..68<br />

Tao Xu


LIST OF SOLICITED ABSTRACT<br />

Chemosensory and Behavior <strong>of</strong> C.elegans is Regulated by An E.Coli Noncoding RNA<br />

..................................72<br />

Implication <strong>of</strong> the env Gene <strong>of</strong> the Human Endogenous Retrovirus W Family in the<br />

Expression <strong>of</strong> BDNF and DRD3<br />

..................................73<br />

Microvavle Immobilization <strong>of</strong> Intact C. Elegans for in vivo Calcium Imaging <strong>of</strong><br />

Neuronal Activities in Response to External Stimuli<br />

..................................74<br />

Inhibitory Effect <strong>of</strong> Ethanol on Gustatory Plasticity in C. Elegans Mediated by the<br />

Serotonin Pathway<br />

..................................75<br />

Is<strong>of</strong>orm-Specific Prolongation <strong>of</strong> KCNQ (Kv7) Potassium Channel Opening Mediated<br />

by a <strong>New</strong> Drug-Channel Binding Site<br />

..................................76<br />

Novel USH2A Compound Heterozygous Mutations Cause RP/USH2 in a Chinese<br />

Family<br />

..................................77<br />

UNC-31/CAPS Docks and Primes Dense Core Vesicles in C. elegans Neurons<br />

..................................78<br />

Cordycepin Protects Against Brain ischemical Reperfusion Injury in Mice by Inhibition<br />

<strong>of</strong> Matrix Metalloproteinases-3 Expression.<br />

..................................79<br />

Calcium Signaling in Astrocytes Induced by Photostimulation with Femtosecond Laser<br />

..................................80<br />

ZD7288 Inhibits the Induction Of LTP in an NMDA Independent Manner at<br />

Hippocampal Schaffer Collateral-CA1 Synapses<br />

..................................81<br />

Isolation, Characterization and Anti-Cancer Activity <strong>of</strong> SK84, a Novel Glycine-Rich<br />

Antimicrobial Peptide from Drosophila Virilis<br />

..................................82<br />

Foamy Virus as Potential Vectors for Gene Therapy in Nervous Disorders and Signal<br />

Pathway in Neural Cell Transduction<br />

..................................83<br />

Role <strong>of</strong> Heat on the Expression and Function <strong>of</strong> KCNQ2<br />

..................................84<br />

Possible Involvement <strong>of</strong> TRPV1 in neonatal Febrile Seizure Mice<br />

..................................85<br />

The Kv1.3 Channel Blocker Scorpion Polypeptide has an Therapeutic effect on EAE<br />

Rats with a Molecule Mechanism<br />

..................................86<br />

IGF-1 Promote BPH-1 Cell Proliferation Primarily via Activating mTOR-dependent<br />

Translational Increases in Cyclin D Proteins


..................................88<br />

GABAB Receptor-Mediated Rap1 Activation and GB1/Rap1GTP Interaction in Neuronal<br />

System<br />

..................................89<br />

Therapeutic Effect <strong>of</strong> Flupirtine on RepetitiveFebrile Seizures Rats<br />

..................................90<br />

Rapid Detection <strong>of</strong> Deletion <strong>of</strong> the PMP22 Gene from a Kindred in China with<br />

Hereditary Neuropathy with Liability to Pressure Palsy by Real-time Quantitative PCR<br />

..................................91<br />

Therapeutic Effect <strong>of</strong> Flupirtine on Repetitive Febrile Seizures Rat<br />

..................................92<br />

TEF3(Transcription Enhancer Factor 3)Induced Angiogenesis through VEGF Pathway<br />

..................................93<br />

Does the Metabotropic Glutamate Receptor 7 (Mglur7) Play a Role in Schizophrenia?<br />

..................................94<br />

Effects <strong>of</strong> Yishendaluo Decoction on Axonal Degeneration, Inflammatory Reaction, and<br />

Neurological Function in a Mouse Model <strong>of</strong> Experimental Autoimmune<br />

Encephalomyelitis<br />

..................................95<br />

Imaging Neuronal Population in Vitro with Acousto-Optic Deflector Based Random<br />

Access Two-Photon Microscopy<br />

..................................96<br />

Inhibition <strong>of</strong> Protein Phosphatase 2A Activity Plays a Key Role for Normal Human<br />

Cells to Acquire TRAIL-Sensitive Phenotype during Tumorigenesis<br />

..................................97<br />

A Novel Mutation <strong>of</strong> PAX3 on a Large Chinese Family with Waardenburg Syndrome<br />

..................................98<br />

Identification a Novel MYO6 Mutation Associated with Autosomal Dominant<br />

Non-Syndromic Hearing Impairment in a Large Chinese Family<br />

..................................99<br />

MAPK Scaffolding by BIT1 in the Golgi Complex Modulates Stress Resistance<br />

................................100<br />

Identification <strong>of</strong> a Novel Genetic Locus on Chromosome 8p21.1-q11.23 for Idiopathic<br />

Basal Ganglia Calcification<br />

................................101<br />

Phosphoinositide-3-Kinase Pathway Activation in PTEN Deficient Prostate Cancer<br />

Cells is Independent <strong>of</strong> Receptor Tyrosine Kinases and Mediated by the p110 or p110<br />

Catalytic Subunits<br />

................................102<br />

Insulin-Like Signaling Pathway Functions in Integrative Response to an Olfactory and<br />

a Gustatory Stimuli in Caenorhabditis elegans<br />

................................103<br />

Akt Overexpression Opposes the Beta Amyloid Toxic Injuries to HEK293/tau Cells<br />

................................104<br />

NMDA Receptors Subunit NR2B Messenger Rnas Expression with Cognitive<br />

Dysfunction after Bilateral Common Carotid Artery Occlusion in the Adult Rats


................................105<br />

Effects <strong>of</strong> ZD7288 on Long-Term Potentiation at Perforant Pathway(PP) Fibers - CA3<br />

Region Synapse Pathway in Rat Hippocampus in Vivo<br />

................................106<br />

Effects <strong>of</strong> Cscl on Synaptic Transmission at Perforant Pathway (PP) Fibers - CA3<br />

Region <strong>of</strong> Hippocampus in Rat<br />

................................107<br />

Embryonic Heart Growth is Regulated by Functional Antagonism Between GRIDLOCK<br />

and GATA5 in Zebrafish<br />

................................108<br />

Quantitative Phosphoproteome Analysis Reveals Signaling Events Involved in<br />

Aroma-Stimulated Mouse Olfactory Bulbs<br />

................................109<br />

Quantitative Phosphoproteomics Analysis <strong>of</strong> GABAB Receptor Signaling<br />

................................110<br />

Autotaxin Signaling via Lysophosphatidic Acid Receptors Contributes to LPC-Induced<br />

Vascular Smooth Muscle Cell Proliferation and Migration<br />

................................111<br />

Morphine Regulated Thioredoxin-1 Expression in Neuroblastoma SH-SY5Y Cells<br />

................................112<br />

Cyclin D1 Promotes Anchorage-independent Cell Survival by Inhibiting<br />

FOXO-Mediated Anoikis<br />

................................113<br />

Investigate the Role <strong>of</strong> Rab27 in the Dcvs Docking and Priming Process in C. elegans<br />

................................114<br />

Biophotons as Neural Communication Signals<br />

................................115<br />

Effects <strong>of</strong> Cyclic Nucleotide Gated Channel Knockout on the Olfactory Bulb Analyzed<br />

by Quantitative Phospho-Proteomics<br />

................................116


INVITED<br />

SPEAKER’S<br />

ABSTRACTS


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Joël Bockaert<br />

Pr<strong>of</strong>essor , Director <strong>of</strong> Institut for Functional<br />

Genomics(IGF)Montpellier<br />

UMR5203 – INSERM U661 – UMI - UMII<br />

141 Rue de la Cardonille 34094<br />

Montpellier Cedex 5 FRANCE<br />

Tel : (+33) 4 67 14 29 30<br />

Fax : (+33) 4 67 14 29 10<br />

E-mail: joel.bockaert@igf.cnrs.fr<br />

Education<br />

1961 Entrance examination for the Ecole Normale d'Instituteurs in Mérignac<br />

1963 Baccalauréat <strong>of</strong> Secondary Education : Experimental Science<br />

1964 Physical, Chemical and Natural Sciences (SPCN) at Université de Bordeaux<br />

1966 Licence ès Sciences<br />

1967 Diplôme d'Etudes Supérieures in Natural Sciences<br />

1968 Agrégation in Natural Sciences<br />

1973 Doctorate in Natural Sciences - Honorable distinction<br />

Selected Publications over 497<br />

1. Sladeczek F., Pin J.P., Récasens M., Bockaert J.S.Weiss (1985) Glutamate<br />

stiumates inositol phosphate formation in striatal neurones. Nature 317: 717-719.<br />

611 citations<br />

2. Harris-Warrick R., Hammond C., Paupardin-Tritsch D., Homburger V., Rouot B. et<br />

Bockaert J. (1988) The a subunit <strong>of</strong> GTP binding protein homologous to<br />

mammalina Goa mediates a dopamine-induced decrease <strong>of</strong> calcium current in snail<br />

neurons. Neuron 1: 27-32 120 citations<br />

3. Dumuis A., Sebben M., Haynes L., Pin .P. et Bockaert J. (1988) NMDA receptors<br />

activate the arachidonic acid cascade system in striatal neurons. Nature 336: 68-70.<br />

479 citations<br />

4. Dumuis A, Pin JP, Oomagari K, Sebben M, Bockaert J (1990) Arachidonic acid<br />

released from striatal neurons by joint stimulation <strong>of</strong> ionotropic and metabotropic<br />

quisqualate receptors. Nature 347: 182-4 203 citations<br />

5. Manzoni,O., Prezeau, L., Marin, P., Bockaert, J, et al. Nitric oxide induced<br />

blockade <strong>of</strong> NMDA receptors Neuron 8 : 653- 662 APR 1992 426 citations<br />

6. Lafon-Cazal M., Pietri S., Culcasi M. et Bockaert J. (1993) NMDA-dependent<br />

superoxide production and neurotoxicity. Nature 364: 535-537. 863 citations<br />

7. Spengler D., Waeber C., Pantaloni C., Holsboer F., Bockaert J., Seeburg P.H. et<br />

Journot L. (1993) Differential signal transduction by five splice variants <strong>of</strong> the<br />

PACAP receptor. Nature 265: 170-175 938 citations<br />

8. Pin J.P., Joly C., Heinemann S.F. et Bockaert J. (1994) Domains involved in the<br />

specificity <strong>of</strong> G-protein activation in phospholipase C coupled metabotropic


glutamate receptors. EMBO J. 13: 342-348.204 citations<br />

9. Chavis, P ;, Fagni, L, Lansman,J.B., Bockaert, J (1996) Functional coupling<br />

between ryanodine receptors and L-type calcium channels in neurons Nature 382 :<br />

719-722 194 citations<br />

10. Ango, F, prézeau, L, Muller, T, Worley, P. Pin, J.P., Bockaert, J, Fagni, L (2001)<br />

Agonistindependent activation <strong>of</strong> mGluRs by intracellular protein Homer. Nature<br />

411 :962-965 164 citations<br />

11. Bécamel C, Alonso G, Galéotti N, Demey E, Jouin P, Ullmer C, Dumuis A,<br />

Bockaert J, Marin P (2002) Synaptic multiprotein complexes associated with<br />

5-HT2c receptors: a proteomic approach. EMBO J 21, 2332-42 70 citations<br />

12. Roussignol G, Ango F, Romorini S, Tu JC, Sala C, Worley PF, Bockaert J, Fagni L<br />

(2005) Shank expres-sion is sufficient to induce functional dendritic spine synapses<br />

in spiny neurons. J Neurosci 25, 3560-70.25<br />

13. Jean A, Conductier G, Manrique C, Bouras C, Berta P, Hen R, Charnay Y,<br />

Bockaert J, Compan V. Anorexia induced by activation <strong>of</strong> serotonin 5-HT4<br />

receptors is mediated by increases in CART in the nucleus accumbens. Proc Natl<br />

Acad Sci U S A. 2007 104(41):16335-40.<br />

14. Delcourt N, Thouvenot E, Chanrion B, Galéotti N, Jouin P, Bockaert J, Marin P.<br />

PACAP type I receptor transactivation is essential for IGF-1 receptor signalling and<br />

antiapoptotic activity in neurons. EMBO J. 2007 26(6):1542-51. 2007<br />

15. Bertaso F, Zhang C, Scheschonka A, de Bock F, FontanaudP, Marin P, Huganir RL,<br />

Betz H, Bockaert J, Fagni L, Lerner- Natoli M. PICK1 uncoupling from mGluR7a<br />

causes absence-like seizures. Nat Neurosci. 2008 (8):940-8 .<br />

16. Barthet G, Carrat G, Cassier E, Barker B, Gaven F, Pillot M, Framery B, Pellissier<br />

LP, Augier J, Kang DS, Claeysen S, Reiter E, Banères JL, Benovic JL, Marin P,<br />

Bockaert J, Dumuis A. Betaarrestin1 phosphorylation by GRK5 regulates G<br />

proteinindependent 5-HT4 receptor signalling.EMBO J. 2009 Sep<br />

16;28(18):2706-18.<br />

17. Mitri C, Soustelle L, Framery B, Bockaert J, Parmentier ML, Grau Y.Plant<br />

insecticide L-canavanine repels Drosophila via the insect orphan <strong>GPCR</strong> DmX.<br />

PLoS Biol. 2009 Jun 30;7(6):e1000147.


ABSTRACT<br />

<strong>New</strong> <strong>Modes</strong> <strong>of</strong> <strong>GPCR</strong> <strong>Signalling</strong><br />

Joel Bockaert, Federica Bertaso, Carine Becamel, Maryline Labasque, Mireille<br />

Lerner-Natoli, Laurent Fagni, Philippe.Marin.<br />

CNRS UMR5203,Institut de Génomique Fonctionnelle,Montpellier,France,INSERM,<br />

U661,Montpellier France and University <strong>of</strong> Montpellier<br />

For a long time, we thought that <strong>GPCR</strong>s were part <strong>of</strong> a two-dimentional signaling<br />

pathways. It was believed that ligand-activated <strong>GPCR</strong>s were only activating one or a<br />

limited number <strong>of</strong> heterotimeric G proteins . However, since according to Paul Valery<br />

« what is simple is certainly wrong » the control <strong>of</strong> cell physiology by <strong>GPCR</strong>s is far<br />

more complex. We will illustrate, with 3 examples, this complexity. The first one<br />

concerns a new type <strong>of</strong> transactivation between <strong>GPCR</strong>s and receptor tyrosine kinases<br />

(RTKs). Classically <strong>GPCR</strong>s are known to be transactivated by RTKs. However, there<br />

are some examples <strong>of</strong> the reverse situation. Indeed we provide data showing that<br />

IGF-Rs transactivate PACAP-Rs in neurons. Phosphorylation <strong>of</strong> PACAP-Rs, induced<br />

by IGF1, lead to un-liganded PACAP-Rs activation and their coupling to Gs. The<br />

second one is the key role <strong>of</strong> <strong>GPCR</strong>-associated signalling in fine-tuning <strong>of</strong> their<br />

signalling. mGluR7 negatively control the P/Q channels <strong>of</strong> pre-synaptic terminals an<br />

effect which require absolutely the physical association <strong>of</strong> their c-terminal PDZ ligand<br />

with PICK1. We have shown, in vivo, that disconnecting mGluR7 from PICK1 (with<br />

cell-permeant dominant-negative peptide and knock-in mice) causes absence-like<br />

seizures. The third one is the illustration that <strong>GPCR</strong>s can signal without any G<br />

activation. This will be illustrated here by showing that long-lasting (one hour)<br />

activation <strong>of</strong> ERK pathway by 5-HT2c-Rs is mediated via a complex formed by the<br />

receptor C-terminal associated and calmodulin and -arrestin.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Kun-Liang Guan<br />

PhD, Pr<strong>of</strong>essor<br />

UCSD Moores Cancer Center,<br />

RM5344, 3855 Health Sciences Dr.,<br />

La Jolla, CA 92093-0815.<br />

Tel: 858-822-7945,<br />

E-mail: kuguan@ucsd.edu<br />

Education<br />

1978-1982 Undergraduate student in the Department <strong>of</strong> Biology.<br />

Hangzhou University, China, B.S.<br />

1982-1983 CUSBEA student, Graduate student in Shanghai Institute <strong>of</strong> Plant<br />

Physiology, Shanghai, China<br />

1983-1989 Graduate student in the Department <strong>of</strong> Biochemistry,<br />

Purdue University, West Lafayette, Indiana, Ph.D. (Dec.17, 89)<br />

1989-1991 Post doctoral fellow in the Department <strong>of</strong> Biochemistry,<br />

Purdue University (Mentor, Jack Dixon)<br />

Academic Appointments<br />

1992-1996 Assistant Pr<strong>of</strong>essor, Department <strong>of</strong> Biological Chemistry,<br />

University <strong>of</strong> Michigan<br />

1996-2000 Associate pr<strong>of</strong>essor, Department <strong>of</strong> Biological Chemistry,<br />

University <strong>of</strong> Michigan<br />

2000-2007 Pr<strong>of</strong>essor, Department <strong>of</strong> Biological Chemistry, University <strong>of</strong> Michigan<br />

2002- Visiting pr<strong>of</strong>essor, Department <strong>of</strong> Chemistry and Biochemistry,<br />

University <strong>of</strong> Colorado at Boulder<br />

2003-2007 Research pr<strong>of</strong>essor, Life Sciences Institute, University <strong>of</strong> Michigan<br />

2003-2007 Halvor N. Christensen Collegiate Pr<strong>of</strong>essor in Life Sciences,<br />

University <strong>of</strong> Michigan<br />

2005- Adjunct pr<strong>of</strong>essor, Fudan University, China<br />

2007- Visiting Research pr<strong>of</strong>essor, Life Sciences Institute,<br />

University <strong>of</strong> Michigan<br />

2007- Pr<strong>of</strong>essor, Department <strong>of</strong> Pharmacology,<br />

Member, Moores Cancer Center, University <strong>of</strong> California, San Diego<br />

Selected Publications over 180<br />

1. Guan, K-L., Broyles, S.S., and Dixon, J.E. (1991) Vaccinia virus encodes a Tyr/Ser<br />

protein phosphatase. Nature. 350, 359-362.<br />

2. Zhang, Z., Wang, Y., Vikis, H. G., Anderson, M.W., Sills, R.C., Holliday, W.,<br />

Devereux, T.R., Guan, K-L., and You, M. (2001) Wild type ras can inhibit lung


carcinogenesis in mice. Nature Genetics. 29, 25-33.<br />

3. Inoki, K., Li, Y., Zhu, T., Wu, J. and Guan, K-L. (2002) TSC2 is phosphorylated<br />

and inhibited by AKT and suppresses mTOR signaling. Nature Cell Biol. 4,<br />

648-657. (Selected by Nature Cell Biol as one <strong>of</strong> the landmark papers).<br />

4. Li, W., Lee, J., Vikis, H., et al, and Guan, K-L. (2004) Activation <strong>of</strong> FAK and<br />

Src are receptor proximal events required for netrin signaling. Nature.<br />

Neuroscience. 7, 1213-1221.<br />

5. Kim, E., Goraksha, P., Li, L., Neufeld, T.P., and Guan, K-L. (2008) Regulation <strong>of</strong><br />

TORC1 by Rag GTPases in nutrient response. Nature Cell Biol. 10, 935-945.<br />

6. Chang, J., Wang, Z., Tang, E., Fan. Z., McCauley, L., Franceschi, R., Guan, K-L.,<br />

Kresbach, PH., and Wang, C-Y. (2009) Inhibition <strong>of</strong> Osteoblast functions by<br />

IKK/NFkB in osteoporosis. Nature.Med. 15, 682-9.<br />

7. Inoki, K., Corradetti, and Guan, K-L. (2005) Disease associated with TOR<br />

Dysregulation. Nature Genetics. 37, 19-24.<br />

8. Guan, K-L. and Dixon, J.E. (1990) Protein tyrosine phosphatase activity <strong>of</strong> an<br />

essential virulence determinant in Yersinia. Science. 249, 553-556.<br />

9. Simon, A. R., Vikis, H.G., Fanburg, B.L., Stewart, S., Cochran, B.H., and Guan,<br />

K.-L. (2000) Regulation <strong>of</strong> STAT3 by direct binding to the small G protein Rac1.<br />

Science. 290, 144-147.<br />

10. Zhao, S., Lin, Y., Xu., W., Jiang, W., Zha, Z., Wang, P., Yu, W., Li, Z., Gong, L.,<br />

Peng, Y., Ding, J., Lei, Q., Guan, K-L., Xiong, Y. (2009) Glioma-derived<br />

Mutations in IDH1 Dominantly Inhibit IDH1 Catalytic Activity and Induce HIF-1.<br />

Science. 324, 261-5.<br />

11. Zhao, S., Xu, W., Jiang, W., Yu, W., Lin, Y., Zhang, T., Yao, J., Zeng, Y., Li, H.,<br />

Li, Y., Shi., J., An, W., Hancock, S.M., He., F., Qin, L., Chin, J., Yang, P., Chen,<br />

X., Lei, Q., Xiong, Y., Guan, K-L., (2010) Regulation <strong>of</strong> cellular metabolism by<br />

protein lysine acetylation. Science. 327, 1000-1004.<br />

12. Wang, Q., Zhang, Y., Yang, C., Xiong, H., Lin, Y., Yao, J., Li, H., Xie, L., Zhao,<br />

W., Yao, Y., Ning, Z-B., Zeng, R., Xiong, Y., Guan, K-L., Zhao, S., Zhao, G-P.,<br />

(2010) Acetylation <strong>of</strong> metabolic enzymes coordinates carbon source utilization and<br />

metabolic flux. Science. 327, 1004-1007.<br />

13. Inoki, K., Zhu, T., and Guan, K-L. (2003) TSC2 mediates cellular energy response<br />

to control cell growth and survival. Cell. 115, 577-590.<br />

14. Inoki, K., Ouyang, H., Zhu, T., Lindvall, C., Wang, Y., Yang, Q., Bennett, C.,<br />

Harada, Y., Stankunas, K., Wang, C., He, H., MacDougald, O., You, M., Williams,<br />

B., and Guan, K-L. (2006) TSC2 integrates Wnt and cellular energy signals through<br />

a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth. Cell.<br />

126, 955-68.<br />

15. Mori, H., Inoki, K., Münzberg, H., Opland, D., Faouzi, M., Villanueva, E.C.,<br />

Ikenoue, T., Kwiatkowski, D., MacDougald, O.A., Myers, M.G. Jr., and Guan, K-L.<br />

(2009) Critical role for hypothalamic mTOR activity in energy balance. Cell<br />

Metabolism. 9, 362-74.<br />

16. Zhao, B., Ye, X., Yu, J., Li, L., Li, W., Li, S., Lin, JD., Wang, C- Y., Chinnaiyan,<br />

AM., Lai, Z-C., and Guan. K-L. (2008) TEAD mediates YAP dependent gene<br />

induction and growth control. Genes & Development. 22, 1962-71.


ABSTRACT<br />

Regulation <strong>of</strong> Cellular Metabolism by Protein Lysine Acetylation<br />

Shimin Zhao, Wei Xu , Wenqing Jiang, Wei Yu, Yan Lin, Tengfei Zhang, Qunying Lei,<br />

Yue Xiong and Kun-Liang Guan<br />

Molecular and Cell Biology Laboratory, Institute <strong>of</strong> Biomedical Sciences, Fudan<br />

University, Shanghai 20032, China<br />

Protein lysine acetylation has emerged as a key posttranslational modification in cellular<br />

regulation, in particular through modification <strong>of</strong> histones and nuclear transcription<br />

regulators. We show that lysine acetylation is a prevalent modification in enzymes that<br />

catalyze intermediate metabolism. Virtually every enzyme in glycolysis,<br />

gluconeogenesis, tricarboxylic acid (TCA) cycle, urea cycle, fatty acid metabolism, and<br />

glycogen metabolism was acetylated in human liver tissue. The concentration <strong>of</strong><br />

metabolic fuels, such as glucose, amino acids, and fatty acids, influenced acetylation<br />

status <strong>of</strong> metabolic enzymes. Acetylation activated enoyl-Coenzyme A<br />

hydratase/3-hydroxyacyl-Coenzyme A dehydrogenase (EHHADH) in fatty acid<br />

oxidation and malate dehydrogenase (MDH) in the TCA cycle, inhibited<br />

argininosuccinate lyase (ASL) in the urea cycle, and destabilized phosphoenolpyruvate<br />

carboxykinase (PEPCK) in gluconeogenesis. Our study reveals that acetylation plays<br />

a major role in metabolic regulation.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Jean-Philippe Pin<br />

PhD, Directeur de Recherche (DR1) CNRS<br />

Inst. for Functional Genomics,<br />

CNRS–INSERM –UM1–UM2<br />

Dept <strong>of</strong> Molecular Pharmacology<br />

141 rue de la Cardonille 34094<br />

Montpellier cedex 5–France<br />

Phone: +33 467 14 2988<br />

FAX: +33 467 54 2996<br />

Email: jppin@igf.cnrs.fr<br />

Education<br />

1987 Diplôme de doctorat (Ph. D.),<br />

University <strong>of</strong> Montpellier.<br />

Centre CNRS-INSERM de Pharmacologie- Endocrinologie.<br />

1984 Diplôme d'Etudes Appr<strong>of</strong>ondies (DEA) <strong>of</strong> Molecular Biology<br />

University <strong>of</strong> Montpellier.<br />

Centre CNRS-INSERM de Pharmacologie- Endocrinologie<br />

1983 Master in Physiology- Certificate <strong>of</strong> Biochemistry<br />

Université Pierre et Marie Curie - Paris VI.<br />

1981 Ecole Normale Supérieure de Paris (45, rue d'Ulm)<br />

Selected Publications<br />

1. Sladeczek F, Pin J-P, Récasens M, Bockaert J and Weiss S (1985) Glutamate<br />

stimulates inositol phosphate formation in striatal neurones. Nature. 317: 717-719.<br />

2. Weiss, S., Pin, J.-P., Sebben, M., Kemp, D., Sladeczek, F., Gabrion, J. and<br />

Bockaert, J. (1986) Synaptogenesis <strong>of</strong> cultured striatal neurones in serum-free<br />

medium: a morphological and biochemical study. Proc. Natl. Acad. Sci. (USA),<br />

83:2238-2242.<br />

3. Dumuis, A., Sebben, M., Haynes, L., Pin, J.-P. and Bockaert, J. (1988) NMDA<br />

receptors activate the arachidonic acid cascade system in striatal neurons. Nature.<br />

336:68-70.<br />

4. Dumuis A, Pin J-P, Oomagari K, Sebben M and Bockaert J (1990) Arachidonic<br />

acid released from striatal neurons by joint stimulation <strong>of</strong> ionotropic and<br />

metabotropic quisqualate receptors. Nature. 347: 182-184.<br />

5. Rassendren F A, Lory P, Pin J-P and Nargeot J (1990) Zinc has opposite effects on<br />

NMDA and non-NMDA responses in Xenopus oocytes injected with rat brain RNA.<br />

Neuron, 4:733-740.<br />

6. Pin J-P, Waeber C, Prézeau L, Bockaert J and Heinemann S F (1992) Alternative<br />

splicing generates metabotropic glutamate receptors inducing different patterns <strong>of</strong><br />

calcium release in Xenopus oocytes. Proc. Natl. Acad. Sci. (USA),<br />

89:10331-10335.<br />

7. Pin J-P, Joly C, Heinemann SF and Bockaert J (1994) Domains involved in the


specificity <strong>of</strong> G protein activation in phospholipase C coupled metabotropic<br />

glutamate receptor. EMBO J 13: 342-348.<br />

8. Ango F, Prézeau L, Muller T, Worley PF, Pin J-P, Bockaert J and Fagni L (2001)<br />

Agonist-independent activation <strong>of</strong> mGluRs by the intracellular interacting protein,<br />

Homer. Nature 411: 962-965.<br />

9. Galvez T, Duthey B, Kniazeff J, Blahos J, Rovelli G, Bettler B, Prézeau L and Pin<br />

J-P (2001) Allosteric interactions between GB1 and GB2 subunits are required for<br />

optimal GABAB receptor function. EMBO J 20: 2152-2159.<br />

10. Bessis A-S, Rondard P, Gaven F, Brabet I, Triballeau N, Prézeau L, Acher F and<br />

Pin J-P (2002) Closure <strong>of</strong> the Venus Flytrap module <strong>of</strong> family 3 <strong>GPCR</strong>s is required<br />

for their activation: insights from mutations converting antagonists into agonists.<br />

Proc. Natl. Acad. Sci (USA), 99:11097-11102.<br />

11. Kniazeff J, Bessis A.-S, Maurel D, Ansanay H, Prezeau L and Pin J-P (2004)<br />

Closed state <strong>of</strong> both binding domains <strong>of</strong> homodimeric mGlu receptors is required<br />

for full activity. Nat. Str. Mol. Biol., 11: 706-713.<br />

12. Goudet C, Gaven F, Kniazeff J, Vol C, Liu J, Cohen-Gonsaud M, Acher F, Prézeau<br />

L and Pin J-P (2004) Heptahelical domain <strong>of</strong> metabotropic glutamate receptor 5<br />

behaves like rhodopsin-like receptors. Proc. Natl. Acad. Sci (USA), 101, 378-383.<br />

13. Hlavackova V, Goudet C, Kniazeff J, Zikova A, Maurel D, Vol C, Trojanova J,<br />

Prézeau L, Pin J-P, Blahos J (2005) Evidence for a single heptahelical domain<br />

being turned on upon activation <strong>of</strong> a dimeric <strong>GPCR</strong>. EMBO J. 24:499-509.<br />

14. Damian M, Martin A, Mesnier D, Pin JP, Banères JL (2006) Asymmetric<br />

conformational changes in a <strong>GPCR</strong> dimer controlled by G-proteins. EMBO J.<br />

25:5693-5702.<br />

15. Rondard P, Huang S, Monnier C, Tu H, Blanchard B, Oueslati N, Malhaire F, Li Y,<br />

Maurel D, Trinquet E, Labesse G, Pin* J-P, Liu J (2008) Functioning <strong>of</strong> the dimeric<br />

GABAB receptor extracellular domain revealed by glycan wedge scanning. EMBO<br />

J 27:1321-1332.<br />

16. Maurel D, Comps-Agrar L, Brock C, Rives M-L, Bourrier E, Ayoub MA, Bazin H,<br />

Tinel N, Durroux T, Prézeau L, Trinquet E, Pin J-P (2008) Cell surface<br />

protein-protein interaction analysis with combined time-resolved FRET and<br />

snap-tag technologies: application to <strong>GPCR</strong> oligomerization. Nat Meth 5:561-567.<br />

17. Rives ML, Vol C, Fukazawa Y, Tinel N, Trinquet E, Ayoub MA, Shigemoto R, Pin<br />

JP, Prezeau L (2009) Crosstalk between GABA(B) and mGlu1a receptors reveals<br />

new insight into <strong>GPCR</strong> signal integration. EMBO J. 28:2195-2208.<br />

18. Bartfai T, Benovic J, Bockaert J, Bond R, Bouvier M, Christopoulos A, Civelli O,<br />

Devi L, George S, Inui A, Kobilka B, Leurs R, Neubig R, Pin J-P, Quirion R,<br />

Roques B, Sakmar T, Seifert R, Stenkamp R, Strange P (2004) Twenty Questions<br />

on <strong>GPCR</strong>s. Nat Rev Drug Discovery. 3:577-626.


