14.01.2013 Views

Visit our Expo - Redox and Inflammation signaling 2012

Visit our Expo - Redox and Inflammation signaling 2012

Visit our Expo - Redox and Inflammation signaling 2012

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.

- 1 -<br />

Luxemb<strong>our</strong>g,<br />

January 25th to 28th, 2006<br />

European Conference Center<br />

Kirchberg - Luxemb<strong>our</strong>g<br />

Cell Signaling World 2006<br />

Signal Transduction Pathways as therapeutic targets<br />

Proceedings<br />

<strong>and</strong><br />

Program<br />

Editor<br />

Marc Diederich<br />

Organized by<br />

Recherches Scientifiques Luxemb<strong>our</strong>g<br />

Printing of the Proceedings sponsored by the<br />

Fondation de Recherche Cancer et Sang (Luxemb<strong>our</strong>g)


- 2 -


Table of content<br />

PREFACE ......................................................................................................................................................................5<br />

ACKNOWLEDGMENTS ...........................................................................................................................................7<br />

GENERAL INFORMATION .....................................................................................................................................8<br />

EXHIBITION ..............................................................................................................................................................11<br />

SCIENTIFIC PROGRAM ........................................................................................................................................14<br />

WEDNESDAY JANUARY 25TH, 2006.......................................................................................................................14<br />

THURSDAY JANUARY 26TH, 2006 (EARLY MORNING SESSION) .........................................................................15<br />

THURSDAY JANUARY 26TH, 2006 (MORNING SESSION)......................................................................................15<br />

THURSDAY JANUARY 26TH, 2006 (EVENING SESSION) .......................................................................................17<br />

FRIDAY JANUARY 27TH, 2006 (EARLY MORNING SESSION) ...............................................................................18<br />

FRIDAY JANUARY 27TH, 2006 (MORNING SESSION) ...........................................................................................18<br />

FRIDAY JANUARY 27TH, 2006 (EVENING SESSION).............................................................................................20<br />

SATURDAY JANUARY 28TH, 2006 (EARLY MORNING SESSION)..........................................................................21<br />

SATURDAY JANUARY 28TH, 2006 (MORNING SESSION)......................................................................................21<br />

ORAL PRESENTATIONS........................................................................................................................................23<br />

WORKSHOP PRESENTATIONS.........................................................................................................................107<br />

POSTER PRESENTATIONS .................................................................................................................................117<br />

SESSION I. PROTEIN DOMAINS ............................................................................................................................118<br />

SESSION II. RECEPTOR SIGNALING AND G PROTEINS.......................................................................................127<br />

SESSION III. PROTEIN KINASE CASCADES AS THERAPEUTIC TARGETS ...........................................................183<br />

SESSION IV. PROTEIN KINASE INHIBITORS: INSIGHTS INTO DRUG DESIGN FROM STRUCTURE....................257<br />

SESSION V. PHOSPHATASES AS KEY CELL SIGNALING INTERMEDIATES.........................................................266<br />

SESSION VI. PROTEASOME DEGRADATION PATHWAYS ....................................................................................276<br />

SESSION VII. STEM CELL SPECIFIC CELL SIGNALING MECHANISMS...............................................................281<br />

SESSION VIII. INFLAMMATION SPECIFIC SIGNALING .......................................................................................284<br />

SESSION IX. NOVEL COMPOUNDS TARGETING INFLAMMATORY SIGNALING PATHWAYS.............................324<br />

SESSION X : CELL DEATH IN CANCER .................................................................................................................333<br />

SESSION XI : CELL DEATH AND CARDIOVASCULAR DISEASES.........................................................................445<br />

SESSION XII : CELL DEATH AND NEURODEGENERATIVE DISEASES ................................................................458<br />

SESSION XIII : CELL SIGNALING PATHWAYS LEADING TO REGULATED CHROMATIN MODIFICATIONS .....488<br />

SESSION XIV : TRANSCRIPTIONAL AND TRANSLATIONAL CONTROL..............................................................503<br />

SESSION XV : REACTIVE OXYGEN SPECIES AND CELL SIGNALING..................................................................585<br />

SESSION XVI : CHEMOPREVENTIVE AGENTS ....................................................................................................592<br />

SESSION XVII : CELL SIGNALING IN HEALTH AND DISEASE ............................................................................607<br />

LIST OF PARTICIPANTS .....................................................................................................................................672<br />

- 3 -


- 4 -


Preface<br />

In 1998, we organized the first specialized meeting in the field of signal tr<strong>and</strong>uction <strong>and</strong> gene<br />

expression in Luxemb<strong>our</strong>g. This type of meeting was originally thought to teach doctoral<br />

students involved in the cellular <strong>and</strong> molecular biology teaching program (DEA de<br />

Pharmacologie moléculaire) of the University of Nancy I (France).<br />

Since then, 3500 fundamental <strong>and</strong> clinical researchers were gathering in Luxemb<strong>our</strong>g for<br />

different meeting to discuss therapeutic applications in the field of signal transduction,<br />

transcription <strong>and</strong> translation. These meeting allowed new insights into a rapidly moving field.<br />

Novel antibodies against receptors, protein kinase inhibitors, antisense oligonucleotides<br />

targeting both signal transduction <strong>and</strong> gene expression will certainly enhance therapeutic<br />

approaches for the next century.<br />

For <strong>our</strong> 2006 edition of meeting, with more than 950 participants on site, I am convinced that<br />

this meeting will be a great success.<br />

Welcome to Luxemb<strong>our</strong>g !<br />

Marc Diederich<br />

- 5 -


- 6 -


Acknowledgments<br />

This meeting has been realized under the aegis of :<br />

Recherches Scientifiques asbl<br />

The Fondation de "Recherche Cancer et Sang"<br />

Assosciation An Haerz fir kriibskrank Kanner<br />

We have the pleasure to acknowledge support from :<br />

Kuwait Petroleum SA - Luxemb<strong>our</strong>g<br />

The City of Luxemb<strong>our</strong>g<br />

The Fondation de Recherche "Cancer et Sang"<br />

Alexis Biochemicals<br />

Novartis Luxemb<strong>our</strong>g<br />

Y<strong>our</strong> onsite organization team :<br />

Franck Morceau, Romain Blasius, Estelle Henry, Marie-Hélène Teiten, Serge Eifes, Simone<br />

Reuter, Silvia Cristofanon, Isabelle Buck, Florence Folmer, Anne-Laure Joly, Delphine<br />

Magard, Emilie C<strong>our</strong>tine, Audrey Richard, Athanase Visvikis <strong>and</strong> Marc Diederich<br />

- 7 -


Meeting Venue<br />

General Information<br />

All meeting sessions are held at the European Congress Center.<br />

European Congress Center - Quartier Européen Sud,<br />

1, rue du Fort Thüngen<br />

L-1499 Luxemb<strong>our</strong>g-Kirchberg<br />

Registration will take place in the area outside of the "Hémicycle" (Level 0) at the registration desk<br />

open daily (9h00-19H30).<br />

Coffee breaks will be served in the exhibit area daily (See general program for details).<br />

Lunch<br />

For the participants that pre-paid lunch, lunch is served from 12h00 - 14h00 at two locations :<br />

Salon Bleu (Level -1) <strong>and</strong> Bar de l'Hémicycle (Level -1)<br />

There is no difference between the two lunch sites, both take about 350 participants. Additional tickets<br />

are NOT available at the registration desk.<br />

Exhibits are open daily form January 25 th to 28 th , 2006.<br />

The Exhibit opens on Wednesday January 25 th (Welcome reception) <strong>and</strong> ends on Friday January 27 st ,<br />

2006 after the afternoon coffee break. Exhibitors are nevertheless welcome to stay on Saturday<br />

January 28th, 2006.<br />

Lecture rooms (see expo map for locations) :<br />

- Main lecture room : Hemicycle : Level 0<br />

- Lecture room Havanne : Level 0<br />

Posters (levels -1 <strong>and</strong> 0)<br />

There will be f<strong>our</strong> poster viewings :<br />

Thursday January 26th<br />

Group A (13h00 - 15h00)<br />

Session I. Protein Domains<br />

Session II. Receptor <strong>signaling</strong> <strong>and</strong> G proteins<br />

Session III. Protein kinase cascades as therapeutic targets<br />

Session IV. Protein kinase inhibitors: insights into drug design from structure<br />

- 8 -


Group B (15h00 - 17h00)<br />

Session V. Phosphatases a key cell <strong>signaling</strong> intermediates<br />

Session VI. Proteasome degradation pathways<br />

Session VII. Stem cell specific cell <strong>signaling</strong> mechanisms<br />

Session VIII. <strong>Inflammation</strong> specific <strong>signaling</strong><br />

Session IX. Novel compounds targeting inflammatory <strong>signaling</strong> pathways<br />

Session XV. Reactive oxygen species <strong>and</strong> cell <strong>signaling</strong><br />

Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease<br />

On Thursday evening, after Group B, the poster boards need to be cleared in order to allow re-<br />

numbering of the boards for the next day. The organizers will otherwise need to take the posters off<br />

<strong>and</strong> will not be responsible for lost or damaged posters.<br />

Friday January 27th<br />

Group C (13h00 - 15h00)<br />

Session X. Cell death in cancer<br />

Session XI. Cell death <strong>and</strong> cardiovascular diseases<br />

Session XII. Cell death <strong>and</strong> neurodegenerative diseases<br />

Group D (15h00 - 17h00)<br />

Session XIII. Cell <strong>signaling</strong> pathways leading to regulated chromatin modifications<br />

Session XIV. Transcriptional <strong>and</strong> translational control<br />

Session XVI. Chemopreventive agents<br />

Welcome reception (expo area)<br />

On Wednesday January 25, 200 from 19H30-20H30 at the expo surface.<br />

Gala Diner<br />

The Gala Diner will take place at the Hotel Le Royal (12, Boulevard Royal, L-2449 Luxemb<strong>our</strong>g) on<br />

Thursday, January 26th starting at 21H00. Additional tickets are NOT available at the registration<br />

desk.<br />

Transportation<br />

Our bus shuttles will bring the participants from the hotels to the meeting center in the morning <strong>and</strong><br />

back to the hotels or city center in the evening. Please note that the busses leave between 6h30 <strong>and</strong><br />

- 9 -


8h00 from y<strong>our</strong> hotel depending on the distance to the meeting center. The busses are red, orange <strong>and</strong><br />

yellow <strong>and</strong> are from the bus company "Demy Cars".<br />

Taxis can be called from the registration desk.<br />

How to reach the congress center ?<br />

- by taxi :<br />

from the airport (Luxemb<strong>our</strong>g - Findel) in about 15 minutes.<br />

from the railroad station (about 15 minutes)<br />

- by bus : take bus line number 16 (every 20 minutes from the city center)<br />

- by car :<br />

from France : Highway A4 from Metz, take Highway A1 (E44) direction Trier Plateau de<br />

Kirchberg Aéroport, choose exit number "8", orient towards "Quartier Europeen Sud Luxemb<strong>our</strong>g-<br />

Centre"<br />

from Belgium : Highway A411 from Brussels, take Highway A6 (E44) direction Trier Plateau<br />

de Kirchberg Aéroport, choose exit number "8", orient towards "Quartier Europeen Sud Luxemb<strong>our</strong>g<br />

Centre"<br />

from Germany : Highway A6 (E44) from Trier, direction Luxemb<strong>our</strong>g, choose exit number<br />

"8", orient towards "Quartier Europeen Sud Luxemb<strong>our</strong>g Centre".<br />

- 10 -


- 11 -<br />

Exhibition


- 12 -


<strong>Visit</strong> <strong>our</strong> <strong>Expo</strong><br />

(in alphabetical order)<br />

Abgent Europe 11<br />

Active Motif 7<br />

ALEXIS Biochemicals 2<br />

amaxa GmbH 37<br />

AMBION 6<br />

Ariadne Genomics 44<br />

AXXORA Platform 3<br />

BD Biosciences 14 <strong>and</strong> 15<br />

Berthold Detection Systems GmbH 29<br />

Biobase GmbH 10<br />

Biochemical J<strong>our</strong>nal/Portl<strong>and</strong> Press 23<br />

Bio-Connect BV 27<br />

Biognost bvba 9<br />

Bio-Rad Laboratories NV/SA 18<br />

Bios<strong>our</strong>ce 20<br />

CCSBE NV Sales 31<br />

Cell Signaling Technology 40<br />

Diagenode 30<br />

Eurogentec 24<br />

GENTAUR Molecular Products 32<br />

Greiner Bio One 21<br />

Invitrogen 19<br />

InvivoGen 5<br />

Lecuit Opto-Technical S.A. 16 <strong>and</strong> 17<br />

Merck Biosciences GmbH 39<br />

Miltenyi Biotec 4<br />

MP Biomedicals 41<br />

nanoTools Antikoerpertechnik 13<br />

New York Academy of Sciences 25<br />

Olympus Belgium NV 43<br />

Perbio Science 35<br />

PerkinElmer NV/SA 42<br />

Polyplus-transfection 22<br />

Prophac 1<br />

R&D Systems Europe Ltd 26<br />

Science/AAAS 36<br />

SEROTEC LTD 28<br />

Sigma-Aldrich 38<br />

tebu-bio 33 <strong>and</strong> 34<br />

VWR International 8<br />

Westburg 12<br />

Internet café <strong>and</strong> wireless internet access generously<br />

offered by Cadolto, prefabricated buildings<br />

http://www.cadolto.com<br />

http://www.prefabriques.lu<br />

- 13 -


Scientific Program<br />

Wednesday January 25th, 2006<br />

9h00 – 14h00 : Registration of the participants<br />

12h00 – 14h30 : Lunch is served<br />

14h00 - 19h00: Keynote session<br />

14h00 – 15h00 : Gregg L. Semenza (Institute for Cell Engineering, The Johns Hopkins<br />