ABSTRACT<br />

Sensing mGlu Receptor Activation<br />

Jean-Philippe Pin 1, Etienne Doumazane 1, Pauline Scholler 1, Siluo Huang 2, Eric<br />

Trinquet 3, Jianfeng Liu 2 & Philippe Rondard 1<br />

1 CNRS UMR5203, INSERM U661, University <strong>of</strong> Montpellier, Department <strong>of</strong> Molecular<br />

Pharmacology, Institute <strong>of</strong> Functional Genomics, Montpellier, France. 2 Huazhong<br />

University <strong>of</strong> Science and Technology, Wuhan, China, 3 CisBio International,<br />

Bagnols/Cèze , France<br />

Metabotropic glutamate receptors play key role in the modulation <strong>of</strong> many synapses,<br />

acting either on the post-synaptic element from where they control post-synaptic<br />

ionotropic receptor activity, or on pre-synaptic elements where they control<br />

neurotransmiter release. Eight genes encoding mGluRs have been identified in<br />

vertebrate genomes, been classified into 3 groups. Group-I mGluRs (mgluR1 and<br />

mGluR5) are post-synaptic receptors coupled to Gq type <strong>of</strong> G-proteins, therefore<br />

activating phospholipase C, whereas group-II (mGluR2 and mGluR3) and group-III<br />

(mGluR4, 6, 7 and 8) are mostly pre-synaptic receptors coupled to Gi/o types <strong>of</strong><br />

G-proteins. These receptors then, represent new promising targets for he development<br />

<strong>of</strong> new drugs for the treatment <strong>of</strong> many neurologic and psychiatric diseases.<br />

These receptors are constitutive dimers, each subunit being made <strong>of</strong> three major<br />

domains: i) the venus flytrap domain (VFT) where glutamate bind; ii) a cystein-rich<br />

domain (CRD); and iii) the 7 transmembrane domain (7TM) responsible for G-protein<br />

activation. How agonist binding leads to the change in conformation in the 7<br />

transmembrane domain remains unclear. The first crystal structures <strong>of</strong> the mGluR1 VFT<br />

with or without bound agonist revealed two major differences: 1) a closed state <strong>of</strong> the<br />

agonist occupied VFT; and 2) a change in the relative position <strong>of</strong> the two VFTs within<br />

the dimeric structure. It was then postulated that the relative movement between the<br />

VFTs constitutes a key step in receptor activation. However, new crystal structures did<br />

not confirm this change in the relative position. Using cell surface labeling <strong>of</strong> each<br />

subunit with FRET compatible fluorophores, we have been able to monitor, in living<br />

cells, any possible movement between the subunits during receptor activation. Such<br />

assay is very sensitive,simple, and can be used to characterize different ligands acting<br />

on mGluRs: agonists, antagonists, positive or negative allosteric modulators. We even<br />

documented the mechanism <strong>of</strong> action <strong>of</strong> partial agonists.<br />

Our data are consistent with the initial hypothesis for receptor activation. This was<br />

further demonstrated using receptor mutants that either prevent the relative movement<br />

<strong>of</strong> the subunits, or mutants in which the two subunits are locked in the active orientation.<br />

These provide important new information on the functioning <strong>of</strong> these receptors, then<br />

<strong>of</strong>fering new possibilities to develop new types <strong>of</strong> drugs modulating their activity.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Philippe Rondard<br />

PhD, Senior researcher,<br />

Institute for Functional Genomic<br />

Department <strong>of</strong> Molecular Pharmacology<br />

CNRS UMR5203 – INSERM U661 – Universités<br />

Montpellier 1 & 2, 141, rue de la Cardonille<br />

34094 Montpellier Cedex 5 France<br />

Tel: 33 (0)4 67 14 2912<br />

Fax: 33 (0)4 67 54 2432<br />

Email: philippe.rondard@igf.cnrs.fr<br />

Education<br />

1993-1998 Ph.D in Biochemistry.<br />

University <strong>of</strong> Paris VII and Institut Pasteur, Paris, France.<br />

1992-1993 Diplôme d'Etudes Appr<strong>of</strong>ondies (DEA)<br />

University <strong>of</strong> Paris VII, France.<br />

Pr<strong>of</strong>essional Experience<br />

1998-2001 Post-doctoral training in the laboratory <strong>of</strong> Dr. Henry BOURNE,<br />

University <strong>of</strong> California San Francisco (UCSF), San Francisco, USA.<br />

1993-1998 Doctoral training in the laboratory <strong>of</strong> Dr. Hugues BEDOUELLE,<br />

Institut Pasteur, Paris, France.<br />

1995-1996 Military Service as Scientific researcher,<br />

Ecole Normale Supérieure and Institut Curie, Paris.<br />

Selected Publications<br />

1. Monnier C., Dodé C., Fabre L., Teixeira L., Labesse G., Pin J.-P., Hardelin J.-P.<br />

and Rondard P. (2009) PROKR2 missense mutations associated with Kallmann<br />

syndrome impair receptor signalling-activity. Hum. Mol. Genet. 18:75-81.<br />

2. Ahier A., Rondard P., Gouignard N., Khayath N., Huang S., Trolet J., Donoghue<br />

DJ, Gauthier M., Pin J.-P. and Dissous C. (2009) A new family <strong>of</strong> receptor tyrosine<br />

kinases with a venus flytrap binding domain in insects and other invertebrates<br />

activated by aminoacids. PLoS One 4(5):e5651.<br />

3. Rondard P., Huang S., Monnier C., Tu H., Blanchard B., Oueslati N., Malhaire, F.,<br />

Oueslati, N., Li Y., Trinquet E., Labesse G., Pin J.-P. and Liu J. (2008) Functioning<br />

<strong>of</strong> the dimeric GABAB receptor extracellular domain revealed by glycan wedge<br />

scanning EMBO J. 27, 1321-1332.<br />

4. Tu. H.*, Rondard P.*, Xu C., Bertaso F., Cao F., Zhang X., Pin J.-P. and Liu J.<br />

(2007) Dominant role <strong>of</strong> GABAB2 and Gαfor GABAB receptor<br />

mediated-ERK1/2/CREB pathway in cerebellar neurons. Cell signal.19:1996-2002.<br />

* Equally contributed<br />

5. Rondard P., Liu J., Huang S., Malhaire F., Vol C., Pinault A., Labesse, G. and Pin<br />

J.-P. (2006) Coupling <strong>of</strong> agonist binding to effector domain activation in


metabotropic glutamate-like receptors. J. Biol. Chem. 281, 24653-24661.<br />

6. Liu J., Maurel D., Etzol S., Brabet I., Ansanay H., Pin J.-P. and Rondard P. (2004)<br />

Molecular determinants involved in the allosteric control <strong>of</strong> agonist affinity in<br />

GABAB receptor by the GABAB2 subunit. J. Biol. Chem. 279, 15824-15830.<br />

7. Pin J.-P., Kniazeff J., Binet V., Liu J., Maurel D., Galvez T., Duthey B.,<br />

Havlickova M., Blahos J., Prézeau L. and Rondard P. (2004). Activation<br />

mechanism <strong>of</strong> the heterodimeric GABAB receptor. Biochem. Pharmacol. 68,<br />

1565-1572.<br />

8. Pin J.-P., Kniazeff J., Goudet C., Bessis A.-S., Liu J., Galvez T., Acher F., Rondard<br />

P. and Prézeau L. (2004) The activation mechanism <strong>of</strong> class-III G-protein coupled<br />

receptors (2004). Biol. Cell 96, 335-342.<br />

9. Bessis A.-S., Rondard P., Gaven F., Brabet I., Triballeau N., Prézeau L., Acher F.<br />

and Pin J.-P. (2002) Closure <strong>of</strong> the Venus flytrap module <strong>of</strong> mGlu8 receptor and the<br />

activation process: Insights from mutations converting antagonists into agonists.<br />

Proc. Natl. Acad. Sci. U. S. A. 99, 11097-11102<br />

10. Rondard P., Iiri T., Srinivasan S., Meng E., Fujita T. and Bourne HR. (2001).<br />

Mutant G protein �subunit activated by G : a model for receptor activation ?<br />

Proc. Natl. Acad. Sci. U. S. A. 98, 6150-6155.<br />

11. Rondard P. and Bedouelle H. (2000) Mutational scanning <strong>of</strong> a hairpin loop in the<br />

tryptophan synthase subunit, implicated<br />

in allostery and substrate channeling.<br />

Biol. Chem. 381, 1185-1193.<br />

12. Rondard P. and Bedouelle H. (1998) A mutational approach shows similar<br />

mechanisms <strong>of</strong> recognition for the isolated and integrated versions <strong>of</strong> a protein<br />

epitope. J. Biol. Chem. 273, 34753-34759.<br />

13. Rondard P., Brégégère F., Lecroisey A., Delepierre M. and Bedouelle H. (1997a)<br />

Conformational and functional properties <strong>of</strong> an undecapeptide epitope fused with<br />

the C-terminal end <strong>of</strong> the maltose binding protein. Biochemistry 36, 8954-8961.<br />

14. Rondard P., Goldberg M.E. and Bedouelle H. (1997b) Mutational analysis <strong>of</strong> an<br />

antigenic peptide shows recognition in a loop conformation. Biochemistry 36,<br />

8962-8968.


ABSTRACT<br />

mGlu Receptors : Complex Allosteric Machines to Tune Up Synaptic<br />

Transmission<br />

Philippe Rondard1, Etienne Doumazane1, Pauline Scholler1, Eric Trinquet2, Sébastien<br />

Granier1 & Jean-Philippe Pin1<br />

1 CNRS UMR5203, INSERM U661, University <strong>of</strong> Montpellier, Department <strong>of</strong> Molecular<br />

Pharmacology, Institute <strong>of</strong> Functional Genomics, Montpellier, France.<br />

2 CisBio International, Bagnols/Cèze , France<br />

The G-protein coupled receptors activated by the neurotransmitter glutamate (mGluRs)<br />

are made up <strong>of</strong> two subunits covalently linked by a disulfide bridge. Each protomer<br />

comprises an extracellular domain that binds agonists and a transmembrane heptahelical<br />

domain responsible for G-protein activation. The general organization <strong>of</strong> mGluRs at the<br />

cell surface, whether they are limited to dimers, or organized into high-order oligomers,<br />

as well as the functioning <strong>of</strong> the dimeric receptors remain unclear.<br />

Here, we first examined whether mGluRs can assemble into heteromeric complexes. To<br />

that aim we describe a new approach enabling the specific labeling <strong>of</strong> two cell surface<br />

proteins carrying SNAP- or CLIP-tags, with two distinct fluorophores compatible with<br />

time-resolved FRET. This approach allowed us to quantify both homomeric and<br />

heteromeric populations. Our data revealed that some, but not all pairs <strong>of</strong> mGluRs can<br />

indeed form heteromeric entities, and this is further supported by biochemical analysis<br />

and functional complementation studies. By FRET competition and saturation analysis,<br />

we show that these complexes are limited to heterodimers. In addition to describing a<br />

new way to analyze cell surface receptor complexes, our data reveal a new possible<br />

level <strong>of</strong> complexity within the mGluR family.<br />

Second, whereas dimerization <strong>of</strong> the extracellular domains is essential for the activation,<br />

it is not known if the function <strong>of</strong> the transmembrane domains also depends on<br />

dimerization. To address this question, we analyzed the G protein coupling <strong>of</strong> purified<br />

mGluR membrane domain reconstituted into phospholipid bilayer nanodisc with a<br />

controlled stoichiometry: one or two protomers per disc. Nanodiscs containing only one<br />

mGluR membrane domain are able to activate G proteins in response to a positive<br />

allosteric modulator, demonstrating that monomeric mGluR membrane domain can fold<br />

and function independently from the rest <strong>of</strong> the receptor. These results suggest that<br />

dimer requirement in mGluR function originate from an obligate communication<br />

between the extracellular and transmembrane domains in the receptor for activation by<br />

glutamate, rather than a necessity for specific association between the two membrane<br />

domains.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Denise Montell<br />

PhD, Pr<strong>of</strong>essor, Director,<br />

Department <strong>of</strong> Biological Chemistry,<br />

Center for Cell Dynamics<br />

Johns Hopkins School <strong>of</strong> Medicine,<br />

855 North Wolfe Street, Baltimore, MD 21205-2185<br />

Phone: (410) 614-2016<br />

Fax: (410) 614-8375<br />

Email: dmontell@jhmi.edu<br />

Education and Training<br />

1979-1983 B. A.,University <strong>of</strong> California, San Diego, Biochemistry and Cell Biology<br />

1983-1988 Ph.D., Stanford University, Neurosciences<br />

1988-1990 Postdoctoral studies, , Carnegie Institution, Developmental Genetics<br />

Pr<strong>of</strong>essional Experience<br />

1990-1992 Staff Associate, Carnegie Institution, Department <strong>of</strong> Embryology<br />

1992-1998 Assistant Pr<strong>of</strong>essor, Department <strong>of</strong> Biological Chemistry<br />

Johns Hopkins University School <strong>of</strong> Medicine<br />

1998-2002 Associate Pr<strong>of</strong>essor, Department <strong>of</strong> Biological Chemistry<br />

Johns Hopkins University School <strong>of</strong> Medicine<br />

2002- Pr<strong>of</strong>essor, Department <strong>of</strong> Biological Chemistr<br />

Johns Hopkins University School <strong>of</strong> Medicine<br />

1999- Director, Graduate Program in Biological Chemistry<br />

2006- Director, Center for Cell Dynamics.<br />

Selected Publications<br />

1. Geisbrecht, E. and Montell, D. J. (2002) Requirement for Myosin VI in<br />

E-cadherin-mediated border cell migration, Nature Cell Biol 4, 616-620. Cover<br />

photo.<br />

2. Montell, D. J. (2003) Border cell migration: the race is on. Nature Reviews<br />

Molecular Cell Biology 4, 13-24.<br />

3. Naora, H. and Montell, DJ. (2005) Ovarian cancer metastasis: Integrating insights<br />

from disparate model organisms. Nature Reviews Cancer, 5, 355-66.<br />

4. Jang, A. C.-C., Chang, Y.-C., Bai, J. and Montell, D.J. (2009) Border cell migration<br />

requires integration <strong>of</strong> spatial and temporal signals by the BTB protein Abrupt.<br />

Nature Cell Biol 11:569-79.<br />

5. Wang, X., He, L., Wu, Y., Hahn, K. and Montell, D. J. Light-mediated activation<br />

reveals a key role for Rac in collective guidance <strong>of</strong> cell movement in vivo, Nature<br />

Cell Biol, in press.<br />

6. Montell, D. J., Keshishian, H., and Spradling, A. C. (1991) Laser ablation studies <strong>of</strong><br />

the role <strong>of</strong> the Drosophila oocyte nucleus in pattern formation. Science 254,


290-293<br />

7. Montell, D. J. (2008) Morphogenetic Cell Movements: Diversity from<br />

Combinatorial Use <strong>of</strong> Modular Mechanical Properties. Science, 322:1502-5.<br />

8. Johnson, D. M., <strong>New</strong>by, R. F. and Bourgeois, S. (1984) Membrane permeability as<br />

a determinant <strong>of</strong> dexamethasone resistance in murine thymoma cells.Cancer<br />

Research 44, 2435-2440.<br />

9. Montell, D. J. and Goodman, C. S. (1988) Drosophila substrate adhesion molecule:<br />

sequence <strong>of</strong> laminin B1 chain reveals domains <strong>of</strong> homology with mouse. Cell 53,<br />

463- 473.<br />

10. Montell, D. J., Rørth, P. and Spradling, A. C. (1992) slow border cells, a locus<br />

required for a developmentally regulated cell migration during oogenesis encodes<br />

Drosophila C/EBP. Cell 71, 51-62.<br />

11. Murphy, A. M., Lee, T., Andrews, C., Shilo, B.-Z. and Montell, D. J. (1995) The<br />

Breathless FGF receptor homolog, a downstream target <strong>of</strong> Drosophila C/EBP in the<br />

developmental control <strong>of</strong> cell migration. Development 121, 2255-2263.<br />

12. Lee, T., Feig, L. and Montell, D. J. (1996) Two distinct roles for Ras in a<br />

developmentally regulated cell migration. Development 122, 409-418.<br />

13. Liu, Y. and Montell, D. J. (1999) Identification <strong>of</strong> Mutations that Cause Cell<br />

Migration Defects in Mosaic Clones. Development 126, 1869-1878.<br />

14. Montell, D. J. (1999) The Genetics <strong>of</strong> Cell Migration in Drosophila melanogaster<br />

and Caenorhabditis elegans Development. Development 126, 3035-3046<br />

15. Bai, J., Uehara, Y. and Montell, D. J. (2000) Regulation <strong>of</strong> invasive cell behavior<br />

by Taiman, a Drosophila protein related to AIB1, a steroid receptor coactivator<br />

amplified in breast cancer. Cell 103, 1047-1058.<br />

16. Liu, Y. and Montell, D. J. (2001) Jing, a downstream target <strong>of</strong> slbo required for<br />

developmental control <strong>of</strong> border cell migration. Development 128, 321-330.<br />

17. Silver, D. and Montell, D. J. (2001) Paracrine Signaling through the JAK/STAT<br />

Pathway Activates Invasive Behavior <strong>of</strong> Ovarian Epithelial Cells in Drosophila,<br />

Cell 107, 831-841<br />

18. Yoshida, H., Cheng, W., Hung, J., Montell, D., Geisbrecht, E., Rosen, D., Liu, J. and Naora, H.<br />

(2004) Lessons from border cell migration in the Drosophila ovary:A role for myosin VI in<br />

migration <strong>of</strong> human ovarian cancer cells, Proc. Natl. Acad. Sci. (USA) 101,<br />

8144-8149.<br />

19. Montell, D. J. (2006) A Kinase Gets Caspases into Shape. Cell 126, 450-2.<br />

20. Silver, D. L., Naora H., Liu, J., Cheng, W. and Montell, D. J. (2004) Activated<br />

STAT3: localization in focal adhesions and function in ovarian cancer cell motility.<br />

Cancer Research 15, 3550-8.


ABSTRACT<br />

Manipulation <strong>of</strong> Rac Activity and Cell Motility in vivo Using Light<br />

Xiaobo Wang, Li He, Yi Wu, Klaus Hahn and Denise J. Montell<br />

Department <strong>of</strong> Biological Chemistry, Center for Cell Dynamics Johns Hopkins School<br />

<strong>of</strong> Medicine, Baltimore, USA<br />

The small GTPase Rac induces actin polymerization, membrane ruffling and focal<br />

contact formation in cultured single cells, can promote growth cone guidance, dynamic<br />

dendrite growth and has been implicated in learning and memory. Recently,<br />

photoactivatable analogues <strong>of</strong> Rac (PA-Rac) have been developed, allowing rapid and<br />

reversible activation or inactivation <strong>of</strong> Rac using light. In cultured single cells,<br />

light-activated Rac leads to focal membrane ruffling, protrusion and migration. Here we<br />

report that focal activation <strong>of</strong> Rac is effective in vivo. Light-mediated activation <strong>of</strong><br />

Rac in one part <strong>of</strong> one cell caused local ruffling and protrusion. Moreover, activation<br />

or inactivation <strong>of</strong> Rac in one cell <strong>of</strong> a cluster caused a dramatic response in the other<br />

cells, and set the direction <strong>of</strong> movement for the whole group. These results suggest<br />

that the cells sense direction as a group according to relative levels <strong>of</strong> Rac activity.<br />

Communication between cells <strong>of</strong> the cluster required Jun N-terminal kinase (JNK) but<br />

not guidance receptor signalling. These studies further suggest that photoactivatable<br />

proteins are likely to be a new and generally effective class <strong>of</strong> tools for manipulating<br />

local protein activities in vivo.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Jianfeng Liu<br />

PhD, Pr<strong>of</strong>essor, Vice director,<br />

Key laboratory <strong>of</strong> molecular biophysics <strong>of</strong> ministry <strong>of</strong><br />

education<br />

Huazhong University <strong>of</strong> Science and Technology (HUST)<br />

1037, Luoyu Road, Hubei, Wuhan, 430074, P.R.China<br />

Tel : 0086-27-87792031<br />

Fax : 0086-27-87792024<br />

Email : jfliu@mail.hust.edu.cn<br />

Education<br />

1994 -1999 Ph.D. Molecular and Cell Biology,<br />

University <strong>of</strong> Paris XII, Paris, France.<br />

1993 -1994 M.S. Biochemistry and Biotechnology,<br />

University <strong>of</strong> Paris XIII, Paris, France.<br />

1989 -1993 B.S. Biochemistry, Department <strong>of</strong> Biochemistry,<br />

Wuhan University, Wuhan, China<br />

Research Experience<br />

2002- Pr<strong>of</strong>essor, College <strong>of</strong> Life Science and Technology,<br />

HuaZhong University <strong>of</strong> Science and technology, Wuhan, China.<br />

2001-2002 Staff Scientist in Aventis Pharma, Bridgewater, <strong>New</strong> Jersey, USA.<br />

1999-2001 Post-doc Fellow<br />

Institut Cochin Génétique Moléculaire (ICGM), Paris, France.<br />

1994-1999 Research Assistant in the Laboratoire de Recherche sur la Croissance<br />

Cellulaire, la Réparation et la Régénération Tissulaire,<br />

University <strong>of</strong> Paris XII, Creteil, France.<br />

1993-1994 Research assistant, Institut d’oncologie Cellulaire et Moléculaire<br />

Humaine,University <strong>of</strong> Paris XIII, Bobigny, France.<br />

Selected Publications<br />

1. Tu HJ, Xu CJ, Zhang WH, Liu QY, Rondard P, Pin JP, Liu JF. (2010) GABAB<br />

receptor activation protects neurons from apoptosis via IGF-I receptor<br />

transactivation. J. Neurosci., 30(2): 749-759.<br />

2. Cao JH, Huang SL, Qian J, Huang JL, Jin L, Su ZX, Yang J, Liu JF. (2009)<br />

Evolution <strong>of</strong> the class C <strong>GPCR</strong> Venus flytrap modules involved positive selected<br />

functional divergence. BMC Evol Biol., 9:67 .<br />

3. Rondard P, Huang SL, Monnier C, Tu HJ, Blanchard B, Oueslati N, Malhaire F,<br />

Maurel D, Trinquet E, Li Y, Labesse G, Pin JP, Liu JF (2008). Functioning <strong>of</strong> the<br />

dimeric GABAB receptor extracellular domain revealed by glycan wedge scanning.<br />

EMBO J, 27(9):1321-32<br />

4. Li X, Cao JH, Li Y, Rondard P, Zhang Y, Yi P, Liu JF, Nan FJ. (2008)


Activity-based Probe for Specific Photo-affinity Labelling <strong>of</strong> GABAB Receptors in<br />

Living Cells: Design, Synthesis and Biological Evaluation. J. Med. Chem<br />

12;51(11):3057-60.<br />

5. Huang S., Liu JF., Labesse G., Pin J.-P. & Rondard P. Rearrangement <strong>of</strong> the<br />

cystein-rich domains during metabotropic glutamate receptors activation. Manuscrit<br />

in preparation<br />

6. Tu HJ, Rondard P, Xu CJ, Bertaso F, Cao FN, Zhang XY, Pin JP, Liu JF. (2007)<br />

GABAB2 subunit and Gßγ are necessary for GABAB receptor mediated-ERK1/2<br />

phosphorylation in cerebellar granule neurons. Cell. Signal., 19(9):1996-2002.<br />

7. Zhang YL, Zhang HQ, Hua SN, Ma LH, Chen C, Liu XY, Jiang LQ, Yang HM,<br />

Zhang PC, Yu DQ, Guo YL, Tan XH, Liu JF. (2007) Identify two herbal<br />

compounds with potential cholesterol-lowing activity. Biochem. Pharmaco.,<br />

74(6):940-7.<br />

8. Rondard P, Liu JF, Huang SL, Malhaire F, Vol C, Pinault A, Pin JP. (2006)<br />

Coupling <strong>of</strong> agonist binding to effector domain activation in metabotropic<br />

glutamate-like receptors. J. Biol. Chem., 281(34):24653-61. (co-first and<br />

corresponding author)<br />

9. Pin JP, Kniazeff J, Liu JF, Binet V, Goudet C, Rondard P, Prézeau L. (2005)<br />

Allosteric functioning <strong>of</strong> dimeric class C G-protein-coupled receptors. FEBS<br />

Journal., 272(12): 2947-2955.<br />

10. Liu JF, Maurel D, Etzol S, Brabet I, Pin JP, Rondard P.(2004) Molecular<br />

determinants involved in the allosteric control <strong>of</strong> agonist affinity in GABAB<br />

receptor by the GABAB2 subunit. J. Biol. Chem. , 279(16):15824-30.<br />

11. Goudet C, Gaven F, Kniazeff J, Vol C, Liu JF, Cohen-Gonsaud M, Acher F,<br />

Prezeau L, Pin JP. (2004) Heptahelical domain <strong>of</strong> metabotropic glutamate<br />

receptor 5 functions like rhodopsin-like receptors. Proc. Natl. Acad. Sci., 101(1):<br />

378-383.<br />

12. Liu JF, Crepin M, Liu JM, Barritault D, Ledoux D. (2002) FGF-2 and TPA induce<br />

matrix metalloproteinase-9 secretion in MCF-7 cells through PKC activation <strong>of</strong> the<br />

Ras/ERK pathway. Biochem. Biophys. Res. Com., 293: 1174-1182.<br />

13. Liu JF, Chevet E, Lemaitre G, Barritault D, Larose L, Crepin M. (1999)<br />

Functional Rac-1 and Nck signaling networks are required for FGF-2-induced DNA<br />

synthesis in MCF-7 cells. Oncogene., 18: 6425-6433.<br />

14. Liu JF, Issad T, Chevet E, Ledoux D, Courty J, Caruelle JP, Barritault D; Crepin M,<br />

Bertin B. (1998) Fibroblast growth factor-2 has opposite effects on human breast<br />

cancer MCF-7 cell growth depending on the activation level <strong>of</strong> the<br />

mitogen-activated protein kinase pathway. Eur. J. Biochem., 258: 271-276.<br />

15. Liu JF, Bagheri-Yarmand R, Xia YL, Crepin M. (1997) Modulations <strong>of</strong> breast<br />

fibroblast and carcinoma cell interactions by a dextran derivative (CMDB7).<br />

Anticancer Res., 17: 253-258.


ABSTRACT<br />

Activation Mechanism <strong>of</strong> GABAB Receptor<br />

Chanjuan Xu, Wenhua Zhang, Xin Lin, Ming Jiang, Haijun Tu, Siluo Huang, Jianfeng<br />

Liu*<br />

Sino-France Laboratory for Drug Screening, Key Laboratory <strong>of</strong> Molecular Biophysics <strong>of</strong><br />

Ministry <strong>of</strong> Education, School <strong>of</strong> Life Science and Technology, Huazhong University <strong>of</strong><br />

Science and Technology, WuHan, 430074<br />

Class-C G-protein coupled receptors (<strong>GPCR</strong>s) represent a distant group among the large<br />

family <strong>of</strong> <strong>GPCR</strong>s. This class includes the receptors for the main neurotransmitters<br />

including glutamate and γ-aminobutyric acid (GABA), and the receptors for Ca 2+ , some<br />

taste and pheromone molecules, as well as some orphan receptors. Class-C <strong>GPCR</strong>s<br />

possess a heptahelical domain (HD) involved in heterotrimeric G-protein activation, but<br />

most <strong>of</strong> them also have a large extracellular Venus Fly Trap (VFT) domain responsible<br />

for agonist recognition and binding. Class -C <strong>GPCR</strong>s are dimers, either homo or<br />

hetero-dimers. GABAB receptor is consisted <strong>of</strong> GABAB1 and GABAB2 subunits and<br />

was the first heteromeric <strong>GPCR</strong> identified. The VFT domain <strong>of</strong> GABAB1 subunit<br />

contains binding sites for neurotransmitter, agonists or antagonists, while the HD<br />

domain <strong>of</strong> GABAB2 subunit is responsible for G-protein coupling. Therefore, both<br />

subunits are necessary for a functional GABAB receptor. In this study, we demonstrated<br />

the specific role <strong>of</strong> each subunit in ligand recognition, intramolecular transduction,<br />

G-protein activation, and allosteric modulation. We also showed that the specific<br />

activation <strong>of</strong> GABAB receptor leads to G-protein activation, which in turn activate<br />

downstream signaling such as MAPK and PI3K/Akt pathway through trans-activation<br />

<strong>of</strong> a receptor tyrosine kinase (RTK). To the best <strong>of</strong> our knowledge, this is the time to<br />

report the trans-activation <strong>of</strong> a RTK by GABAB receptor.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Yong Shen<br />

PhD, Senior scientist/Pr<strong>of</strong>essor<br />

Haldeman Laboratory <strong>of</strong> Molecular and Cellular<br />

Neurobiology<br />

Sun Health Research Institute<br />

10515 West Santa Fe Drive, Sun City, AZ, 85351, U.S.A.<br />

Phone: (623)876-5456<br />

Fax: (623)876-6688<br />

Email: Yong.Shen@bannerhealth.com<br />

Education<br />

1983 Nanjing University, China BSc. Physiology<br />

1986 Shanghai Inst. <strong>of</strong> Physiology, Academy <strong>of</strong> Sciences MSc. Neurophysiology<br />

1989 State University <strong>of</strong> <strong>New</strong> York (SUNY) Ph.D. Neuroscience<br />

Selected Publications<br />

1. Ewers M, Cheng X, Nural HF, Walsh C, Meindl T, Teipel SJ, Buerger K, Shen Y,<br />

Hampel H. (2010). CSF- BACE1 activity predicts hippocampus atrophy in<br />

Alzheimer’s disease. J Alzheimers Dis, Apr 26.<br />

2. Cheng X., Yang L., He P, Li R., Shen Y. (2010). Differential activation <strong>of</strong> TNF<br />

receptors distinguishes Alzheimer from normal brains. J Alzheimers Dis,<br />

19(2):621-30.<br />

3. Ghosal P., Vogt D, Man L, Shen Y., Lamb B., Pimplikar, SW (2009). Alzheimer's<br />

disease-like pathological features in transgenic mice expressing the APP<br />

intracellular domain. Proc Natl Acad Sci U S A. 106(43): 18367-18377.<br />

4. He, P., Staufenbiel M and Shen, Y. (2008). Interruption <strong>of</strong> β-catenin signaling<br />

reduces neurogenesis in Alzheimer’s disease’s brains. Nature Proceedings,<br />

September, and J.Neurosci. (2009), 29(20):6545-57.<br />

5. The paper is selected as World Faculty 1000 and Feature Article Xia W., Yang T.,<br />

Imelda M. Smith I. M., Shen Y., Walsh D. M. and Dennis J. Selkoe, D. J. (2009). A<br />

specific ELISA for measuring amyloid β-protein oligomers in human plasma and<br />

the brains <strong>of</strong> Alzheimer patients. Archives <strong>of</strong> Neurology, 66(2):190-9.<br />

6. Nural F., He P, Beach T, Sue L, Xia W, and ShenY. (2009). Disassembled DJ-1<br />

high molecular weight complex in cortex in cortex mitochondria from Parkinson’s<br />

disease Patients. Molecular Neurodegeneration.<br />

7. Kozikowski, A. P., Chen, Y., Subahasish, T., Zhong, Z., Melissa A. D’Annibale,<br />

M.A., Shen, Y. and Langley, B.C. (2010). PKC activation and HDAC inhibition -A<br />

dual drug approach to Alzheimer’s disease that reduces Aβ production while<br />

blocking oxidative stress. Proc Natl Acad Sci U S A.. Submitted.<br />

8. Chirapu, S. R, Pachaiyappan, B, Zhong, Z., Abdul-Hay, S. O., Yuan, H., Thatcher,<br />

G., Shen, Y., Kozikowski, A. P, Petukhov, P. A. (2008). Molecular modeling,<br />

Synthesis and Activity Studies <strong>of</strong> Inhibitors Targeting Alzheimer’s disease


β-Secretase (BACE1). Biological Medicinal Chemistry Letters, 19(1): 264-74.<br />

9. Ewers, M., Zhong, Z., Teipel, S., Büger, K., Wallin, A., Blennow, K., and Shen, Y*.<br />

and Hampel, H*.(2008). Increased CSF-BACE1 activity is associated with<br />

ApoE-ε4 genotype in subjects with mild cognitive impairment and Alzheimer’s<br />

disease, Brain, 131:1252-8.<br />

10. Zhong, Z., Ewers, M., Teipel, S., Büger, K., Wallin, A., Blennow, K., Hampel, H.,<br />

and Shen, Y. (2007). High Levels <strong>of</strong> Beta-Secretase (BACE1) in Cerebrospinal<br />

Fluid as a Predictor <strong>of</strong> Risk in Mild Cognitive Impairment, the early stage <strong>of</strong><br />

Alzheimer ’s disease, Archive <strong>of</strong> General Psychiatry, 64(6):718-26.<br />

11. He P, Zhong Z, Lindholm K, Berning L, Lee W, Lemere C, Staufenbiel M, Li R<br />

and ShenY. (2007). Deletion <strong>of</strong> TNF death receptor inhibits amyloid beta<br />

generation and prevents learning and memory deficits in Alzheimer’s mice.<br />

Journal <strong>of</strong> Cell Biology, 178(5): 829-841. Highlighted as “Death receptor takes<br />

central stage” by Nature Neuroscience,October, 2007; Hot story by Science, 2007.<br />

12. Tesco G, Koh YH, Kang EL, Cameron AN, Das S, Sena-Esteves M, Hiltunen M,<br />

Yang SH, Zhong Z, Shen Y, Simpkins JW, Tanzi RE. (2007). Depletion <strong>of</strong> GGA3<br />

stabilizes BACE and enhances beta-secretase activity. Neuron.54(5):721-37.<br />

13. Kim DY, Carey BW, Wang H, Ingano LA, Binshtok AM, Wertz MH, Pettingell<br />

WH, He P, Lee VM, Woolf CJ, Kovacs DM. (2007). BACE1 regulates<br />

voltage-gated sodium channels and neuronal activity. Nature Cell Biol.<br />

9(7):755-64.<br />

14. Yue X, Lu M, Lancaster T, Cao P, Honda S, Staufenbiel M, Harada N, Zhong Z,<br />

Shen Y, Li R. (2005). Brain estrogen deficiency accelerates Abeta plaque formation<br />

in an Alzheimer's disease animal model. Proc Natl Acad Sci U S A.<br />

102(52):19198-203.<br />

15. Li R., Yang LB, Lindholm K, Yan R, Citron M, Beach T, Sue L, Subbagh M., Cai<br />

H., Wong P, Price D, Shen Y.(2004). Aβ load is correlated with elevated BACE<br />

activity in sporadic Alzheimer patients. Proc Natl Acad Sci U S A.),101(10),<br />

3632-3637.<br />

16. Li R., Lindholm K, Yang LB, Konishi Y, Hampel H, Zhang, D. Shen Y. (2004).<br />

TNF death receptor signaling cascade is required for amyloid-β-protein induced<br />

neuron death. J.Neurosci, 28(7), 1760-1771.<br />

17. Yang LB, Lindholm K, Yan R, Citron M, Xia W, Beach T, Sue L, Wong P, Price D,<br />

Li R, Shen Y. (2003). Elevated beta-secretase expression and enzymatic activity<br />

detected in sporadic Alzheimer disease. Nature.Med, 9(1): 3-4<br />

18. Yang, L-B., Lindholm, K., Konishi, Y., Li, R. and Shen, Y. (2002) Targeting<br />

deletion <strong>of</strong> TNF receptor subtypes reveals hippocampal neuron survival and death<br />

through distinct signal transduction pathways. J.Neurosci, 22(8), 3025-3032.<br />

19. Yang, L-B., Li, R., Meri, S., Rogers, J. and Shen, Y. (2000) Deficiency <strong>of</strong><br />

complement protective protein, CD59 in Alzheimer’s brains may contribute to<br />

neurodegeneration. J.Neurosci, 20(20), 7505-7509.