University School of Medicine, Baltimore, USA):<br />

Keynote: Hypoxia Signal Transduction in Health <strong>and</strong> Disease<br />

15h00 – 15h30 : Michael B. Yaffe (Center for Cancer Research, Department of Biology<br />

Massachusetts Institute of Technology, Cambridge, MA, USA):<br />

Phosphoserine/threonine-binding domains: Integrators of protein kinase <strong>signaling</strong> pathways in<br />

cell cycle control <strong>and</strong> cancer<br />

15h30 – 16h00 : Mathieu Bollen (Afdeling Biochemie, Faculteit Geneeskunde, Katholieke<br />

Universiteit Leuven, Leuven, Belgium):<br />

Targeting of Polycomb repressive complexes by the moonlighting protein NIPP1<br />

16h00 – 16h30 : Coffee break <strong>and</strong> visit of the expo<br />

16h30 – 17h00 : Jurg Tschopp (Department of Biochemistry, University of Lausanne, BIL<br />

Biomedical Research Center, Epalinges, Switzerl<strong>and</strong>):<br />

Conserved <strong>signaling</strong> mechanisms in plant <strong>and</strong> vertebrate innate immunity<br />

17h00 – 17h30 : Roger J. Davis (Howard Hughes Medical Institute, UMASS Medical<br />

School, Worcester, USA):<br />

Signal transduction by stress-activated MAP kinases<br />

17h30 – 18h00 : Laurent Meijer (CNRS, Roscoff, France):<br />

Multiple intracellular effects of pharmacological inhibitors of cyclin-dependent kinases<br />

18h00 – 18h30 : François Fuks (Free University of Brussels, Faculty of Medicine,<br />

Laboratory of Molecular Virology, Brussels, Belgium) :<br />

Mechanisms of DNA methylation in mammals<br />

19h00 : Official inauguration <strong>and</strong> welcome reception<br />

- 14 -


Thursday January 26th, 2006 (Early morning session)<br />

7h30 – 8h30 : Receptor Signaling : Short communications<br />

7h30 – 7h40 : Jan. S. Jepsen (Danish Cancer Society, Copenhagen, Denmark ) :<br />

Protein Kinase C alpha as a potential target in the treatment of acquired antiestrogen<br />

resistance of human breast cancer.<br />

7h40 – 7h50 : Raymond R. Mattingly (Department of Pharmacology <strong>and</strong> Karmanos Cancer<br />

Institute, Wayne State University, Detroit, Michigan, U.S.A.)<br />

Increased EGF Receptor expression <strong>and</strong> activity is induced by deregulated N-Ras <strong>and</strong> may<br />

provide a therapeutic target in NF1 neurofibrosarcoma<br />

7h50 – 8h00 : Shyamala Maheswaran (Department of Surgical Oncology, Massashusetts<br />

General Hospital, Boston, MA , USA) :<br />

Loss of the Antiproliferative Gene BTG2 in Estrogen Receptor Positive Breast Carcinoma is<br />

Associated with Tumor Grade <strong>and</strong> Overexpression of Cyclin D1 Protein<br />

8h00 – 8h10 : Xiao C. Li (Division of Hypertension <strong>and</strong> Vascular Research, Henry Ford<br />

Hospital, Detroit, MI 48202, USA ) :<br />

Cross-talk between angiotensin II <strong>and</strong> glucagon receptor <strong>signaling</strong> mediates activation of<br />

mitogen-activated protein kinases ERK 1/2 in rat glomerular mesangial cells.<br />

8h10 – 8h20 : Metoda Lipnik-Stangelj (Department of Pharmacology <strong>and</strong> Experimental<br />

Toxicology, Faculty of Medicine, University of Ljubljana, Slovenia) :<br />

Multiple role of histamine H1 receptor-PKC-MAPK signalling pathway in histaminestimulated<br />

nerve growth factor secretion from glial cells<br />

8h20 – 8h30 : Ying E. Zhang (Laboratory of Cellular <strong>and</strong> Molecular Biology, Center for<br />

Cancer Research, National Cancer Institute, National Institutes of Health,<br />

Bethesda, Maryl<strong>and</strong>, USA) :<br />

Ubiquitin ligase Smurf1 controls osteoblast activity <strong>and</strong> bone homeostasis by targeting<br />

MEKK2-JNK pathway<br />

Thursday January 26th, 2006 (Morning session)<br />

8h30 - 10h30: Receptor <strong>signaling</strong><br />

8h30 – 9h00 : Walter Birchmeier (Max-Delbruck-Center for Molecular Medicine, Berlin,<br />

Germany):<br />

The role of Met-Gab1 Signalling in vivo.<br />

9h00 – 9h30 : Ulf R. Rapp (Institut fuer Medizinische Strahlenkunde und Zellforschung<br />

(MSZ), Wuerzburg, Germany):<br />

Role of Prohibtin <strong>and</strong> E-cadherin in Raf mediated oncogenesis<br />

9h30 – 10h00 : Johannes L. Bos (Dept of Physiological Chemistry, Utrecht, The<br />

Netherl<strong>and</strong>s):<br />

Epac <strong>and</strong> Rap1 in the control of cell adhesion<br />

10h00 – 10h10 : Jean-Paul Borg (Molecular Pharmacology, Marseille Cancer Institute,<br />

UMR599 Inserm-Institut Paoli-Calmettes, Marseille, France) :<br />

- 15 -


The mammalian tumor suppressor Scribble has a key role in cell migration<br />

10h10 – 10h20 : Tarik Issad (CNRS UMR8104, INSERM U567, Université René Descartes<br />

Paris 5, Institut Cochin, Paris, France) :<br />

The use of BRET (Bioluminescence Resonance Energy Transfer) for the study of tyrosinekinase<br />

receptors<br />

10h20 – 10h30 : Atsufumi Kawabata (Division of Physiology <strong>and</strong> Pathophysiology, School<br />

of Pharmaceutical Sciences, Kinki University, Japan) :<br />

Cell <strong>signaling</strong> by proteinase-activated receptor-2 for interleukin-8 formation in human lung<br />

epithelial cells<br />

10h30 – 11h00 : Coffee break <strong>and</strong> visit of the expo<br />

11h00 - 13h00: Protein kinase cascades as therapeutic targets<br />

11h00 – 11h30 : Dario Alessi (MRC Protein Phosphorylation Unit, School of Life Sciences,<br />

MSI/WTB Complex, University of Dundee, Scotl<strong>and</strong>):<br />

Regulation <strong>and</strong> function of the WNK1 <strong>and</strong> WNK4 protein kinases that are mutated in<br />

Gordon’s hypertension syndrome.<br />

11h30 – 12h00 : Jacques Pouysségur (Institute of Signaling, Developmental Biology <strong>and</strong><br />

Cancer Research CNRS UMR 6543, Centre A. Lacassagne, Nice, France):<br />

Nutrient stress <strong>signaling</strong> <strong>and</strong> tumor progression<br />

12h00 – 12h30 : Brian Hemmings (Friedrich Miescher Institute for Biomedical Research,<br />

Basel, Switzerl<strong>and</strong>):<br />

The NDR kinase family: Regulators of cell proliferation <strong>and</strong> morphogenesis<br />

12h30 – 13h00 : Ying Xia (Center for Environmental Genetics <strong>and</strong> Department of<br />

Environmental Health, University of Cincinnati Medical Center, Cincinnati, USA):<br />

The MEKK1-JNK Pathway Regulates Mouse Eyelid Morphogenesis<br />

12h00 – 14h30 : Lunch is served<br />

13h00 – 17h30 : Workshops<br />

Thursday January 26th, HEMICYCLE room<br />

13h00-14h00 : Becton-Dickinson<br />

14h00-15h00 : Ambion 1<br />

15h00-16h30 : Biobase 1<br />

16h30-17h30 : Amaxa<br />

Thursday January 26th, HAVANNE room<br />

13h00-14h00 : CST 1<br />

14h00-15h00 : CST 2<br />

15h00-16h00 : Miltenyi Biotech 1<br />

13h00 – 15h00 : Poster session I<br />

15h00 – 17h00 : Poster session II<br />

17h00 – 17h30 : Coffee break <strong>and</strong> visit of the expo<br />

- 16 -


Thursday January 26th, 2006 (Evening session)<br />

17h30 - 20h00: Cell death in cancer<br />

17h30 – 18h00 : Caroline Dive (Cellular <strong>and</strong> Molecular Pharmacology Group, Paterson<br />

Institute for Cancer Research, Christie Hospital NHS Trust, Manchester, United Kingdom):<br />

title to be announced<br />

18h00 – 18h30 : Peter H. Krammer (Tumorimmunology Program, German Cancer<br />

Research Center, Heidelberg, Germany):<br />

Signaling of life <strong>and</strong> death in T cells<br />

18h30 – 19h00 : Guido Kroemer (CNRS, UMR8125, Institut Gustave Roussy, Villejuif,<br />

France):<br />

Mechanisms of caspase-independent cell death<br />

19h00 – 19h30 : Michael J. Pinkoski (MRC Toxicology Unit, Leicester, United Kingdom):<br />

Live <strong>and</strong> let die by 73 - mechanisms of degradation <strong>and</strong> chemoresistance<br />

19h30 - 19h45 : Sidhartha Ray (Arnold & Marie Schwartz College of Pharmacy & Health<br />

Sciences Div. of Pharmaceutical Scs/Long Isl<strong>and</strong> University, Brooklyn, NY, USA):<br />

How Antitoxic <strong>and</strong> Anticancer signals Maneuver Programmed <strong>and</strong> Unprogrammed Cell<br />

Death In Vivo?<br />

19h45 – 20h00 : Lina Ghibelli (University Tor Vergata, Rome, Italy):<br />

Oxidative Bax activation as the trigger of the stress-induced intrinsic apoptotic pathway<br />

21-00 – 24h00 : Gala diner at Hotel Le Royal<br />

- 17 -


Friday January 27th, 2006 (Early morning session)<br />

7h30 – 8h30 : Cell Signaling in health <strong>and</strong> disease : short communications I<br />

7h30 – 7h40 : Bernd Schmeck (Department of Internal Medicine/Infectious Diseases,<br />

Charité, Universitätsmedizin Berlin, Germany ) :<br />

Listeria monocytogenes induced Rac1-dependent endothelial histone modification <strong>and</strong><br />

cytokine release<br />

7h40 – 7h50 : Mayra Solis (Terry Fox Molecular Oncology Group, Lady Davis Institute<br />

McGill University, Montreal Canada) :<br />

Distinct roles of IRF-3 <strong>and</strong> IRF-7 in the activation of anti-tumor properties of human<br />

macrophages<br />

7h50 – 8h00 : Maria C. Albertini (Istituto di Chimica Biologica A. Fornaini, Universita' di<br />

Urbino, Urbino <strong>and</strong> Universita' di Roma Tor Vergata, Rome, Italy) :<br />

Mechanisms involved in the anti-apoptotic effect of magnetic fields<br />

8h00 – 8h10 : Michal Hayun (Safdié Institute for AIDS <strong>and</strong> Immunology Research, Faculty<br />

of Life Sciences, Israel) :<br />

The immunomodulator AS101 induces growth arrest <strong>and</strong> apoptosis in Multiple Myeloma cells<br />

via the PI3-K/Akt pathway.<br />

8h10 – 8h20 : Livio Mallucci (Cell Signaling Laboratory, Pharmaceutical Sciences Research<br />

Division, King's College London, Franklin Wilkins Building, London, UK) :<br />

PI3K Targeting by the Beta-GBP Cytokine. Mitogenic Input <strong>and</strong> Therapeutic Response<br />

8h20 – 8h30 : Chantal Schwartz (Laboratoire d’Hémato-Cancérologie Expérimentale, CRP-<br />

Santé, Luxemb<strong>our</strong>g) :<br />

Survey on in vitro cell death induction of multiple myeloma cells upon valproic acid<br />

treatment.<br />

Friday January 27th, 2006 (Morning session)<br />

8h30 - 10h30: <strong>Inflammation</strong> specific <strong>signaling</strong><br />

8h30 – 9h00 : Bharat B. Aggarwal (Cytokine Research Section, The University of Texas M.<br />

D. Anderson Cancer Center, Houston, USA):<br />

Targeting Inflammatory Cell <strong>signaling</strong> pathway for Prevention <strong>and</strong> Treatment of cancer<br />

9h00 – 9h30 : Mark Ginsberg (Department of Medicine, University of California San<br />

Diego, La Jolla, CA, USA):<br />

Integrin Signaling in Cancer <strong>and</strong> <strong>Inflammation</strong><br />

9h30 – 10h00 : Young-Joon Surh (National Research Laboratory of Molecular<br />

Carcinogenesis <strong>and</strong> Chemoprevention, College of Pharmacy, Seoul National University,<br />

South Korea):<br />

<strong>Redox</strong>-Sensitive Transcription Factors as Prime Targets for Chemoprevention <strong>and</strong><br />

Cyoprotection<br />

10h00 – 10h15 : Irfan Rahman (Department of Environmental Medicine, Lung Biology <strong>and</strong><br />

Disease Program, University of Rochester Medical Center, Rochester, NY, USA):<br />

- 18 -


Regulation of inflammation <strong>and</strong> glucocorticoid <strong>signaling</strong> by curcumin <strong>and</strong> resveratrol<br />

10h15 – 10h30 : Moncef Zouali (INSERM U606, Centre Viggo Petersen, Hôpital<br />

Lariboisière, Paris, France):<br />

Signaling pathways controlling self-tolerance: On the road to systemic autoimmune diseases<br />

10h30 – 11h00 : Coffee break <strong>and</strong> visit of the expo<br />

11h00 - 13h00: Cell <strong>signaling</strong> in health <strong>and</strong> disease<br />

11h00 – 11h30 : Bernd Groner (Georg-Speyer-Haus, Institute for Biomedical Research,<br />

Frankfurt am Main, Germany):<br />

Signaling events in mammary epithelial cell differentiation <strong>and</strong> breast cancer<br />

11h30 – 12h00 : John Hiscott (Molecular Oncology Group Lady Davis Institute for Medical<br />

Research, Jewish General Hospital, Montreal, Canada):<br />

TLR-dependent <strong>and</strong> -independent pathways leading to the interferon antiviral response<br />

12h00 – 12h20 : Anna Krichevsky (Center for Neurologic Diseases, Brigham <strong>and</strong> Women's<br />

Hospital<br />

Harvard Medical School, Boston, USA):<br />

A role of microRNAs in brain tumors (preliminary title)<br />

12h20 – 12h40 : Dominique Stehelin (CNRS UMR 8526, Institut de Biologie de Lille,<br />

France):<br />

New steps toward the use of parvovirus H1 as a therapeutic anti-cancer drug.<br />

12h40 – 13h00 : Jacques Emile Dumont (Institut de Recherche Interdisciplinaire (IRIBHM),<br />

Universite Libre de Bruxelles (ULB), Belgique):<br />

Molecular Biology of Thyroid Cancer<br />

12h00 – 14h30 : Lunch is served<br />

13h00 – 17h30 : Workshops<br />

Friday January 27th, HEMICYCLE<br />

13h00-14h00 : GenOway<br />

14h00-15h00 : Ambion 2<br />

15h00-16h30 : Biobase 2<br />

16h30-17h30 : Polyplus Transfection<br />

Friday January 27th, HAVANNE<br />

13h00-15h00 : Ariadne Genomics 1<br />

13h00-15h00 : Ariadne Genomics 2<br />

15h00-16h00 : IBA 1<br />

16h00-17h00 : IBA 2<br />

13h00 – 15h00 : Poster session III<br />

15h00 – 17h00 : Poster session IV<br />

17h00 – 17h30 : Coffee break <strong>and</strong> visit of the expo<br />

- 19 -


Friday January 27th, 2006 (Evening session)<br />

17h30 – 19h00 : Cell <strong>signaling</strong> pathways leading to regulated chromatin modifications<br />

17h30 – 17h50 : Richard Pestell (Lombardi Comprehensive Cancer Center, Georgetown<br />

University, Washington, USA):<br />

Cyclins <strong>and</strong> HDAC/HATs<br />

17h50 – 18h10 : Luciano Di Croce (ICREA <strong>and</strong> Centre de Regulacio Genomica (CRG),<br />

Barcelona, Spain):<br />

Targeting chromatin machines to promoters in leukemias<br />

18h10 – 18h30 : Willem Voncken (Research Institute Growth <strong>and</strong> Development, University<br />

Maastricht, The Netherl<strong>and</strong>s):<br />

Talking to chromatin: Silencers Silenced.<br />

18h30 – 18h50 : Guy Haegeman (Laboratory for Eukaryotic Gene Expression <strong>and</strong> Signal<br />

Transduction, Department of Molecular Biology, Ghent University, Belgium):<br />

Chromatin impact on NFkB-driven gene expression<br />

23h00 – 3h00 : Science Party at Melusina Night Club<br />

- 20 -


Saturday January 28th, 2006 (Early morning session)<br />

7h30 – 8h30 : Cell <strong>signaling</strong> un health <strong>and</strong> disease : short communications II<br />

7h30 – 7h40 : Réjane Paumelle (Département d’Athérosclerose, U545 Inserm, Institut<br />

Pasteur de Lille <strong>and</strong> Faculté de Pharmacie, Université de Lille II, Lille, France) :<br />

Acute anti-inflammatory properties of statins involve Peroxisome Proliferator-Activated<br />

Receptor-alpha via inhibition of the PKC signalling pathway<br />

7h40 – 7h50 : Alex<strong>and</strong>er L. Pukhalsky (Research Centre for Medical Genetics, Moscow ) :<br />

Alkylating drugs applied in non-cytotoxic doses as a novel compounds targeting<br />

inflammatory signal pathways<br />

7h50 – 8h00 : Eric Adriaenssens (Institut Pasteur, Lille, France.) : A new antisense<br />

transcript is involved in the genomic imprinting control at the H19/IGF2 locus.<br />

8h00 – 8h10 : Caroline Rouaux (Laboratoire de Signalisation Moléculaire et<br />

Neurodégénérescence – INSERM U692 - Faculté de Médecine, Strasb<strong>our</strong>g, France) :<br />

Inhibition of histone deacetylase activity as a therapeutic tool in amyotrophic lateral sclerosis.<br />

8h10 - 8h20 : Peter J. van den Elsen (Leiden University Medical Center, Leiden, The<br />

Netherl<strong>and</strong>s ) :<br />

Epigenic control of MHC2TA transcription: varying contributions of DNA <strong>and</strong> histone<br />

modifications in cell-activation <strong>and</strong> interferon-gamma-mediated induction of gene expression<br />

in cancer cells<br />

8h20 – 8h30 : Joydeb Kumar Kundu (National Research Laboratory of Molecular<br />

Carcinogenesis <strong>and</strong> Chemoprevention, College of Pharmacy, Seoul National University,<br />

South Korea) :<br />

Resveratrol inhibits phorbol ester-induced expression of COX-2 through inactivation of NF-<br />

!B <strong>and</strong> AP-1 in mouse skin: role of I!B kinase <strong>and</strong> MAP kinases<br />

Saturday January 28th, 2006 (Morning session)<br />

8h30 - 10h30: Transcriptional control<br />

8h30 – 9h00 : Yinon Ben-Neriah (The Lautenberg Center for Immunology, Hebrew-<br />

University-Hadassah Medical School, Jerusalem, Israel):<br />

Tissue specific control of NF-kappaB activation<br />

9h00 – 9h30 : Sankar Ghosh (Yale University School of Medicine, New Haven, CT , USA):<br />

Regulation of NF-kB-dependent transcription<br />

9h30 – 10h00 : Moshe Oren (Department of Molecular Cell Biology, The Weizmann<br />

Institute, Rehovot, Israel):<br />

Oncogenic <strong>signaling</strong> by mutant p53<br />

10h00 – 10h10 : Hyeyoung Kim (Department of Pharmacology <strong>and</strong> Brain Korea 21 Project<br />

for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Korea) :<br />

Crosstalk between NF-kB <strong>and</strong> AP-1 in Helicobacter pylori-mediated inflammation in human<br />

gastric epithelial cells<br />

- 21 -


10h10 – 10h20 : Gerhard Müller-Newen (Institut für Biochemie, Universitätsklinikum der<br />

RWTH Aachen, Aachen, Germany) :<br />

Real-time analysis of Jak/STAT signal transduction in single cells.<br />

10h20 – 10h30 : Claude P. Muller (Institute of Immunology, Laboratoire National de Santé,<br />

Luxemb<strong>our</strong>g) :<br />

Transcriptional control of differential glucocorticoid receptor expression: determining tissue<br />

specific expression, <strong>and</strong> protein isoforms.<br />

10h30 – 11h00 : Coffee break <strong>and</strong> visit of the expo<br />

11h00 - 13h00: Transcriptional control<br />

11h00 – 11h30 : Iris Behrman (University of Luxemb<strong>our</strong>g):<br />

Jaks <strong>and</strong> cytokine receptors - an intimate relationship.<br />

11h30 – 12h00 : Jacques Piette (Laboratory of Virology & Immunology, Institute of<br />

Pathology B23, University of Liege, Liege, Belgium):<br />

The phosphatase SHIP-1 is an important mediator in the oxidative stress-induced NF-kB<br />

activation.<br />

12h00 – 12h30 : Peter Angel (Deutsches Krebsforschungszentrum, DKFZ, Division of<br />

Signal Transduction <strong>and</strong> Growth Control, Heidelberg, Germany):<br />

Regulation of gene expression in skin remodeling <strong>and</strong> tum<strong>our</strong>igenesis<br />

12h30 – 12h40 : Michèle J. Hoffmann (Urologische Klinik und Institut für Pathologie*,<br />

Heinrich-Heine-Universität Düsseldorf, Germany) :<br />

Epigenetic Regulation of Stem Cell Maintenance Genes in Prostate Carcinoma<br />

12h40 – 12h50 : Jose Aramburu (Departament de Ciències Experimentals i de la Salut,<br />

Universitat Pompeu Fabra, Barcelona, Spain) :<br />

Regulation of the cell cycle by the transcription factor NFAT5 in response to osmotic <strong>and</strong><br />

genotoxic stress.<br />

12h50 – 13h00 : Wim V<strong>and</strong>en Berghe (Laboratory for Eukaryotic Gene Expression <strong>and</strong><br />

Signal Transduction (LEGEST), Department of Molecular Biology, Ghent University,<br />

Belgium ) :<br />

Attenuation of MSK1-driven NF-kB gene expression by soy isoflavones does not require<br />

estrogenic activity<br />

13h00: End of the meeting<br />

12h00 – 14h30 : Lunch is served<br />

- 22 -


Oral presentations<br />

(by alphabetical order)<br />

- 23 -


A new antisense transcript is involved in the genomic imprinting control at the<br />

H19/IGF2 locus<br />

Nathalie Berteaux 1 , Nicole Aptel 2 , Thierry Dugimont 1 , Guy Cathala 2 , Céline Janton 1 ,<br />

Jean Coll 3 , Hubert Hondermarck 1 , Jean-Jacques Curgy 1 , Thierry Forné 2 <strong>and</strong> Eric<br />

Adriaenssens 1 .<br />

1 ERI-8 INSERM, UPRES-EA 1033, IFR 118, USTL, 59655 Villeneuve d’Ascq cedex,<br />

France ; 2 Institut de Génétique Moléculaire, UMR 5535 CNRS, Université Montpellier<br />

II, IFR 122, Montpellier, France ; 3 UMR 8527 CNRS, IBL, Institut Pasteur, Lille,<br />

France.<br />

E-mail : eric.adriaenssens@univ-lille1.fr<br />

Natural antisense transcripts have been implicated in many aspects of eucaroytic gene<br />

expression including genomic imprinting <strong>and</strong> RNA interference. Our group has identified a<br />

new antisense transcript at the human <strong>and</strong> mouse H19/IGF2 locus, that we called 91H. This<br />

locus belongs to an imprinting domain located in human chromosome 11p15.5 (homologue to<br />

the mouse distal chromosome 7). H19 is expressed from the maternal allele <strong>and</strong> Igf-2 is<br />

paternally expressed. Both genes share common regulatory sequences including the H19 5’<br />

differentially methylated domain ICR (Imprinting Control Region) <strong>and</strong> the 3’ downstream<br />

enhancer sequences.<br />

In human, 91H is an 120 kb-long transcript, antisense to H19, spanning the entire H19<br />

transcribed region <strong>and</strong> extending 69Kb <strong>and</strong> 39 kb downstream <strong>and</strong> upstream, respectively.<br />

This includes the ICR <strong>and</strong> the 3’ enhancer regions but not the IGF2 transcription unit. The<br />

promoter region is contained in the first intron of the MrpL23 gene (the H19 3’neighb<strong>our</strong>ing<br />

gene). In addition, we show that in mouse 91H is a biallelic transcript.<br />

We demonstrate that this antisense RNA is a nuclear <strong>and</strong> short-lived transcipt in human <strong>and</strong><br />

mouse normal cells, but we observe its stabilization <strong>and</strong> accumulation in human breast<br />

epithelial cells. Consistently, an in vivo study confirms these data revealing a 91H<br />

overexpression in breast cancer biopsies. Furthermore, the developmental expression of 91H<br />

is very similar to those of the H19 RNA.<br />

Knock-down of 91H by RNA interference leads to loss of silencing of the Igf-2 gene on the<br />

paternal chromosome <strong>and</strong> a down-regulation of Igf-2 expression. We investigate the effect of<br />

91H on DNA methylation in ICR <strong>and</strong> differentially methylated region of this locus but also in<br />

heterochromatin formation leading to gene silencing.<br />

Our results indicate that 91H plays a key role in genomic imprinting at the H19/IGF2 locus<br />

<strong>and</strong> we discuss the role of long intergenic antisense transcript in chromatin remodelling <strong>and</strong><br />

imprinting maintenance.<br />

Publications:<br />

1: Berteaux N, Lottin S, Monte D, Pinte S, Quatannens B, Coll J, Hondermarck H, Curgy JJ, Dugimont T,<br />

Adriaenssens E. H19 mRNA-like noncoding RNA promotes breast cancer cell proliferation through positive<br />

control by E2F1. J Biol Chem. 2005, 280:29625-36.<br />

2: Lottin S, Adriaenssens E, Berteaux N, Lepretre A, Vilain MO, Denhez E, Coll J, Dugimont T, Curgy JJ. The<br />

human H19 gene is frequently overexpressed in myometrium <strong>and</strong> stroma during pathological endometrial<br />

proliferative events. Eur J Cancer. 2005, 41:168-77.<br />

3: Berteaux N, Lottin S, Adriaenssens E, Van Coppenolle F, Leroy X, Coll J, Dugimont T, Curgy JJ. Hormonal<br />

regulation of H19 gene expression in prostate epithelial cells. J Endocrinol. 2004, 183:69-78.<br />

- 24 -


4: : Lottin S, Vercoutter-Edouart AS, Adriaenssens E, Czeszak X, Lemoine J, Roudbaraki M, Coll J,<br />

Hondermarck H, Dugimont T, Curgy JJ. Thioredoxin post-transcriptional regulation by H19 provides a new<br />

function to mRNA-like non-coding RNA. Oncogene. 2002, 21:1625-31.<br />

5: Yazidi-Belk<strong>our</strong>a IE, Adriaenssens E, Vercoutter-Edouart AS, Lemoine J, Nurcombe V, Hondermarck H.<br />