ABSTRACT<br />

From Bench to Bedside:<br />

BACE1, the Beta-Secretase Enzyme, in Basic Science and Clinical<br />

Investigation<br />

Yong Shen<br />

Robert Haldeman Laboratory <strong>of</strong> Molecular and Cellular Neurobiology<br />

Banner Sun Health Research Institute<br />

β-Secretase, or β-site amyloid precursor protein cleaving enzyme 1 (BACE1), has been<br />

identified as the rate-limiting enzyme for amyloid-beta (Aβ) peptide production that<br />

occurs primarily in neurons in the brain. Aβis the main component <strong>of</strong> amyloid plaques<br />

and vascular deposits in Alzheimer's disease (AD) brains, and it is believed to initiate<br />

the deadly amyloid cascade that leads to neurodegeneration and dementia. BACE1 is<br />

the principle beta-secretase, as its knock-out completely prevents Aβ generation.<br />

BACE1 is likely to process a number <strong>of</strong> different substrates, and consequently, it may<br />

exert several independent physiological functions in neurons. Currently, however, the<br />

physiological functions <strong>of</strong> BACE1 are not clear. Multiple cellular mechanisms for<br />

BACE1 regulation, including transcriptional regulation, neuronal excitability, receptor<br />

mediation, and identification <strong>of</strong> new molecules as biomarkers for AD and molecular<br />

targeting for Aβ lowering therapies will be discussed.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Craig Montell<br />

PhD, Pr<strong>of</strong>essor, Director<br />

Departments <strong>of</strong> Biological Chemistry and Neuroscience<br />

The Johns Hopkins University School <strong>of</strong> Medicine<br />

855 N. Wolfe St. Baltimore, MD 21205<br />

Tel: (410) 955-1199;<br />

FAX: (410) 287-7672<br />

E-mail: cmontell@jhmi.edu<br />

Education<br />

1978 B.A., Department <strong>of</strong> Bacteriology, University <strong>of</strong> California, Berkeley<br />

1983 Ph.D., Department <strong>of</strong> Microbiology, University <strong>of</strong> California, Los Angeles<br />

Honorary Degree<br />

2010 March Catholic University, Leuven, Belgium<br />

Appointments<br />

1983-1988 Postdoctoral Fellow, Dept. <strong>of</strong> Biochemistry,<br />

University <strong>of</strong> California, Berkeley, CA.<br />

1988-1994 Assistant Pr<strong>of</strong>essor,Dept. <strong>of</strong> Biological Chemistry &Neuroscience,<br />

The Johns Hopkins University, School <strong>of</strong> Medicine, Baltimore, MD.<br />

1994-1998 Associate Pr<strong>of</strong>essor, Dept. <strong>of</strong> Biological Chemistry &Neuroscience,<br />

The Johns Hopkins University, School <strong>of</strong> Medicine, Baltimore, MD.<br />

2000-2006 Director, BCMB Graduate Program<br />

1998- Pr<strong>of</strong>essor, Dept. <strong>of</strong> Biological Chemistry &Neuroscience,<br />

The Johns Hopkins University, School <strong>of</strong> Medicine, Baltimore, MD.<br />

Selected Publications over 100<br />

1. Montell, C., Fisher, E.F., Caruthers, M.H. and Berk, A.J. 1982. Resolving the<br />

functions <strong>of</strong> overlapping viral genes by site-specific mutagenesis at a mRNA splice<br />

site. Nature 295, 380-384.<br />

2. Montell, C., Fisher, E.F., Caruthers, M.H. and Berk, A.J. 1983. Inhibition <strong>of</strong> RNA<br />

cleavage but not polyadenylation by a point mutation in mRNA 3' consensus<br />

sequence AAUAAA. Nature 305, 600-605.<br />

3. Montell, C. 2000. PLC fills a GAP in G protein coupled signaling. Nature Cell<br />

Biol. 2, E82-83.<br />

4. Montell, C. 2000. A PDZ protein Ushers in new links. 2000. Nature Genet. 26,<br />

6-7.<br />

5. Montell, C., Courtois, G., Eng, C., and Berk, A.J. 1984. Complete transformation<br />

by adenovirus 2 requires both E1A proteins. Cell 36, 951-961.<br />

6. Montell, C. and Rubin, G. 1988. The Drosophila ninaC locus encodes two


photoreceptor cell specific proteins with domains homologous to protein kinases<br />

and the myosin heavy chain head. Cell 52, 757-782.<br />

7. Bloomquist, B. T., Shortridge, R. D., Schneuwly, S., Perdew, M., Montell, C.,<br />

Steller, H., Rubin, G. and Pak, W. L. 1988. Isolation <strong>of</strong> a putative phospholipase C<br />

gene <strong>of</strong> Drosophila, norpA, and its role in phototransduction. Cell 54, 723-733.<br />

8. Xu, X.Z., Li, H.S., Guggino, W.B. and Montell, C. 1997. Coassembly <strong>of</strong> TRP and<br />

TRPL produces a distinct store-operated conductance. Cell 89, 1155-1164.<br />

9. Montell, C., Birnbaumer, L. and Flockerzi, V. 2002. The TRP channels, a<br />

remarkably functional family. Cell 108, 595-598.<br />

10. Montell, C. 2005. The latest waves in calcium signaling. Cell 122, 157-163.<br />

11. Montell, C. 2007. Dynamic regulation <strong>of</strong> the INAD signaling scaffold becomes<br />

crystal clear. Cell 131, 19-21.<br />

12. Venkatachalam, K., Long, A.A., Elsaesser, R., Nikolaeva, D., Broadie, K. and<br />

Montell, C. 2008. Motor deficit in a Drosophila model <strong>of</strong> mucolipidosis type IV<br />

due to defective clearance <strong>of</strong> apoptotic cells. Cell 135, 838-851.<br />

13. Montell, C. 2010. Preventing a perm with TRPV3. Cell (in press)<br />

14. Montell, C., Jones, K., Hafen, E., and Rubin, G. 1985. Rescue <strong>of</strong> the Drosophila<br />

phototransduction mutation trp by germline transformation. Science 230,<br />

1040-1043.<br />

15. Porter, J.A., Yu, M., Doberstein, S.K., Pollard, T.S. and Montell, C. 1993.<br />

Dependence <strong>of</strong> calmodulin localization in the retina on the NINAC unconventional<br />

myosin. Science 262, 1038-1042.<br />

16. Arnon, A., Cook, B., Montell, C., Selinger, Z. and Minke, B. 1997. Calmodulin<br />

regulation <strong>of</strong> calcium stores in phototransduction <strong>of</strong> Drosophila. Science 275,<br />

1119-1121.<br />

17. Shen, W., Kwon, Y. and Montell, C. 2010. Rhodopsin required for temperature<br />

discrimination in Drosophila. Science (in revision)<br />

18. Kwon, Y, Shim, H.S., Wang, X. and Montell, C. 2008. Control <strong>of</strong> thermotactic<br />

behavior via coupling <strong>of</strong> a TRP channel to a phospholipase C signaling cascade.<br />

Nat. Neurosci. 11, 871-873.<br />

19. Lee, Y., Moon, S.J. and Montell, C. 2009. Multiple gustatory receptors required for<br />

the caffeine response in Drosophila. Proc. Natl. Acad. Sci. U.S.A. 106, 4495-4500.<br />

20. Jiao, Y., Moon, S.J. and Montell, C. 2007. A Drosophila gustatory receptor<br />

required for the responses to sucrose, glucose, and maltose identified by mRNA<br />

tagging. Proc. Natl. Acad. Sci. U.S.A. 104, 14110-14115<br />

21. Liu, Z, Wang, X, Yu, Y, Li, X., Wang, T., Jang, H., Ren, Q., Jiao, Y., Sawa, A.,<br />

Moran, T., Ross, C.A., Montell, C. and Smith, W.W. 2008. A Drosophila model for<br />

LRRK2-linked Parkinsonism. Proc. Natl. Acad. Sci. U.S.A. 105, 2693-2698.<br />

22. Elsaesser, R., Kalra, D., Li, R. and Montell, C. 2010. Light-induced translocation <strong>of</strong><br />

Drosophila visual Arrestin2 depends on Rac2. Proc. Natl. Acad. Sci. U.S.A. 107,<br />

4740-4745.<br />

23. Kim, S. H., Lee, Y., Akitake, B., Woodward, O. M., Guggino, W. B. and Montell,<br />

C. 2010. Drosophila TRPA1 channel mediates chemical avoidance in gustatory<br />

receptor neurons. Proc. Natl. Acad. Sci. U.S.A. (in press)


ABSTRACT<br />

TRPA1: a Sentinel for Noxious Chemicals and Temperatures<br />

Craig Montell<br />

Department <strong>of</strong> Biological Chemistry, The Johns Hopkins University School <strong>of</strong> Medicine<br />

TRP channels endow us with the ability to sense tastants, mechanical stimuli, a variety<br />

<strong>of</strong> chemicals, distinct temperatures and in some animals, light and pheromones. In this<br />

seminar, I will begin with the identification <strong>of</strong> the founding member <strong>of</strong> the TRP family,<br />

which functions in fly phototransduction, and the realization that TRP channels are<br />

global and evolutionarily conserved mediators <strong>of</strong> sensory signaling in all animals.<br />

I will describe our recent studies demonstrating that the ability <strong>of</strong> Drosophila larva to<br />

discriminate between the optimal temperature <strong>of</strong> 18°C and slightly higher temperatures<br />

depends on the TRPA1 channel, which functions downstream <strong>of</strong> a<br />

phototransduction-like cascade. We propose that activation <strong>of</strong> TRPA1 via a signaling<br />

cascade, rather than through direct thermal activation, promotes amplification <strong>of</strong> small<br />

differences in temperature and facilitates adaptation to temperatures within the<br />

comfortable range.<br />

I will summarize our work demonstrating that insect TRPA1 is a target for insect<br />

repellents. Our studies identifies insect TRPA1 as a potential target for developing<br />

improved repellents to reduce insect-borne diseases such as malaria, which cause<br />

significant morbidity and mortality.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Huaxi Xu<br />

PhD, Pr<strong>of</strong>essor and Acting Director<br />

Neurodegenerative Disease Program<br />

Center for Neuroscience, Stem Cell and Aging Research<br />

Burnham Institute for Medical Research<br />

10901 N. Torrey Pines Road<br />

La Jolla, California 92037.<br />

Tel: 858-795-5246<br />

Fax:858-597-5273<br />

E-mail: xuh@burnham.org<br />

Education<br />

1981-1985 Xiamen University, China. B.S. in Biochemistry.<br />

1987-1988 Visiting Scholar. Division <strong>of</strong> Rheumatology.<br />

University <strong>of</strong> Connecticut Health Center, Farmington, CT.<br />

1988-1993 Ph.D. Dept. <strong>of</strong> Anatomy and Structural Biology.<br />

Albert Einstein College <strong>of</strong> Medicine, <strong>New</strong> York.<br />

1993-1994 Post-Doctoral Fellow. Dept.<strong>of</strong> Developmental and Molecular Biology,<br />

Albert Einstein College <strong>of</strong> Medicine.<br />

1994-1998 Post-Doctoral Associate, Research Associate.<br />

The Rockefeller University, <strong>New</strong> York, NY.<br />

Joint Appointment in Dept. <strong>of</strong> Neurology & Neuroscience,<br />

Cornell University Medical College, <strong>New</strong> York, NY.<br />

1998-2003 Assistant Pr<strong>of</strong>essor, Fisher Center for Research on Alzheimer Disease,<br />

The Rockefeller University, <strong>New</strong> York, NY.<br />

2003-2008 Associate Pr<strong>of</strong>essor, Neurodegenerative Disease Program<br />

Burnham Institute for Medical Research<br />

2008- Pr<strong>of</strong>essor and Acting Director,Neurodegenerative Disease Program<br />

Burnham Institute for Medical Research<br />

Selected Publications<br />

1. Xu, H., Sweeney, D., Greengard, P., and Gandy, S. (1996). Metabolism <strong>of</strong><br />

Alzheimer ß-Amyloid Precursor Protein: Regulation by Protein Kinase A in Intact<br />

Cells and in a Cell-Free System. Proc. Natl. Acad. Sci. USA. 93:4081-4084.<br />

2. Xu, H., Sweeney, D., Wang, R., Thinakaran, G., Lo, A. C. Y., Sisodia, S.,<br />

Greengard, P., and Gandy, S. (1997) Generation <strong>of</strong> Alzheimer ß-amyloid<br />

Protein in the trans-Golgi Network in the Apparent Absence <strong>of</strong> Vesicle Formation.<br />

Proc. Natl. Acad. Sci. USA. 94:3748-3752.<br />

3. Xu, H., Gouras, G. K., Greenfield, J. P., Vincent, B, Naslund, J., Mazzarelli, L,<br />

Jovanovic, J. N, Seeger, M, Relkin, N. R, Liao, F., Checler, F., Buxbaum, J. D,<br />

Chait, B. T., Thinakaran, G., Sisodia, S., Wang, R., Greengard, P., and Gandy, S.<br />

(1998). Estrogen Reduces Neuronal Generation <strong>of</strong> Alzheimer ß-amyloid Peptides.<br />

Nature.Med 4:447-451.


4. Greenfield, J. P., Tsai, J., Gouras, G. K., Hai, B., Thinakaran, G., Checler, F.,<br />

Sisodia, S. S., Greengard, P., and Xu, H. (1999). Endoplasmic Reticulum and<br />

trans-Golgi Network Generate Distinct Populations <strong>of</strong> Alzheimer ß-Amyloid<br />

Peptides. Proc. Natl. Acad. Sci. USA 96:742-747.<br />

5. Gouras, G. K.*, Xu, H.*, Gross, R., Greenfield, J. P., Hai, B., Wang, R., and<br />

Greengard, P. (2000) Testosterone Reduces Neuronal Secretion <strong>of</strong> Alzheimer<br />

ß-amyloid Peptides. Proc. Natl. Acad. Sci. USA 97:1202-1205. (*, equal<br />

contribution).<br />

6. Dou, F., Netzer, W. J., Tanemura, K., Li, F., Hartl, F.U., Takashima, A., Gouras<br />

G.K., Greengard, P., and Xu, H. (2003) Chaperones increase association <strong>of</strong> tau<br />

protein with microtubules. Proc. Natl. Acad. Sci. USA 100:721-726.<br />

7. Netzer, W.J., Dou, F., Cai, D., Veach, D., Jean, S., Li, Y., Bornmann, W.G.,<br />

Clarkson, B., Xu, H., and Greengard, P. (2003) Gleevec inhibits -amyloid<br />

production but not Notch cleavage. Proc. Natl. Acad. Sci. USA<br />

100(21):12444-12449..<br />

8. Cai, D., Zhong, M., Wang, R., Netner, W.J., Shields, D., Zheng, H., Sisodia S.S.,<br />

Foster, D.A., Gorlick, S.F., Xu, H.*, and Greengard, P*. (2006) Phospholipase D1<br />

corrects impaired APP trafficking and neurite outgrowth in familial Alzheimer’s<br />

disease-linked presenilin-1 mutant neurons. Proc. Natl. Acad. Sci. 103:1936-1940.<br />

9. Li, H.L., Wang, H.H., Liu, S., Deng, Y.Q., Zhang, Y.J., Tian, Q., Wang, X.C.,<br />

Chen, X.Q., Yang, Y., Zhang, J.Y., Wang, Q., Xu, H., Liao, F.F., and Wang, J.Z.<br />

(2007) Phosphorylation <strong>of</strong> tau antagonizes apoptosis by stabilizing -catenin. Proc.<br />

Natl. Acad. Sci. 104:3591-3596.<br />

10. Zhang, Y-w., Wang, R., Liu, Q., Zhang, H., Liao, F-F., and Xu, H. (2007)<br />

Presenilin/ -secretase-dependent processing <strong>of</strong> APP regulates EGFR expression.<br />

Proc. Natl. Acad. Sci. 104:10613-10618.<br />

11. Liu, Y., Zhang, Y-w., Wang, X., Zhang, H., You, X., Liao, F-F. and Xu, H. (2009)<br />

Intracellular trafficking <strong>of</strong> Presenilin 1 is regulated by -amyloid precursor protein<br />

and phospholipase D1. J. Biol. Chem. 284(18):12145-52.<br />

12. Ma, T., Zhao, Y.B., Kwak, Y-D., Thompson, R., Luo, Z., Xu, H., and Liao, F-F.<br />

(2009) Statin's excitoprotection is mediated by sAPP and the subsequent attenuation<br />

<strong>of</strong> calpain-induced truncation events likely via Rho-ROCK signaling. J. Neurosci.<br />

29:11226-36.<br />

13. Zhang, Y-w., Liu, L., Zhang, X., Li, W-B., Chen, Y., Huang, X., Sun, L., Luo,<br />

W-j., Netzer, W.J., Threadgill, R., Wiegand, G., Wang, R., Cohen, S.N.,<br />

Greengard, P., Liao, F-F., Li, L., and Xu, H. (2009) A Functional Mouse<br />

Retroposed Gene Rps23r1 Inhibits Alzheimer’s -Amyloid Generation and Tau<br />

Phosphorylation. Neuron. 64(3):328-340.<br />

14. Dunys, J., Sevalle, J., Giaimei, E., Pardossi-Piquard, R., Vitek, M.P., Renbaum,<br />

P., Levy-Lahad, E., Zhang, Y-w., Xu, H., Checler, F., and Alves da Costal, C.<br />

(2009) p53-dependent control <strong>of</strong> PEN-2 promoter transactivation by presenilins. J.<br />

Cell. Sci. 122:4003-4008.<br />

15. Zhang, Y-w., and Xu, H. (2010) Substrate check for gamma-secretase. Nat. Struct.<br />

& Mol. Biol. 17(2):140-141.


ABSTRACT<br />

Novel Genes and Pathways that Regulate Alzheimer’s Disease<br />

Pathogenesis<br />

Huaxi Xu<br />

Neurodegenerative Disease Research Program, Sanford-Burnham Medical Research<br />

Institute,10901 N. Torrey Pines Rd.,La Jolla, CA 92037<br />

Alzheimer's disease (AD) is the most common neurodegenerative disorder, afflicting<br />

millions <strong>of</strong> aged people in the world. Extraneuronal plaques consisting <strong>of</strong> β-amyloid<br />

(Aβ) peptides, intracellular neur<strong>of</strong>ibrillary tangles (NFTs) composed <strong>of</strong><br />

hyperphosphorylated paired helical filaments <strong>of</strong> the tau protein, and extensive neuronal<br />

loss are major pathological hallmarks <strong>of</strong> AD. Multiple lines <strong>of</strong> evidence suggest that<br />

overproduction/aggregation <strong>of</strong> the neurotoxic Aβ peptide in the brain is a prime culprit<br />

in AD pathogenesis, causing dramatic neuronal death directly responsible for the<br />

cognitive decline in AD patients. Hyperphosphorylated tau has also been found in many<br />

other neurodegenerative diseases (tauopathies). Hence, knowledge <strong>of</strong> mechanisms<br />

underlying Aβ generation, tau hyperphosphorylation, and neuronal cell death is crucial<br />

for disease intervention; identification <strong>of</strong> new genes/proteins involved in these<br />

processes is a major goal <strong>of</strong> AD research. Our group has recently identified two new<br />

players important in AD: 1) RPS23R1, whose gene originated through retroposition <strong>of</strong><br />

the ribosomal protein S23 gene, can reduce Aβ level and tau phosphorylation in cell<br />

cultures and alleviate these AD-like pathologies in the APP/PS1/tau triple transgenic<br />

AD mouse model. Detailed mechanistic studies revealed that RPS23R1 can interact<br />

with adenylate cyclases to increase cAMP synthesis, which upregulates PKA activity,<br />

inhibiting GSK-3 activity which is important in modulating Aβ generation and tau<br />

phosphorylation. 2) Appopstosin is an APP-interacting protein that we identified<br />

through yeast-two-hybrid screens. When overexpressed, appoptosin can induce intrinsic<br />

caspase-dependent apoptosis. Importantly, we find that the level <strong>of</strong> appoptosin is<br />

upregulated in neurons treated with Aβ, glutamate, or ischemia insults as well as in<br />

AD brains. Furthermore, downregulation <strong>of</strong> appoptosin can protect neurons against<br />

Aβ- and glutamate-induced neurotoxicity.<br />

Together our studies demonstrate that RPS23R1 is beneficial for inhibiting Aβ<br />

generation and tau phosphorylation, whereas appoptosin-mediated apoptosis plays an<br />

important role in neurotoxicity-induced cell death and neurodegenerative diseases.<br />

These studies will provide new therapeutic possibilities for AD treatment.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Jiu-ping Ding<br />

PhD, Pr<strong>of</strong>essor,<br />

Key laboratory <strong>of</strong> Molecular Biophysics <strong>of</strong> the Ministry <strong>of</strong><br />

Education, College <strong>of</strong> Life Science and Technology<br />

Huazhong University <strong>of</strong> Science and Technology<br />

1037 Luoyulu St.,Wuhan, Hubei, 430074 P.R.China<br />

Tel: 86-27-8779-2153<br />

Fax: 86-27-8779-2024<br />

Email: jpding@mail.hust.edu.cn<br />

Education<br />

1982 M.S (Astrophysics), Nanjing University, Nanjing, China.<br />

1968 B.S. (Electronics), University <strong>of</strong> Science and Technology <strong>of</strong> China, Beijing,<br />

China.<br />

Working experiences<br />

1985-1987 Associated Pr<strong>of</strong>essor, Nanjing Railway Medical College.<br />

1987-1989 Visiting Scholar, SUNY at Buffalo, NY, USA<br />

1989-1992 Research Associated, Dept. <strong>of</strong> biology,<br />

Washington University, St. Louis, MO, USA<br />

1992-2002 Research Associated, Dept <strong>of</strong> Anaesthesiology,<br />

Washington University School <strong>of</strong> Medicine, St. Louis, MO, USA<br />

2002- Pr<strong>of</strong>essor,<br />

Huazhong University <strong>of</strong> Science and Technology, Wuhan, China.<br />

Research Experience<br />

1992- Research associate in Dept. <strong>of</strong> Anesthesiology,<br />

Washington University School <strong>of</strong> Medicine, St. Louis. MO.<br />

1989-1992 Research associate in Dept. <strong>of</strong> Biology,<br />

Washington University, St. Louis, MO.<br />

1987-1989 Visiting Scholar in Biophysics,<br />

State University <strong>of</strong> <strong>New</strong> York at Buffalo, NY.<br />

Selected Publications<br />

1. Liang Sun, Yu Xiong, Xuhui Zeng, Ying Wu, Na Pan, Christopher J. Lingle, Anlian<br />

Qu and Jiuping Ding. Differential Regulation <strong>of</strong> Action Potentials by Inactivating<br />

and Noninactivating BK Channels in Rat Adrenal Chromaffin Cells. Biophys. J.,<br />

2009, (In press).<br />

2. Ying Wu, Yu Xiong, Sheng Wang, Hong Yi, Hui Li, Na Pan, Frank T Horrigan<br />

Yingliang Wu and Jiuping Ding. Intersubunit coupling in the Pore <strong>of</strong> BK channels.<br />

J. Biol. Chem., 2009,284(35): 23353-23363.<br />

3. Zhaohua Guo, Caixia Lv, Hong Yi, Yu Xiong, Yingliang Wu, Wenxin Li, Tao Xu


and Jiuping Ding. A residue at the cytoplasmic entrance <strong>of</strong> BK-type channels<br />

regulating single-channel opening by its hydrophobicity. Biophys. J., 2008, 4 (9):<br />

3714-3725.<br />

4. Lv, C.X., Chen, M.R., Gan, G.L., Wang, L.F. Xu, T. and Ding, J.P.. Four-turn a<br />

helical segment prevents surface expression <strong>of</strong> an auxiliary hβ2 subunit <strong>of</strong> BK-type<br />

channel. J. Biol. Chem., 2008, 283(5): 2709-2715.<br />

5. Geliang Gan, Hong Yi, Maorong Chen, Liang Sun, Wenxin Li, Yingliang Wu and<br />

Jiuping Ding. Structural basis for toxin-resistance <strong>of</strong> 4 associated<br />

calcium-activated potassium (BK) channels. J. Biol. Chem., 2008, 283:<br />

24177-24184.<br />

6. Maorong Chen, Geliang Gan, Ying Wu, Lu Wang, Yingliang Wu and Jiuping Ding.<br />

Lysine-rich extracellular gate formed by hβ2 subunits reduces the single-channel<br />

conductance <strong>of</strong> BK channels. PLoS ONE,2008,<br />

7. Shang-bang GAO, Ying WU, Cai-xia LÜ, Zhao-hua GUO, Chen-hong LI, Jiu-ping<br />

DING. Slack and Slick KNa channels are required for the depolarizing<br />

afterpotential <strong>of</strong> acutely isolated, medium diameter rat dorsal root ganglion neurons.<br />

Acta Pharmacologica Sinica, 2008, 29(8): 899-905.<br />

8. Xiao-Hui Sun, Jiu-Ping Ding, Hui Li, Na Pan, Lu Gan, Xiang-Liang Yang, and<br />

Hui-Bi Xu. Activation <strong>of</strong> Large-Conductance Calcium-Activated Potassium<br />

Channels by Puerarin: The Underlying Mechanism <strong>of</strong> Puerarin-Mediated<br />

Vasodilation. J Pharmacol Exp Ther, 2007, 323(1): 391-397.<br />

9. Li, H., Yao, J., Tong, X.T., Wu, Y., Sung, L., Pan, N., Guo, Z.H., Wu, H. M., Xu, T.<br />

and Ding, J.P.. Interaction Sites Between the Slo1 Pore and the N-terminus <strong>of</strong> the<br />

β2 Subunit, Probed with a Three Residue Sensor. J. Biol. Chem.,2007,282(24):<br />

17720-8.<br />

10. Li, W., Gao, S.B., Lv, C.X., Wu, Y., Guo, Z.H., Ding, J.P. and Xu, T.<br />

Characterization <strong>of</strong> voltage- and Ca 2+ -activated K + channels in rat dorsal root<br />

ganglion neurons. J. Cell Physiol.,2007,212: 348-57.