Proteomics of breast cancer: outcomes <strong>and</strong> prospects.Technol Cancer Res Treat. 2002, 1:287-96.<br />

6: Lottin S, Adriaenssens E, Dupressoir T, Berteaux N, Montpellier C, Coll J, Dugimont T, Curgy JJ.<br />

Overexpression of an ectopic H19 gene enhances the tumorigenic properties of breast cancer cells.<br />

Carcinogenesis. 2002, 23:1885-95.<br />

7 Hondermarck H, Dolle L, El Yazidi-Belk<strong>our</strong>a I, Vercoutter-Edouart AS, Adriaenssens E, Lemoine J.<br />

Functional proteomics of breast cancer for signal pathway profiling <strong>and</strong> target discovery. J Mammary Gl<strong>and</strong> Biol<br />

Neoplasia. 2002, 7:395-405.<br />

8: Adriaenssens E, Lottin S, Berteaux N, Hornez L, Fauquette W, Fafeur V, Peyrat JP, Le B<strong>our</strong>his X,<br />

Hondermarck H, Coll J, Dugimont T, Curgy JJ. Cross-talk between mesenchyme <strong>and</strong> epithelium increases H19<br />

gene expression during scattering <strong>and</strong> morphogenesis of epithelial cells. Exp Cell Res. 2002, 275:215-29.<br />

9: Adriaenssens E, Lemoine J, El Yazidi-Belk<strong>our</strong>a I, Hondermarck H. Growth <strong>signaling</strong> in breast cancer cells:<br />

outcomes <strong>and</strong> promises of proteomics. Biochem Pharmacol. 2002, 64:797-803.<br />

10: Adriaenssens E, Lottin S, Dugimont T, Fauquette W, Coll J, Dupouy JP, Boilly B, Curgy JJ. Steroid<br />

hormones modulate H19 gene expression in both mammary gl<strong>and</strong> <strong>and</strong> uterus. Oncogene. 1999, 18:4460-73.<br />

11: Adriaenssens E, Dumont L, Lottin S, Bolle D, Lepretre A, Delobelle A, Bouali F, Dugimont T, Coll J,<br />

Curgy JJ. H19 overexpression in breast adenocarcinoma stromal cells is associated with tumor values <strong>and</strong> steroid<br />

receptor status but independent of p53 <strong>and</strong> Ki-67<br />

expression. Am J Pathol. 1998, 153:1597-607.<br />

12: Dugimont T, Montpellier C, Adriaenssens E, Lottin S, Dumont L, Iotsova V, Lagrou C, Stehelin D, Coll J,<br />

Curgy JJ. The H19 TATA-less promoter is efficiently repressed by wild-type tumor suppressor gene product<br />

p53. Oncogene. 1998, 16:2395-401.<br />

- 25 -


Targeting Inflammatory Transcription Factor by Dietary Agents for<br />

Prevention <strong>and</strong> Treatment of Cancer<br />

Bharat B. Aggarwal, Ph.D.<br />

Cytokine Research Laboratory, Department of Experimental Therapeutics, The<br />

University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, U.S.A.<br />

NF-kB, a transcription factor, is present normally in the cytoplasm as an inactive<br />

heterotrimer consisting of p50, p65 <strong>and</strong> IkBa subunits. When activated, NF-kB translocates<br />

to the as a p50-p65 heterodimer. This factor regulates the expression of various genes that<br />

control apoptosis, viral replication, tumorigenesis, various autoimmune diseases, <strong>and</strong><br />

inflammation. NF-kB has been linked to the development of carcinogenesis for several<br />

reasons. First, various carcinogens <strong>and</strong> tumor promoters have been shown to activate NF-kB.<br />

Second, activation of NF-kB has been shown to block apoptosis <strong>and</strong> promote proliferation.<br />

Third, the tumor microenvironment can induce NF-kB activation. F<strong>our</strong>th, constitutive<br />

expression of NF-kB is frequently found in tumor cells. Fifth, NF-kB activation induces<br />

resistance to chemotherapeutic agents. Fifth, several genes involved in tumor initiation,<br />

promotion, <strong>and</strong> metastasis are regulated by NF-kB. Sixth, various chemopreventive agents<br />

have been found to downregulate the NF-kB activation. All these observation suggest that<br />

NF-kB could mediate tumorigenesis <strong>and</strong> thus can be used as a target for chemoprevention <strong>and</strong><br />

for the treatment of cancer. Besides NF-kB, we have also targeted AP-1 <strong>and</strong> STAT3, other<br />

transcription factors that mediate tumorigenesis. We will present the data which shows that<br />

phytochemicals derived from fruits, vegetables <strong>and</strong> spices are important inhibitors of NF-kB<br />

activation, <strong>and</strong> can suppress the expression of genes involved in carcinogenesis <strong>and</strong><br />

tumorigenesis in vivo.<br />

Recent references:<br />

1 Shishodia, S., <strong>and</strong> Aggarwal B.B. Diosgenin Inhibits Osteoclastogenesis, Invasion,<br />

<strong>and</strong> Proliferation Through the Downregulation of Akt, IkB Kinase Activation <strong>and</strong> NF-kB-<br />

Regulated Gene Expression, Oncogene (in press)<br />

2 Aggarwal BB, Shishodia S., Takada Y., Banerjee S., Newman RA, Bueso-Ramos CE,<br />

<strong>and</strong> Price JE Curcumin Suppresses the Paclitaxel-induced NF-!B Pathway in Breast Cancer<br />

Cells <strong>and</strong> Inhibits Lung Metastasis of Human Breast Cancer in Nude Mice, Clinical Cancer<br />

Research (in press).<br />

3 Aggarwal BB, Ichikawa H. Molecular Targets <strong>and</strong> Anticancer Potential of Indole-3-<br />

Carbinol <strong>and</strong> Its Derivatives. Cell Cycle. 2005 Sep 6;4(9) 1201-1215<br />

4 Takada Y, Kobayashi Y, <strong>and</strong> Aggarwal BB. Evodiamine abolishes constitutive <strong>and</strong><br />

inducible NF-kappa B activation by inhibiting Ikappa Balpha kinase activation, thereby<br />

suppressing NF-kappa B-regulated antiapoptotic <strong>and</strong> metastatic gene expression, upregulating<br />

apoptosis, <strong>and</strong> inhibiting invasion.J. Biol Chem. 2005 Feb 14; 2005;280(17):17203-12<br />

5 Takada Y, Andreeff M, Aggarwal BB. Indole-3-carbinol suppresses NF-{kappa}B<br />

<strong>and</strong> I{kappa}B{alpha} kinase activation causing inhibition of expression of NF-{kappa}Bregulated<br />

antiapoptotic <strong>and</strong> metastatic gene products <strong>and</strong> enhancement of apoptosis in<br />

myeloid <strong>and</strong> leukemia cells. Blood. 2005 Apr 5; 2005 Jul 15;106(2):641-9.<br />

6 Siwak DR, Shishodia S, Aggarwal BB, Kurzrock R. Related Articles, Links Abstract<br />

Curcumin-induced antiproliferative <strong>and</strong> proapoptotic effects in melanoma cells are associated<br />

with suppression of IkappaB kinase <strong>and</strong> nuclear factor kappaB activity <strong>and</strong> are independent of<br />

- 26 -


the B-Raf/mitogen-activated/extracellular signal-regulated protein kinase pathway <strong>and</strong> the<br />

Akt pathway. Cancer. 2005 Aug 15;104(4):879-90.<br />

7 Takada Y., <strong>and</strong> Aggarwal B. B., Zerumbone Abolishes NF-kappaB <strong>and</strong> IkappaB<br />

Kinase Activation Leading to Suppression of Antiapoptotic <strong>and</strong> Metastatic Gene Expression,<br />

Upregulation of Apoptosis, <strong>and</strong> Downregulation of Invasion, ONCOGENE (in press)<br />

8 Haruyo Ichikawa, Yasunari Takada, Akira Murakami <strong>and</strong> Bharat B. Aggarwal<br />

Identification of a Novel Blocker of I!B" Kinase That Enhances Cellular Apoptosis <strong>and</strong><br />

Inhibits Cellular Invasion Through Suppression of NF-kB Regulated Gene Products, J<strong>our</strong>nal<br />

of Immunology (2005 Jun 1;174(11):7383-92.<br />

9 Shishir Shishodia, Hesham M. Amin, Raymond Lai, <strong>and</strong> Bharat B. Aggarwal,<br />

Curcumin (Diferuloylmethane) Inhibits Constitutive NF-!B Activation, Induces G1/S Arrest,<br />

Suppresses Proliferation, <strong>and</strong> Induces Apoptosis in Mantle Cell Lymphoma, Biochemical<br />

Pharmacology (2005 Sep 1;70(5):700-13.<br />

10 Yan, C., Jamaluddin Md S., Aggarwal, B.B., Myers JN, Boyd D.D., Gene Expression<br />

Profiling Identifies ATF3 as a Novel Mediators of the Anti-Tumor Effect of Curcumin,<br />

Molecular Cancer Therapeutics 2005 Feb;4(2):233-41.<br />

11 Aggarwal B. B., Nuclear Factor NF-!B: Enemy Within, Cancer Cell 2004<br />

Sep;6(3):203-8.<br />

12 Takada Y., Bhardwaj A., Potdar P., <strong>and</strong> Aggarwal B. B., Nonsteroidal<br />

Antiinflammatory Agents Differ in Their Ability To Suppress NF-!B Activation, Inhibition<br />

of Expression of Cyclooxygenase-2 <strong>and</strong> Cyclin D1, <strong>and</strong> Abrogation of Tumor Cell<br />

Proliferation, ONCOGENE (2004) 23, 9247-9258)<br />

13 Takada Y, Aggarwal B. B. Evidence that Genetic Deletion of the TNF Receptor p60<br />

or p80 in Macrophages Modulates RANKL-Induced Signaling. BLOOD. 2004 Dec<br />

15;104(13):4113-21<br />

14 Aggarwal B. B., Takada Y.<strong>and</strong> Ommen O. V. From Chemopreventive to<br />

chemotherapy: common targets <strong>and</strong> common goals. Expert Opinion in Investigational<br />

Drugs 13, (10), 2004, 1-12<br />

15 Shishodia S. <strong>and</strong> Aggarwal B. B., Guggulsterone Inhibits NF-!B <strong>and</strong> I!B" Kinase<br />

Activation, Suppresses Expression of Antiapoptotic Gene Products <strong>and</strong> Enhances Apoptosis;<br />

J<strong>our</strong>nal of Biological Chemistry. 2004 Nov 5;279(45):47148-58.<br />

16 Shishodia S., <strong>and</strong> Aggarwal B.B., Nuclear Factor-!B: A Friend or a Foe in Cancer?<br />

Biochemical Pharmacology 2004 Sep 15;68(6):1071-80.<br />

17 Aggarwal, B.B., A. Kumar, <strong>and</strong> A. Bharti. Therapeutic potential of curcumin derived<br />

from turmeric (Curcuma longa), Herbal <strong>and</strong> Traditional Medicine: Molecular Basis of<br />

Health (ed by Lester Packer, Choon Nam Ong <strong>and</strong> Barry Halliwell)<br />

18 Aggarwal B. B., A. Kumer, M.S. Aggarwal, S. Shishodia, Curcumin derived from<br />

turmeric (Curcuma longa): A spice for all seasons, in Phytochemicals in Cancer<br />

Chemoprevention (ed by Debasis Bagchi, Ph.D., <strong>and</strong> Harry G. Preuss, M.D.) CRC Press.<br />

19 Dorai T., <strong>and</strong> Aggarwal B. B., Role of Chemopreventive Agents in Cancer Therapy.<br />

Cancer Letters 2004 Nov 25;215(2):129-40.<br />

20 Li, L., B.B. Aggarwal, S. Shishodia, J. Abbruzzese <strong>and</strong> R. Kurzrock. Nuclear factor-<br />

!B <strong>and</strong> I!B kinase are constitutively active in human pancreatic cells <strong>and</strong> their downregulation<br />

by curcumin (diferuloyl methane) is associated with suppression of proliferation<br />

<strong>and</strong> induction of apoptosis Cancer 2004 Nov 15;101(10):2351-62<br />

21 Aggarwal B.B., A. Bhardwaj, R.S. Aggarwal, N.P. Seeram, S. Shishodia, <strong>and</strong> Y.<br />

Takada, Role of resveratrol in prevention <strong>and</strong> therapy of cancer: preclinical <strong>and</strong> clinical<br />

studies Invited review. Anticancer Research 2004 Sep-Oct;24(5A):2783-840<br />

- 27 -


Mechanisms involved in the anti-apoptotic effect of magnetic fields<br />

Maria C. Albertini, Claudia Cerella *, Sonia Cordisco*, Augusto Accorsi, Antonio<br />

Bergamaschi#, Maria D'Alessio*, Milena De Nicola*, Silvia Nuccitelli*, Lina Ghibelli*<br />

Istituto di Chimica Biologica A. Fornaini, Universita' di Urbino, Urbino, Italy<br />

*Dipartimento di Biologia, #Cattedra di Medicina del Lavoro, Universita' di Roma Tor<br />

Vergata, Rome, Italy 00133; *. E-mail: cemetbio@uniurb.it<br />

Concern for possible health hazard due to increasing exposure to magnetic fields (MFs), came<br />

from epidemiological reports indicating a correlation between exposure to MFs <strong>and</strong><br />

development of some types of tumors. This led to great interest in the interaction of MFs with<br />

living matter, especially focusing on possible interference on cell metabolism that may<br />

account for a pro-tumoral role of MFs. Research on this issue is made quite difficult since<br />

different types of MFs (i.e., static vs oscillating; different oscillation frequencies; different<br />

intensities; different vector directions; etc.) may differently interact with cells, thus giving rise<br />

to controversial results. We showed that 0.3-60 mTesla (mT) static (i.e., magnete-induced)<br />