ABSTRACT<br />

Structural Feature and Function <strong>of</strong> hβ-subunits <strong>of</strong> BK Channel<br />

Jiuping Ding<br />

Key Laboratory <strong>of</strong> Molecular Biophysics Ministry <strong>of</strong> Education, College <strong>of</strong> Life<br />

Science and Technology, Huazhong University <strong>of</strong> Science and Technology, Wuhan,<br />

Hubei, 430074, China<br />

The functional diversity <strong>of</strong> large conductance Ca 2+ - and voltage-dependent K + (BK)<br />

channels arises mainly from co-assembly <strong>of</strong> the pore-forming mSlo α subunits with four<br />

tissue enriched auxiliary β subunits. The structural basis <strong>of</strong> the interaction between α<br />

subunits with β subunits is not well understood.<br />

Using immun<strong>of</strong>luorescence and patch clamp techniques, we demonstrated that 1)<br />

four-turn α-helical segment prevents surface expression <strong>of</strong> the auxiliary hβ2 subunit <strong>of</strong><br />

BK-type channel; 2) lysine-rich extracellular rings formed by hβ2 subunits confer the<br />

outward rectification <strong>of</strong> BK channels; 3) structural basis for toxin resistance <strong>of</strong><br />

β4-associated calcium-activated potassium (BK) channels. Those findings provide a<br />

better understanding <strong>of</strong> the structure and the diverse interactions between α and β<br />

subunits and will improve the design <strong>of</strong> channel inhibitors.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Jianzhi Wang<br />

PhD, Pr<strong>of</strong>essor, Director,<br />

Pr<strong>of</strong>essor and Director <strong>of</strong> Pathophysiology<br />

Pathophysiology Department,<br />

Tongji Medical College,<br />

Huazhong University <strong>of</strong> Science and Technology<br />

Wuhan, Hubei Province, 430074 P.R.China<br />

Email: wangjz@mails.tjmu.edu.cn<br />

Education and Training<br />

1975-1978 General Medicine,<br />

Tongji (Wuhan) Medical College, Wuhan, China<br />

1985-1988 Post Graduate Training in Biochemistry,<br />

Tongji Medical University, Wuhan, China<br />

1992-1993 Postdoctoral Research in Biochemistry, Siena University, Siena, Italy<br />

1993-1996 Postdoctoral Research in Alzheimer disease,<br />

NYS Institute for Basic Research, SI, NY, USA<br />

Selected Publications<br />

1. Zhu LQ, Zheng HY, Peng CX, Liu D, Li HL, Wang Q and Wang JZ*. Protein<br />

Phosphatase 2A Facilitates Axonogenesis by Dephosphorylating CRMP2. J<br />

Neurosci. 30(10):3839 –48, 2010<br />

2. Zhu LQ, Liu D, Hu J, Cheng J, Wang SH, Wang Q, Wang F, Chen JG, and Wang<br />

JZ*. GSK-3 inhibits presynaptic<br />

vesicle exocytosis by phosphorylating P/Q-type<br />

calcium channel and interrupting SNARE complex formation. J Neurosci.<br />

30(10):3849 –55, 2010<br />

3. Wang JZ*, Liu F. Microtubule-associated protein tau in development, degeneration<br />

and protection <strong>of</strong> neurons. Prog Neurobiol. 85:148-75, 2008 An invited review<br />

4. Li HL, Wang HH, Liu SJ, Deng YQ, Zhang YJ, Tian Q, Wang XC, Chen XQ,<br />

Yang Y, Zhang JY, Wang Q, Xu H, Liao F and Wang JZ*. Phosphorylation <strong>of</strong> tau<br />

antagonizes apoptosis by stabilizing -catenin, a mechanism involved in<br />

Alzheimer’s neurodegeneration. Proc Natl Acad Sci U S A. 104(9): 3591-3596,<br />

2007<br />

5. Zhu LQ, Wang SH, Yin YY, Liu D, Zheng HY, Shi HR, Tian Q, Wang XC, Wang<br />

Q, Guo L, Wang JZ*. Activation <strong>of</strong> glycogen synthase kinase-3 inhibits long term<br />

potentiation with synapse-associated impairments. J Neurosci. 27,12211–20, 2007<br />

6. Wang D, Qian L, Xiong H, Liu J, Neckameyer WS, Oldham S, Xia K, Wang J,<br />

Bodmer R, Zhang Z.Antioxidants protect PINK1-dependent dopaminergic neurons<br />

in Drosophila. Proc Natl Acad Sci U S A. 103:13520-5,2006<br />

7. Liu SJ, Zhang JY, Li HJ, Fang ZY, Wang Q, Deng HM, Gong C, Iqbal K, Wang<br />

JZ*. Tau becomes a more favorable substrate for GSK-3 when it is


prephosphorylated by PKA in rat brain. J Biol Chem. 279(48):50078-88, 2004.<br />

8. Wang JZ, Grundke-Iqbal I, Iqbal K. Glycosylation <strong>of</strong> microtubule-associated<br />

protein tau: an abnormal posttranslational modification in Alzheimer disease. Nat<br />

Med. 2(8): 870-6, 1996


ABSTRACT<br />

The Synaptic Mechanisms <strong>of</strong> Glycogen Synathese Kinase-3 in Regulating<br />

Leaning and Memory<br />

Jian-Zhi Wang, Ling-Qiang Zhu, Dan Liu, Juan Hu, Qun Wang<br />

Pathophysiology Department, Key Laboratory <strong>of</strong> Education Committee on<br />

Neurological Disorders, Tongji Medical College, Huazhong University <strong>of</strong> Science and<br />

Technology, Wuhan, China<br />

Previous studies have demonstrated that upregulation <strong>of</strong> glycogen synathese kinase-3<br />

(GSK-3 ) impairs learning<br />

and memory in rats and transgenic mouse models, and<br />

ctivation <strong>of</strong> GSK-3 inhibits long term potentiation with mechanisms involving a<br />

decreased presynaptic glutamate release. To further verify the mechanisms underlying<br />

the impaired synaptic vesicle exocytosis, we have recently demonstrated that activation<br />

<strong>of</strong> GSK-3 could inhibit calcium influx through phosphorylating the intracellular loop<br />

connecting domains II and III (LII-III) <strong>of</strong> P/Q-type Ca 2+ channels, which leads to<br />

decrease <strong>of</strong> intracellular Ca 2+ influx through the P/Q-type voltage-dependent calcium<br />

channel (VDCC). GSK-3 interferes with the formation <strong>of</strong> SNARE complex through (i)<br />

weakening the association <strong>of</strong> synaptobrevin with SNAP25 and syntaxin; (ii) reducing<br />

the interaction between the phosphorylated LII-III and synaptotagmin, SNAP25 and<br />

syntaxin; and (iii) inhibiting dissociation <strong>of</strong> synaptobrevin from synaptophysin I, all <strong>of</strong><br />

which are required for SNARE complex formation and thus for an efficient exocytosis.<br />

These results suggest that GSK-3 regulates<br />

synaptic vesicle cycle by affecting<br />

multiple steps in vesicular exocytosis.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. XianZhong Xu<br />

PhD, Assistant Pr<strong>of</strong>essor<br />

Dept <strong>of</strong> <strong>of</strong> Molecular & Integrative Physiology<br />

Life Sciences Institute, 6115A<br />

The University <strong>of</strong> Michigan<br />

Ann Arbor, MI 48109<br />

Tel: (734)615-9311<br />

Email: shawnxu@umich.edu<br />

Education<br />

1987-1991 B.S in Biochemistry , Wuhan University, Wuhan, China ,<br />

1991-1994 M.S in Biochemistry , Wuhan University, Wuhan, China,<br />

1995-2000 MD, Ph.D in Neuroscience& Biochem.<br />

Johns Hopkins University, Baltimore,<br />

2001-2005 Postdoc Neuroscience & Genetics<br />

California Institute <strong>of</strong> Technology, Pasadena, CA,<br />

Positions and Employment<br />

1991-1993 Guest researcher, National Laboratory <strong>of</strong> Biomacromolecules,<br />

Institute <strong>of</strong> Biophysics, Chinese Academy <strong>of</strong> Sciences, Beijing, China.<br />

2005- Assistant Pr<strong>of</strong>essor, Dept <strong>of</strong> Molecular & Integrative Physiology &<br />

Life Sciences Institute, The University <strong>of</strong> Michigan, Ann Arbor, MI.<br />

Selected Publications<br />

1. Xu, X.Z.S.*, Li, H. S.*, Guggino, W.B., and Montell, C. (1997). Coassembly <strong>of</strong><br />

TRP and TRPL produces a distinct store operated conductance. Cell 89, 1155<br />

1164.<br />

2. Xu, X.Z.S., Choudhury, A., Li, X., and Montell, C. (1998). Coordination <strong>of</strong> an<br />

array <strong>of</strong> signaling proteins through homo- and heteromeric interactions between<br />

PDZ domains and target proteins. Journal <strong>of</strong> Cell Biology 142, 545-555<br />

3. Xu, X.Z.S., Wes, P., Li, H. S., Yu, M., Morgan, S., Liu Y., and Montell, C. (1998)<br />

Retinal targets for calmodulin include proteins implicated in synaptic transmission.<br />

J. Biol.Chem 273, 31297-31307.<br />

4. Wes P. D*., Xu, X.Z.S.*, Li, H.-S., Chen, F., Doberstein, S. K., and Montell, C.<br />

(1999) Termination <strong>of</strong> phototransduction requires binding <strong>of</strong> the NINAC myosin<br />

III and the PDZ protein INAD. Nature Neuroscience 2, 447-453.<br />

5. Li, H.-S., Xu, X.Z.S., and Montell, C. (1999) Activation <strong>of</strong> a TRPC3-dependent<br />

cation current through the neurotrophin BDNF. Neuron 24, 261-273.<br />

6. Xu, X.Z.S., Chien, F., Butler, A., Salk<strong>of</strong>f, L., and Montell, C. (2000) TRP , a<br />

Drosophila TRP-related subunit, forms a regulated cation channel with TRPL.<br />

Neuron 26, 647-657.<br />

7. Xu, X.Z.S., Moebius, F., Gill, D.L., and Montell, C. (2001) Regulation <strong>of</strong>


melastatin, a TRP-related protein,through interaction with a cytoplasmic is<strong>of</strong>orm.<br />

Proc. Natl. Acad. Sci. U.S.A. 98,10692-10697.<br />

8. Xu, X.Z.S., and Sternberg, P.W. (2003) A C. elegans sperm TRP protein required<br />

for sperm-egg interactions during fertilization. Cell 114, 285-297.<br />

9. Li, W., Feng, Z., Sternberg, P.W., and Xu, X.Z.S. (2006) A C. elegans stretch<br />

receptor neuron revealed by a mechanosensitive TRP channel homologue.<br />

Nature 440, 684-687.<br />

10. Feng, Z.*, Li, W.*, Ward, A., Piggott, B.J., Larkspur, E., Sternberg, P.W., and Xu,<br />

X.Z.S. (2006) A C. elegans model <strong>of</strong> nicotine-dependent behavior: regulation by<br />

TRP family channels. Cell 127, 621-633<br />

11. Hsu, A.L.*, Feng, Z.*, Hsieh, M.-Y., and Xu, X.Z.S. (2009) Identification by<br />

machine vision <strong>of</strong> the rate <strong>of</strong> motor activity decline as a lifespan predictor in C.<br />

elegans. Neurobiology <strong>of</strong> Aging. 30, 1498-1503. (Epub 2008 Feb 5).<br />

12. Ward, A.*, Liu, J.*, Feng, Z., and Xu, X.Z.S. (2008) Light-sensitive neurons and<br />

channels mediate phototaxis in C. elegans. Nature Neuroscience 11, 916-922.<br />

13. Xiao, R., and Xu, X.Z.S. (2009) Function and regulation <strong>of</strong> TRP family channels in<br />

C. elegans. Pflugers Arch. 458, 851–860. (review)<br />

14. Ward, A., Walker, V.J., Feng, Z., and Xu, X.Z.S. (2009) Cocaine modulates<br />

locomotion behavior in C. elegans. PLoS ONE 4, e5946. PMCID2691951<br />

15. Liu, J.*, Ward, A.*, Gao, J., Dong, Y., Nishio, N., Inada, H., Kang, L., Yu, Y., Ma.,<br />

D., Xu, T., Mori, I., Xie, Z., and Xu, X.Z.S. (2010) C. elegans phototransduction<br />

requires a G protein-mediated cGMP pathway and a taste receptor homolog.<br />

Nature Neuroscience (in press)


ABSTRACT<br />

Sensory Transduction in C. elegans: What Can't a Worm Sense?<br />

Shawn X. Xu<br />

Life Sciences Institute, University <strong>of</strong> Michigan, Ann Arbor, USA<br />

The ability to sense and respond to sensory inputs is essential for life. There are five<br />

common senses in mammals: vision, smell, taste, hearing and touch. In addition, we<br />

rely on proprioception, which is <strong>of</strong>ten referred to as the sixth sense, to control body<br />

posture, balance and movement. Among the most common sensory stimuli are<br />

chemicals (smell and taste), mechanical forces (touch, hearing and proprioception) and<br />

light (vision). We are particularly interested in understanding how neurons detect and<br />

transduce mechanical and light stimuli, and how gene networks (e.g. receptors, ion<br />

channels and signaling molecules) regulate these processes. Currently, we focus on<br />

mechanotransduction and phototransduction. We have reported that proprioception is<br />

present in C. elegans. We have also made the surprising discovery that C. elegans, an<br />

organism that is generally believed to be blind (light-insensitive), in fact possesses a<br />

simple visual system, senses light and engages in phototaxis behavior. Our data reveal<br />

conservations in mechanotransduction and phototransduction between worms and<br />

mammals. These studies establish C. elegans as a powerful genetic model for studying<br />

the mechanisms <strong>of</strong> mechanotransduction and phototransduction.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Jean-Louis Besserau<br />

Ph.D, M.D, Pr<strong>of</strong>esosor<br />

Genetique et Neurobiologie de C. elegans<br />

INSERM U. 789<br />

Ecole Normale Superieure<br />

46, rue d'Ulm,75 005 Paris,France<br />

Lab phone: 33 (0)1 44 32 23 05<br />

Fax: 33 (0)1 44 32 36 54<br />

Email: jlbesse@wotan.ens.fr<br />

Education<br />

1994 M.D., Specialization in Neurology.<br />

1995 Ph.D,Molecular and cellular pharmacology,<br />

University <strong>of</strong> Paris VI, France.<br />

1995 Researcher position at the INSERM,<br />

Laboratory <strong>of</strong> Pr Changeux- Pasteur Institute, Paris.<br />

1997 Post-doctoral position in Erik Jorgensen laboratory,<br />

University <strong>of</strong> Utah, USA.<br />

2000 Junior group leader, Ecole Normale Supérieure-Paris.<br />

2005 INSERM Research Director at the Ecole Normale Supérieure-Paris.<br />

Selected Publications<br />

1. Gendrel M et al.: 2009. A secreted complement-control-related protein ensures<br />

acetylcholine receptor clustering. Nature 461: 992-996.<br />

2. Jospin M et al.: 2009. A Neuronal Acetylcholine Receptor Regulates the Balance <strong>of</strong><br />

Muscle Excitation and Inhibition in Caenorhabditis elegans. PLoS Biol 7:<br />

e1000265.<br />

3. Robert VJ and Bessereau JL: 2009. Manipulating the Caenorhabditis elegans<br />

genome using mariner transposons. Genetica.<br />

4. Boulin T et al.: 2008. Eight genes are required for functional reconstitution <strong>of</strong> the<br />

Caenorhabditis elegans levamisole-sensitive acetylcholine receptor. Proc Natl<br />

Acad Sci U S A 105: 18590-18595.<br />

5. Nilsson L et al.: 2008. Caenorhabditis elegans num-1 Negatively Regulates<br />

Endocytic Recycling. Genetics 179: 375-387.<br />

6. Robert V et al.: 2008. Gene conversion and end-joining repair double-strand breaks<br />

in the C. elegans germline. Genetics 180: 673-679.<br />

7. Ruaud AF et al.: 2008. The C. elegans P4-ATPase TAT-1 regulates lysosome<br />

biogenesis and endocytosis. Traffic 10: 88-100.<br />

8. Eimer S et al.: 2007. Regulation <strong>of</strong> nicotinic receptor trafficking by the<br />

transmembrane Golgi protein UNC-50. EMBO J 26: 4313-4323.<br />

9. Ruaud AF and Bessereau JL: 2007. The P-type ATPase CATP-1 is a novel


egulator <strong>of</strong> C. elegans developmental timing that acts independently <strong>of</strong> its<br />

predicted pump function. Development 134: 867-879.<br />

10. Robert V and Bessereau JL: 2006. Targeted engineering <strong>of</strong> the Caenorhabditis<br />

elegans genome following Mos1-triggered chromosomal breaks. EMBO J 26:<br />

170-183.<br />

11. Ruaud AF and Bessereau JL: 2006. Activation <strong>of</strong> nicotinic receptors uncouples a<br />

developmental timer from the molting timer in C. elegans. Development 133:<br />

2211-2222.<br />

12. Weimer RM et al.: 2006. UNC-13 and UNC-10/rim localize synaptic vesicles to<br />

specific membrane domains. J Neurosci 26: 8040-8047.<br />

13. Williams DC et al.: 2005. Characterization <strong>of</strong> Mos1-mediated mutagenesis in<br />

Caenorhabditis elegans: a method for the rapid identification <strong>of</strong> mutated genes.<br />

Genetics 169: 1779-1785.<br />

14. Yeh E et al.: 2005. Identification <strong>of</strong> genes involved in synaptogenesis using a<br />

fluorescent active zone marker in Caenorhabditis elegans. J Neurosci 25:<br />

3833-3841.<br />

15. Gally C et al.: 2004. A transmembrane protein required for acetylcholine receptor<br />

clustering in Caenorhabditis elegans. Nature 431: 578-582.<br />

16. Rostaing P et al.: 2004. Preservation <strong>of</strong> immunoreactivity and fine structure <strong>of</strong> adult<br />

C. elegans tissues using high-pressure freezing. Journal <strong>of</strong> Histochemistry &<br />

Cytochemistry 52: 1-12.<br />

17. Bosher JM et al.: 2003. The Caenorhabditis elegans vab-10 spectraplakin is<strong>of</strong>orms<br />

protect the epidermis against internal and external forces. J Cell Biol 161: 757-768.<br />

18. Gally C and Bessereau JL: 2003. GABA is dispensable for the formation <strong>of</strong><br />

juncational GABA receptor clusters in Caenorhabditis elegans. J.Neurosci 23:<br />

2591-2599.<br />

19. Gally C and Bessereau JL: 2003. C. elegans: <strong>of</strong> neurons and genes. M S-Medecine<br />

Sciences 19: 725-734.<br />

20. Bessereau J-L et al.: 2001. Mobilization <strong>of</strong> a Drosophila transposon in the<br />

Caenorhabditis elegans germ line. Nature 413: 70-74.


ABSTRACT<br />

Spatial Control <strong>of</strong> Synaptic Transmission at a Cholinergic Synapse in<br />

C. elegans<br />

Jean-Louis BESSEREAU<br />

Ecole Normale Superieure, INSERM U789, Paris, France<br />

Chemical synapses are specialized junctions that ensure precise and efficient signal<br />

transduction between excitable cells. Specifically, synaptic vesicles fuse with the<br />

pre-synaptic membrane at the active zone, in register with a specialized domain <strong>of</strong> the<br />

post-synaptic membrane containing a high density <strong>of</strong> neurotransmitter receptors.<br />

To analyze the subcellular organization <strong>of</strong> synapses at high resolution in a near to<br />

living-state, we physically immobilized freely moving nematodes C. elegans by instant<br />

freezing under high-pressure. After freeze-substitution and resin embedding, we used<br />

electron microscopy (EM) tomography to improve 3D resolution as compared to<br />

classical transmission-EM. We observed that at cholinergic neuromuscular junctions<br />

synaptic vesicles were interconnected by filaments similar to the cytomatrix <strong>of</strong><br />

vertebrates. Furthermore, vesicles docked at the plasma membrane were retained at the<br />

active zone by physical contacts with the presynaptic dense projection. In unc-10/RIM<br />

and syd-2/liprin mutants, docked vesicles mislocalized, which provided structural basis<br />

for the functional defects observed in these mutants.<br />

On the post-synaptic membrane, ionotropic acetylcholine receptors (AChRs) are<br />

clustered at high density in front <strong>of</strong> ACh release sites. We previously demonstrated that<br />

clustering depend on the muscle-secreted protein LEV-9 and the transmembrane protein<br />

LEV-10 that assemble in an extracellular scaffold with AChRs. In a forward genetic<br />

screen, we identified a gene coding for a small secreted protein containing only one<br />

immunoglobulin domain. This protein localizes at cholinergic neuromuscular junctions<br />

and is necessary to stabilize the AChR/LEV-9/LEV-10 complex at the synapse.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Jian-guo Chen<br />

M.D, Pr<strong>of</strong>essor<br />

Dept. <strong>of</strong> Pharmacology, Tongji Medical College,<br />

Huazhong University <strong>of</strong> Science Technology,<br />

#13 Hangkong Rd, Wuhan, 430030, China<br />

Phone: 0086-27-83692636<br />

Fax: 0086-27-83692608<br />

E-mail: chenj@mails.tjmu.edu.cn<br />

Education and Academic Appointments<br />

2002- Pr<strong>of</strong>essor, Chairman, Dept. <strong>of</strong> Pharmacology, Tongji Medical College,<br />

Huazhong University <strong>of</strong> Science & Technology<br />

1998-2001 Postdoctoral Associate, Dr. T. Hoshi’s laboratory<br />

The University <strong>of</strong> Iowa College <strong>of</strong> Medicine,USA.<br />

1996-1998 Visiting scholar, Dr. J. Sandkuhler’s laboratory, II.<br />

The Medical College, University <strong>of</strong> Heidelberg ,Germany.<br />

1995-1996 Visiting scholar, Pr<strong>of</strong>. Dr. J. W. Deitmer’s laboratory,<br />

The University <strong>of</strong> Kaiserslautern ,Germany.<br />

1990-1995 Lecturer, Dept. <strong>of</strong> Pharmacology,<br />

Tongji Medical University ,China.<br />

1987-1990 MD, Pr<strong>of</strong>. ZW Li’s laboratory<br />

The Center <strong>of</strong> Experimental Medical Researches,<br />

Tongji Medical University ,China.<br />

1980-1985 Bachelor <strong>of</strong> Medcine, Xianning Medical College ,China.<br />

Selected Publications<br />

1. Huang C, Hu ZL, Wu WN, Yu DF, Xiong QJ, Song JR, Shu Q, Fu H, Wang F and<br />

Chen JG. Existence and distinction <strong>of</strong> acid-evoked currents in rat astrocytes. Glia<br />

(2010, accepted, in press)<br />

2. Liu C, Wu JL, Xu K, Cai F, Gu J, Ma LQ and Chen JG. Neuroprotection by<br />

baicalein in ischemic brain injury involves PTEN/AKT pathway. J<br />

Neurochemistry, (2010, accepted, in press)<br />

3. Yang MJ, Wang F, Wang JH, Wu WN, Hu ZL, Cheng J, Yu DF, Long LH, Fu H, Xie<br />

N, Chen JG*. PI3-k integrates the effects <strong>of</strong> insulin and leptin on large-conductance<br />

Ca 2+ -activated K + channels in neuropeptide Y neurons <strong>of</strong> the hypothalamic arcuate<br />

nucleus. Am J Physiol Endocrinol Metab. 2010, 298(2)E: 193-201.<br />

4. Wang T, Gu J, Wu PF, Wang F, Xiong Z, Yang YJ, Wu WN, Dong LD and Chen JG.<br />

Protection by Tetrahydroxystilbene glucoside against cerebral ischemia:<br />

Involvement <strong>of</strong> JNK, SIRT1, NF-κB pathways and inhibition <strong>of</strong> intracellular


ROS/RNS generation. Free Radical Biology & Medicine,2009 Aug<br />

1;47(3):229-40.<br />

5. Mao LM, Wang W, Chu XP, Zhang GC, Liu XY, Yang YJ, Haines M, Papasian CJ,<br />

Fibuch EE, Buch S, Chen JG, and Wang JQ. Stability <strong>of</strong> NMDA receptors at the<br />

plasma membrane controls synaptic and behavioral adaptations to amphetamine,<br />

Nature Neuroscience, 2009 May;12(5):602-10.<br />

6. Liu J, Wang W, Wang F, Cai F, Hu ZL, Yang YJ, Chen J, Chen JG.<br />

Phosphatidylinositol-linked novel D(1) dopamine receptor facilitates long-term<br />

depression in rat hippocampal CA1 synapses. Neuropharmacology. 2009 Aug;<br />

57(2):164-71.<br />

7. Cai F, Wang F, Lin FK, Liu C, Ma LQ, Liu J, Wu WN, Wang W, Wang JH, Chen JG.<br />

Redox modulation <strong>of</strong> long-term potentiation in the hippocampus via regulation <strong>of</strong><br />

the glycogen synthase kinase-3beta pathway. Free Radical Biology & Medicine,<br />

2008 Oct 1;45(7):964-70.<br />

8. Ma LQ, Liu C, Wang F, Xie N, Gu J, Fu H, Wang JH, Cai F, Liu J and Chen JG,<br />

Activation <strong>of</strong> phosphatidylinositol-linked novel D1 dopamine receptors inhibits<br />

high voltage activated Ca 2+ currents in primary cultured striatal neurons. Journal <strong>of</strong><br />

Neurophysiology, 2009 May;101(5):2230-8.<br />

9. Zhang WT, Jiang FC, Wu PF, Ruan JL, Zhang LN, Fang W, Chen XL, Wang Y, Cao<br />

BS, Chen GY, Zhu YJ, Gu J and Chen JG*. Design, synthesis, and cytoprotective<br />

effect <strong>of</strong> 2-Aminothiazole Analogs as potent poly (ADP-ribose) polymerase-1<br />

inhibitors. Journal <strong>of</strong> Medicinal Chemistry, 2009 Feb 12;52(3):718-25.<br />

10. Cai F, Wang F, Lin FK, Liu C, Ma LQ, Liu J, Wu WN, Wang W, Wang JH, Chen JG.<br />

Redox modulation <strong>of</strong> long-term potentiation in the hippocampus via regulation <strong>of</strong><br />

the glycogen synthase kinase-3beta pathway. Free Radical Biology & Medicine,<br />

2008 Oct 1;45(7):964-70.<br />

11. Wang JH, Wang F, Yang MJ, Yu DF, Wu WN, Liu J, Ma LQ, Cai F, Chen JG. Leptin<br />

regulated calcium channels <strong>of</strong> neuropeptide Y and proopiomelanocortin neurons by<br />

activation <strong>of</strong> different signal pathways. Neuroscience. 2008 Sep 22;156(1):89-98.<br />

12. Lin F, Xin Y, Wang J, Ma L, Liu J, Liu C, Long L, Wang F, Jin Y, Zhou J, Chen J<br />

(2007). Puerarin facilitates Ca 2+ -induced Ca 2+ release triggered by<br />

KCl-depolarization in primary cultured rat hippocampal neurons. European<br />

Journal <strong>of</strong> Pharmacology, 570:43-49<br />

13. Lin FK, Xin Y, Gao DM, Xiong Z and Chen JG *(2007). Effects <strong>of</strong> electrical<br />

stimulation <strong>of</strong> the parafascicular nucleus neuronal activities <strong>of</strong> the subthalamic<br />

nucleus and the ventromedial nucleus in rats. Acta Physiologica Sinica<br />

59(1):79-85.<br />

14. Moore SA, Saito F, Chen J, Michele DE, Henry MD, Messing A, Cohn RD,<br />

Ross-Barta SE, Westra S, Willamson R, Hoshi T and Campbell KP. 2002.<br />

Deletion <strong>of</strong> brain dystroglycan recapitulates aspects <strong>of</strong> congenital muscular<br />

dystrophy. Nature, Vol. 418: 422-425.


ABSTRACT<br />

The Function <strong>of</strong> Phosphatidylinositol-Linked Novel D1 Dopamine<br />

Receptor in Central Nervous System<br />

Jian-Guo Chen, Fang Wang, Jue Liu and Li-Qun Ma<br />

Department <strong>of</strong> Pharamcology and the Key Laboratory <strong>of</strong> Neurological Disease <strong>of</strong><br />

Ministry <strong>of</strong> Education <strong>of</strong> China , Tongji Medical College, Huazhong University <strong>of</strong><br />

Science and Technology, Wuhan, 430030, China<br />

The physiological actions <strong>of</strong> dopamine (DA) are mediated via five distinct subtypes<br />

(D1-D5) <strong>of</strong> G protein-coupled receptors. Stimulation (D1, D5 subtypes) or inhibition<br />

(D2-D4 subtypes) <strong>of</strong> adenylyl cyclase through Gs/olf or Gi/o proteins by DA receptors<br />

is the traditional signaling pathway. In addition to the Gs/olf-coupled classical D1 DA<br />

receptor that increases the formation <strong>of</strong> cAMP, a D1-like DA receptor that couples to<br />

Gq protein, stimulates phospholipase Cb (PLCb) and results in hydrolysis <strong>of</strong><br />

phosphoinositide has been described and named as PI-linked DA receptor. SKF83959, a<br />

recently identified selective agonist for the PI-linked DA receptor, provided a powerful<br />

tool for exploring the function <strong>of</strong> this novel signal pathway in brain. However, the<br />

physiological and pathological roles <strong>of</strong> SKF83959 are unclear. In the recent work from<br />

our laboratory, it has been shown that activation <strong>of</strong> D1 receptor by SKF83959 mediates<br />

a dose-dependent mobilization <strong>of</strong> [Ca 2+ ]i via the PLC signaling pathway in cultured rat<br />

prefrontal cortical neurons and astrocytes. It was also inhibited HVA Ca2+ currents in<br />

cultured rat striatal neurons by PLC-IP3-Ca2+-calcineurin signal pathway. Furthermore,<br />

SKF83959 facilitated a postsynaptically PLC-dependent form <strong>of</strong> LTD at the Schaffer<br />

collater-CA1 synapses. In the pathological condition, SKF83959 mediates more<br />

effective functions <strong>of</strong> anti-Parkinsonian symptom with less dyskinesia. Our results<br />

provide further understanding for the role <strong>of</strong> PI-coupled dopamine system that may<br />

mediate important physiological or pathological challenges in the brain.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Xi He<br />

PhD, Pr<strong>of</strong>essor <strong>of</strong> Neurology, Harvard Medical School<br />

Children's Hospital Boston<br />

300 Longwood Avenue,Boston, MA, 02115<br />

Phone: 617-919-2257<br />

Fax: 617-730-1953<br />

Email: xi.he@childrens.harvard.edu<br />

Education<br />

1982-1985 Master’sdegree, Department <strong>of</strong> Biomedical Engineering,<br />

Huazhong University <strong>of</strong> Science and Technology, China.<br />

1985-1986 Research Associate, Department <strong>of</strong> Biomedical Engineering,<br />

Huazhong University <strong>of</strong> Science and Technology, China.<br />

1986-1992 Graduate Studies with Dr. Michael G. Rosenfeld, HHMI,<br />

School <strong>of</strong> Medicine, University <strong>of</strong> California at San Diego ,La Jolla.<br />

1992-1994 Postdoctoral Fellow with Dr. Luis F. Parada, NCI/NIH ,Frederick.<br />

1994-1996 Postdoctoral Fellow with Dr. Harold E.Varmus, NCI/NIH ,Bethesda.<br />

Pr<strong>of</strong>essinal Experience<br />

1997 Assistant Pr<strong>of</strong>essor, Neuroscience, Children’s Hospital,<br />

Harvard Medical School (Boston, MA)<br />

2002 Associate Pr<strong>of</strong>essor, Neuroscience, Children’s Hospital,<br />

Harvard Medical School (Boston, MA)<br />

2007 Pr<strong>of</strong>essor, Neurology/Neuroscience, Children’s Hospital,<br />

Harvard Medical School (Boston, MA)<br />

Selected Publications<br />

1. He X, Treacy M, Simmons D, Ingraham H, Swanson L, Rosenfeld Mg. Expression<br />

<strong>of</strong> a Large Family <strong>of</strong> POU-domain Regulatory Genes in Mammalian Brain<br />

Development. Nature 1989; 340:35-42.<br />

2. He X, Saint-Jeannet JP, Woodgett JR, Varmus HE, Dawid IB. Glycogen synthase<br />

kinase-3 and dorsoventral patterning in Xenopus embryos. Nature 1995;<br />

374:617-622.<br />

3. He X, Saint-Jeannet JP, Nathas J, Wang Y, Dawid IB, Varmus HE. A member <strong>of</strong><br />

the frizzled protein family mediating axis induction by Wnt-5A. Science 1997;<br />

275:1652-1654.<br />

4. Tamai K, Semenov M, Kato Y, Spokony R, Liu C, Katsuyama Y, Hess F,<br />

Saint-Jeannet J-P, He X. LDL receptor-related proteins in Wnt signal transduction.<br />

Nature 2000; 407: 530-535.<br />

5. Semenov M, Tamai K, Brott B, Kuhl M, Sokol S, He X. Head inducer Dickkopf-1


is a ligand for Wnt coreceptor LRP6. Current Biology 2001; 11: 951-961.<br />

6. Habas R, Kato Y, He X. Wnt/Frizzled activation <strong>of</strong> Rho regulates vertebrate<br />

gastrulation and requires a novel Formin homology protein Daam1. Cell 2001; 107,<br />

843-854.<br />

7. Liu C, Li Y, Semenov M, Baeg G-H, Han C, Tan Y, Zhang Z, Lin X, He X.<br />

Control <strong>of</strong> beta-catenin phosphorylation-degradation by a dual-kinase mechanism.<br />

Cell 2002; 108, 837-847.<br />

8. Kato Y, Habas R, Katsuyama Y, Naar A, He X. A component <strong>of</strong> the ARC/Mediator<br />

complex required for TGF-beta/Nodal signaling. Nature 2002; 418: 641-646.<br />

9. Capelluto DGS, Kutateladze TG, Habas R, Finkielstein CV, He X, Overduin M.<br />

DIX domain targets Dishevelled to actin stress fibers and vesicular membranes.<br />

Nature 2002; 419, 726-729.<br />

10. Habas, R., Dawid, I., and He, X. Co-activation <strong>of</strong> Rac and Rho by Wnt/Frizzled<br />

signaling is required for vertebrate gastrulation. Genes Dev. 2003; 17, 295-309.<br />

11. He, X. Wnt signaling went derailed again: a new track via the LIN-18 receptor?<br />

Cell 2004, 118, 668-670.<br />

12. He, X. Antagonizing Wnt and FGF receptors: an enemy from within (the ER). Cell<br />

2005 120, 156-158.<br />

13. Zeng X, Tamai K, Doble B, Li S, Huang H, Habas R, Okamura H, Woodgett J and<br />

He X. A dual-kinase mechanism for Wnt coreceptor phosphorylation and activation.<br />

Nature, 2005, 438, 873-877.<br />

14. He X and Axelrod J. A WNTer wonderland in Snowbird. Development, 2006, 133,<br />

2597-2603.<br />

15. He X. Unwinding a path to nuclear beta-catenin. Cell, 2006, 127, 40-42.<br />

16. MacDonald B, Semenov M and He X. Wnt/beta-catenin signaling (SnapShot). Cell<br />

2007, 131, 1204-1205.<br />

17. Semenov M, Habas R, MacDonald B, and He X. Non-canonical Wnt signaling<br />

(SnapShot). Cell 2007, 131, 1378-1379.<br />

18. Zeng X, Huang H, Tamai K, Zhang X, Harada, Y, Yokota, C, Almeida, KL, Wang<br />

J, Doble B, Woodgett J, Wynshaw-Boris A, Hsieh J-C, He X. Initiation <strong>of</strong> Wnt<br />

signaling: Control <strong>of</strong> Wnt coreceptor LRP6 phosphorylation/activation via<br />

Frizzled/Dishevelled/Axin Functions. Development .<br />

19. Choi YJ, Di Nardo A, Kramvis I, Meikle L, Kwiatkowski DJ, Sahin M, He X.<br />

Tuberous Sclerosis Complex proteins control axon formation. Genes & Dev. 2008,<br />

22, 2485-2495.<br />

20. Wu G, Huang H, Abreu JG, He X. Inhibition <strong>of</strong> GSK3 phosphorylation <strong>of</strong> b-catenin<br />

via phosphorylated PPPSPXS motis <strong>of</strong> Wnt receptor LRP6. PLoS One 2009, 4,<br />

e4926.