MFs may well act as tumor promoting agents, tumor promotion being the epigenetic effect<br />

that allows tumor growth by favoring the growth/survival of transformed cells. Indeed, MFs<br />

strongly reduce the extent of damage-induced apoptosis on a set of cell lines, thus increasing<br />

survival of possibly transformed cells (Fanelli et al., FASEB J 1999, 133:95-102). We<br />

describe here the events triggered by exposure to MFs on a reporter cell line (U937), which<br />

lead to impairment of damage-induced apoptosis, compared to an insensitve one (Jurkat). We<br />

show that MFs anti-apoptotic effect requires a correct redox equilibrium, a working<br />

phospholipase C (PLC, a key actor of receptor-mediated signal transduction), <strong>and</strong> nitric oxide<br />

syntase (NOS, a key actor of intracellular inflammatory signal transduction), the antiapoptotic<br />

effect being abolished in the presence of reducing (DTT) or oxidizing (BSO)<br />

agents; of PLC inhibitors (neomycin <strong>and</strong> U73122); <strong>and</strong> NOS inhibitors (L-NAME),<br />

respectively. Accordingly, the sensitive cells show increased Ca2+ influx, increased IP3<br />

production, plasma membrane hyperpolarization <strong>and</strong> alterations of free radicals <strong>and</strong><br />

glutathione levels. Jurkat cells may be rendered sensitive upon activation with<br />

phytohemagglutinin; the normal/activated Jurkat provide an ideal system where to look for<br />

the primary determinants for the sensitivity to MFs anti-apoptotic effect. Intriguingly,<br />

extremely low frequency (50Hz) electroMFs (ELF) exert similar effects, reducing apoptosis<br />

in a Ca2+ dependent fashion.<br />

LIST OF RECENT PAPERS<br />

Piacentini MP, Piatti E, Fraternale D, Ricci D, Albertini MC, Accorsi A.<br />

Phospholipase C-dependent phosphoinositide breakdown induced by ELF-EMF in Peganum<br />

harmala calli.<br />

Biochimie. 2004 Apr-May;86(4-5):343-9.<br />

Ghibelli L, Teodori L, Cerella C, De Nicola M, D'Alessio M, Clavarino G, Cordisco S,<br />

Albertini MC, Accorsi A, Magrini A, Bergamaschi A.<br />

Epigenetic role of magnetic field exposure in tumor progression: fine-tuning experimental<br />

models]<br />

G Ital Med Lav Ergon. 2003 Jul-Sep;25 Suppl(3):277-8.<br />

- 28 -


Albertini MC, Accorsi A, Citterio B, Burattini S, Piacentini MP, Uguccioni F, Piatti E.<br />

Morphological <strong>and</strong> biochemical modifications induced by a static magnetic field on Fusarium<br />

culmorum.<br />

Biochimie. 2003 Oct;85(10):963-70.<br />

Albertini MC, Teodori L, Piatti E, Piacentini MP, Accorsi A, Rocchi MB.<br />

Automated analysis of morphometric parameters for accurate definition of erythrocyte cell<br />

shape.<br />

Cytometry A. 2003 Mar;52(1):12-8.<br />

Piatti E, Albertini MC, Baffone W, Fraternale D, Citterio B, Piacentini MP, Dacha M,<br />

Vetrano F, Accorsi A.<br />

Antibacterial effect of a magnetic field on Serratia marcescens <strong>and</strong> related virulence to<br />

Hordeum vulgare <strong>and</strong> Rubus fruticosus callus cells.<br />

Comp Biochem Physiol B Biochem Mol Biol. 2002 Jun;132(2):359-65.<br />

Colussi C, Albertini MC, Coppola S, Rovidati S, Galli F, Ghibelli L.<br />

H2O2-induced block of glycolysis as an active ADP-ribosylation reaction protecting cells<br />

from apoptosis.<br />

FASEB J. 2000 Nov;14(14):2266-76.<br />

- 29 -


Regulation <strong>and</strong> function of the WNK1 <strong>and</strong> WNK4 protein kinases that are mutated in<br />

Gordon’s hypertension syndrome.<br />

Dario R Alessi<br />

MRC Protein Phosphorylation Unit, University of Dundee, Dundee DD1 5EH, UK<br />

The WNK family of protein kinases comprise 4 members (WNK1, WNK2, WNK3 <strong>and</strong><br />

WNK4) <strong>and</strong> were originally identified as serine-threonine protein kinases that lack a<br />

conserved Lys residue normally found in subdomain II of the catalytic domain. Subsequent<br />

studies, identified mutations in the genes encoding WNK1 <strong>and</strong> WNK4, in families with an<br />

inherited hypertension <strong>and</strong> hyperkalemia (elevated plasma K + ) disorder, called<br />

pseudohypoaldosteronism type II (PHAII, also known as Gordon’s syndrome). WNK<br />

isoforms are large protein kinases (WNK1-2382 residues, WNK4-1243 residues), in which<br />

the catalytic domain is located at the N-terminus (residues 221 to 479 for WNK1 <strong>and</strong> 174 to<br />

432 for WNK4). Apart from two putative coiled-coil domains, the remainder of the WNK<br />

polypeptides possess no obvious structural features. Mutations in the WNK1 gene found in<br />

PHAII subjects, are deletions in intron-1, which reportedly elevate the expression of the<br />

WNK1 protein, indicating that hypertension could result from increased expression of<br />

WNK1. Consistent with this notion, mice lacking one allele of WNK1, had lower blood<br />

pressure. The WNK1 knockout is an embryonic lethal, indicating that WNK1 is also required<br />

for normal development. Thus far, the mutations in the WNK4 gene found in PHAII subjects,<br />

lie distal to both of the putative coiled-coil domains. Little is known about the molecular<br />

mechanism by which WNK isoforms regulate cellular processes. In my talk I will present <strong>our</strong><br />

recent results that indicate that the WNK protein kinases are activated by osmotic stress <strong>and</strong><br />

phosphorylate <strong>and</strong> activate protein kinases of the STE20 family, termed STE20/SPS1-related<br />

Proline-Alanine-rich Kinase (SPAK) <strong>and</strong> the Oxidative Stress Response kinase-1 (OSR1).<br />

SPAK <strong>and</strong> OSR1 were previously reported to be activated in cells in response to osmotic<br />

stress such as high salt <strong>and</strong> phosphorylate <strong>and</strong> regulate the activity of ion transporters such as<br />

NKCC1. It had also been previously shown that treatment of cells with high salt concentration<br />

activated WNK1. I will discuss the possibility that osmotic stress might control the activity of<br />

ion transporters through the WNK-SPAK/OSR1 pathway. It is possible that mutations in<br />

WNK1 <strong>and</strong> WNK4 genes in humans disrupt this pathway, thereby affecting ion transporter<br />

activity in organs such as the kidney, which could account for the development of<br />

hypertension in subjects with these mutations.<br />

- 30 -


AP-1-dependent gene expression in skin remodelling <strong>and</strong> tum<strong>our</strong>igenesis<br />

Peter Angel<br />

Deutsches Krebsforschungszentrum, Division Signal Transduction <strong>and</strong> Growth Control<br />

(A100), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany.<br />

The transcription factor AP-1, which is composed of members of the Fos, Jun <strong>and</strong> ATF<br />

protein families participates in physiological <strong>and</strong> pathophysiological processes due to its<br />

central role as a cellular switch of genetic programs in response to extracellular signals. The<br />

distinct expression pattern of AP-1 subunits in the skin, the loss of chemically induced<br />

carcinogenesis in AP-1 compromised mice, <strong>and</strong> the large number of putative AP-1 target<br />

genes in epidermal cells, indicate that AP-1 members play a pivotal role in epidermal<br />

organisation, skin homeostasis <strong>and</strong> tum<strong>our</strong>igenesis (1, 2). We have employed a heterologous<br />

organotypic coculture system containing fibroblasts from c-Jun or JunB-deficient mice to<br />

identify trans-regulatory activities of c-Jun <strong>and</strong> JunB in the paracrine interaction between<br />

keratinocytes <strong>and</strong> dermal fibroblasts (3). Large-scale expression profiling of these cultures<br />

revealed novel AP-1 target genes in fibroblasts including cytokines, growth factors <strong>and</strong><br />

chemoattractants (4). Mice lacking the AP-1 member JunB in skin exhibit defects in wound<br />

healing, which is associated with aberrant expression of these newly identified target genes<br />

(5). The mouse skin was also the model of choice to study the regulation <strong>and</strong> function of AP-1<br />

subunits in tum<strong>our</strong>igenesis in vivo (1). Upon combination of the well-established chemically<br />

induced mouse model of epithelial skin tum<strong>our</strong>s <strong>and</strong> expression profiling we have identified a<br />

novel signalling pathway activating AP-1, which is initiated by S100A8 <strong>and</strong> S100 A9, two<br />

members of the S100 family of Ca2+ binding proteins (6), which are also upregulated in a c-<br />

Jun/JunB-dependent mouse model of human psoriasis (7). Both genes are also overexpressed<br />

in human epithelial tum<strong>our</strong>s of the skin <strong>and</strong> other organs, suggesting an important role of this<br />

pathway in tum<strong>our</strong> progression <strong>and</strong> metastasis.<br />

1. Angel, P., Szabowski, A., <strong>and</strong> Schorpp-Kistner M. (2001). Function <strong>and</strong> regulation of AP-1 subunits in skin<br />

physiology <strong>and</strong> pathology. Oncogene 20, 2413-23<br />

2. Hess, J., Angel, P. <strong>and</strong> Schorpp-Kistner, M. (2004) Functions of AP-1 subunits: Quarrel <strong>and</strong> Harmony among<br />

Siblings. J. Cell Sci 117:5965-73<br />

3. Szabowski, A., Maas-Szabowski, N., Andrecht, S., Kolbus, A., Schorpp-Kistner, M., Fusenig, N.E. <strong>and</strong><br />

Angel, P. (2000) c-Jun <strong>and</strong> JunB antagonistically control cytokine regulated mesenchymal-epidermal interaction<br />

in skin.” Cell 103, 745-55<br />

4. Florin, L., Hummerich, L., Dittrich, B., Kokocinski, F., Wrobel, G., Gack, S., Schorpp-Kistner, M., Werner,<br />

S., Hahn, M., Lichter, P., Szabowski, A., Angel, P. (2004) Identification of novel AP-1 target genes in<br />

fibroblasts regulated during cutaneous wound healing. Oncogene 23:7005-17<br />

5. Florin, L., Knebel, J., Zigrino, P., Vonderstrass, B., Mauch, C., Schorpp-Kistner, M., Szabowski, A. <strong>and</strong><br />

Angel P. (2005) Delayed wound healing <strong>and</strong> epidermal hyperproliferation in mice lacking JunB in the skin. J Inv<br />

Dermatol, in press<br />

6. Gebhardt, C., Breitenbach, U., Tuckermann, J. P., Dittrich, B. T., Richter, K. H., <strong>and</strong> Angel, P. (2002).<br />

Calgranulins S100A8 <strong>and</strong> S100A9 are negatively regulated by glucocorticoids in a c-Fos-dependent manner <strong>and</strong><br />

overexpressed throughout skin carcinogenesis. Oncogene 21, 4266-4276.<br />

7. Zenz R., Eferl R., Kenner L., Florin L., Hummerich, Mehic D., Scheuch H., Angel P., Tschachler E. <strong>and</strong><br />

Wagner, E.F. (2005) Psoriasis-like skin disease <strong>and</strong> arthritis caused by inducible epidermal deletion of Jun<br />

proteins. Nature (2005) 437, 369-75<br />

- 31 -


Regulation of the cell cycle by the transcription factor NFAT5 in response to osmotic<br />

<strong>and</strong> genotoxic stress.<br />

Jose Aramburu, Katherine Drews, Beatriz Morancho <strong>and</strong> Cristina López-Rodriguez.<br />

Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, 08003<br />

Barcelona, Spain. E-mail: jaramburu@imim.es; katherine.drews@upf.edu;<br />

beatriz.morancho@upf.edu; cristina.lopez-rodriguez@upf.edu.<br />

The transcription factor NFAT5 belongs to the Rel family, that comprises NF-kappaB <strong>and</strong> the<br />

calcineurin-activated NFATc proteins (1). NFAT5 is activated by hypertonicity <strong>and</strong> regulates<br />

the expression of osmoprotective genes, but also responds to mitogenic signals <strong>and</strong> is<br />

expressed in proliferating cells (2, 3). As osmotic stress can cause DNA breaks, <strong>and</strong> activation<br />

of NFAT5 by hypertonicity is regulated by PI3-kinase related kinases (PIKK) (4), which<br />

control cellular responses to DNA damage, we hypothesized that NFAT5 might play a role in<br />

the cellular response to genotoxic stress. Cell cycle arrest is a hallmark of genotoxic stress <strong>and</strong><br />

is often exacerbated by the disruption of proteins involved in DNA damage sensing <strong>and</strong><br />

repair. We have studied whether the inhibition of NFAT5 had an effect in the cell cycle of<br />

cells exposed to osmotic <strong>and</strong> genotoxic stress. Our results showed that proliferating NFAT5-/-<br />

T cells did not display substantial cell cycle defects under non-stress conditions, but arrested<br />

at G1 <strong>and</strong> G2/M when subjected to hypertonicity <strong>and</strong> ionizing radiation doses that did not<br />

arrest wild-type T lymphocytes. Likewise, suppression of NFAT5 with shRNA in HEK293<br />

cells caused them to accumulate in G2/M when exposed to hypertonic stress or ionizing<br />

radiation. We found that ionizing radiation <strong>and</strong> the genotoxic drugs etoposide <strong>and</strong><br />

hidroxyurea caused NFAT5 to accumulate in the nucleus in HEK293 cells <strong>and</strong> that its<br />

overexpression rendered cells more resistant to cell cycle arrest when exposed to<br />

hypertonicity or ionizing radiation. Our results indicate that NFAT5 can regulate the cell<br />

cycle in response to genotoxic stress, <strong>and</strong> we are currently exploring the mechanisms<br />

underlying this function.<br />

References.<br />

1) López-Rodríguez C, Aramburu J, Rakeman AS, Copel<strong>and</strong> NG, Gilbert DJ, Thomas S,<br />