ABSTRACT<br />

Mechanisms <strong>of</strong> Wnt Signaling and Regulation<br />

Xi He, Xinjun Zhang, Jose Garcia Abreu, Chika Yokota, Bryan MacDonald, He Huang<br />

The F. M Kirby Center for Neurobiology, Children’s Hospital Boston, CLS-12064,<br />

Harvard Medical School, Boston, MA 02115, USA.<br />

Canonical Wnt/β-catenin signaling is initiated by the action <strong>of</strong> the Frizzled (Fz) receptor<br />

and its coreceptor LDL receptor-related-protein 6 (LRP6). Wnt signaling induces LRP6<br />

phosphorylation at conserved PPPSPxS motifs, which serve as docking sites for the<br />

scaffolding protein Axin, thereby allowing the Wnt receptor complex to inhibit<br />

b-catenin phosphorylation and degradation. LRP6 phosphorylation is mediated via the<br />

action <strong>of</strong> glycogen synthase kinase 3 (GSK3) and casein kinase 1 (CK1), and requires<br />

the function <strong>of</strong> the Fz receptor and its downstream partner Dishevelled (Dvl).<br />

I’ll discuss our study on the regulation <strong>of</strong> LRP6 phosphorylation, and the role <strong>of</strong><br />

LRP6-Axin interaction in the initiation and amplification <strong>of</strong> Wnt signaling at the plasma<br />

membrane. I’ll also discuss some biochemical experiments aimed to understand the<br />

mechanism by which phosphorylated LRP6 leads to inhibition <strong>of</strong> b-catenin<br />

phosphorylation. Finally I’ll describe a novel transmembrane protein, Tiki1, that<br />

appears to regulate the Wnt morphogen via an unexpected mechanism during vertebrate<br />

anterior patterning.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. John Wemmie<br />

MD, PhD, Assistant Pr<strong>of</strong>essor<br />

University <strong>of</strong> Iowa, Roy J and Lucille A Carver<br />

College <strong>of</strong> Medicine, Department <strong>of</strong> Psychiatry<br />

500 EMRB, Iowa City IA 52242<br />

Tel: 319 335 6540<br />

Fax: 319 335 7623<br />

Email: john-wemmie@uiowa.edu<br />

Education<br />

1989 BA, Central College, , Chemistry, Pella, IA<br />

1988 Trinity College, Foreign Study<br />

Carmarthen, Wales, UK Program<br />

1992 St. Petersburg State University, Intensive Russian,<br />

St. Petersburg, Russia, Language<br />

1996 PhD, University <strong>of</strong> Iowa Department <strong>of</strong> Physiology and Biophysics, Iowa City,<br />

1996 M.D, University <strong>of</strong> Iowa College <strong>of</strong> Medicine, Iowa City,<br />

2000 Residency Psychiatry,University <strong>of</strong> Iowa Hospitals and Clinics<br />

Selected Publications<br />

1. Wemmie J, Chen J, Askwith C, Hruska-Hageman A, Price M, Nolan B, Yoder P,<br />

Lamani E, Hoshi T, Freeman J, Welsh M: The Acid-Activated Ion Channel ASIC<br />

Contributes to Synaptic Plasticity, Learning, and Memory. Neuron (34): 463-477,<br />

2002.<br />

2. Wemmie J, Askwith C, Lamani E, Cassell M, Freeman J, Welsh M: ASIC1 is<br />

Localized in Brain Regions with High Synaptic Density and Contributes to Fear<br />

Conditioning. J Neurosci 23:5496-5502, 2003.<br />

3. Leonard AS, Yermolaieva O, Hruska-Hageman A, Askwith CC, Price MP,<br />

Wemmie JA, Welsh MJ: cAMP-dependent protein kinase phosphorylation <strong>of</strong> the<br />

acid-sensing ion channel-1 regulates its binding to the protein interacting with<br />

C-kinase-1. Proc Natl Acad Sci U S A. Feb 18;100 (4):2029-34, 2003.<br />

4. Xiong Z, Zhu X, Chu X, Minami M, Hey J, Wemmie JA, Price MP, Welsh MJ,<br />

Simon RP, Neuroprotection in ischemia: blocking calcium-permeable acid-sensing<br />

ion channels, Cell 118(6):687-698, 2004.<br />

5. Wemmie JA, Coryell MW, Askwith CC, Lamani E, Leonard AS, Sigmund CD,<br />

Welsh MJ: Overexpression <strong>of</strong> acid-sensing ion channel 1a in transgenic mice<br />

increases acquired fear-related behavior. Proc Natl Acad Sci USA. 2004 Mar<br />

9;101(10):3621-6. Epub 2004 Feb 26.<br />

6. Chu XP, Wemmie JA, Wang WZ, Zhu XM, Saugstad JA, Price MP, Simon RP,<br />

Xiong ZG: Subunit-dependent high-affinity zinc inhibition <strong>of</strong> acid-sensing ion<br />

channels. J Neurosci 2004 Oct 6;24(40):8678-89.


7. Zha X-M, Wemmie JA, Welsh MJ (2006) ASIC1a is a postsynaptic proton receptor<br />

that influences the density <strong>of</strong> dendritic spines. Proc Natl Acad Sci USA<br />

103:16556-16561.<br />

8. Coryell M, Espie-Ziemann A, Westmoreland P, Haenfler J, Kurjakovic Z, Zha X,<br />

Price M, Schnizler M, Wemmie J. Targeting ASIC1a reduces innate fear and<br />

alters neural activity in the fear circuit. Biological Psychiatry. 2007 Jul 27;<br />

9. Friese MA, Craner MJ, Etzensperger R, Vergo S, Wemmie JA, Welsh MJ, Vincent<br />

A, Fugger L. Acid-sensing ion channel-1 contributes to axonal degeneration in<br />

autoimmune inflammation <strong>of</strong> the central nervous system. Nat Med. 2007<br />

Dec;13(12):1483-9. Epub 2007 Nov 11.<br />

10. Ziemann AE, Schnizler MK, Albert GW, Severson MA, Howard III MA, Welsh<br />

MJ, Wemmie JA. Seizure termination by acidosis depends on ASIC1a. Nat<br />

Neurosci. 2008 Jul;11(7):816-22. Epub 2008 Jun 8.<br />

11. Coryell M, Wunsch A, Haenfler J, Allen J, McBride J, Davidson B, Wemmie J,<br />

Restoring Acid-Sensing Ion Channel-1a in the Amygdala <strong>of</strong> Knockout Mice<br />

Rescues Fear Memory but not Unconditioned Fear Responses. J Neurosci. 2008<br />

Dec 17;28(51):13738-41.<br />

12. Oxidant regulated inter-subunit disulfide bond formation between ASIC1a subunits.<br />

Zha XM, Wang R, Collier DM, Snyder PM, Wemmie JA, Welsh MJ. Proc Natl<br />

Acad Sci U S A. 2009 Mar 3;106(9):3573-8.<br />

13. Acid-sensing ion channel-1a in the amygdala, a novel therapeutic target in<br />

depression-related behavior. Coryell MW, Wunsch AM, Haenfler JM, Allen JE,<br />

Schnizler M, Ziemann AE, Cook MN, Dunning JP, Price MP, Rainier JD, Liu Z,<br />

Light AR, Langbehn DR, Wemmie JA. J Neurosci. 2009 Apr 29;29(17):5381-8<br />

14. Ziemann AE, Allen JE, Dahdaleh NS, Drebot II, Coryell MW, Wunsch AM, Lynch<br />

CM, Faraci FM, Howard III MA, Welsh MJ, Wemmie JA. The Amygdala Is a<br />

Chemosensor that Detects Hypercarbia and Acidosis to Elicit Fear Behavior. Cell,<br />

2009 Volume 139, Issue 5, 1012-1021, 25 November 2009<br />

15. Wemmie JA, Price MP, Welsh MJ (2006) Acid-sensing ion channels: advances,<br />

questions and therapeutic opportunities. Trends Neurosci 29:578-586.<br />

16. Kathleen A. Sluka, Olivia C. Winter, John A. Wemmie. Acid-Sensing Ion<br />

Channels: A <strong>New</strong> Target for Pain and CNS Diseases. Curr Opin Drug Discov<br />

Devel. 2009 Sep;12(5):693-704.<br />

17. J. Render1, K. Howe, A. Wunsch, S. Guionaud, P. Cox, J. Wemmie Histologic<br />

Examination <strong>of</strong> the Eye <strong>of</strong> Acid-Sensing Ion Channel 1a Knockout Mice, Int J<br />

Physiol Pathophysiol Pharmacol 2010;2(1):69-72


ABSTRACT<br />

Acid Sensing Ion Channels: A Novel Therapeutic Target for Anxiety and<br />

Depression?<br />

John Wemmie<br />

University <strong>of</strong> Iowa Carver College <strong>of</strong> Medicine, USA<br />

Acid sensing ion channels (ASICs) are members <strong>of</strong> the degenerin/epithelial sodium<br />

channel family that are activated by low extracellular pH. Several ASIC subunits are<br />

expressed in the brain including ASIC1a, 2a, and 2b. <strong>of</strong> these, our work suggests that<br />

ASIC1a is critical for acid-evoked currents in brain neurons. Disrupting ASIC1a in<br />

mice eliminated currents in CNS neurons evoked by pH as low as 5.0. Subcellular<br />

localization suggests ASIC1a is distributed to the neuron cell body, dendrites, and<br />

dendritic spines, where it confers pH sensitivity, increases intracellular Ca 2+ , and<br />

promotes synaptic plasticity. ASIC1a protein was found to be particularly abundant in<br />

brain structures that underlie fear, anxiety, and depression-related behaviors, including<br />

the basolateral amygdala. Consistent with these observations, loss <strong>of</strong> ASIC1a<br />

disrupted fear-related learning and memory. Loss <strong>of</strong> ASIC1a also reduced<br />

unconditioned fear, anxiety, and depression-related behaviors in mice. Supporting a<br />

role for ASIC1a as a brain pH sensor, we found that ASIC1a in the amygdala mediates<br />

fear and anxiety responses to CO2 inhalation and brain acidosis. This observation that<br />

suggests that ASICs may underlie the well-described ability <strong>of</strong> CO2 to triggers panic<br />

attacks in panic disorder patients. Together, these findings raise the possibility that<br />

ASIC1a might provide a novel and effective therapeutic target for anxiety disorders and<br />

depression.


INVITED SPEAKER’S INFO<br />

Pr<strong>of</strong>. Tao Xu<br />

PhD, Pr<strong>of</strong>essor, Director<br />

Institute <strong>of</strong> Biophysics,<br />

Chinese Academy <strong>of</strong> Sciences<br />

Beijing 100101, P.R. China<br />

Phone: +86-10-64888469<br />

Fax: +86-10-64871293<br />

E-mail: xutao@ibp.ac.cn<br />

Education<br />

1992-1996 Ph.D. (Biophysics), Institute <strong>of</strong> Biophysics & Biochemistry,<br />

Huazhong University <strong>of</strong> Science and Technology, China.<br />

1988-1992 B.S. (Engineering),<br />

Huazhong University <strong>of</strong> Science and Technology, China.<br />

Reaearch Experience<br />

2007- Pr<strong>of</strong>essor, Director, Institute <strong>of</strong> Biophysics,<br />

Chinese Academy <strong>of</strong> Sciences, Beijing, China<br />

2003-2007 Pr<strong>of</strong>essor, Deputy Director, Institute <strong>of</strong> Biophysics,<br />

Chinese Academy <strong>of</strong> Sciences, Beijing, China<br />

2000-2003 Pr<strong>of</strong>essor, Institute <strong>of</strong> Biophysics and Biochemistry,<br />

Huazhong University <strong>of</strong> Science and Technology, China.<br />

1999-2000 Senior Fellow, Department <strong>of</strong> Physiology and Biophysics,<br />

University <strong>of</strong> Washington, USA.<br />

1996-1999 Postdoctoral Fellowship,<br />

Max-Planck-Institute for Biophysical Chemistry, Germany.<br />

Selected Publications<br />

1. Wei Ji, Pingyong Xu, Zhengzheng Li, Jingze Lu, Lin Liu, Yi Zhan, Yu Chen, Bertil<br />

Hille, Tao Xu*, And Liangyi Chen*. Functional Stoichiometry <strong>of</strong> The Unitary<br />

Calcium-Release-Activated Calcium Channel. Proc. Natl. Acad. Sci. (USA).<br />

105(36): 13668–13673. 2008.<br />

2. Zixuan He, Junmei Fan, Lijun Kang, Jingze Lu, Yanhong Xue, Pingyong Xu, Tao<br />

Xu* And Liangyi Chen*. Ca 2+ Triggers A Novel Clathrin-Independent But<br />

Actin-Dependent Fast Endocytosis In Pancreatic Beta Cells. Traffic. 9: 910–923.<br />

2008.<br />

3. Li Jiang, Junmei Fan, Li Bai, Yan Wang, Yu Chen, Lu Yang, Liangyi Chen, And<br />

Tao Xu*. Direct Quantification <strong>of</strong> Fusion Rate Reveals A Distal Role For As160 In<br />

Insulin-Stimulated Fusion <strong>of</strong> Glut4 Storage Vesicles. J. Biol. Chem. 283(13):<br />

8508–8516. 2008.<br />

4. Guo Z, Lv C, Yi H, Xiong Y, Wu Y, Li W, Xu T*, Ding J*. A Residue At The


Cytoplasmic Entrance <strong>of</strong> Bk-Type Channels Regulating Single-Channel Opening<br />

By Its Hydrophobicity. Biophys J. 94(9): 3714-25. 2008.<br />

5. Li Bai, Yan Wang, Junmei Fan, Yu Chen, Wei Ji, Anlian Qu, Pingyong Xu*, David<br />

E. James, And Tao Xu*. Dissecting Multiple Steps <strong>of</strong> Glut4 Trafficking And<br />

Identifying The Sites <strong>of</strong> Insulin Action. Cell Metabolism. 5: 47-57. 2007.<br />

6. Ke-Ming Zhou, Yong-Ming Dong, Qian Ge, Dan Zhu, Wei Zhou, Xian-Guang Lin,<br />

Tao Liang, Zheng-Xing Wu*, Tao Xu*. Pka Activation Bypasses The Requirement<br />

For Unc-31 In The Docking <strong>of</strong> Dense Core Vesicles From C.Elegans Neurons.<br />

Neuron. 56: 657-669. 2007.<br />

7. Zhengzheng Li, Jingze Lu, Pingyong Xu, Xiangyang Xie, Liangyi Chen*, And Tao<br />

Xu*. Mapping The Interacting Domains <strong>of</strong> Stim1 And Orai1 In<br />

Ca 2+ Release-Activated Ca 2+ Channel Activation. J. Biol. Chem. 282(40):<br />

29448–29456. 2007.<br />

8. Wei Li, Shang-Bang Gao, Cai-Xia Lv, Ying Wu, Zhao-Hua Guo, Jiu-Ping Ding*,<br />

And Tao Xu*. Characterization <strong>of</strong> Voltage- And Ca 2+ -Activated K + Channels In<br />

Rat Dorsal Root Ganglion Neurons. Journal <strong>of</strong> Cellular Physiology. 212(2):<br />

348-357. 2007.<br />

9. Lijun Kang, Zixuan He, Pingyong Xu, Junmei Fan, Andrea Bet, Nils Brose, And<br />

Tao Xu*. Munc13-1 Is Required For The Sustained Release <strong>of</strong> Insulin From<br />

Pancreatic Beta Cells. Cell Metabolism. 3: 463-468. 2006<br />

10. Li Bai, Dan Zhu, Keming Zhou, Wei Zhou, Dongdong Li, Yan Wang, Rongying<br />

Zhang And Tao Xu*. Differential Properties <strong>of</strong> Gtp- And Ca 2+ -Stimulated<br />

Exocytosis From Large Dense Core Vesicles. Traffic. 7: 416-428. 2006.<br />

11. Xie L, Zhang M, Zhou W, Wu Zx, Ding Jp, Chen Ly*, And Xu T*. Extracellular<br />

Atp Stimulates Cells.�Exocytosis Via Localized Ca 2+ Release From Acidic Stores In<br />

Rat Pancreatic. Traffic. 7: 429-439. 2006.<br />

12. Dongfang Liu, Liang Xu, Fan Yang, Dongdong Li, Feili Gong, And Tao Xu*.<br />

Rapid Biogenesis And Sensitization <strong>of</strong> Secretory Lysosomes In NK Cells Mediated<br />

By Target-Cell Recognition. Proc Natl Acad Sci U S A. 102(1): 123-127. 2005.<br />

13. Jie Liu, Qunfang Wan, Xianguang Lin, Hongliang Zhu, Kirill Volynski, Yuri<br />

Ushkaryov And Tao Xu*. Α-Latrotoxin Modulates The Secretory Machinery Via<br />

Receptor-Mediated Activation <strong>of</strong> Protein Kinase C. Traffic. 6: 756- 765. 2005.<br />

14. Zhu H, Hille B, Xu T*. Sensitization <strong>of</strong> Regulated Exocytosis By Protein Kinase C.<br />

Proc Natl Acad Sci U S A. 99(26): 17055-17059. 2002.<br />

15. Xu T, Bajjalieh SM. SV2 Modulates The Size <strong>of</strong> The Readily Releasable Pool <strong>of</strong><br />

Secretory Vesicles. Nat Cell Biol. 3(8): 691-698. 2001.<br />

16. Xu T, Rammner B, Margittai M, Artalejo AR, Neher E, Jahn R. Inhibition <strong>of</strong><br />

SNARE Complex Assembly Differentially Affects Kinetic Components <strong>of</strong><br />

Exocytosis. Cell. 99(7): 713-722. 1999.<br />

17. Xu T, Ashery U, Burgoyne RD, Neher E. Early Requirement For Alpha-SNAP And<br />

NSF In The Secretory Cascade In Chromaffin Cells. EMBO J. 18(12): 3293-304.<br />

1999.<br />

18. Xu T, Binz T, Niemann H, Neher E. Multiple Kinetic Components <strong>of</strong> Exocytosis<br />

Distinguished By Neurotoxin Sensitivity. Nat Neurosci. 1(3): 192-200. 1998.


ABSTRACT<br />

Sequential Activation <strong>of</strong> Orai1 Subunits by Different Numbers <strong>of</strong> STIM1<br />

Leads to Multistate Opening <strong>of</strong> the CRAC Channel<br />

Zhengzheng Li 1, Lin Liu 2, Yongqiang Deng 1, Wei Ji 1, Wen Du 1, Pingyong Xu 1 Liangyi<br />

Chen 1, and Tao Xu 1,2<br />

1National Key Laboratory <strong>of</strong> Biomacromolecules, Institute <strong>of</strong> Biophysics, Chinese<br />

Academy <strong>of</strong> Sciences, Beijing 100101, P. R. China.<br />

2College <strong>of</strong> Life Science and Technology, Huazhong University <strong>of</strong> Science and<br />

Technology, Wuhan 430074, P. R. China.<br />

STIM1 and Orai1 are essential proteins <strong>of</strong> the Ca 2+ -release-activated Ca 2+ (CRAC)<br />

channel which mediates store-operated Ca 2+ entry. STIM1 is localized at the<br />

endoplasmic reticulum (ER) and functions as the sensor <strong>of</strong> ER luminal Ca 2+<br />

concentration, and Orai1 is the channel pore subunit at the plasma membrane. STIM1<br />

aggregates underneath the plasma membrane after store depletion and activates the<br />

CRAC channel by directly binding to Orai1. It remains a challenge to determine how<br />

many STIM1 molecules are required to open a CRAC channel. Here, by linking<br />

different numbers <strong>of</strong> Orai1 subunits with functional cytoplasmic domains <strong>of</strong> STIM1<br />

(residues 336-485, designated S domains), we provide evidence that each CRAC<br />

channel can be fully activated by eight S domains. Further experiments revealed that<br />

two S domains can fully activate one Orai1 subunit and the CRAC channel can<br />

progressively open as more Orai1 subunits are activated. Our data suggest that full<br />

activation <strong>of</strong> one CRAC channel complex requires eight STIM1 molecules, and the<br />

activation <strong>of</strong> the channel is not an “all-or-none” process but undergoes multiple<br />

intermediate states corresponding to different numbers <strong>of</strong> Orai1 subunits being<br />

activated.


SOLICITED<br />

ABSTRACTS


Chemosensory and Behavior <strong>of</strong> C.elegans is Regulated by An E.Coli<br />

Noncoding RNA<br />

Jinjing Wu, Jing Ren, Ge Shan<br />

Key Laboratory <strong>of</strong> Molecular Biophysics, Ministry <strong>of</strong> Education College <strong>of</strong> Life Science<br />

and Technology, Huazhong University <strong>of</strong> Science and Technology, Wuhan 430074,<br />

People’s Republic <strong>of</strong> China.<br />

It has been known that food as one environmental factor can affect the expression <strong>of</strong><br />

genes in animal cells, although we still lack examples <strong>of</strong> this kind <strong>of</strong> effect in which the<br />

detailed molecular mechanism is well understood. Here, we found that the expression <strong>of</strong><br />

a C. elegans gene, che-2, is down regulated by oxidative stressed E. coli. We<br />

demonstrated that this down regulation is mediated by an E. coli noncoding RNA, OxyS<br />

RNA. Oxidative stress induces the expression <strong>of</strong> OxyS in E. coli, which through a<br />

mechanism <strong>of</strong> feeding RNA interference, suppresses the expression <strong>of</strong> che-2 in C.<br />

elegans with a 17nt complementary sequence between OxyS and che-2 mRNA.<br />

Agonate and Dicer, Key components in the microRNA/ RNA interference pathway in C.<br />

elegans were shown to be required for this regulation <strong>of</strong> gene expression by E. coli<br />

OxyS RNA. Behavior analysis indicated that oxidative stressed E. coli "utilizes" OxyS<br />

noncoding RNA as a "repellent" to protect itself from bacterial forager such as C.<br />

elegans. These results indicate a co-evolutionary relationship between C. elegans<br />

and a natural food <strong>of</strong> this animal, E. coli.


Implication <strong>of</strong> the env Gene <strong>of</strong> the Human Endogenous Retrovirus W<br />

Family in the Expression <strong>of</strong> BDNF and DRD3<br />

Shan Li, Honglian Yu, Lijuan Liu, Wang Zheng, Chunlan Liu, Jie Zeng, Yabing Wang,<br />

and Fan Zhu*<br />

School <strong>of</strong> Basic Medical Science, Wuhan University<br />

It has been reported that HERVs play functional roles in some diseases, such as tumor,<br />

some types <strong>of</strong> schizophrenia, bipolar disorder, and so on. However, how HERVs<br />

contribute to these diseases is far from clear. We found that HERV-W env could<br />

activate BDNF pathways that might result in the development <strong>of</strong> schizophrenian. In this<br />

study, U251 Cells were used to study whether HERV env protein could active the<br />

BDNF pathways on the molecular level.


Microvavle Immobilization <strong>of</strong> Intact C. Elegans for in vivo Calcium<br />

Imaging <strong>of</strong> Neuronal Activities in Response to External Stimuli<br />

Jingjing Wang, Xiaojun Feng, Wei Du, Bi-Feng Liu*<br />

Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for<br />

Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory,<br />

Department <strong>of</strong> Systems Biology, College <strong>of</strong> Life Science and Technology, Huazhong<br />

University <strong>of</strong> Science and Technology, Wuhan 430074, People’s Republic <strong>of</strong> China.<br />

Aim To immobilize intact young adult C. elegans for imaging and explore neuronal<br />

activities in response to the changing environments. Methods We adopted a harmless<br />

and innocuous method to immobilize awake behaving C. elegans in a micr<strong>of</strong>luidic<br />

device for imaging in vivo neuronal responses to controllable drug stimuli. A<br />

comb-shaped micr<strong>of</strong>luidic valve was developed for the immobilization <strong>of</strong> living C.<br />

elegans with minimal influence on its activity for a long period <strong>of</strong> time. Results Our<br />

micr<strong>of</strong>luidic device achieved the complete immobilization effect <strong>of</strong> living C. elegans,<br />

similar to the glue-based method. In coupling to a single-drug delivery system, we<br />

investigated the neuronal response and adaptation <strong>of</strong> the amphid polymodal ASH<br />

sensory neurons to repeated rapid, sharp stimuli such as high concentrations <strong>of</strong> glycerol.<br />

With a multi-drug delivery system, we sequentially stimulated the same animal with<br />

three different chemical reagents, resulting in successful differentiation between<br />

mutants and wild type. Conclusion The PDMS comb-shaped microvalve demonstrates<br />

the complete immobilization <strong>of</strong> awake behaving C. elegans. Micr<strong>of</strong>luidic devices also<br />

provide micrometer-sized scales for rapid and sharp change in microenvironment to<br />

observe neuron responses to controllable drug stimuli. Thus, sensitive imaging <strong>of</strong><br />

neuronal activities can be conducted in vivo. We expect that micr<strong>of</strong>luidic chips can be<br />

used to study different aspects <strong>of</strong> nervous system in C. elegans.<br />

Key Words: C. elegans, micr<strong>of</strong>luidic, comb-shaped valve, neuronal activities<br />

*Corresponding author<br />

Email: bfliu@mail.hust.edu.cn Tel: 86-27-87792203


Inhibitory Effect <strong>of</strong> Ethanol on Gustatory Plasticity in C. Elegans<br />

Mediated by the Serotonin Pathway<br />

Ying Wang, Lichun Tang, Wei Du, Bi-Feng Liu*<br />

Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for<br />

Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory,<br />

Department <strong>of</strong> Systems Biology, College <strong>of</strong> Life Science and Technology, Huazhong<br />

University <strong>of</strong> Science and Technology, Wuhan 430074, People’s Republic <strong>of</strong> China.<br />

Aim Ethanol can affect the formation <strong>of</strong> learning and memory in many species.<br />

However, the molecular mechanisms underlying the behavioral effects <strong>of</strong> ethanol are<br />

still poorly understood. The results from the invertebrate model organisms contribute to<br />

and accelerate the research in higher animal species. So the effect <strong>of</strong> acute ethanol<br />

exposure on learning and memory in invertebrates yet needs to be studied more in-depth.<br />

Methods In C. elegans, gustatory plasticity is a simple learning paradigm, in which<br />

animals after prolonged pre-exposure to a chemo-attractive salt show chemo-aversion to<br />

this salt. We are using this learning model to study the effect <strong>of</strong> ethanol on learning and<br />

memory in C. elegans. Results Our results show that ethanol exhibits an acute<br />

inhibitory effect on gustatory plasticity in well-fed C. elegans. Genetic analysis revealed<br />

that mutant animals with defects in serotonin synthetic enzyme tph-1 failed to exhibit<br />

ethanol’s inhibitory effect on gustatory plasticity, however this inhibitory effect could<br />

be restored by expression <strong>of</strong> tph-1 in the NSM neurons. In addition, tph-1 acts as a<br />

partial downstream target <strong>of</strong> the BK potassium channel slo-1, regulating acute<br />

intoxicating effects <strong>of</strong> ethanol in locomotion and egg-laying. Furthermore, we show that<br />

G protein-coupled receptors <strong>of</strong> serotonin SER-4, SER-7 as well as G-protein α subunits<br />

GPA-3, GPA-11 possibly function in the same genetic pathway to modulate this<br />

response to ethanol. Results <strong>of</strong> calcium imaging indicate that ethanol directly changes<br />

the activity <strong>of</strong> ASER neuron that plays a dominant role in chemotaxis to salt.<br />

Conclusion Together, our results demonstrate the distinct role <strong>of</strong> serotonin pathway in<br />

modulation <strong>of</strong> acute response to ethanol in gustatory plasticity in C. elegans. Also, the<br />

effect <strong>of</strong> ethanol on gustatory plasticity explored in our study is a novel example <strong>of</strong> the<br />

effect <strong>of</strong> ethanol on learning behavior.<br />

Key Words: Caenorhabditis elegans, ethanol, gustatory plasticity, serotonin pathway<br />

*Corresponding author<br />

Tel: 86-27-87792203 , Email: bfliu@mail.hust.edu.cn


Is<strong>of</strong>orm-Specific Prolongation <strong>of</strong> KCNQ (Kv7) Potassium Channel<br />

Opening Mediated by a <strong>New</strong> Drug-Channel Binding Site<br />

Zhaobing Gao1, 5, Tangzhi Zhang2, Meng Wu1, Qiaojie Xiong1, 3, Haiyan Sun1, 4,<br />

Yinan Zhang2, Liansuo Zu2, Wei Wang2,* and Min Li1,*<br />

1Department <strong>of</strong> Neuroscience, High Throughput Biology Center and Johns Hopkins<br />

Ion Channel Center, School <strong>of</strong> Medicine, Johns Hopkins University, 733 North<br />

Broadway, Baltimore, MD 21205, 2Department <strong>of</strong> Chemistry & Chemical Biology,<br />

University <strong>of</strong> <strong>New</strong> Mexico, Albuquerque, NM 87131, 3 Current address: Cold Spring<br />

Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, NY 11724, 4 Current<br />

address: Corning Inc. One Science Center Road, Corning, NY 14831, 5 Current address:<br />

Shanghai Institute <strong>of</strong> Materia Medica, Shanghai Institutes for Biological Sciences,<br />

Chinese Academy <strong>of</strong> Sciences, Shanghai 201203<br />

Kv7 channels, especially Kv7.2 (KCNQ2) and Kv7.3 (KCNQ3), are key determinants<br />

for membrane excitability. Some chemical modulators for KCNQ are being approved<br />

for therapeutic use as anti-epileptic drugs, such as retigabine ((D-23129,<br />

N-(2-amino-4-(4-fluorobenzylamino)-phenyl). Therefore, a better understanding <strong>of</strong><br />

Compound-channel interaction is an area <strong>of</strong> intense interest. Of particular interest, a<br />

single residue change <strong>of</strong> W236L in KCNQ2 abolishes its sensitivity to Retigabine. We<br />

have conducted a screen <strong>of</strong> 20,000 compounds for KCNQ potentiators using rubidium<br />

flux combined with atomic absorption spectrometry (AAS). Here, we report<br />

characterization <strong>of</strong> a series <strong>of</strong> new structures with structural similarity to ICA-27243.<br />

We found that they display is<strong>of</strong>orm specificity and induce a marked reduction <strong>of</strong><br />

deactivation distinct from that <strong>of</strong> retigabine. Furthermore, KCNQ2(W236L) remains<br />

fully sensitive to these compounds. This result together with mutagenesis studies and<br />

analyses <strong>of</strong> combinatorial drug treatments suggests ZTZ compounds confers a new<br />

mode <strong>of</strong> action presumably by recognizing a new site on channel protein. Our results<br />

argue for a rationale for a deliberate search for channel potentiators with preferential<br />

effects <strong>of</strong> inhibiting channel close.<br />

Key Words: Potassium Channel, KCNQ, Activator, High Throughput Screening<br />

* To whom the correspondence should be addressed:<br />

Wei Wang, Ph.D., Department <strong>of</strong> Chemistry & Chemical Biology, University <strong>of</strong> <strong>New</strong><br />