Disteche C, Jenkins NA, <strong>and</strong> Rao A. 1999. NF-AT5: the NF-AT family of transcription<br />

factors exp<strong>and</strong>s in a new direction. Cold Spring Harb Symp Quant Biol. 1999; 64:517-526.<br />

2) López-Rodríguez C, Aramburu J, Jin L, Rakeman AS, Michino M, Rao A. 2001. Bridging<br />

the NFAT <strong>and</strong> NF-kappaB families: NFAT5 dimerization regulates cytokine gene<br />

transcription in response to osmotic stress. Immunity. 15:47-58.<br />

3) López-Rodríguez C, Antos C. L, Shelton J, Richardson J. A, Fangming L, Novobrantseva<br />

T. I, Bronson R.T, Igarashi P, Rao A, <strong>and</strong> Olson E. N. 2004. Loss of NFAT5 results in renal<br />

atrophy <strong>and</strong> lack of tonicity-responsive gene expression. Proc. Natl Acad of Sci USA. 101;<br />

2392-2397.<br />

4) Irarrazabal C.E, Liu J.C, Burg M.B, <strong>and</strong> Ferraris J.D. 2004. ATM, a DNA damageinducible<br />

kinase, contributes to activation by high NaCl of the transcription factor<br />

TonEBP/OREBP. Proc. Natl. Acad. Sci. U. S. A 101, 8809-8814.<br />

- 32 -


Jaks <strong>and</strong> cytokine receptors – an intimate relationship<br />

Claude Haan 1 , Simone Radtke 2 , Bernd Giese 2 , Christiane Margue-Wurth 1 , Andreas<br />

Herrmann 2 , Gerhard Müller-Newen 2 , Serge Haan 2 , Peter C. Heinrich 2 , Iris Behrmann 1<br />

1 Laboratoire de Biologie et Physiologie Intégrée, University of Luxemb<strong>our</strong>g, 162a, av. de<br />

la Faïencerie, 1511 Luxemb<strong>our</strong>g, Gr<strong>and</strong>-Duchy of Luxemburg,<br />

2 Institute of Biochemistry, RWTH-Aachen Medical School, 52074 Aachen, Germany<br />

E-mail: iris.behrmann@uni.lu<br />

Many cytokines signal via Janus kinases (Jaks). These tyrosine kinases are associated with<br />

cytokine receptor subunits, they become activated upon receptor triggering <strong>and</strong> subsequently<br />

activate downstream signalling events, e. g. the phosphorylation of STAT transcription<br />

factors.<br />

Using gp130 (the common receptor subunit for IL-6-type cytokines) <strong>and</strong> Jak1 as an example,<br />

we analysed the structural requirements for association between Jaks <strong>and</strong> cytokine receptors,<br />

as well as the dynamics of this interaction. Subdomains F1 <strong>and</strong> F2 of the N-terminal FERM<br />

domain of Jak1 are crucially involved in binding to gp130 as well as to other cytokine<br />

receptors. On the receptor side, the conserved box1 <strong>and</strong> box2 regions as well as the<br />

intervening sequences are crucial for Jak association. The receptor plays an active role in<br />

activation of the bound Jaks, as shown by the mutant gp130-W652A that does not elicit<br />

signalling in spite of associating Jaks (1,2).<br />

Jaks are found exclusively in membrane fractions. The plasma membrane localization of Jaks<br />

is dependent on their ability to associate with cytokine receptors since the non-receptor<br />

binding Jak1 mutant L80A/Y81A is only found within the cytoplasm. Using FRAP analysis<br />

with fluorescent gp130 <strong>and</strong> Jak1 proteins we show that Jak1 has the same diffusion behavi<strong>our</strong><br />

as the transmembrane protein gp130, <strong>and</strong> that there is no rapid exchange of Jaks between<br />

different receptors. Thus, this cytokine receptor/Jak complex can be regarded to be equivalent<br />

to a receptor tyrosine kinase (3,4).<br />

To assess the role of the predicted SH2 domain in Jak1 we exchanged the conserved arginine<br />

residue by lysine, a mutation known to abrogate the phosphotyrosine binding. This R466K<br />

mutation did not affect the signal transducing capacities of the kinase, nor its localization,<br />

indicating that the Jak1-SH2 domain does not fulfil the “classical” SH2 domain function.<br />

However, it may play a structural role for association with the oncostatin M receptor<br />

(OSMR)(5).<br />

Jaks are not only crucial for signal transduction of cytokines, but they are also involved in the<br />

regulation of the surface expression of at least some cytokine receptors, including the OSMR.<br />

We identified three dileucine motifs within the interbox1/2 region that prevent efficient<br />

OSMR surface expression in the absence of associated Jaks, possibly by lysosomal targeting<br />

<strong>and</strong> destabilization of the receptor. This may provide a quality control ensuring that<br />

signalling-competent receptors (i. e. those with an associated Jak) would be enriched at the<br />

cell surface (6).<br />

Refs.:<br />

(1) Haan et al., 2001, JBC 276: 37451<br />

(2) Haan et al., 2002, Biochem. J. 361: 105<br />

(3) Behrmann et al., 2004, JBC 279: 35486<br />

(4) Giese et al., 2003, JBC 278: 39205<br />

(5) Radtke et al., 2005, JBC 280: 25760<br />

(6) Radtke et al., 2005, JBC online.<br />

- 33 -


Tissue specific control of NF-kB activation<br />

Yinon Ben-Neriah1, Jongdae Lee2, Elad Horwitz1, Gady Cojocaru1, Naama Kanarik1,<br />

Matti Davis1, Irit Alkalay1, Kyoko Katakura2, Lars Eckmann2, Eli Pikarsky3 <strong>and</strong> Eyal<br />

Raz2<br />

1The Lautenberg Center for Immunology <strong>and</strong> 3Dept of Pathology, Hebrew-University-<br />

Hadassah Medical School Jerusalem 91120, Israel; 2Department of Medicine University<br />

of California San Diego, La Jolla, CA 92093-0663<br />

Canonical NF-kB activation targets IkB <strong>and</strong> p105/NF-kB1 to degradation via the ubiquitin<br />

proteasome system. We <strong>and</strong> others have previously shown that #-TrCP is a key regulator in<br />

this system; its two isoforms #-TrCP1 <strong>and</strong> 2 are both necessary <strong>and</strong> sufficient to control IkB<br />

stability. Ongoing experiments in <strong>our</strong> lab attempt to unveil the unique functions of each #-<br />

TrCP isoform in different tissues <strong>and</strong> determine the consequence of specific isoform<br />

inhibition. So far, #-TrCP-controlled degradation has been mainly studied in st<strong>and</strong>ard tissue<br />

culture conditions, yet many tissues <strong>and</strong> particularly epithelial cells are configured in vivo in a<br />

specific orientation. We will describe a cellular mechanism whereby the polarity of gut<br />

epithelium dictates the outcome of NF-kB <strong>signaling</strong>, thereby playing a major role in colonic<br />

homeostasis. TLR9 activation via apical <strong>and</strong> basolateral surface domains have distinct<br />

transcriptional responses. Whereas basolateral TLR9 signals NF-kB activation, apical TLR9<br />

invokes intracellular tolerance against subsequent basolateral TLR9 <strong>and</strong> other TLRs'<br />

challenges. TLR9-deficient mice lacking the tolerizing receptor are triggered much more<br />

rapidly for NF-kB activation <strong>and</strong> are highly susceptible to experimental colitis. Our data<br />

provide a case for an organ-specific innate immunity where TLR regulation evolved to<br />

maintain colonic homeostasis. This is achieved by a novel cellular mechanism, where the<br />

different surface domains of a polarized cell dictate an opposing <strong>signaling</strong> outcome to a<br />

similar stimulus.<br />

- 34 -


The Role of Met-Gab1 Signalling in Vivo<br />

Walter BIRCHMEIER, Jolanta CHMIELOWIEC, <strong>and</strong> Ute SCHAEPER<br />

Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13125 Berlin,<br />

Germany<br />

We examined wound healing in conditional mutant mice, in which the Met tyrosine kinase,<br />

the receptor of HGF/SF, was ablated in the epidermis. In the Met-deficient epidermis,<br />

keratinocytes were completely unable to contribute to re-epithelialization of the wounds.<br />

However, wound closure could occur by fast proliferation <strong>and</strong> migration of the few (5%)<br />

remaining wild-type keratinocytes. These results show that the HGF/SF <strong>and</strong> Met signalling<br />

system, which is not important for normal skin formation <strong>and</strong> maintenance, is crucial for skin<br />

regeneration.<br />

Gab1 is a signalling protein that belongs to the class of docking proteins, which function<br />

downstream of tyrosine kinases. Gab1 was discovered as a direct target of the tyrosine kinase<br />

c-Met, the receptor for SF/HGF. It was not known, which downstream signalling pathways<br />

are important for Gab1 function in vivo, <strong>and</strong> what are the in vivo contributions of the c-Met<br />

<strong>and</strong> Grb2 binding sites for coupling Gab1 to specific tyrosine kinases. To address these<br />

questions, we generated Gab1 knock-in mice that express Gab1 cDNAs, which carry<br />

mutations in the c-Met, Grb2, PI3K or Shp-2 binding sites. Mutant mice display distinct, but<br />

also overlapping phenotypes, which demonstrate that Gab1 acts in several tyrosine kinase<br />

signalling pathways. Of particular importance in vivo is the binding site of Gab1 to the Shp-2<br />

downstream substrate, which is essential for the migration of muscle precursor cells into the<br />

limbs.<br />

- 35 -


Targeting of Polycomb repressive complexes by the moonlighting protein NIPP1<br />

Mathieu Bollen<br />

Division of Biochemistry, Faculty of Medicine, Catholic University of Leuven, B-3000<br />

Leuven, Belgium. E-mail: Mathieu.Bollen@med.kuleuven.be<br />

NIPP1 is a nuclear protein that is ubiquitously expressed in metazoans <strong>and</strong> plants. The<br />

knockout of NIPP1 in mice is embryonic lethal before gastrulation <strong>and</strong> NIPP1-/- cell lines are<br />

not viable. NIPP1 interacts in vivo with a protein kinase (MELK), a protein phosphatase<br />

(PP1), two pre-mRNA splicing factors (CDC5L, SAP155) <strong>and</strong> two transcriptional repressors<br />

(EED, EZH2). We previously reported that NIPP1 is associated with storage sites for splicing<br />

factors <strong>and</strong> is required for a late step of spliceosome assembly. In addition, NIPP1 functions<br />

as a transcriptional repressor by a mechanism that does not require functional interaction sites<br />

for splicing factors, MELK nor PP1, but depends on the presence of EED <strong>and</strong> EZH2, two core<br />

components of the Polycomb Repressive Complexes 2-4 (PRC2-4). The latter complexes<br />

initiate the heritable repression of genes that are important for development <strong>and</strong> cell<br />

proliferation, at least in part by the EZH2-mediated trimethylation of histone H3 on Lys27<br />

(H3K27). Consistent with a role for NIPP1 in Polycomb signalling, we found that NIPP1 is<br />

associated with H3K27-trimethylated chromatin <strong>and</strong> that NIPP1-/- mouse blastocyst<br />

outgrowths <strong>and</strong> cultured cells with a decreased concentration of NIPP1 are severely deficient<br />

in H3K27 trimethylation, but are normally trimethylated on H3K9. We also show that NIPP1<br />

is a nucleic-acid binding protein <strong>and</strong> interacts strongly with intron-1 of the MSMB gene,<br />

which encodes the prostatic tumor growth suppressor PSP94. We identified the MSMB gene<br />

as a novel Polycomb target gene <strong>and</strong> show that the siRNA-mediated knockdown of NIPP1 is<br />

associated with a decreased recruitment of EZH2 to this gene. These findings are consistent<br />

with the notion that NIPP1 is involved in the recruitment of PRC2 to its target genes. Our data<br />

suggest that NIPP1 functions as a moonlighting protein <strong>and</strong> has independent functions in premRNA<br />

splicing <strong>and</strong> transcription.<br />

- 36 -


The mammalian tumor suppressor Scribble has a key role in cell migration<br />

Sébastien Nola, Marie-Josée Santoni, Claire N<strong>our</strong>ry, Christelle Navarro <strong>and</strong> Jean-Paul<br />

Borg.<br />

Molecular Pharmacology, Marseille Cancer Institute, UMR599 Inserm-Institut Paoli-<br />

Calmettes, Marseille, France 13009. E-mail : borg@marseille.inserm.fr<br />

Genetic studies in Drosophila <strong>and</strong> mice have shown that the PDZ protein Scribble has a<br />

crucial role in many aspects of cell polarity. Loss of function of Scribble in Drosophila<br />

provokes profound defects in epithelial apico-basal polarity, as well as the formation of<br />

neoplastic tumors. Mice deficient for Scribble show abnormal planar cell polarity, <strong>and</strong> die<br />

within a few days after birth from craniorachischisis, a dramatic neural tube defect. We have<br />

shown that mammalian Scribble is a cytoplasmic protein tighly associated to the plasma<br />

membrane, <strong>and</strong> that it is part of a protein complex containing betaPIX, an exchange factor for<br />

Rac <strong>and</strong> Cdc42, <strong>and</strong> GIT1, a GTPase Activating Protein (GAP) for Arf6. The PDZ domains<br />

of Scribble associate directly to the carboxy-terminus of betaPIX, <strong>and</strong> beta-PIX is bound to<br />

GIT1. Here, we demonstrate that PAK, a serine-threonine kinase, is also associated to<br />

Scribble in epithelial cells. The Scribble-betaPIX-GIT1-PAK protein complex is located at the<br />

leading edge of migratory T47D epithelial cells, <strong>and</strong>, using siRNA <strong>and</strong> dominant-negative<br />

constructs, we demonstrate that localization of betaPIX is dependant on the correct<br />

positioning of Scribble at the plasma membrane. Using a modified Boyden chamber assay, we<br />

show that Scribble, as well as the members of the associated protein complex, are required for<br />

the effective migration of T47D cells towards a chemoattractant stimulus. In conclusion,<br />

Scribble <strong>and</strong> the associated betaPIX-GIT1-PAK protein complex is required for cell migration<br />

of epithelial cells. The unproper positioning of the betaPIX-GIT1-PAK signalling machinery<br />

in Scribble deficient cells probably accounts for the cell migration defect in T47D cells <strong>and</strong><br />

may explain the neural tube defect observed in Scribble deficient mice.<br />

N<strong>our</strong>ry C, Grant SG, Borg JP (2003). PDZ domain proteins: plug <strong>and</strong> play! Science’s STKE<br />

179: RE7.<br />

Legouis R, Jaulin-Bastard F, Schott S, Navarro C, Borg J-P, <strong>and</strong> Labouesse M (2003).<br />

Basolateral targeting by Leucine Rich Repeat domains in epithelial cells. EMBO Reports 4:<br />

1096–1100.<br />

Audebert S., Navarro C., N<strong>our</strong>ry C., Chasserot-Golaz S., Lécine P., Bellaiche Y., Dupont J.-<br />

L., Premont R.T., Sempéré C., Strub J.-M., Van Dorsselaer A., Vitale N. <strong>and</strong> Borg J.-P.<br />

Mammalian Scribble forms a tight complex with the bPIX exchange factor (2004). Current<br />

Biology 14: 987-95.<br />

Navarro C., Nola S., Audebert S., Santoni M.-J., Arsanto J.-P., Ginestier C., Marchetto S.,<br />

Jacquemier J., Isnardon D., Le Bivic A., Birnbaum D., <strong>and</strong> Borg J.-P. Junctional recruitment<br />

of mammalian Scribble relies on E-cadherin engagement. (2005) Oncogene, 24: 4330-4339.<br />

O.Lahuna*, Quellari M., Achard C., Nola S., Méduri G., Navarro C., Vitale N., Borg J.-P.*<br />

<strong>and</strong> Misrahi M*. Thyrotropin receptor trafficking relies on the hScrib-bPIX-GIT1-ARF6<br />

<strong>signaling</strong> pathway. (2005) The EMBO J<strong>our</strong>nal, 24: 1364-1374. (*) co-corresponding authors.<br />

Margolis B. <strong>and</strong> Borg J.-P. Apico-basal polarity complexes. J Cell Science, 118: 5157-5159.<br />

- 37 -


Epac <strong>and</strong> Rap1 in the control of cell adhesion<br />

Johannes L. Bos<br />

Department of Physiological Chemistry <strong>and</strong> Centre for Biomedical Genetics, University<br />

Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherl<strong>and</strong>s<br />

Rap1 is a Ras-like small GTPase originally identified as a revertant of Ras-induced cell<br />

transformation, but current studies in both lower eakaryotes <strong>and</strong> mammalian cells have<br />

revealed that the main function of Rap in the control of cell adhesion. Rap1 is activated by a<br />

variety of different stimuli, some of which are mediated by cAMP. We have identified<br />

previously two guanine nucleotide exchange factors (GEFs) for Rap1, Epac1 <strong>and</strong> Epac2.<br />

These two GEFs have a cAMP-binding domain <strong>and</strong> respond to cAMP. Structural analyses<br />

revealed that the cAMP binding domain functions as an auto-inhibitory domain, preventing<br />

binding of Rap by steric hindrance. By structure-based drug design we developed a cAMP<br />

analog 8-pCPT-2'OMe-cAMP (007) that efficiently activates Epac but not protein kinase A<br />

(PKA). Indeed in various cell lines 007 was able to induce Rap1 activation, but not PKA. 007<br />

did not induce the activation of ERK, challenging the model that cAMP-induced activation of<br />

ERK is mediated by Rap1. Using this analog we could show that both Epac <strong>and</strong> Rap1<br />

regulates integrin-mediated cell adhesion <strong>and</strong> E-cadherin <strong>and</strong> VE-cadherin-mediated cell<br />

junction formation. Our results suggest that Rap1 may have a more general function in cell<br />

adhesion, perhaps in the recruitment of cell adhesion molecules.<br />

- 38 -


SIGNAL TRANSDUCTION BY STRESS-ACTIVATED MAP KINASES<br />

ROGER J. DAVIS<br />

Howard Hughes Medical Institute & University of Massachusetts Medical School,<br />

Worcester, MA 01605 USA, E-mail : Roger.Davis@Umassmed.Edu<br />

The JNK group of stress-activated MAP kinases consists of ten protein kinases that<br />

phosphorylate the NH2-terminal activation domain of c-Jun on Ser-63 <strong>and</strong> Ser-73 causing<br />

increased transcriptional activity. JNK protein kinase activity is increased in response to<br />

treatment of cells with pro-inflammatory cytokines or exposure to environmental stress.<br />

Activated JNK is phosphorylated on Thr <strong>and</strong> Tyr within the tripeptide motif Thr-Pro-Tyr<br />

located in kinase sub-domain VIII. Mutational analysis demonstrates that JNK activation<br />

requires the phosphorylation of both Thr <strong>and</strong> Tyr within this motif. This phosphorylation is<br />

mediated by dual specificity protein kinases, including MKK4 <strong>and</strong> MKK7.<br />

The function of the JNK <strong>signaling</strong> pathway has been studied using a combination of<br />

biochemical <strong>and</strong> genetic approaches. Genetic analysis of JNK <strong>signaling</strong> in Drosophila<br />

demonstrates that JNK is required for early embryonic morphogenesis. Similarly, disruption<br />

of the JNK <strong>signaling</strong> pathway in mice using homologous recombination demonstrates that<br />

JNK is required for embryonic viability. In contrast, mice with genetically engineered<br />

selective defects in JNK <strong>signaling</strong> are viable, but exhibit changes in stress-induced gene<br />

expression <strong>and</strong> apoptosis. These studies provide insight into the role of the JNK stressactivated<br />

MAP kinase pathway in the cellular response to environmental stress, including<br />

apoptosis <strong>and</strong> cell survival.<br />

- 39 -


Targeting chromatin machines to promoters in leukemias<br />

R. Villa, L. Morey, M. Buschbeck, A. Gutierrez, <strong>and</strong> L. Di Croce<br />

ICREA <strong>and</strong> Center for Genomic Regulation (CRG), Barcelona, Spain<br />

Many human cancers are characterized by alterations in the balance of DNA methylation. In<br />

cancer cells, a large part of the genome undergoes dramatic hypomethylation, which is often<br />

linked to genome instability, concomitantly with regional gain of methylated sequences at<br />

sites usually unmethylated (CpG isl<strong>and</strong>s). The major outcome of promoter hypermethylation<br />

appears to be long-term silencing of the associated gene. Silencing of tumor suppressors due<br />

to such a mechanism can provide a growth advantage to cancer cells.<br />

The ability of the PML-RARa fusion protein to block hematopoietic differentiation <strong>and</strong> to<br />

induce acute promyelocytic leukemia is based on aberrant gene repression. Mechanistically,<br />

PML-RARa inactivates its target genes by recruiting histone deacetylase (HDAC) <strong>and</strong> DNA<br />

methyltransferase activities to the promoters.<br />

We will show that MBD1, a member of a conserved family of proteins able to bind<br />

methylated DNA, cooperates with PML-RARa in transcriptional repression <strong>and</strong> cellular<br />

transformation. PML-RARa recruits MBD1 to its target promoter through an HDAC3mediated<br />

mechanism. Binding of HDAC3 <strong>and</strong> MBD1 is not confined to the promoter region<br />

but instead is spread over the locus. Retroviral expression of dominant negative mutants of<br />

MBD1 in hematopoietic precursors compromised the ability of PML-RARa to block their<br />

differentiation <strong>and</strong> thus restored cell differentiation. Our results demonstrate that PML-RARa<br />

functions by recruiting an HDAC3-MBD1 complex that contributes to the establishment <strong>and</strong><br />

maintenance of the silenced chromatin state. Our results identify MBD1 as a critical mediator<br />

of PML-RARa-induced gene silencing subsequent to promoter hypermethylation.<br />

Further characterization of the PML-RARa!co-repressor complex, which establishes <strong>and</strong><br />

allows spreading of the silenced state via Polycomb complex, will be additionally discussed,<br />

together with the implication of crosstalk among the different epigenetic layers to the<br />

molecular pathology of leukemia.<br />

- 40 -


Title to be announced<br />

Caroline Dive<br />

Cellular <strong>and</strong> Molecular Pharmacology Group, Paterson Institute for Cancer Research,<br />

Christie Hospital NHS Trust, Manchester, United Kingdom<br />

(no abstract provided)<br />

- 41 -


MOLECULAR BIOLOGY OF THYROID CANCER<br />

Det<strong>our</strong>s, V., Delys, L., Hebrant, A., Weiss, D., Maenhaut, C., Dumont, J.E.<br />

IRIBHM, University of Brussels, School of Medicine, Campus Erasme, B - 1070<br />

Brussels.<br />

Thyroid tumors are interesting for several reasons :<br />

1) there are several well defined tumors from benign to very malignant, some former<br />

evolving in the latter<br />

2) the primary cause of the majority of these tumors is known<br />

3) the signal transduction pathways involved have been well studied<br />

4) there are good in vivo <strong>and</strong> in vitro models.<br />

The types, pathogeneses of the various tumors are described.<br />

Gene expression has been studied in human tumors, in mice transgenic models <strong>and</strong> in<br />

vitro models (primary cultures of human <strong>and</strong> dog thyroid cells, human cancer cell lines). The<br />

comparison of the results on the various models allows to draw several general conclusions on<br />

the evolution in vitro of all lines, the role of negative feedback in tumorigenesis <strong>and</strong> the<br />

importance of taking into account cell populations in the interpretation of results of<br />

microarray studies on solid tumors. A pathogenic scheme of the evolution of papillary<br />

carcinomas into anaplastic carcinomas is presented.<br />

- 42 -


Mechanisms of DNA methylation in mammals<br />

FRANÇOIS FUKS<br />

Free University of Brussels, Faculty of Medicine, 808 route de Lennik, 1070 Brussels,<br />

Belgium.<br />

In mammals, DNA methylation plays an important role in development <strong>and</strong> is associated with<br />

transcriptional silencing. The goal of <strong>our</strong> current work is to elucidate the mechanisms by<br />

which the DNA methylation machinery - the DNA methyltransferases (DNMTs) <strong>and</strong> the<br />

MBDs- functions. In particular, we wish to address two questions: 1. How do the DNMTs <strong>and</strong><br />

the MBDs lead to gene silencing? 2. How is the DNMT enzymatic activity regulated?<br />

1. One mechanism by which the DNA methylation machinery brings about transcriptional<br />

repression is through recruitment of HDAC <strong>and</strong> histone H3 Lys9 methyltransferase activities<br />

(1, 2). Our recent work indicates that DNMTs are mechanistically linked to the Polycomb<br />

Group (PcG) proteins (3). We find that the PcG protein EZH2 interacts with DNA<br />

methyltransferases <strong>and</strong> associates with DNA methyltransferase activity in vivo. ChIPs<br />

indicate that binding of DNMTs to several EZH2-repressed genes depends on the presence of<br />

EZH2. Furthermore, we show that EZH2 is required for DNA methylation of EZH2-target<br />

promoters. Our results suggest that EZH2 serves as a recruitment platform for DNA<br />

methyltransferases. They highlight a previously unrecognized direct connection between two<br />

fundamental epigenetic repression systems.<br />

2. A key question still poorly understood is how are the DNMTs, <strong>and</strong> in particular their<br />

enzymatic activity, regulated. Data will be presented that suggest a new mechanism for the<br />

regulation of DNA methylation by post-translational modification.<br />

References<br />

1. Fuks F. Curr Opin Genet Dev. 2005 Oct;15(5):490-5.<br />

2. Brenner C, Deplus R, Didelot C, Loriot A, Vire E, De Smet C, Gutierrez A, Danovi D,<br />

Bernard D, Boon T, Pelicci PG, Amati B, Kouzarides T, de Launoit Y, Di Croce L, Fuks F.<br />

(2005). EMBO J. 24:336-46.<br />

3. Vire E, Brenner C, Deplus R, Blanchon L, Fraga M, Didelot C, Morey L, Van Eynde A,<br />

Bernard D, V<strong>and</strong>erwinden JM, Bollen M, Esteller M, Di Croce L, de Launoit Y, Fuks F.<br />

Nature. 2005 Dec 14 [Epub ahead of print].<br />

- 43 -


Oxidative Bax activation as the trigger of the stress-induced intrinsic apoptotic pathway<br />

Lina Ghibelli<br />

Dipartimento di Biologia, Universita' di Roma Tor Vergata<br />

Bax translocation is the most upstream event of the intrinsic apoptotic pathway so far<br />

described. Despite the importance of the issue, the mechanisms that push Bax to mitochondria<br />

are still unknown. The intrinsic pathway of apoptosis is mostly triggered by cell damage,<br />

involving gross environmental alterations such as uncontrolled Ca2+ or ROS overload. This<br />

suggests that its activation may be due not to sophisticated interactions (such as those<br />

involved in the receptor-mediated extrinsic apoptotic pathway), but rather to sudden<br />

environmental changes, thus rendering quite difficult to catch the mechanisms involved. The<br />

idea was to check whether Bax may be a protein "sensing" intracellular chemical/physical<br />

alterations, rather that being activated by specific molecular interactions. Bax translocation to<br />

mitochondria is induced by conformational changes that may be caused by Bax<br />

homodimerisation. We show by computer simulation that the 2 cysteine residues of Bax may<br />

form disulfide bridges, producing conformational changes that fav<strong>our</strong> Bax translocation.<br />