Mexico, Albuquerque, NM 87131. 505-277-2609 (fax). wwang@unm.edu<br />

Min Li, Ph.D., Department <strong>of</strong> Neuroscience, Johns Hopkins University School <strong>of</strong><br />

Medicine, BRB311, 733 North Broadway, Baltimore, MD 21205. 410-614-1001 (fax).<br />

minli@jhmi.edu


Novel USH2A Compound Heterozygous Mutations Cause RP/USH2 in a<br />

Chinese Family<br />

Xiaowen Liu,1,2 Zhaohui Tang,2 Chang Li,2 Kangjuan Yang,3 Guanqi Gan,2 Zibo<br />

Zhang,3 Jingyu Liu,2 Fagang Jiang,1 Qing Wang,2 and Mugen Liu2<br />

1The Union Hospital, Huazhong University <strong>of</strong> Science and Technology, Wuhan, Hubei,<br />

P.R. China, 2Key Laboratory <strong>of</strong> Molecular Biophysics <strong>of</strong> Ministry <strong>of</strong> Education, College<br />

<strong>of</strong> Life Science and Technology, Center for Human Genome Research, Huazhong<br />

University <strong>of</strong> Science and Technology, Wuhan, China, 3Departments <strong>of</strong> Cell Biology<br />

and Medical Genetics, Yanbian University, Yanji, China<br />

Purpose: To identify the disease-causing gene in a four-generation Chinese family<br />

affected with retinitis pigmentosa (RP). Methods: Linkage analysis was performed with<br />

a panel <strong>of</strong> microsatellite markers flanking the candidate genetic loci <strong>of</strong> RP. These loci<br />

included 38 known RP genes. The complete coding region and exon-intron boundaries<br />

<strong>of</strong> Usher syndrome 2A (USH2A) were sequenced with the proband DNA to screen the<br />

disease-causing gene mutation. Restriction fragment length polymorphism (RFLP)<br />

analysis and direct DNA sequence analysis were done to demonstrate co-segregation <strong>of</strong><br />

the USH2A mutations with the family disease. One hundred normal controls were used<br />

without the mutations. Results: The disease-causing gene in this Chinese family was<br />

linked to the USH2A locus on chromosome 1q41. Direct DNA sequence analysis <strong>of</strong><br />

USH2A identified two novel mutations in the patients: one missense mutation<br />

p.G1734R in exon 26 and a splice site mutation, IVS32+1G>A, which was found in the<br />

donor site <strong>of</strong> intron 32 <strong>of</strong> USH2A. Neither the p.G1734R nor the IVS32+1G>A<br />

mutation was found in the unaffected family members or the 100 normal controls. One<br />

patient with a homozygous mutation displayed only RP symptoms until now, while<br />

three patients with compound heterozygous mutations in the family <strong>of</strong> study showed<br />

both RP and hearing impairment. Conclusions: This study identified two novel<br />

mutations: p.G1734R and IVS32+1G>A <strong>of</strong> USH2A in a four-generation Chinese RP<br />

family. In this study, the heterozygous mutation and the homozygous mutation in<br />

USH2A may cause Usher syndrome Type II or RP, respectively. These two mutations<br />

expand the mutant spectrum <strong>of</strong> USH2A.<br />

Correspondence to:<br />

Dr. Mugen Liu, Huazhong University <strong>of</strong> Science and Technology Human Genome<br />

Research Center, Wuhan, Hubei, 430074, P.R. China; Phone: 86-27-87794549; FAX:<br />

86-27-87794549; email: lium@mail.hust.edu.cn


UNC-31/CAPS Docks and Primes Dense Core Vesicles in C. elegans<br />

Neurons<br />

Xian-Guang Lin, Min Ming, Mao-Rong Chen, Wei-Pin Niu, Yong-Deng Zhang, Bei Liu,<br />

Ya-Ming Jiu, Jun-Wei Yu, Tao Xu,* and Zheng-Xing Wu*<br />

Key Laboratory <strong>of</strong> Molecular Biophysics <strong>of</strong> Ministry <strong>of</strong> Education, School <strong>of</strong> Life<br />

Science and Technology, Huazhong University <strong>of</strong> Science and Technology, Wuhan,<br />

430074 China<br />

UNC-31 or its mammalian homologue, Ca 2+ -dependent activator protein for secretion<br />

(CAPS), is indispensable for exocytosis <strong>of</strong> dense core vesicle (DCV) and synaptic<br />

vesicle (SV). From N- to the C-terminus, CAPS contains putative functional domains,<br />

including dynactin 1 binding domain (DBD), C2, PH, (M)UNC-13 homology domain<br />

(MHD) and DCV binding domain (DCVBD), the last four we examined in this study.<br />

We employed UNC-31 null mutant C. elegans worms to examine whether UNC-31<br />

functions could be rescued by ectopic expression <strong>of</strong> full-length UNC-31 vs each <strong>of</strong><br />

these four domain-deleted mutants. Full length UNC-31 cDNA rescued the phenotypes<br />

<strong>of</strong> C. elegans null mutants in response to Ca 2+ -elevation in ALA neurons. Surprisingly,<br />

MHD deletion also rescued UNC-31 exocytotic function in part because the relatively<br />

high Ca 2+ level (pre-flash Ca 2+ was 450 nM) used in the capacitance study could bypass<br />

the MHD defect. Nonetheless, the three other domain-truncation cDNAs had almost no<br />

rescue on Ca 2+ evoked secretion. Importantly, this genetic null mutant rescue strategy<br />

enabled physiological studies at levels <strong>of</strong> whole organism to single cells, such as<br />

locomotion assay, pharmacological study <strong>of</strong> neurotransmission at neuromuscular<br />

junction, in vivo neuropeptide release measurement and analysis <strong>of</strong> vesicular docking,<br />

Using this battery <strong>of</strong> physiological assays, we found that all four UNC-31 domains are<br />

independently indispensible for UNC-31 to effect its function. The rescue <strong>of</strong> the MHD<br />

deletion was an in vitro but non-physiological phenomenon which was not recapitulated<br />

in the physiological assays Our results suggest that each <strong>of</strong> these UNC-31 domains<br />

support distinct sequential molecular actions <strong>of</strong> UNC-31 in vesicular exocytosis,<br />

including steps in vesicle tethering and docking that bridge vesicle with plasma<br />

membrane, and subsequently priming vesicle by initiating the formation <strong>of</strong> soluble<br />

N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) core complex.


Cordycepin Protects Against Brain ischemical Reperfusion Injury in<br />

Mice by Inhibition <strong>of</strong> Matrix Metalloproteinases-3 Expression.<br />

Zhenyong Cheng , Lianjun Guo*,Qing Lv, Xulin Xu<br />

Department <strong>of</strong> Pharmacology ,Tongji medical college,Huazhong University <strong>of</strong> Science<br />

and Technology, WuHan, China 430030<br />

Aim: the neuronal death following transient global ischemia - reperfusion in the mouse<br />

takes to occur, Since matrix metalloproteinase-3 (MMP-3) enhances inflammation and<br />

promotes apoptosis in ischemia – reperfusion. providing a potential neuroprotective<br />

effect <strong>of</strong> cordycepin. Methods: Kun ming male mice (18-25g) were randomly divided<br />

into five groups:control group, brain ischemic- reperfusion injury group, cordycepin<br />

10mg/kg, 20mg/kg and extract <strong>of</strong> cordyceps militaris(CME) treatmented groups. The<br />

bilateral common carotid artery was occlusion (2VO) for 15 min, and then subjected to<br />

a subsequent 4h period <strong>of</strong> ischemia reperfusion. The brain protein was extracted and<br />

the expression <strong>of</strong> matrix metalloproteinases-3(MMP-3) was measured by western blot<br />

analysis. Results: The expression <strong>of</strong> MMP-3 in the cordycepin- treated group(10mg/kg)<br />

and CME group were decreased by 45.8% and 41.6% contrast with model<br />

group(P


Calcium Signaling in Astrocytes Induced by Photostimulation with<br />

Femtosecond Laser<br />

Yuan Zhao, Yuan Zhang, Wei Zhou, Shaoqun Zeng*<br />

Britton Chance Center for Biomedical Photonics<br />

Wuhan National Laboratory for Optoelectronics-Huazhong University <strong>of</strong> Science and<br />

Technology,Wuhan, 430074, China<br />

Astrocytes have been proved to actively contribute to brain functions, communicating<br />

with neurons and other brain cells. However, conventional stimulation approaches are<br />

hard to be feasibly utilized to precisely activate astrocytes for delicate investigations.<br />

Here, we developed non-invasive photostimulation with high accuracy to evoke calcium<br />

(Ca 2+ ) signaling in astrocytes. The near infrared (800 nm) femtosecond laser was<br />

focused onto the cell membrane <strong>of</strong> an astrocyte. Ca 2+ elevation was immediately evoked<br />

in the stimulated cell and subsequently in surrounding cells, forming a radial Ca 2+ wave<br />

throughout the network. Localized transient photoporation was demonstrated to be<br />

induced on the cell membrane by laser irradiation. Extracellular Ca 2+ entry into the<br />

cytoplasm through this tiny poration was necessary for astrocytic Ca 2+ signaling.<br />

Repetitive photostimulation was actually performed on one cell with well-controlled<br />

laser intensity and targeting, resulting in similar responses. Further, different patterns <strong>of</strong><br />

Ca 2+ elevation in the stimulated astrocyte were observed by varying femtosecond laser<br />

power. Generally, the amplitude <strong>of</strong> Ca 2+ response in the stimulated cell was increased<br />

with enhanced stimulating laser power, and concomitantly, the Ca 2+ wave was widened.<br />

Therefore, noncontact photostimulation <strong>of</strong> astrocytes with femtosecond laser is<br />

demonstrated to be non-disruptive, reproducible, and with high spatiotemporal precision.<br />

Photogenerated Ca 2+ response follows a femtosecond laser-power-dependent manner, so<br />

that distinct Ca 2+ signaling ranges can be feasibly provided for specific studies. This<br />

versatile tool is thus promisingly efficient and helpful for resolving problems that<br />

conventional stimulations are difficult to touch, especially for in vivo studies.<br />

Key Words: astrocyte, Ca 2+ signaling, photostimulation, femtosecond laser,<br />

photoporation<br />

*Corresponding author : Email: sqzeng@mail.hust.edu.cn


ZD7288 Inhibits the Induction Of LTP in an NMDA Independent<br />

Manner at Hippocampal Schaffer Collateral-CA1 Synapses<br />

Wei He, Zhenyong Cheng, Shibin Li, Xulin Xu, Qing Lu, Lianjun Guo*<br />

Department <strong>of</strong> Pharmacology, Tongji Medical College <strong>of</strong> Huazhong University <strong>of</strong><br />

Science and Technology, Wuhan, 430030, China<br />

Aim: To investigate the effect <strong>of</strong> ZD7288 on synaptic transmission and high frequency<br />

stimulation (HFS)-induced long-term potentiation(LTP) in the Schaffer collateral-CA1<br />

synapse <strong>of</strong> mouse hippocampal slices, and identify whether the effect <strong>of</strong> ZD7288 on the<br />

induction <strong>of</strong> LTP can be explained by inhibition <strong>of</strong> N-methyl-D-aspartate (NMDA) type<br />

<strong>of</strong> glutamate receptors. Methods: Extracellular field excitatory postsynaptic<br />

potentials(fEPSPs) recordings were used to examine the effect <strong>of</strong> ZD7288 on basal<br />

synaptic transmission and HFS-induced LTP in the Schaffer collateral-CA1 synapse <strong>of</strong><br />

mouse hippocampal slices, and measured the evoked depolarisations that by direct<br />

agonist NMDA receptors. Result: Bath application <strong>of</strong> 10 µM ZD7288 had no effect on<br />

basal synaptic transmission but inhibited the induction <strong>of</strong> HFS-induced LTP. In addition,<br />

10 µM ZD7288 had no effect on the responses to exogenous NMDA. Conclusion: The<br />

results suggested that ZD7288 has the ability to prevent the induction <strong>of</strong> LTP at the<br />

Schaffer collateral-CA1 synapse <strong>of</strong> mouse hippocampus in vitro, and this inhibitory<br />

effect is an NMDA receptor independent manner.<br />

Key Words: ZD7288; synaptic transmission; long-term potentiation; NMDA<br />

Acknowledgments: This work was supported by grants from the National Natural<br />

Science Foundation <strong>of</strong> China (NSFC, No. 30772559) to Lianjun Guo<br />

First author : Wei He Tel: +86 27 83691762,E-mail: hewei8286291@yahoo.com.cn<br />

* Correspondence author :Tel: +86 27 83691763, E-mail: gljyl@yahoo.com.cn


Isolation, Characterization and Anti-Cancer Activity <strong>of</strong> SK84, a Novel<br />

Glycine-Rich Antimicrobial Peptide from Drosophila Virilis<br />

Jie Lu*, Mu-ya Shu, Ming-lei Leng, Zheng-wang Chen<br />

Institute <strong>of</strong> Biophysical & Biochemistry, College <strong>of</strong> Life Science and<br />

Technology,Huazhong University <strong>of</strong> Science & Technology, Wuhan, 430074, China<br />

We report herein the isolation and characterization <strong>of</strong> a novel glycine-rich antimicrobial<br />

peptide purified from the larvae <strong>of</strong> Drosophila virilis. A range <strong>of</strong> chromatographic<br />

methods was used for isolation and its antibacterial activity against Bacillus subtilis was<br />

employed to screen for the most active fractions. The peptide, termed SK84 due to its<br />

N-terminal serine, C-terminal lysine and a total <strong>of</strong> 84 residues, was completed<br />

sequenced using RT-PCR cDNA cloning. SK84 contains a high level <strong>of</strong> glycine (15.5%)<br />

and a hexaglycine cluster motif in the N-terminal part. SK84 displayed antibacterial<br />

activity against the tested Gram-positive bacteria (Bacillus subtilis, Bacillus<br />

thuringiensis and Staphylococcus aureus), but had no effect on Gram-negative bacteria<br />

(Pseudomonas aeruginosa, Escherichia coli) and fungi (Saccharomyces cerevisiae,<br />

Candida albicans). SK84 had specific inhibitory effects on the proliferation <strong>of</strong> several<br />

cancer cell lines (Human leukemia THP-1, liver cancer HepG2, and breast cancer<br />

MCF-7 cells), but no hemolytic activity. The results from scanning electron microscopy<br />

observations revealed that SK84 killed THP-1 cells by destroying the cell membranes.<br />

Alignment results show that SK84 is a mature protein processed from the pseudoprotein<br />

GJ19999 from Drosophila virilis, and is very similar to several pseudoproteins from<br />

different Drosophila species. SK84 induces the apoptosis <strong>of</strong> HepG2 cells by stimulating<br />

the caspase-3 activation. Our results show that SK84 represents a novel glycine-rich<br />

peptide family in Drosophila species with antimicrobial and anti-cancer cell activities.<br />

Key Words:antimicrobial peptide; glycine-rich peptide; Drosophila virilis;<br />

anti-cancer-cell activity;apoptosis.<br />

*Corresponding author to:<br />

Tel: 86-18971142265;Fax: 86-027-87792024;E-mail:lujie.jane@163.com


Foamy Virus as Potential Vectors for Gene Therapy in Nervous Disorders<br />

and Signal Pathway in Neural Cell Transduction<br />

Ying-Ying ZHANG#, Yong-Juan LIU#, Guo-Guo ZHU, Yan-Yan QIU, Biwen PENG,<br />

Jun YIN, Wan-Hong LIU* and Xiao-Hua HE*<br />

School <strong>of</strong> Medicine, Wuhan University, Wuhan 430071, China<br />

# Equal contribution to this work<br />

Foamy viruses are a type <strong>of</strong> retrovirus with a wide range <strong>of</strong> hosts and non-pathogenicity.<br />

Due to their unique structures, safety and the large packaging capacities, foamy viruses<br />

have been developed into an effective vector system for gene therapy. However, it is<br />

still a controversial issue whether foamy virus vectors could be as exogenous gene<br />

conveyance in the nervous system located a large number <strong>of</strong> growth-arrested cells. In<br />

this review, we focus on this issue and analyze the reasons for their potentiality <strong>of</strong><br />

infecting the growth-arrested cells. Several evidences in papers (from 1997 to 2009)<br />

concerning transduction <strong>of</strong> the growth-arrested cells and neurons by foamy virus vectors<br />

were compared and analyzed. It is demonstrated that foamy virus vectors can transduce<br />

some growth-arrested cells and neurons. Assuredly, further pro<strong>of</strong>s are needed in the<br />

future. However, these facts suggested that foamy viruses may become potential<br />

transfer vectors for gene therapy in nervous disorders and even for signal pathway in<br />

neural cell transduction.<br />

Key Words: foamy virus vectors; growth-arrested cells; neurons; gene therapy;<br />

neuro-signalling transduction<br />

Acknowledgment: This work was supported by National Natural Sciences Foundation<br />

<strong>of</strong> China (No. 30870856 and 30970145), Program for <strong>New</strong> Century Excellent Talents,<br />

Wuhan University (No. NCET-07-0630),the Scientific Research Foundation for the<br />

Returned Overseas Chinese Scholars in State Education Ministry, Research Fund for the<br />

Doctoral Program <strong>of</strong> Higher Education <strong>of</strong> China (No. 20090141110010) and the<br />

Independent Scientific Research Foundation <strong>of</strong> Wuhan University (No.3081002).<br />

*Corresponding author<br />

Tel: 86-27-68759985, Fax: 86-27-68759991.<br />

E-mail: liuwanhong@whu.edu.cn or hexiaohua@whu.edu.cn


Role <strong>of</strong> Heat on the Expression and Function <strong>of</strong> KCNQ2<br />

Fang Yu#, Yuncui Wang#, Yanlan Liu, Wanhong Liu, Biwen Peng* and Xiaohua,He*<br />

School <strong>of</strong> Medicine, Wuhan University, Wuhan 430071, China<br />

# Equal contribution to this work<br />

Objective: KCNQ2 (Kv7.2), Voltage-dependent potassium channels, plays a key role in<br />

regulating neuronal excitability. Mutations in neuronal KCNQ2/3 subunits, the<br />

molecular correlates <strong>of</strong> M current, have previously been linked with benign familial<br />

neonatal epilepsy (BFNC). In this study, we measured the temperature sensitivity <strong>of</strong><br />

KCNQ2 channel and determined whether temperature affected the excitability <strong>of</strong> the<br />

nervous system by the regulation <strong>of</strong> KCNQ2 channels.Methods: The wild type and<br />

mutant cDNAs <strong>of</strong> human KCNQ2, R214W and Y284C genes were subcloned into<br />

pEGFP-C1 plasmid, and then were transfected into 293T cells respectively. The<br />

KCNQ2 protein expression at different temperature was detected by fluorescence<br />

confocal microscopy and Western-blotting. While whole-cell patch clamp was<br />

performed to record the effect <strong>of</strong> temperature electrophysiological activity <strong>of</strong> KCNQ2<br />

potassium channel. Results: Total fluorescence intensity values <strong>of</strong> wtKCNQ2,R214W<br />

and Y284C were enhanced with the temperature increasing from 37℃ to 40 ℃<br />

(P


Possible Involvement <strong>of</strong> TRPV1 in neonatal Febrile Seizure Mice<br />

Yuan Xiao-ran Yu Hong-mei He Xiao-hua and Peng Bi-wen<br />

Department <strong>of</strong> physiology, School <strong>of</strong> Basic Medical Science, Wuhan University<br />

Febrile seizures (FSs) are acute symptomatic seizures that occur in response to a fever<br />

in an age-specific manner. Although FS are largely benign, complex FS increase the<br />

risk to develop temporal lobe epilepsy (TLE). TRPV1 is a capsaicin/vanilloid-gated<br />

cation channel expressed predominantly by primary nociceptive neurons and can<br />

alternatively be activated by elevated temperatures (>43℃). Studies involving systemic<br />

or intrahypothalamic capsaicin administration have suggested a role for TRPV1 in body<br />

temperature control. To explore whether TRPV1 is involved in febrile seizure in<br />

immature brain, behavioral responses were observed on neonatal C57BL6/J mice and<br />

TRPV1 -/- mice. Hyperthermia was induced by warm air in 14-day-old mice. P14 pups in<br />

TRPV1 -/- group showed prolonged latency, shorten duration and lower degree <strong>of</strong><br />

seizures compared to wild type mice. Y-maze and Open field test did not show much<br />

difference in two groups at the age <strong>of</strong> P30. Western-blot and immunohistochemistry<br />

were also used to find out the different expression <strong>of</strong> TRPV1 between the control group<br />

and FS-model group in wild type mice. These results suggest that TRPV1 probably<br />

participate in the generation <strong>of</strong> fever seizure and thus may provide new leads for<br />

treatment <strong>of</strong> children at risk for complex FS and TLE.<br />

Key Words: TRPV1; Febrile Seizure; behavior response; neonatal<br />

Acknowledgment: This work was supported by National Natural Sciences Foundation<br />

<strong>of</strong> China (No. 30970994 and 30770734), Program for <strong>New</strong> Century Excellent Talents,<br />

Wuhan University (No. NCET-07-0630),the Scientific Research Foundation for the<br />

Returned Overseas Chinese Scholars in State Education Ministry, and Research Fund<br />

for the Doctoral Program <strong>of</strong> Higher Education <strong>of</strong> China (No. 20090141110010).<br />

*Corresponding author: Email: pengbiwen@whu.edu.cn


The Kv1.3 Channel Blocker Scorpion Polypeptide has an Therapeutic<br />

effect on EAE Rats with a Molecule Mechanism<br />

Zhi Li1#, Wanhong Liu1#, Song Han2, Liang Li1, Yuanteng Fan1, Zhihao Wang1, Bo<br />

Yang1, Wu Yingliang2*, Xiaohua He1* and Wenxin Li2<br />

1School <strong>of</strong> Medicine, Wuhan University, Wuhan 430071, China<br />

2College <strong>of</strong> Life sciences, Wuhan University, Wuhan 430072, China<br />

# Equal contribution to this work<br />

Objective: Experimental autoimmune encephalomyelitis(EAE), an ideal model <strong>of</strong><br />

human multiple sclerosis(MS), is a kind <strong>of</strong> autoimmune disease in central nervous<br />

system .We are to investigate the efficacy <strong>of</strong> the Kv1.3 channel blocker scorpion<br />

polypeptide on the inhibition <strong>of</strong> T cell proliferation in vitro and the therapy <strong>of</strong> EAE rats<br />

in vivo .Furthermore, to study the T cell activation signaling transduction pathways<br />

which are associated with the Kv1.3 channel.Method: Female inbred Sprague<br />

Dawley(SD) rats with age <strong>of</strong> 8-10weeks were immunized by their own cerebral and<br />

spinal cord homogenate in Freund's complete adjuvant with subcutaneous injection <strong>of</strong><br />

0.06g/100g antigen. The administration <strong>of</strong> polypeptide was 100ug/kg .i.p. for 14days,<br />

rats were observed for 16days and the clinical score <strong>of</strong> EAE were recorded. The lesion<br />

tissues were Hematoxylin -Eosin(H.E.) stained to observe the inflammatory infiltration<br />

under the microscope. By transmission electron microscopy (TEM),the structure <strong>of</strong><br />

loose myelin and the axonal damage <strong>of</strong> neurons were observed. The cytokine levels <strong>of</strong><br />

IL-2,IFN-γ,TNF-α from the rats serum were detected by the ELISA assay. In vitro T<br />

cells proliferation assay, spleen cells from EAE rats were cultured in a 96-well plates<br />

and activated by antigen, treated with different doses <strong>of</strong> drugs, cells were cultured for<br />

72h and were added with [ 3 H]-thymidine for 18h before being harvested , the cpm<br />

values(the [ 3 H]-thymidine incorporation )were measured by the liquid<br />

scintillation .Result: From the H.E. sections , the polypeptide can efficiently attenuate<br />

the inflammatory infiltration into the central nervous system. From the ultra structure <strong>of</strong><br />

the tissues, the myelin and axon from polypeptide treatment group were basically<br />

normal.IL-2 and TNF-α <strong>of</strong> EAE group were significantly higher than the control<br />

(* P


pathways, and the change <strong>of</strong> signal molecules after the intervention <strong>of</strong> scorpion<br />

polypeptides.<br />

Key Words: EAE , Kv1.3 channel, T cells, signaling pathways<br />

Acknowledgment: This work was supported by Program for <strong>New</strong> Century Excellent<br />

Talents, Wuhan University (No. NCET-07-0630),the Scientific Research Foundation for<br />

the Returned Overseas Chinese Scholars in State Education Ministry, and Research<br />

Fund for the Doctoral Program <strong>of</strong> Higher Education <strong>of</strong> China (No. 20090141110010).<br />

*Corresponding author<br />

E-mail: hexiaohua@whu.edu.cn or liwxlab@whu.edu.cn


IGF-1 Promote BPH-1 Cell Proliferation Primarily via Activating<br />

mTOR-dependent Translational Increases in Cyclin D Proteins<br />

Ke Gong, Chao Chen and Wenhua Li*<br />

Department <strong>of</strong> Cell Biology, College <strong>of</strong> Life Sciences, Wuhan University, Wuhan<br />

430072, PR China.<br />

Aim: Insulin-like growth factor-1 (IGF-1) and its binding protein (IGFBP-3) play a<br />

crucial role in the initiation and progression <strong>of</strong> prostate and benign prostatic hyperplasia<br />

(BPH). We have previously report that stromally expressed c-Jun protein modulates the<br />

epithelial proliferation via regulating production and paracrine signals <strong>of</strong> IGF-1, which<br />

promote the BPH-1 cells proliferation through upregulate cyclin D protein levels.<br />

Methods: Western blots analyses for protein levels and cell cycle array <strong>of</strong> FACS were<br />

performed. Results: In this study, the exact molecular mechanisms <strong>of</strong> these regulations<br />

were investigated. We found that IGF-1 increases cyclin D proteins and promotes cell<br />

cycle transition from G0 to S phase, bringing in cell proliferation. IGF-1 almost didn’t<br />

affects cyclin D mRNA levels and it primarily stimulates post-transcriptional increases<br />

in cyclin D proteins. IGF-1 stimulation increases cyclin D protein in translation levels<br />

via activation mammalian target <strong>of</strong> rapamycin (mTOR) pathway in BPH-1 cells. Two<br />

vital factors <strong>of</strong> mTOR-downstream, the eukaryotic translation initiation factor-4E<br />

binding protein-1 (4EBP1) and 70-kD ribosomal protein S6 kinase (p70S6K), were<br />

activated after IGF-1 treatment. Blocking mTOR will decrease cyclin D levels and<br />

<strong>of</strong>fset IGF-1 stimulation. Moreover, we found that, in BPH-1cells, activated mTOR by<br />

IGF-1 increases cyclin D protein expression is mediated through activation <strong>of</strong> the<br />

PI3K/Akt pathways cells, but not MAP kinase pathway. Also, our data indicate that<br />

IGF-1 probably influences the combination <strong>of</strong> cyclin D and ubiquitin, and then stabilize<br />

the cyclin D protein. Conclution: These results elucidate the molecular transduction<br />

pathways <strong>of</strong> IGF-1 promote BPH-1 cell proliferation, which will be help to improve our<br />

understanding <strong>of</strong> BPH and develop new clinical therapy for it.<br />

Key Words: IGF-1; BPH; mTOR; cyclinD<br />

*Corresponding Authors:<br />

Wenhua Li, Email: whli@whu.wdu.cn, Tel: 86-27-68756711


GABAB Receptor-Mediated Rap1 Activation and GB1/Rap1GTP<br />

Interaction in Neuronal System<br />

Zongyong Zhang, Siluo Huang,Ninghua Sun,Lin Wu,Yilei Zhang,Jianfeng Liu, Li Su<br />

Sino-France Laboratory for Drug Screening, Key Laboratory <strong>of</strong> Molecular Biophysics <strong>of</strong><br />

Ministry <strong>of</strong> Education, School <strong>of</strong> Life Science and Technology, Huazhong University <strong>of</strong><br />

Science and Technology, Wuhan, 430074 ,China<br />

Rap1, a Ras-related small GTPase, binds to either GTP(Rap1GTP active form ) or GDP<br />

(Rap1GDP inactive form) to regulate cell proliferation, adhesion, or migration.<br />

Gamma-aminobutyric acid (GABA) B receptors, which are GB1/GB2 heterodimer, play<br />

an important role in many physiological activities in neuron. We found that activation<br />

<strong>of</strong> GABAB receptor stimulated with Bacl<strong>of</strong>en or CGP7930, analogs <strong>of</strong> GABA, activated<br />

Rap1 signaling both in cultured mice cerebellar granule neurons (CGNs) and in<br />

HEK293 cell. This activation can be blocked by depleting <strong>of</strong> Ca 2+ in the medium with<br />

Ca 2+ chelator BAPTA. We further found the active Rap1 translocated to premembrane<br />

region and interacted directly with GB1 C-terminal via its hypothetical inter-protein<br />

β-sheet structure.<br />

Key Words : Rap1, GABA, GABAB receptor, protein-protein interaction


Therapeutic Effect <strong>of</strong> Flupirtine on RepetitiveFebrile Seizures Rats<br />

Yanlan Liu, Fang Yu, Biwen Peng, Yuncui Wang, Wanhong Liu, Xiaohua,He*<br />

School <strong>of</strong> Medicine, Wuhan University, Wuhan 430071, China<br />

Aim:KCNQ2 and KCNQ3 encode subunits <strong>of</strong> neuronal M-type K + channels,which are<br />

key regulators <strong>of</strong> brain excitability.This study tests whether Flupirtine , a selective<br />

KCNQ2 activator ,is effective in the experimental model <strong>of</strong> repetitive febrile seizures.<br />

Methods: Our study includes 40 male Sprague-Dawley (SD) rats at postnatal day 10<br />

(P10) divided into normal、febrile seizures、phenobarbital and flupirtine groups. We<br />

induced repetitive febrile seizures starting at postnatal day 10 (P10) with hot water<br />

bath(45.0 ℃ ) for 8 consecutive days. In phenobarbital and flupirtine groups,<br />

phenobarbital and flupirtine at the dose 30mg/kg b.m. was administered respectively 2<br />

hours before febrile seizures . febrile seizures group and normal group rats received<br />

only normal saline. At P28, we induced febrile seizures again to test the rats'<br />

susceptivity to febrile seizures. At P30, the Morris water maze were applied to measure<br />

the rats’ learning and memory abilities in each group. Thereafter, hippocampal slices<br />

were prepared for histological and morphometric examination.<br />

Results: Both Flupirtine and phenobarbital can significantly increase the latency and<br />

decrease the rate and duration <strong>of</strong> febrile seizures when administered before FS(P


Rapid Detection <strong>of</strong> Deletion <strong>of</strong> the PMP22 Gene from a Kindred in China<br />

with Hereditary Neuropathy with Liability to Pressure Palsy by Real-time<br />

Quantitative PCR<br />

Yuan He1, Qiang Wu2, Qianqian Wang1, Wanhong Liu1* and Xiaohua He1*<br />

1School <strong>of</strong> Medicine, Wuhan University, Wuhan 430071, China<br />

2Wuhan General Hospital, Wuhan 430072, China<br />

The hereditary neuropathy with liability to pressure palsy (HNPP) is an<br />

autosomal-dominantly inherited peripheral neuropathy, and the deletion <strong>of</strong> the<br />

peripheral myelin protein (PMP22) gene in chromosome 17p11.2-12 is the main cause<br />

for it, accounting for approximately 70% <strong>of</strong> all cases. In this study we reported the<br />

clinical phenotypes and electrophysiological findings from one patient in a kindred in<br />

china who was affected by HNPP. At the same time, the deletion <strong>of</strong> PMP22 gene which<br />

came from two patients and six asymptomatic normal members in the same kindred was<br />

studied by real-time quantitative PCR using SYBR Green I. The copy number <strong>of</strong> the<br />

PMP22 gene was determined by the comparative threshold cycle method and the<br />

albumin was used as a reference gene. This study disclosed that the PMP22 gene from<br />

two patients did deleted, which was the same as the previous report in other nations, and<br />

six asymptomatic normal members had no deletion except two members. We conclude<br />

the two asymptomatic members with deletion <strong>of</strong> the PMP22 gene might develop some<br />

relative symptoms one day.<br />

Key Words: HNPP; Peripheral neuropathy; PMP22; Real-time quantitative PCR<br />

Acknowledgment: This work was supported by Program for <strong>New</strong> Century Excellent<br />

Talents, Wuhan University (No. NCET-07-0630),the Scientific Research Foundation for<br />

the Returned Overseas Chinese Scholars in State Education Ministry, and Research<br />

Fund for the Doctoral Program <strong>of</strong> Higher Education <strong>of</strong> China (No. 20090141110010).<br />

*Corresponding author<br />

E-mail: liuwanhong@whu.edu.cn or hexiaohua@whu.edu.cn


Therapeutic Effect <strong>of</strong> Flupirtine on Repetitive Febrile Seizures Rats<br />

Yanlan Liu, Fang Yu, Biwen Peng, Yuncui Wang, Wanhong Liu, Xiaohua,He*<br />

School <strong>of</strong> Medicine, Wuhan University, Wuhan 430071, China<br />

Aim:KCNQ2 and KCNQ3 encode subunits <strong>of</strong> neuronal M-type K + channels,which are<br />

key regulators <strong>of</strong> brain excitability.This study tests whether Flupirtine , a selective<br />

KCNQ2 activator ,is effective in the experimental model <strong>of</strong> repetitive febrile seizures.<br />

Methods: Our study includes 40 male Sprague-Dawley (SD) rats at postnatal day 10<br />

(P10) divided into normal、febrile seizures、phenobarbital and flupirtine groups. We<br />

induced repetitive febrile seizures starting at postnatal day 10 (P10) with hot water<br />

bath(45.0 ℃ ) for 8 consecutive days. In phenobarbital and flupirtine groups,<br />

phenobarbital and flupirtine at the dose 30mg/kg b.m. was administered respectively 2<br />

hours before febrile seizures . febrile seizures group and normal group rats received<br />

only normal saline. At P28, we induced febrile seizures again to test the rats'<br />

susceptivity to febrile seizures. At P30, the Morris water maze were applied to measure<br />

the rats’ learning and memory abilities in each group. Thereafter, hippocampal slices<br />

were prepared for histological and morphometric examination.<br />

Results: Both Flupirtine and phenobarbital can significantly increase the latency and<br />

decrease the rate and duration <strong>of</strong> febrile seizures when administered before FS(P


TEF3(Transcription Enhancer Factor 3)Induced Angiogenesis through<br />

VEGF Pathway<br />

Xin Liu1, Cheng Qiao1, and Dezheng Zhao2,4 Huiyan Zeng2,3,4*,<br />

1.Department <strong>of</strong> Microbiological and Biochemical Pharmacy, College <strong>of</strong> Pharmacy ,<br />

Wuhan University, Wuhan 430071, PR China 2 .Departments <strong>of</strong> Pathologyand<br />

Medicine (Division <strong>of</strong> Molecular & Vascular Medicine), 3. Center for vascular Biology<br />

Research 4.Beth Israel Deaconess Medical Center and Harvard Medical School, Boston<br />

MA 02215, USA<br />

Aims: Transcription Enhancer Factor-3 (TEF3, RTEF-1, TEAD4) belongs to a family<br />

<strong>of</strong> transcription factor proteins. Although it has been extensively studied, the signaling<br />

pathways mediating its biological functions during angiogenensis are still not<br />

understood. Recently we identified a novel VEGF targeted gene, Down Syndrome<br />

Candidate Region 1 is<strong>of</strong>orm 1L (DSCR1-1L) that is important for the function <strong>of</strong> VEGF<br />

in pathological angiogenesis and is expressed in human tumor vasculature, not in<br />

normal vasculature, rendering it a specific target for tumor angiogenesis therapy. TEF3<br />

was required for DSCR1-1L expression through binding to the M-CAT site in its<br />

promoter and could be an attractive target for anti-angiogenesis therapy. It was also<br />

reported that TEF3 expression is induced by hypoxia and TEF3 overexpression<br />

increases VEGF gene transcription and in vitro under hypothexia condition. Ours aims<br />

are to investigate the role <strong>of</strong> TEF3 in angiogenesis. Methods: We overexpressed TEF3<br />

in HUVEC cell and we used the yeast two-hybrid system to find the proteins interaction<br />

with the TEF3. Results: We reported previously that another DSCR1 is<strong>of</strong>orm,<br />

DSCR1-1L, was also upregulated by VEGF-A 165 in cultured endothelial cells and in<br />

several in vivo models <strong>of</strong> pathological angiogenesis and that different from DSCR1-4,<br />

DSCR1-1L overexpression alone induced cultured endothelial cell proliferation and<br />

promoted angiogenesis in Matrigel assays. We also demonstrated that TEF3 directly<br />

interacted with the putative M-CAT site in the DSR1-1L promoter in vitro and in vivo.<br />

Finally, Overexpression <strong>of</strong> TEF3 induces HUVEC proliferation, migration and tube<br />

formation. Conclusion: TEF3 upregulatecd DSCR1-1l expression with the VEGF<br />

stimulation,and TEF3 itself can promote angiogenesis in endothelial cells. Then it was<br />

tested that TEF3 also acts as a novel factor in VEGF-induced angiogenesis signaling<br />

pathway.<br />

Key Words: VEGF; TEF3; angiogenesis<br />

Authors:<br />

Xin Liu ,Email: lx@whu.edu.cn;<br />

Huiyan Zeng, Email: hzeng@caregroup.harvard.edu Tel: 86-27-68759006


Does the Metabotropic Glutamate Receptor 7 (Mglur7) Play a Role in<br />

Schizophrenia?<br />

Yali Zhao, Yu Fan, Qi Xu*, Yan Shen*<br />

National Laboratory <strong>of</strong> Medical Molecular Biology, Institute <strong>of</strong> Basic Medical Sciences<br />

Chinese Academy <strong>of</strong> Medical Sciences and Peking Union Medical College, Tsinghua<br />

University. No. 5, Dong Dan San Tiao, Beijing, China, 100005<br />

Background: The metabotropic glutamate receptor 7 (mGluR7) is a class C G-protein<br />

coupled receptor found in the pre-synaptic elements <strong>of</strong> various types <strong>of</strong> neurons, which<br />

plays a critical role in regulating the glutamate transmission. Glutamate is the<br />

predominant excitatory neurotransmitter in the central nervous system. Glutamate<br />

blocking drugs such as phencyclidine can mimic the psychosis symptoms and cognitive<br />

problems associated with schizophrenia, which is a common yet severe psychiatric<br />

disorder and characterized by hallucinations, delusions and cognitive deficits with a<br />

lifetime risk <strong>of</strong> about 1%. Aim: The purpose <strong>of</strong> this study is to investigate the<br />

contribution <strong>of</strong> mGluR7 to schizophrenia susceptibility. We genotyped 14 single<br />

nucleotide polymorphisms (SNPs) located in the functional region <strong>of</strong> mGluR7 among<br />

549 patients with schizophrenia and 562 controls origining from Chinese utilizing<br />

Sequenom MassARRAY iPLEX Gold platform (Sequenom, San Diego, California).<br />

The differences <strong>of</strong> allelic and genotypic frequencies between patients and controls were<br />

examined using Unphased s<strong>of</strong>tware (v 3.0.12). Results: We observed no significant<br />

allelic associations with the disease in any <strong>of</strong> the 14 SNPs. We also observed no<br />

significant genotypic differences between patients and controls. Conclusion: These<br />

results suggested that mGluR7 may not play a major role in schizophrenia pathogenesis<br />

in the Chinese population as it is a multifactor disease.<br />

Key Words: metabotropic glutamate receptor 7; Glutamate; schizophrenia;<br />

Author: Yali Zhao, Tel: 86-10-65296432; Email: hollyzyl@126.com;<br />

Corresponding author: Qi Xu, Tel: 86-10-65296432; Email: qixu@vip.sina.com


Effects <strong>of</strong> Yishendaluo Decoction on Axonal Degeneration, Inflammatory<br />

Reaction, and Neurological Function in a Mouse Model <strong>of</strong> Experimental<br />

Autoimmune Encephalomyelitis<br />

Xiaoling Shang1, Ying Gao2, Ling Yin3, Jintao Zhang4, Shuoren Wang2<br />

1Department <strong>of</strong> Basic Theory <strong>of</strong> Traditional Chinese Medicine, Basic Medical College,<br />

Changchun University <strong>of</strong> Chinese Medicine,Changchun 130117, Jilin Province, China, 2.<br />

Key Laboratory <strong>of</strong> Chinese Internal Medicine, Ministry <strong>of</strong> Education, Dongzhimen<br />

Hospital <strong>of</strong> Beijing University <strong>of</strong> Chinese Medicine, Beijing, 100700, China, 3.Center for<br />

Neuroinformatics, General Hospital <strong>of</strong> Chinese PLA, Beijing 100853, China,<br />

4.Department <strong>of</strong> Neurology, the 88 Hospital <strong>of</strong> Chinese PLA, Taian 271000, Shandong<br />

Province, China<br />

Aim: To investigate the effects <strong>of</strong> Yishendaluo decoction on a mouse model <strong>of</strong> experimental<br />

autoimmune encephalomyelitis. Methods: A total <strong>of</strong> 96 healthy, female, SJL/J mice, aged 8-12<br />

weeks, were equally and randomly assigned to normal, model, hormone, and Chinese medicine<br />

groups. A total <strong>of</strong> 0.2 mL antigen preparation, supplemented with 150 μg PLP139-151 and<br />

400 μg H37RA, was subcutaneously injected into the upper abdomen <strong>of</strong> mice from the model,<br />

hormone, and Chinese medicine groups. Mouse models <strong>of</strong> experimental autoimmune<br />

encephalomyelitis were established by intravenous injection <strong>of</strong> 0.1 mL Bordetella pertussis<br />

solution containing 0.6 × 106 Bordetella pertussis at days 1 and 3. Mice from the model,<br />

Chinese medicine, and hormone groups were respectively subjected to 0.2 mL saline, 2 g/kg<br />

Yishendaluo decoction, and 0.078 mg/kg prednisone acetate, once daily for 14 consecutive days.<br />

Mice from the normal group were left intact. Results: A few inflammatory cell infiltration,<br />

nerve fiber breakage and slight demyelination were detected in the central nervous system <strong>of</strong><br />

mice from the Chinese medicine and hormone groups compared with the model group.<br />

Expression <strong>of</strong> β-amyloid precursor protein and p38 protein was significantly diminished in the<br />

central nervous system <strong>of</strong> mice from the Chinese medicine and hormone groups compared with<br />

the model group (P < 0.05 or P < 0.01), and the decrease was greatest in the Chinese medicine<br />

group. The decrease in mouse weight was not significant, and neurological function scores were<br />

less in the Chinese medicine and hormone groups compared with the model group (P < 0.05 or<br />

P < 0.01). Interferon-γ levels were significantly reduced (P < 0.01), and interleukin-4 levels<br />

were significantly increased (P < 0.01) in the brains <strong>of</strong> the Chinese medicine and hormone<br />

groups, compared with the model group. Conclusion: Yishendaluo decoction improved<br />

neurological function in mice with experimental autoimmune encephalomyelitis by<br />

downregulating β-amyloid precursor protein expression, resisting axonal degeneration, and<br />

relieving inflammatory reaction. The anti-inflammatory mechanism was regulated by inhibition<br />

<strong>of</strong> the p38 mitogen-activated protein kinase signal pathway.<br />

Key Words: autoimmune encephalomyelitis; multiple sclerosis; Yishendaluo decoction; β<br />

-amyloid precursor protein


Imaging Neuronal Population in Vitro with Acousto-Optic Deflector<br />

Based Random Access Two-Photon Microscopy<br />

Shaoqun Zeng, Wei R Chen, Qingming Luo<br />

Britton Chance Center for Biomedical Photonics,Wuhan National Laboratory for<br />

Optoelectronics-Huazhong University <strong>of</strong> Science and Technology, Wuhan 430074,<br />

China<br />

Two-photon microscopy has grown up to be an important technique in biology research<br />

especially in the field <strong>of</strong> neuroscience for its high penetration depth and<br />

three-dimensional selectivity. However, its imaging rate is limited by the mechanic<br />

scanning mechanism and cannot satisfy the requirement for imaging the encoding<br />

pattern <strong>of</strong> brunches <strong>of</strong> dendrite or cell populations in brain tissue. Laser scanning with<br />

two sequential orthogonally oriented (2D) acousto-optical deflectors (AODs) does not<br />

involve actual mechanical movement and thus provides a fast scanning rate, high<br />

precision, and stability. 2D AOD scanning also allows random access to each pixel in<br />

the field <strong>of</strong> view (FOV). Random scanning in regions <strong>of</strong> interest can devote dwell time<br />

to pixels <strong>of</strong> interest and increase the signal-to-noise ratio and the frame-capture rate, and<br />

would provide unique applications in neuroscience research. However this random<br />

scanning two-photon microscopy with femtosecond laser is frustrated by the temporal<br />

and spatial dispersion. With a special dispersion compensation scheme, we have<br />

constructed a random scanning two-photon microscope and are able to track the fast<br />

neuronal activity which is not available with other techniques. In this presentation, we<br />

will show 1) the evolution <strong>of</strong> the femtosecond laser pulse after passing the AOD<br />

scanner, 2) a technique to compensate the spatial and temporal dispersion<br />

simultaneously and facilitate two-dimensional random access two-photon microscopy,<br />

and 3) a special optimization algorithm which is particularly effective to reconstruct the<br />

firing pattern from calcium signal <strong>of</strong> low signal to noise ratio. The whole system is<br />

validated with calcium signals recorded from neurons <strong>of</strong> brain slices.<br />

Key Words: neuronal activity, two-photon microscopy, acousto-optic deflector, firing<br />

pattern<br />

*Corresponding author : Email: sqzeng@mail.hust.edu.cn


Inhibition <strong>of</strong> Protein Phosphatase 2A Activity Plays a Key Role for Normal<br />

Human Cells to Acquire TRAIL-Sensitive Phenotype during Tumorigenesis<br />

Yansheng Li1, Yajun Xiao1, Xuegang Wang1, Shaoyong Chen2, Siping Zeng1, Changshuai<br />

Shao1, Xiaohui Qian1,Rong Wang1, Xuanyu Chen1, Quansheng Du3, Aria F. Olumi4,<br />

Hongmei Yang5, Xiaoping Zhang1§<br />

1. Department <strong>of</strong> Urology, Union Hospital, Tongji Medical School, Huazhong University <strong>of</strong><br />

Science and Technology, Wuhan 430022, China, 2.Cancer Biology Program,<br />

Hematology-Oncology Division, Department <strong>of</strong> Medicine, Beth Israel Deaconess Medical<br />

Center, Harvard Medical School, Boston, Massachusetts 02215, USA 3.Cancer Center,<br />

Medical College <strong>of</strong> Georgia, Augusta, GA 30912, USA4. Department <strong>of</strong> Urology,<br />

Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.<br />

5.Department <strong>of</strong> Pathogen Biology, Tongji Medical School, Huazhong University <strong>of</strong> Science<br />

and Technology, Wuhan 430030, China<br />

Aim: Tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) is a promising<br />

anticancer agent because it induces apoptosis in most cancer cells but spares normal cells. The<br />

aim <strong>of</strong> the study is to determine how normal human cells acquire TRAIL-sensitive phenotype<br />

during the process <strong>of</strong> malignant transformation. Methods: An experimental cell system was<br />

developed by sequential introduction <strong>of</strong> hTERT, SV40 LT, SV40 ST and H-Ras into normal<br />

human prostatic epithelial cells. Then these cells were treated with TRAIL by using MTT assay.<br />

After determining which cells became sensitive to TRAIL, the underlying mechanisms were<br />

explored. Results: For the first time, we demonstrate that introduction <strong>of</strong> SV40 ST into cells<br />

changes TRAIL phenotype from TRAIL-resistant to TRAIL-sensitive, activity inhibition <strong>of</strong> the<br />

protein phosphatase 2A (PP2A) either by SV40 ST or okadaic acid (OA) sensitizes normal<br />

human prostatic epithelial cells to TRAIL-induced apoptosis during premalignant period <strong>of</strong><br />

tumorigenesis and tumor suppressor gene PP2A may exert its antiapoptotic by negatively<br />

regulating c-Fos/AP-1. In addition, low-dose OA treatment sensitizes TRAIL-resistant cancer<br />

cells to TRAIL, which indicates PP2A inhibitors such as OA can potentially used as an<br />

enhancer <strong>of</strong> apoptosis induced by TRAIL or TRAIL-like agents. Conclusion: Inhibition <strong>of</strong><br />

PP2A activity is a key step for normal human cells to acquire TRAIL-sensitive phenotype<br />

during tumorigenesis, and lower PP2A activity might be a major determinant for cancer cells to<br />

be sensitive to TRAIL-induced apoptosis.<br />

Key Words: TRAIL, the protein phosphatase 2A (PP2A), Apoptosis, SV40 ST, Okadaic acid<br />

(OA)<br />

Grant Support: the National Natural Science Foundation <strong>of</strong> China (NSFC) (30572139),<br />

(30872924) and Program for <strong>New</strong> Century Excellent Talents in University from Department <strong>of</strong><br />

Education <strong>of</strong> China (NCET-08-0223) to XZ<br />

§ Corresponding Authors:<br />

Xiaoping Zhang:1277 Jiefang Dadao,Wuhan 430022,Hubei Province,China, Department <strong>of</strong><br />

Urology, Union Hospital, Email: xiaoping.zhang2008@gmail.com


Phone: 86-27-85351625<br />

A Novel Mutation <strong>of</strong> PAX3 on a Large Chinese Family with<br />

Waardenburg Syndrome<br />

Juan Liu1, Xiangfu Meng2, Caixia Sun2, Jingmin Wen1, Yong Gao1, Jing Yu Liu1*<br />

1Key Laboratory <strong>of</strong> Molecular Biophysics <strong>of</strong> the Ministry <strong>of</strong> Education, College <strong>of</strong> Life<br />

Science and Technology, Center for Human Genome Research, Huazhong University<br />

<strong>of</strong> Science and Technology, 430074, Wuan, Hubei, P. R. China;2 People Hospital <strong>of</strong><br />

Xiajin, Xiajin, Shandong, 253200, China<br />

Objectives: Waardenburg syndrome (WS) is an autosomal dominant and genetically<br />

heterogeneous disorder characterized by manifests with sensorineural deafness and<br />

pigmentation defects <strong>of</strong> the hair, skin, and iris. It is classified into four types depending<br />

on the presence or absence <strong>of</strong> additional symptoms, WS1, WS2, WS3 and WS4. WS1<br />

and 3 are due to mutations in the PAX3 gene. WS2 are associated with mutations <strong>of</strong><br />

MITF, SNAI2, and SOX10, in addition, there are two loci, 1p21-p13.3 and 8p23. WS4<br />

can result from mutations in EDNRB and EDN3.Methods: Linkage analysis was used<br />

to identify the chromosomal location <strong>of</strong> the disease gene, direct DNA sequence analysis<br />

was used for mutation detection and restriction fragment length polymorphism (RFLP)<br />

analysis was checked this mutation in the family. Results: A large five-generation<br />

Chinese family from Shandong Provience with autosomal dominant WS1 have<br />

identified and characterized. Linkage analysis was identified with marker D2S126,<br />

which is located near Pax3 gene. The PAX3 gene is closely linked to the disease <strong>of</strong> this<br />

family. Mutational analysis <strong>of</strong> all exons and exon-intron boundaries <strong>of</strong> PAX3, a novel<br />

mutation c.272A>G in the proband was found. RFLP analysis showed that the mutation<br />

is in all patients, but not in all normal individuals and 200 normal controls. The<br />

mutation occurs at a highly conserved residue in the paired domain (PD) <strong>of</strong> PAX3,<br />

which is the DNA binding domain. Conclusion: These results indicate that the<br />

c.272A>C mutation <strong>of</strong> PAX3 gene probably causes WS in the family and expand the<br />

mutation spectrum <strong>of</strong> PAX3 gene.<br />

Key Words: Waardenburg syndrome, PAX3, Linkage analysis, Mutation analysis<br />

*Correspondence to: Dr. J. Y. Liu, liujy@mail.hust.edu.cn


Identification a Novel MYO6 Mutation Associated with Autosomal<br />

Dominant Non-Syndromic Hearing Impairment in a Large Chinese<br />

Family<br />

Yulei Li1,2, Zhenping Xu1,2, Xiangyang Zhang1, Juan Li1, Jing Yu Liu1*<br />

1Key Laboratory <strong>of</strong> Molecular Biophysics <strong>of</strong> the Ministry <strong>of</strong> Education, College <strong>of</strong> Life<br />

Science and Technology, Center for Human Genome Research, Huazhong University<br />

<strong>of</strong> Science and Technology, 430074, Wuan, Hubei, P. R. China; 2Department <strong>of</strong> Life<br />

Science and Technique, Xinxiang Medical University, Xinxiang, 453003, China)<br />

Objectives: Hearing impairment is a disorder <strong>of</strong> partial or complete hearing loss, it is<br />

caused by organic or functional pathological changes in the human auditory system. Hearing<br />

impairment have an estimated incidence rate <strong>of</strong> approximately 1 in 500 newborns with bilateral<br />

congenital sensorineural hearing loss≥40dB in developed countries. And the ratio up to 2.7‰<br />

for the age <strong>of</strong> 5. In the adolescent, this number rises to 3.5‰. Genetic factor is the most<br />

important nosogenesis among all the factors. Most <strong>of</strong> the inherited cases are non-syndromic,<br />

with the only symptom <strong>of</strong> auditory dysfunction, not accompanied with other malformation or<br />

disorder. Most <strong>of</strong> the non-syndromic hearing impairment (NSHI) is caused by monogenic<br />

mutation, and they are genetically heterogeneity. Many autosomal dominant NSHI genes have<br />

been identified. Most <strong>of</strong> these genes have a high expression or specially expressed in the inner<br />

ear cells. By the molecular basis <strong>of</strong> these studies, it gives us a further understands <strong>of</strong> the<br />

physiological mechanism <strong>of</strong> auditory system and the pathological basis <strong>of</strong> hearing<br />

impairment.Methods: Linkage analysis was carried out by STR Markers flanking all known<br />

autosomal dominant NSHI genes and direct DNA sequence for the proband was used to<br />

mutation analysis by ABI PRISM 3100 Genetic Analyzer.Results: A large Chinese Han family<br />

with an autosomal dominant non-syndromic hearing impairment have been identified and<br />

characterized. Linkage analysis <strong>of</strong> genotyping data from the family showed the DFNA22 locus<br />

can not be excluded. There is only one candidate gene MYO6 in this locus which is responsible<br />

for NSHI. Mutational analysis showed a novel missense mutation c.599A>G in exon 8 was<br />

found in proband, it was co-segregated with the affected individuals in the family and did not<br />

exist in the unaffected family members and 100 unrelated normal controls. The mutation results<br />

in a substitution <strong>of</strong> an asparagine residue by a serine residue at codon 200 (p.N200S). The<br />

p.N200S occurred in the motor domain near the ATP binding site <strong>of</strong> myosin VI, and the N200<br />

residue is evolutionally highly conserved from flagellum to human.Conclusion: It is presumed<br />

that the mutation p.N200S in the motor domain maybe affect the generation <strong>of</strong> mechanical force<br />

in myosin VI, and lead to the dysfunction <strong>of</strong> inner ear hair cell, and eventually result in the<br />

hearing impairment.<br />

Key Words: non-syndromic hearing impairment, mutation analysis, myosin VI, p.N200S,<br />

motor domain<br />

*Correspondence to: Dr. J. Y. Liu, liujy@mail.hust.edu.cn


MAPK Scaffolding by BIT1 in the Golgi Complex Modulates Stress<br />

Resistance<br />

Ping Yi1,2,3*, Duc Thang Nguyên4*, Arisa Higa-Nishiyama1,2, Patrick Auguste2,4,<br />

Marion Bouchecareilh1, Michel Dominguez5, Regula Bielmann4, Sandrine Palcy2,<br />

Jian Feng Liu3 and Eric Chevet1,2,3 ‡<br />

1Avenir, INSERM U889, Bordeaux, France; 2Université Victor Segalen Bordeaux 2, IFR<br />

66, F-33076, Bordeaux, France; 3Key Laboratory <strong>of</strong> Molecular Biophysics <strong>of</strong> Ministry <strong>of</strong><br />

Education, School <strong>of</strong> Life Science and Technology, Huazhong University <strong>of</strong> Science and<br />

Technology, Wuhan, Hubei, China; 4Department <strong>of</strong> Surgery, McGill University,<br />

Montreal, QC, Canada; 5HyperOmics Farma, Montreal, QC, Canada;<br />

*These authors contributed equally to this work<br />

The endoplasmic reticulum (ER) is an essential organelle whose major functions are to<br />

ensure proper secretory protein folding and trafficking. These mechanisms involve the<br />

activation <strong>of</strong> specific ER-resident molecular machines, which might be regulated by<br />

their membranous environments. Based on this observation, we aimed to characterize<br />

the proteome <strong>of</strong> ER-membrane microdomains to identify new components <strong>of</strong> the ER<br />

that have a role in secretory pathway-associated functions. Using this approach with dog<br />

pancreatic rough microsomes, we found that mitochondrial Bcl-2 inhibitor <strong>of</strong><br />

transcription (BIT1) localized in the early secretory pathway and accumulated in the<br />

Golgi complex. Using both a chimeric protein <strong>of</strong> the luminal and transmembrane<br />

domains <strong>of</strong> ER-resident TRAPa and the cytosolic domain <strong>of</strong> BIT1, and silencing <strong>of</strong><br />

BIT1 expression, we perturbed endogenous BIT1 oligomerization and localization to<br />

the Golgi. This led to enhanced ERK signaling from the Golgi complex, which resulted<br />

in improved stress resistance. This work provides the first evidence for the existence<br />

<strong>of</strong> ER microdomains that are involved in the regulation <strong>of</strong> BIT1 structure and<br />

trafficking, and identifies BIT1 as a negative regulator <strong>of</strong> the ERK-MAPK signaling<br />

pathway in the Golgi.<br />

Key Words: BIT1, MAPK, Secretory pathway, Stress<br />

‡Author for correspondence: Eric Chevet, eric.chevet@u-bordeaux2.fr


Identification <strong>of</strong> a Novel Genetic Locus on Chromosome 8p21.1-q11.23<br />

for Idiopathic Basal Ganglia Calcification<br />

Xiaohua Dai,1# Yong Gao,1# Zhenping Xu,1,2# Xiaoniu Cui,1 Juan Liu,1 Yulei Li,1,2<br />

Haibo Xu,3 Mugen Liu,1 Qing K Wang,1,4,* and Jing Yu Liu 1,*<br />

1Key Laboratory <strong>of</strong> Molecular Biophysics <strong>of</strong> the Ministry <strong>of</strong> Education, College <strong>of</strong> Life<br />

Science and Technology, Center for Human Genome Research, Huazhong University<br />

<strong>of</strong> Science and Technology, 430074, Wuan, Hubei, P. R. China;2Department <strong>of</strong> Life<br />

Science and Technique, Xinxiang Medical University, Xinxiang, 453003,<br />

China;3Department <strong>of</strong> Radiology, Union Hospital <strong>of</strong> Huazhong University <strong>of</strong> Science<br />

and Technology, Wuhan, 430074, China;4Department <strong>of</strong> Molecular Cardiology, Lerner<br />

Research Institute, Cleveland Clinic, Cleveland 44195, USA<br />

#Contributed equally to this work.<br />

Objectives:Idiopathic basal ganglia calcification (IBGC) is a neurodegenerative<br />

disorder that is characterized by basal ganglia and extrabasal ganglia calcification, and<br />

usually inherited in an autosomal dominant pattern. To date, two genetic loci for IBGC<br />

were identified on chromosomes 14q and 2q, but further genetic heterogeneity clearly<br />

exists. Methods: Linkage analysis was used to identify the chromosomal location <strong>of</strong> the<br />

disease gene.Results: A large five-generation Chinese family from Henan Provience<br />

with an autosomal dominant inheritance, family IBGC have identified and characterized.<br />

Linkage analysis excluded the 14q13 and 2q37 loci and other known related genes with<br />

extrapyramidal disease in the nervous system, including ApoE, Tau, ACT, BChE-K,<br />

APP, PS1, PS2 and VLDL-R. Using the family, significant linkage was identified with<br />

markers on chromosome 8p21.1-q11.23 with a maximum LOD score <strong>of</strong> 4.10. Fine<br />

mapping defined the new genetic locus within a 25.0 cM region between markers<br />

D8S1809 and D8S1833. Interpretation: Future studies <strong>of</strong> the candidate genes at the<br />

8p21.1-q11.23 locus may lead to identification <strong>of</strong> a disease-causing gene with IBGC.<br />

Key Words: Idiopathic basal ganglia calcification (IBGC); Linkage analysis; Genetics;<br />

Calcification; LOD score<br />

*Correspondence to:<br />

Dr. J. Y. Liu, liujy@mail.hust.edu.cn<br />

Dr. Q. K. Wang, qkwng@mail.hust.edu.cn


Phosphoinositide-3-Kinase Pathway Activation in PTEN Deficient<br />

Prostate Cancer Cells is Independent <strong>of</strong> Receptor Tyrosine Kinases and<br />

Mediated by the p110 or p110 Catalytic Subunits<br />

Xinnong Jiang1,2, Sen Chen1, John Asara1, Steven P. Balk1,*<br />

1Division <strong>of</strong> Hematology-Oncology, Beth Israel Deaconess Medical Center/Harvard<br />

Medical School, Boston, MA, USA<br />

2 Huazhong University <strong>of</strong> Science and Technology, College <strong>of</strong> Life Science and<br />

Technology 1037 Luoyu Road, Wuhan, Hubei<br />

Class IA phosphoinositide-3-kinase (PI3K) p110 catalytic subunits are activated upon<br />

SH2 domain mediated binding <strong>of</strong> their p85 regulatory subunits to tyrosine<br />

phosphorylated pYxxM motifs in receptor tyrosine kinases (RTKs) or adaptor proteins.<br />

The PI3K pathway is activated by PTEN loss in the majority <strong>of</strong> prostate cancers (PCa),<br />

but the contribution <strong>of</strong> upstream RTKs that may be targeted therapeutically has not been<br />

assessed. Immunoblotting <strong>of</strong> p85 associated proteins in serum starved PTEN deficient<br />

LNCaP and C4-2 PCa cells showed a small set <strong>of</strong> discrete tyrosine phosphorylated<br />

proteins, but these proteins were not recognized by an anti-pYxxM motif antibody and<br />

were not observed in PTEN deficient PC3 PCa cells. LC/MS/MS and immunoblotting<br />

showed that p85 was associated primarily with p110b and p110d. An interaction with<br />

ErbB3 was also detected in serum starved cells LNCaP cells, but was independent <strong>of</strong><br />

ErbB3 tyrosine phosphoryation and was not required for basal PI3K activity. Basal<br />

tyrosine phosphorylation <strong>of</strong> p110 and p110 could be blcoked by c-Src inhibitors,<br />

but this did not suppress basal PI3K activity, which was similarly independent <strong>of</strong> Ras.<br />

Using siRNA specific for each p110 is<strong>of</strong>orm we found that basal PI3K activity was<br />

mediated by p110 in PC3 cells, and by both p110 �and p110 �in LNCaP cells, while<br />

p110 was required for PI3K activation in response to RTK stimulation by<br />

heregulin- 1. Taken together, these findings indicate that basal PI3K activity in PTEN<br />

deficient PCa cells is RTK independent and mediated by p110 and p110.


Insulin-Like Signaling Pathway Functions in Integrative Response to an<br />

Olfactory and a Gustatory Stimuli in Caenorhabditis elegans<br />

Ya-Ming Jiu1, Zheng-Xing Wu1 , Tao Xu 1,2,<br />

1Key Laboratory <strong>of</strong> Molecular Biophysics, Ministry <strong>of</strong> Education, and Institute <strong>of</strong><br />

Biophysics and Biochemistry, Huazhong University <strong>of</strong> Science and Technology, Luoyu<br />

Road 1037#, Wuhan 430074, PR China<br />

2 National Laboratory <strong>of</strong> Biomacromolecules, Institute <strong>of</strong> Biophysics, Chinese<br />

Academy <strong>of</strong> Sciences, Beijing 100101, PR China<br />

Animals integrate various environmental stimuli within the nervous system to generate<br />

proper behavioral responses. However, the underlying neural circuits and molecular<br />

mechanisms are largely unknown. The insulin-like signaling pathway is known to<br />

regulate dauer formation, fat metabolism, and longevity in Caenorhabditis elegans (C.<br />

elegans). Here, we show that this highly conserved signaling pathway also functions in<br />

the integrative response to an olfactory diacetyl and a gustatory Cu 2+ stimuli. Worms <strong>of</strong><br />

wild-type N2 Bristol displayed a strong avoidance to the Cu 2+ barrier in the migration<br />

pathway to the attractive diacetyl. Mutants <strong>of</strong> daf-2 (insulin receptor), daf-18 (PTEN<br />

lipid phosphatase), pdk-1 (phosphoinositide-dependent kinase), akt-1/-2 (Akt/PKB<br />

kinase) and sgk-1 (serum- and glucocorticoid-inducible kinase) show severe defects in<br />

the elusion from the Cu 2+ . Mutations in DAF-16, a forkhead-type transcriptional factor,<br />

suppress the integrative defects <strong>of</strong> daf-2 and akt-1/-2 mutants. We further report that<br />

neither cGMP nor TGFβ pathways, two other dauer formation regulators, likely plays a<br />

role in the integrative learning. These results suggest that the insulin-like signaling<br />

pathway constitutes an essential component for sensory integration and decision-making<br />

behavior plasticity.<br />

Key Words: C. elegans, insulin signaling pathway, sensory integration, decision<br />

making behavior<br />

Author: Ya-Ming Jiu, Email: hustjym@163.com, Tel: 15110039473


Akt Overexpression Opposes the Beta Amyloid Toxic Injuries to<br />

HEK293/tau Cells<br />

Yin Gang,Zhou xin-wen,Wang jian-zhi<br />

Department <strong>of</strong> pathology and pathophysiology, the key laboratory <strong>of</strong> neurological<br />

diseases <strong>of</strong> the ministry <strong>of</strong> education ,tongji medical college,huazhong university <strong>of</strong><br />

science and technology,wuhan 430030,china<br />

Aim:To study the effect <strong>of</strong> a direct akt overexpression on Aβ-induced toxic injuries to<br />

neuronal cells.. Methods:To evaluate the effect <strong>of</strong> Aβ-induced toxic injuries on<br />

neuronal cells,we treated HEK293/tau neurons with several doses <strong>of</strong> Aβ for 24h.cells<br />

were simultaneously transfected with several doses <strong>of</strong> a gene akt for 48h. CCK-8 KIT<br />

assay the cell viability,and Reactive oxygen species assay kit assay the level <strong>of</strong> the level<br />

<strong>of</strong> the Reactive oxygen species.the apoptosis was detected by flow cytometry.Results:<br />

CCK-8 KIT assay showed that Aβ decreased neuronal cell viability in a<br />

concentration-dependent manner and also that akt overexpression effectively prevented<br />

Aβ-induced neuronal cell death. Aβ treatments increased ROS production in<br />

HEK293/tau cells. Overexpression <strong>of</strong> Akt but not Akt mutant in the HEK293/tau cells<br />

significantly attenuated Aβ induced ROS production and lipids peroxidation.<br />

Furthermore, overexpression <strong>of</strong> Akt also attenuated Aβ-induced mitochondrial<br />

dysfunction and apoptosis, and promoted cell survival in HEK293/tau cells.Conclusion:<br />

the neurotoxic effect <strong>of</strong> Aβ can be partially prevented by akt overexpression.<br />

Key Words: amyloid-beta;akt overexpression; toxic injury;oxidative stress<br />

Author: Yin gang (1977-) was born in xiantao hubei,now I am study in Huazhong<br />

University <strong>of</strong> Science and Technology for Ph.D, The direction <strong>of</strong> reseach is the<br />

diagnosis and treatment <strong>of</strong> alzheimer’s disease. Email: 7706190@163.com, Tel:<br />

13986672302


NMDA Receptors Subunit NR2B Messenger Rnas Expression with<br />

Cognitive Dysfunction after Bilateral Common Carotid Artery Occlusion<br />

in the Adult Rats<br />

Lianjun Guo, Yang Cheng, Bo Zhao<br />

Department <strong>of</strong> Pharmacology, Tongji Medical College, Huazhong University <strong>of</strong> Science<br />

and Technology, Wuhan 430030, P.R. China<br />

Aim: To explore whether and how NR2B mRNA may be altered in global incomplete<br />

chronic cerebral ischemic rat model. Methods: Chronic cerebral hypoperfusion in Male<br />

SD rats was performed by permanent occlusion <strong>of</strong> the bilateral common carotid artery<br />

(2VO) for 30 days. Morris water maze was used to measure spatial learning and<br />

memory performance. Electrophysiological equipments were used to record long-term<br />

potentiation (LTP). In situ hybridization and reverse transcriptase polymerase chain<br />

reaction (RT-PCR) assays were used to investigate NR2B mRNA may be altered in<br />

global incomplete chronic cerebral ischemic rat model. Results: the spatial learning<br />

and memory function <strong>of</strong> rats attenuated by longer escape latency, shorter time spent in<br />

the target quadrant and impaired long term potentiation (LTP) after chronic cerebral<br />

ischemia. In the in situ hybridization experiment, NR2B declined to 58.82% and<br />

45.60% <strong>of</strong> the control values in the neocortex and hippocampal region separately after<br />

chronic cerebral ischemia. NR2B mRNA in the hippocampal region and neocortex was<br />

markedly down regulated by ischemia, reaching 43.57% and 30. 05% <strong>of</strong> the control<br />

values respectively in the semi-quantitative RT-PCR experiment. Conclusions: Our data<br />

suggest that NR2B subunit plays an important role for the spatial learning and memory<br />

function <strong>of</strong> rats.<br />

Key Words: NR2B messenger RNA; hippocampal neurons; cerebral ischemia<br />

Acknowledgments: This work was supported by grants from the National Natural<br />

Science Foundation <strong>of</strong> China (NSFC, No. 30772559) to Lianjun Guo<br />

Author: Lianjun Guo , Tel:02783691763, Email: gljyl@yahoo.com.cn


Effects <strong>of</strong> ZD7288 on Long-Term Potentiation at Perforant Pathway(PP)<br />

Fibers - CA3 Region Synapse Pathway in Rat Hippocampus in Vivo<br />

Xulin Xu1, Min Zheng1, 2 , Lian-Jun Guo1*,<br />

1Department <strong>of</strong> Pharmacology, Tongji Medical College, Huazhong University <strong>of</strong><br />

Science & Technology, 13 Hongkong Road, Wuhan 430030, China.<br />

2 Department <strong>of</strong> Pharmacology,Xianning College, 88 Xianning Thoroughfare,<br />

Xianning 437100, China.<br />

Aim: To investigate effects <strong>of</strong> a hyperpolarization-activated cyclic nucleotide-gated cation<br />

(HCN) channel specific blocker ZD7288 to LTP at perforant pathway (PP) fibers-CA3 region<br />

synapse pathway in rat hippocampus. Methods: The field excitatory postsynaptic potentials<br />

(fEPSPs) <strong>of</strong> PP—CA3 region synapse in rats was recorded by using the extracellular<br />

recording techniques in vivo. Tetanic stimulation (TS; consisted <strong>of</strong> 50 trains at 0.5 Hz each<br />

composed <strong>of</strong> 4 pulses at 500Hz) was administrated to observe the induction and maintenance <strong>of</strong><br />

LTP. We observed the effects <strong>of</strong> ZD7288 on the induction and sustain <strong>of</strong> LTP in PP—CA3<br />

pathway. Simultaneously, the synapse ultrastructural at hippocampal CA3 region was observed<br />

by transmission electron microscope. Results: ①The fEPSPs amplitude persistently stabilized<br />

at 281.8 ± 6.6% <strong>of</strong> baseline in 90min after giving tetanic stimulation <strong>of</strong> PP fibers in<br />

LTP-induction group and significantly higher than that in control group(97.4 ±1.8%)(P < 0.01).<br />

② Local administration <strong>of</strong> ZD7288 100nmol/L before TS, the induction <strong>of</strong> LTP was obviously<br />

suppressed. The relative fEPSPs amplitudes in each time point were all significantly lower than<br />

that in LTP-induction group (n=10, P 0.05).③Local administration <strong>of</strong> ZD7288<br />

100nmol/L at 30min after TS, the fEPSPs amplitudes at each time point in 1h after<br />

administration were all significantly lower than LTP-induction group (n=10, P < 0.01), and the<br />

average values were 95.8 ± 6.4% near to control group(P > 0.05). ④ In the LTP induction group,<br />

the cell ultramicrostructure at hippocampus CA3 region showed that there were many synaptic<br />

vesicles in presynaptic endochylema and many synaptic vesicles like thyrsiform occurred in the<br />

presynaptic end in this synapses. ⑤In the ZD7288 -TS group, the cell ultramicrostructure at<br />

hippocampus CA3 region showed that there were no synaptic glomerulus and the synaptic<br />

vesicles were decreased compared with the LTP induction group. Conclusions: It is suggest<br />

that ZD7288 could produce inhibitive effects on the induction and maintenance <strong>of</strong> long-term<br />

potentiation at PP-CA3 synapse pathway and this action may be associated with altering the<br />

release <strong>of</strong> neurotransmitter.<br />

Key Words: ZD7288; HCN channel; PP—CA3 pathway ; LTP<br />

Acknowledgments: This work was supported by grants from the National Natural Science<br />

Foundation <strong>of</strong> China (NSFC, No. 30772559) to Lianjun Guo<br />

Author: Xulin Xu , Email: xulinxu@yahoo.com , Tel: 027-83691762;<br />

* Correspondence author: Tel: +86 27 83691762, E-mail: gljyl@yahoo.com.cn


Effects <strong>of</strong> Cscl on Synaptic Transmission at Perforant Pathway (PP)<br />

Fibers - CA3 Region <strong>of</strong> Hippocampus in Rat<br />

Min Zheng1, 2 ,Xulin Xu1, Lian-Jun Guo1*,<br />

1Department <strong>of</strong> Pharmacology, Tongji Medical College, Huazhong University <strong>of</strong> Science &<br />

Technology, 13 Hongkong Road, Wuhan 430030, China.<br />

2 Department <strong>of</strong> Pharmacology,Xianning College, 88 Xianning Thoroughfare, Xianning<br />

437100, China.<br />

Aim: To investigate the contribution <strong>of</strong> a HCN channel nonspecific blocker CsCl to synaptic<br />

transmission at PP-CA3 pathway in rat hippocampus. Methods: The population spike (PS) <strong>of</strong><br />

PP-CA3 region in rats was recorded by using the extracellular recording techniques in vivo.<br />

Tetanic stimulation(TS; consisted <strong>of</strong> 50 trains at 0.5 Hz each composed <strong>of</strong> 4 pulses at 500Hz)<br />

was administrated to observe the induction and maintenance <strong>of</strong> long-term potentiation(LTP). We<br />

observed the effects <strong>of</strong> CsCl on the induction and sustain <strong>of</strong> LTP in PP-CA3 pathway.<br />

Simultaneously, the synapse ultrastructural at hippocampal CA3 region was observed by<br />

transmission electron microscope. Results: ①The PS amplitude level persistently stabilized at<br />

281.8 ± 6.6% <strong>of</strong> baseline in 90min after giving tetanic stimulation <strong>of</strong> PP fibers in LTP-induction<br />

group and significantly higher than that in control group(97.4 ±1.8%)(n=10, P < 0.01); ②<br />

Local administration <strong>of</strong> CsCl 5µmol /L before TS, the relative PS amplitudes in each time point<br />

were all significantly lower than that in LTP-induction group (n=10, P


Embryonic Heart Growth is Regulated by Functional Antagonism<br />

Between GRIDLOCK and GATA5 in Zebrafish<br />

Haibo Jia, Sameer Chopra, Mingwei Ni, xiaoling Jiang, Xiaoqun Guan, Terri T. Ni, Sam<br />

Wells and Tao P. Zhong<br />

Departments <strong>of</strong> Medicine and Cell & Developmental Biology, Vanderbilt Medical<br />

School, Nashville, TN 37232<br />

Embryonic heart growth is revealed primarily by the proliferation <strong>of</strong> cardiomyocytes<br />

and an increase in their cell size, but the mechanisms that control heart growth during<br />

development are not well understood. In our current study we present evidence that<br />

the Hairy-related bHLH transcription factor Gridlock (Grl) negatively regulates heart<br />

growth in the zebrafish embryo. We show that a reduction in Grl activity by mutation<br />

causes an increase in myocardial gene expression and cardiomyocyte number, resulting<br />

in the development <strong>of</strong> hyperplastic hearts. Conversely, elevation <strong>of</strong> Grl activity causes<br />

differentiating cardiomyocytes to fail to divide and grow, thereby reducing the heart size<br />

in the embryo. These Grl-dependent cardiac growth effects are counterbalanced by<br />

Gata5, a transcriptional activator that promotes cardiomyocyte expansion. The<br />

antagonistic relationship between Grl and Gata5 is mediated through protein<br />

interactions that squelch the Gata5 transcriptional activity to inhibit myocardial gene<br />

expression. Hence, the opposing effects <strong>of</strong> Grl and Gata5 serve to regulate embryonic<br />

heart growth during development


Quantitative Phosphoproteome Analysis Reveals Signaling Events<br />

Involved in Aroma-Stimulated Mouse Olfactory Bulbs<br />

Lei-Ke Zhang1, Fei Liu2, Xiao-Ping Rao2, Fu-Qiang Xu2, Lin Guo1<br />

1College <strong>of</strong> Life Sciences, Wuhan University, Wuhan 430072, P. R. China<br />

2 State Key Laboratory <strong>of</strong> Magnetic Resonance and Atomic and Molecular Physics,<br />

Wuhan Centre for Magnetic Resonance, Wuhan Institute <strong>of</strong> Physics and Mathematics,<br />

Chinese Academy <strong>of</strong> Sciences,Wuhan 430071, P.R.China<br />

Stimulation <strong>of</strong> odorant molecules is transduced by olfactory receptor neurons (ORNs)<br />

located in the olfactory epithelium <strong>of</strong> the nasal cavities. Each ORN projects a single<br />

axon to the main olfactory bulb (MOB), where it terminates on the second-order<br />

neurons <strong>of</strong> the MOB in glomeruli, the initial site for the coding and processing <strong>of</strong> odor<br />

information. In the glomeruli, ORN axons terminate on the dendrites <strong>of</strong> mitral/tufted<br />

cells, which in turn project into higher cortical structures. The activity in the MOB is<br />

determined by the input from the SONs, the centrifugal inputs that include regulatory<br />

and feedback fibers from many brain regions, and the nerurocircuits within the MOB.<br />

Finding the proteins that are involved in responding to olfactory stimulation can help to<br />

reveal the mechanism <strong>of</strong> sensory information processing. Here, signaling events in<br />

odor-stimulated MOBs were explored using quantitative phosphoproteomic analysis.<br />

Our preliminary results showed that a 10 mg protein mixture from the MOB lysate<br />

resulted in the identification and quantification <strong>of</strong> >1000 unique phosphorylation sites<br />

and ~700 unique phosphoproteins. Further analysis revealed that many up- or<br />

down-regulated phosphorylation events were known having functions in signal<br />

transductuon after odor stimulating. For example, Thr-34 <strong>of</strong> DCLK1 is<strong>of</strong>orm 1<br />

(accession number: IPI00468380) was up-regulated 8 fold, and Ser-181 <strong>of</strong> Pgrmc1<br />

(accession number: IPI00319973) was down-regulated 2.13 fold.


Quantitative Phosphoproteomics Analysis <strong>of</strong><br />

GABAB Receptor Signaling<br />

Meng FU1, Chanjuan XU2, Jianfeng LIU2,, Lin GUO1,<br />

1College <strong>of</strong> Life Science, Wuhan University, Wuhan, Hubei, P. R. China, 430072<br />

2College <strong>of</strong> Life Science and Technology, Huazhong University <strong>of</strong> Science and<br />

Technology, Wuhan, Hubei, P. R. China, 430074<br />

The G-protein-coupled receptors (<strong>GPCR</strong>s) play important role in cell-cell interaction.<br />

GABAB receptor, one <strong>of</strong> the best known <strong>GPCR</strong> family members, is activated by the<br />

neurotransmitter GABA and is expressed in most neurons. In the current study,<br />

primary cells from cerebellar granule neurons (CGNs) and cerebra cortex were cultured<br />

using SILAC (stable isotope labeling by amino acid in cell culture) method, with<br />

‘SILAC-Light’ as the control group, and ‘SILAC-Heavy’ treated with bacl<strong>of</strong>en, an<br />

agonist specific to mammalian GABAB receptors. After treatment, cell lysate were<br />

mixed 1:1 (protein ratio), and phosphopeptides were enriched by immobilized metal<br />

affinity chromatography (IMAC) method. A hydrophilic interaction chromatography<br />

(HILIC) fractionation step was used before phosphopeptide sequencing and<br />

quantification by liquid chromatography tandem mass spectrometry (LC-MSMS)<br />

analysis. In the preliminary data, we obtained quantitative information for 727<br />

phosphopeptides from cerebra cortex and 254 phosphopeptides from CGNs.<br />

Key wors: Phosphoproteomics, GABAB Receptor, Cell signaling,


Autotaxin Signaling via Lysophosphatidic Acid Receptors Contributes to<br />

LPC-Induced Vascular Smooth Muscle Cell Proliferation and Migration<br />

Bao liang, Zhao Gui-jun, Zhang qian,Yang wen-hua, Yang De-zhi, Li Ying-hui,Alatan<br />

Gaole*<br />

(Department <strong>of</strong> Biochestry and Molecular Biology ,College <strong>of</strong>Life Sciences,Inner<br />

Mongolia University,Hohhot,010021)<br />

Background: Lysophosphatidylcholine (LPC) is a major bioactive phospholipid<br />

component <strong>of</strong> oxidized low-density lipoprotein (Ox-LDL) and is implicated as a critical<br />

factor in the atherogenic activity <strong>of</strong> Ox-LDL. LPC is believed to play an important role<br />

in atherosclerosis and inflammatory diseases by altering various functions in a number<br />

<strong>of</strong> cell-types. We have examined that LPC stimulated the proliferation and migratory<br />

responses <strong>of</strong> cultured human vascular smooth muscle cells(VSMC),and these actions<br />

were inhibited by Ki16425,a receptor antagonist for the ATX enzymatic product<br />

lysophosphatidic acid (LPA) . We also find that ATX were strongly expressed in the<br />

cells. However, the ATX product mediating the increase in cellular proliferation and<br />

motility remain to be identified. Here,we reported that ATX expressed in the cells<br />

contributes to Lpc-induced VSMC Proliferation and migration .Results: LPC and LPA<br />

stimulated hVSMCs proliferation and migration in a PTX-sensitive manner. These<br />

actions were suppressed by using SiRNA sepecific to ATX and LPA1 receptor,Ki16425.<br />

Furthermore, The LPC- and LPA-induced proliferation <strong>of</strong> hVSMCs was markedly<br />

inhibited by a potent calcineurin inhibitor cyclosporine A and PD98059,a ERK inhibitor.<br />

Instead, an epidermal growth factor (EGF) receptor tyrosine kinase inhibitor and P38<br />

inhibitor markedly suppressed the migration response to LPC and LPA without having<br />

any significant effect on proliferation <strong>of</strong> VSMC. .Conclusion: The LPC-induced<br />

stimulation <strong>of</strong> proliferation in VSMC is mediated by the ATX enzymatic product LPA<br />

through LPA1receptors / Gi/o-proteins/ ERK- MAP kinase /Ca 2+ /calcineurin pathways .<br />

The migration response to LPC and LPA are mediated by the ATX enzymatic product<br />

LPA through LPA1-receptors / Gi/o-proteins/ P38- MAP kinase signaling pathway and<br />

also mediated by transactivation <strong>of</strong> EGF receptor pathways .<br />

Key words: Autotaxin, Lysophosphatidylcholine, Vascular smooth muscle<br />

cells(VSMC), LPA-receptors signaling pathway<br />

Acknowledge: Supported by the National Science Foundation <strong>of</strong> China (Grant No.<br />

30760052), was also supported in part by Fostering Talents in Basic Research <strong>of</strong> the<br />

National Natural Science Foundation <strong>of</strong> China (Grant No.J0730648 ).<br />

Author*: Alatan gaole, PhD, Pr<strong>of</strong>esser , Department <strong>of</strong> Biochemstry and Molecular<br />

Biology , College <strong>of</strong> Life Sciences , Inner Mongolia University. Adress:No.235,West<br />

University Street, Hohhot, Inner Mongolia, 010021,Tel:0471-4993197, E-mail:<br />

bigaole@imu.edu.cn


Morphine Regulated Thioredoxin-1 Expression<br />

in Neuroblastoma SH-SY5Y Cells<br />

Yuemei Feng, Kui Li, Shengdong Wang, Yanhui Li, Junying Song, Fucheng Luo, Jie<br />

Bai*<br />

College <strong>of</strong> life science and technology, Kunming University <strong>of</strong> Science and Technology,<br />

Kunming 650224, China<br />

Aim: Thioredoxin-1 (TRX-1) plays multiple roles in regulation <strong>of</strong> redox, inhibition <strong>of</strong><br />

apoptosis and regulation <strong>of</strong> transcription factors. Recent studies showed that morphine<br />

could cause oxidative stress in cells and tissues. The aim <strong>of</strong> current study is to<br />

investigate the expression <strong>of</strong> TRX-1 under morphine administration in human<br />

neuroblastoma SH-SY5Y cells. Methods: Cell culture, Western blotting. Results: The<br />

expression <strong>of</strong> TRX-1 was decreased after 1-2h then increased from 4 -24h after<br />

morphine treatment in human neuroblastoma SH-SY5Y. The decreased expression <strong>of</strong><br />

TRX-1 by morphine was restored by an inhibitor <strong>of</strong> GSK3β, lithium chloride. The<br />

increased expression <strong>of</strong> TRX-1 induced by morphine was suppressed by LY294002,<br />

which is one <strong>of</strong> the PI3K inhibitor and was suppressed by the antagonist <strong>of</strong> opioid<br />

receptor, naloxone. The increased expression <strong>of</strong> TRX-1 recovered to normal level after<br />

morphine withdrawal. These data suggested that TRX-1 was induced through the opioid<br />

receptor and the PI3K/GSK3β pathway. The expression <strong>of</strong> TRX-1 is dependent on the<br />

morphine administration. Conclusion: Our study suggested that TRX-1 might be<br />

associated with morphine tolerance and abuse.<br />

Key words: thioredoxin-1, morphine, neuroblastoma SH-SY5Y cells<br />

*Author for correspondence: Tel.: +86 13354980126; fax: +86 8713801191, E-mail:<br />

jiebai662001@yahoo.com.cn


Cyclin D1 Promotes Anchorage-independent Cell Survival by Inhibiting<br />

FOXO-Mediated Anoikis<br />

Lu Gan 1, Xiangliang Yang 1, Huibi Xu 1, Haojie Huang 2<br />

1. College <strong>of</strong> Life Science and Technology, Huazhong University <strong>of</strong> Science and<br />

Technology, Wuhan 430074, China<br />

2. Masonic Cancer Center, University <strong>of</strong> Minnesota, Minneapolis, MN 55455, USA<br />

Aim: Adhesion to an appropriate extracellular matrix (ECM) is important for normal<br />

cells to survive, and detachment from such supportive matrices usually triggers a<br />

specific type <strong>of</strong> apoptosis termed anoikis. However, most cancer cells are commonly<br />

resistant to anoikis and can survive in the presence <strong>of</strong> an inappropriate ECM or in the<br />

absence <strong>of</strong> anchorage to the ECM. O-class Forkhead box (FOXO) transcription factors<br />

are critical regulators <strong>of</strong> diverse cellular processes, including apoptosis, cell-cycle arrest,<br />

DNA damage repair and oxidative stress resistance. In this study we determined<br />

whether FOXOs are involved in anoikis and its mechanism. Methods: Annexin V-PE<br />

apoptosis detection kit and s<strong>of</strong>t agar colony formation assay were used to measure the<br />

anoikis in prostate cancer cells. FOXOs activities were determined using a generic<br />

FOXO luciferase reporter gene that contains three copies <strong>of</strong> forkhead response elements.<br />

Immunoprecipitation was used to determine the endogenous and exogenous interaction<br />

between cyclin D1 and FOXO. In vitro protein binding assay was used to confirm the<br />

interaction between the two proteins. In vitro kinase activity determined whether<br />

FOXO1 was phosphorylated by CDK4/CDK6. Results: FOXO1 and FOXO3a play an<br />

essential role in promoting cell detachment-induced anoikis. In contrast, elevated cyclin<br />

D1 inhibits anoikis. We further show that cyclin D1 interacts with FOXO proteins and<br />

impedes their transcriptional regulatory and anoikis-promoting functions. This effect <strong>of</strong><br />

cyclin D1 requires its transcription repression domain but is independent <strong>of</strong><br />

cyclin-dependent kinases CDK4 and CDK6. Moreover, we demonstrate that<br />

cancer-derived mutants <strong>of</strong> cyclin D1 are much more stable than wild-type cyclin D1<br />

under anchorage-independent conditions and possess a greater antagonistic effect on<br />

FOXO-regulated anoikis and anchorage-independent growth <strong>of</strong> cancer cells.<br />

Conculsion: These data suggest that cyclin D1 may play a critical role in tumorigenesis<br />

and cancer metastasis by inhibiting the anoikis-promoting function <strong>of</strong> FOXO proteins.<br />

Keywords: Foxo; cyclin D1; Anoikis; protein-protein interaction<br />

Author: Lu Gan,PhD, College <strong>of</strong> Life Science and Technology, Huazhong University<br />

<strong>of</strong> Science and Technology, Wuhan 430074, China, Tel: 86-27-87792147<br />

E-mail: lugan@mail.hust.edu.cn


Investigate the Role <strong>of</strong> Rab27 in the Dcvs Docking and Priming Process<br />

in C. elegans<br />

Wan-juan Feng<br />

Key Laboratory <strong>of</strong> Molecular Biophysics, Ministry <strong>of</strong> Education, College <strong>of</strong> Life Science<br />

& Technology, Huazhong University <strong>of</strong> Science & Technology<br />

Objective Rab27 is a member <strong>of</strong> a large family <strong>of</strong> Ras-related small GTPases. Recent<br />

studies have suggested that Rab27 play an important role in dense-core vesicle<br />

exocytosis. However, which step does Rab27 take part in and what is the precise<br />

mechanisms by which the GTPases regulate dense-core vesicle exocytosis are not clear.<br />

Methods We have succeeded in monitoring the docking and fusion <strong>of</strong> single dense core<br />

vesicles (DCVs) employing total internal reflection fluorescence microscopy (TIRFM)<br />

in primary cultured C. elegans neurons. Results Using these approaches, we provide<br />

direct evidence that Rab27 regulates the docking step <strong>of</strong> DCVs at the plasma<br />

membrane in C. elegans.<br />

Keywords: Rab27, total internal reflection fluorescence microscopy(TIRFM), dense<br />

core vesicle(DCV)<br />

Author: E-mail: wanjuanfeng@gmail.com


Biophotons as Neural Communication Signals<br />

Yan Sun, Chao-Wang, Jia-pei Dai *<br />

Wuhan Institute for Neuroscience and Neuroengineering, South-Central<br />

University for Nationalities, Wuhan 430074, China<br />

Aim: Cell to cell communication by biophotons has been demonstrated in plants,<br />

bacteria and certain animal cells. Whether such a signal communication way is existed<br />

in neural cells is unclear. Methods: Here by developing a new method, named as in situ<br />

biophoton autography, we investigated the biophotonic activity in rat spinal nerve roots<br />

in vitro. Results: We found that different spectral light stimulations (infrared, red,<br />

yellow, blue, green and white) can generate biophotons that conduct along the neural<br />

fibers. Such an effect can be significantly inhibited by procaine (a neural conduction<br />

blocker) or classic metabolic inhibitors, suggesting that biophotons may serve as neural<br />

communication signals. The mechanism <strong>of</strong> biophotonic conduction along neural fibers<br />

may be mediated via protein-protein biophotonic interactions. Conclusion: Biophtonic<br />

activities might be an important way for neural information exchange (biophotonic<br />

transmission) besides well-demonstrated electrical and chemical transmission. This<br />

study may provide a new thought for better understanding <strong>of</strong> the fundamental<br />

mechanisms <strong>of</strong> neural communications and the functions <strong>of</strong> nervous system such as<br />

vision, learning and memory, as well as the mechanisms in human neurological<br />

diseases.<br />

Keywords: Biophotons, in situ biophoton autography, neural communication signals,<br />

spinal nerve root.<br />

* Corresponding author<br />

E-mail: jdai@mail.scuec.edu.cn; Tel: 0086-27-67840917


Effects <strong>of</strong> Cyclic Nucleotide Gated Channel Knockout on the Olfactory<br />

Bulb Analyzed by Quantitative Phospho-Proteomics<br />

Fei Liu 1 , Xiao-Ping Rao, Lei-Ke Zhang 2 , Lin Guo 2 , Fuqiang Xu 1<br />

1State Key Laboratory <strong>of</strong> Magnetic Resonance and Atomic and Molecular<br />

Physics, Wuhan Centre for Magnetic Resonance, Wuhan Institute <strong>of</strong> Physics<br />

and Mathematics Chinese Academy <strong>of</strong> Sciences, Wuhan 430071, P. R. China<br />

2College <strong>of</strong> Life Sciences, Wuhan University, Wuhan 430072, P. R. China<br />

When odorant molecules enter nasal cavity and are dissolved in the mucus covering the<br />

olfactory epithelium, they bind with odorant receptor proteins in the cilia <strong>of</strong> the<br />

olfactory sensory neurons, leading to a series <strong>of</strong> signal transduction processes:<br />

activation <strong>of</strong> trimeric G-proteins and then nucleotide cyclase, to increase cAMP<br />

concentration, opening <strong>of</strong> cyclic nucleotide gated (CNG) channel, and at last, generating<br />

action potentials that are sent to the olfactory bulb. CNG channel, mainly expressed in<br />

visual and olfactory systems, is non-selective to cations. It is necessary to lead to<br />

depolarization <strong>of</strong> the olfactory sensory neurons. Knockout <strong>of</strong> one <strong>of</strong> its subunit, cnga2,<br />

give rise to anosmic mice. The transgenic animals have much lower surviving rate,<br />

thinner olfactory epithelium, smaller olfactory bylb and less aggressive behavior. To<br />

investigate the effects <strong>of</strong> long term absence <strong>of</strong> peripheral stimulation on the olfactory<br />

bulb, we used quantitative phosphor-proteomics to analyze the change <strong>of</strong> protein<br />

phosphorylation. We found hundreds <strong>of</strong> phosphorylated proteins and even more<br />

phosphorylated sites. Generally, proteins with significant changes in phosphorylation<br />

are these related to signal transduction, neuronal transmission, homeostasis and<br />

neurogenesis. For examples, GAD1, a GABA synthase, is decrease to 19%; CAMK2b, a<br />

kinase, to 33%; and Slc4a4, a electrogenic sodium bicarbonate cotransporter, to 17%<br />

<strong>of</strong> the control, while Hsp90aa1, heat shock protein HSP 90-alpha, increased by a<br />

101%; and Alpl, a tissue-nonspecific alkaline phosphatase by 346%. The exact roles <strong>of</strong><br />

these proteins in the olfactory bulb are under investigation.<br />

Key words: olfactory bulbs, transgenic mice, phosphoproteins, IMAC/LC-MS

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

Saved successfully!

Ooh no, something went wrong!