Oxidative, non apoptogenic treatments produce an upshift of Bax migration compatible with<br />

homodimerisation that is reverted by reducing agents; this is accompanied by translocation to<br />

mitochondria. Dimers also appear in pure cytosolic fractions of cell lysates treated with<br />

H2O2, showing that Bax dimerisation may take place in the cytosol. Bax dimers-enriched<br />

lysates support Bax translocation to isolated mitochondria much more efficiently than<br />

untreated lysates, indicating that dimerisation may promote Bax translocation. The absence of<br />

apoptosis in <strong>our</strong> system allows to demonstrate that Bax moves because of oxidations even in<br />

the absence of apoptosis. This provides the first evidence that Bax dimerisation <strong>and</strong><br />

translocation respond to oxidative stimuli, suggesting a novel role for Bax as a sensor of<br />

redox imbalance.<br />

- 44 -


Blocking the "4 Integrin-Paxillin Interaction Selectively Impairs Mononuclear<br />

Leukocyte Recruitment to an Inflammatory Site<br />

Chloé C. Féral*, David M. Rose*, Jaewon Han, Norma Fox, Gregg J. Silverman,<br />

Kenneth Kaushansky, <strong>and</strong> Mark H. Ginsberg.<br />

The University of California San Diego, La Jolla, CA 92093.<br />

"4 integrin antagonists show promise for several autoimmune <strong>and</strong> inflammatory diseases but<br />

may exhibit mechanism-based toxicities. We tested the capacity of blockade of "4 integrin<br />

<strong>signaling</strong> to perturb functions involved in inflammation, while limiting potential adverse<br />

effects. We generated <strong>and</strong> characterized mice bearing an "4(Y991A) mutation that blocks<br />

paxillin binding <strong>and</strong> inhibits "4 signals that support leukocyte migration. In contrast to the<br />

embryonic-lethal phenotype of "4 null mice, mice bearing the "4(Y991A) mutation were<br />

viable <strong>and</strong> fertile; however, they exhibited defective recruitment of mononuclear leukocytes<br />

into thioglycollate-induced peritonitis. "4 integrins are essential for definitive hematopoiesis;<br />

however the "4(Y991A) mice had intact lympho-hematopoiesis <strong>and</strong>, with the exception of<br />

reduced Peyer’s patches, normal architecture <strong>and</strong> cellularity of secondary lymphoid tissues.<br />

We conclude that interference with "4 integrin <strong>signaling</strong> can selectively impair mononuclear<br />

leukocyte recruitment to sites of inflammation while sparing vital functions of "4 integrins in<br />

development <strong>and</strong> hematopoiesis.<br />

- 45 -


Regulation of NF-kB-dependent transcription<br />

Sankar Ghosh<br />

Yale University School of Medicine, New Haven, CT , USA<br />

(no abstract provided)<br />

- 46 -


Regulation of NF-kB-dependent transcription<br />

Sankar Ghosh<br />

Yale University School of Medicine, New Haven, CT , USA<br />

(no abstract provided)<br />

- 47 -


Chromatin impact on NF-kB-driven gene expression<br />

Guy HAEGEMAN, Wim VANDEN BERGHE, Ruben HOYA ARIAS, ‘Matladi<br />

NDLOVU & Linda VERMEULEN<br />

Laboratory for Eukaryotic Gene Expression <strong>and</strong> Signal Transduction (LEGEST),<br />

Department of Molecular Biology, Ghent University, Ledeganckstraat 35, 9000 GENT<br />

(Belgium)<br />

E-mail: guy.haegeman@ugent.be<br />

Interleukin-6 (IL6) is a pluri-potent cytokine, of which the expression needs to be tightly<br />

regulated in order to keep the organism in a balanced <strong>and</strong> homeostatic condition. Indeed,<br />

dysregulation of IL6 leads to a variety of affections, like chronic inflammation, autoimmune<br />

<strong>and</strong> cardiovascular diseases, cancer progression, osteoporosis, etc.<br />

The IL6 gene promoter contains a plethora of transcription factor-binding sites, among which<br />

NF-kB is the most important factor for induction of the gene by infammatory stimuli, like e.g.<br />

TNF. NF-kB is in majority a cytoplasmic protein complex (composed of a p50 <strong>and</strong> a p65<br />

subunit), which upon inflammatory stimulation migrates to the nucleus <strong>and</strong> occupies its<br />

position on a variety of gene promoters. In previous work, we have shown that, in addition to<br />

this cytoplasmic activation step, the transcriptional potency of NF-kB is codetermined by the<br />

activated MAP kinase pathway, more particularly by the nuclear kinase MSK1, that<br />

phosphorylates the p65 subunit at Ser 276 (to generate a fully transcription-competent<br />

enhanceosome), as well as the Histon-3 tails at Ser 10 (which is the onset of chromatin<br />

relaxation).<br />

As it was recently published that IKK-a is the Histon-3 phosphorylating kinase after TNF<br />

induction, we investigated the role <strong>and</strong> impact of these two different kinases for IL6 gene<br />

expression. We found that MSK1 is the primary initiating kinase for temporary chromatin<br />

opening <strong>and</strong> relaxation (even in the absence of activated NF-kB), whereas IKK-a (only<br />

released after TNF induction) takes over <strong>and</strong> continues the status of chromatin relaxation,<br />

when activated MSK1 levels off. Sustained chromatin opening <strong>and</strong> arrival of NF-kB at the<br />

gene promoter upon TNF induction thus lead to abundant IL6 gene expression, whereas<br />

enhanced IL6 gene expression <strong>and</strong>/or temporary chromatin relaxation by MSK1 extinguishes<br />

in the absence of TNF signalization.<br />

Not only the local (i.e. the gene-specific) chromatin relaxation, but also the entire<br />

nucleosomal arrangement along the chromatin fiber is determinative for the (cell-specific)<br />

levels of gene expression. Indeed, upon comparison of the IL6 gene promoter status in two<br />

different breast cancer cell lines, i.e. the benign, low IL6-expressing cell type MCF-7 <strong>and</strong> the<br />

aggressive tumor cell line MDA-MB-231 that shows abundant IL6 expression, we found that,<br />

upon DNaseI digestion <strong>and</strong> restriction enzyme accessibility assays, high IL6 expression<br />

correlates with an increased number of hypersensitive sites, constitutive NF-kB/DNA binding,<br />

<strong>and</strong> elevated MSK1 activity. Moreover, chromatin of the low-expressing cell line can be<br />

converted to a more open configuration upon treatment of the cells with inhibitors of DNA<br />

methylation, thus leading to augmented levels of IL6 gene expression in the restrictive cell<br />

type MCF-7.<br />

- 48 -


The immunomodulator AS101 induces growth arrest <strong>and</strong> apoptosis in Multiple<br />

Myeloma cells via the PI3-K/Akt pathway.<br />

Michal Hayun1, Merav Weil1, Yaniv Naor1, Yona Kalechman1, Michael Albeck2,<br />

Nechama Haran-Ghera3 <strong>and</strong> Benjamin Sredni1<br />

1Safdié Institute for AIDS <strong>and</strong> Immunology Research, Faculty of Life Sciences,<br />

2Department of Chemistry, Bar-Ilan University, Ramat-Gan, Israel; 3Department of<br />

Immunology, The Weizman Institute of Science, Rehovot, Israel<br />

Multiple Myeloma (MM) is a clonal B-cell malignancy affecting both the immune <strong>and</strong> the<br />

skeletal systems, <strong>and</strong> accounts for 10% of all hematological cancers. The immunomodulator<br />

ammonium trichloro (dioxoethylene-o,o') tellurate (AS101) is a non toxic compound which<br />

has been shown to have direct anti-tumoral properties in several tumor models. The present<br />

study examined the anti-tumoral activity of AS101 compound by targeting certain <strong>signaling</strong><br />

pathway (PI3-K/Akt) crucial for survival of MM cells. We showed that AS101 induced a<br />

significant inhibition of cell proliferation in MM cells which was time-<strong>and</strong> dose-dependent.<br />

AS101 induced G2/M arrest, an effect associated with an increase of the p53 tumor<br />

suppressor gene, cyclin-dependent kinase inhibitor p21WAF1, <strong>and</strong> CDK1 phosphorylation.<br />

Longer incubation of MM cells with AS101 resulted in an increase in the early apoptotic cell<br />

population <strong>and</strong> caspase 9 activity. PI3-K/Akt pathway is of particular interest because of its<br />

role in inhibiting apoptosis <strong>and</strong> promoting cell proliferation. Downregulation of Akt, decrease<br />

in Survivin expression <strong>and</strong> suppression of the antiapoptotic effect of exogenously addition of<br />

IGF-1 were observed following AS101 treatment. AS101 was also found to sinergize with<br />

Rapamycin in inducing G2/M arrest in MM cells. Rapamycin is a selective inhibitor of the<br />

phosphoprotein mammalian target of rapamycin (mTOR), a downstream target of pAkt. Our<br />

results indicate that the immunomodulator AS101, currently being used in clinical studies<br />

alone or combined with Rapamycin may be effective in the treatment of MM patients.<br />

- 49 -


THE NDR KINASE FAMILY: REGULATORS OF CELL PROLIFERATION AND<br />

MORPHOGENESIS<br />

Hemmings, B.A., Hergovich, A., Schmitz, D., Kohler, R., Vichalkovski, A., Cornils, H.,<br />

Stegert, M.R., Tamaskovic, R., <strong>and</strong> Bichsel, S.J.<br />

Friedrich Miescher Institute for Biomedical Research, Basel, Switzerl<strong>and</strong><br />

The NDR (nuclear-Dbf2-related) family of proteins is a group of serine/threonine kinases<br />

conserved from yeast to mammals which are important regulators of cell morphogenesis <strong>and</strong><br />

proliferation. They are members of AGC kinases (protein kinases A, G, <strong>and</strong> C) <strong>and</strong> include<br />

the following protein kinases NDR, LATS, Cbk1, Orb6, Cot-1 <strong>and</strong> Dbf2. Although, the<br />

precise function(s) of the mammalian NDR kinases still needs to be defined, evidence<br />

suggests that NDR might be involved in tumor progression. NDR1 is hyper-activated in<br />

several S100B-positive melanoma cell lines, <strong>and</strong> thus given that S100B is over expressed in<br />

more than 80% of metastatic melanomas, NDR1 could potentially influence tumor metastasis<br />

potential. Moreover, disruption of the murine NDR2 gene by insertional mutagenesis was<br />

found to result in B-cell lymphomas, suggesting that both mammalian forms of NDR kinase<br />

might be involved in tumor initiation or progression.<br />

Interestingly, in Saccharomyces cerevisiae, the NDR relative Dbf2 constitutes an integral part<br />

of mitotic exit network (MEN), whereas the other NDR homologue in budding yeast, Cbk1, is<br />

required for regulating morphological changes (RAM network). Recent genetic studies of<br />

yeast have unraveled many key players in processes regulating these NDR-related proteins.<br />

Importantly, these studies demonstrated that the budding yeast relatives of human NDR,<br />

interact with Mob1 (Mps one binder 1) <strong>and</strong> Mob2, respectively. A crucial interaction required<br />

for the activity <strong>and</strong> biological functions of these yeast kinases.<br />

Recent work from <strong>our</strong> laboratory has shown human NDR1 <strong>and</strong> NDR2 to be regulated in a<br />

similar fashion. Both proteins are efficiently activated upon treatment of cells with the protein<br />

phosphatase 2A inhibitor, okadaic acid (OA). Upon activation, phosphorylation occurs on the<br />

activation segment site, Ser281 (Ser282 for NDR2), <strong>and</strong> the hydrophobic motif site, Thr444<br />

(Thr442 for NDR2). Phosphorylation of Ser281 occurs by autophosphorylation, whereas<br />

Thr444 is targeted by an upstream kinase, recently identified as the Ste20-like kinase MST3.<br />

Significantly, phosphorylation of both sites is crucial for NDR activity in vitro <strong>and</strong> in vivo.<br />

Further, we found evidence that human MOB1 (hMOB1), the closest relative of yeast Mob1<br />

<strong>and</strong> Mob2, stimulates NDR kinase activity <strong>and</strong> interacts with NDR both in vivo <strong>and</strong> in vitro.<br />

hMOB1 activates NDR1 at the membrane <strong>and</strong> membrane association of NDR1 was dependent<br />

on the domain required for hMOB1 binding.<br />

Recent developments regarding the regulation <strong>and</strong> function of this emerging <strong>signaling</strong><br />

pathway will be presented.<br />

- 50 -


Signaling pathways leading to the activation of the interferon antiviral response<br />

John Hiscott, Molecular Oncology Group<br />

Lady Davis Institute –McGill University, Montreal Canada H3T 1E2<br />

IRF-3 <strong>and</strong> IRF-7 transcription factors are essential for the induction of type I interferon (IFN)<br />

<strong>and</strong> development of the innate antiviral response. Retinoic acid inducible gene I (RIG-I)<br />

contributes to virus-induced IFN production independent of the Toll-like receptor pathways in<br />

response to RNA viruses <strong>and</strong> dsRNA. We recently demonstrated that the NF-kB inducible,<br />

anti-apoptotic protein A20 functioned as a negative regulator of RIG-I <strong>and</strong> blocked RIG-Imediated<br />

NF-kB <strong>and</strong> IRF gene activation, but only weakly interfered with TLR-3 mediated<br />

IFN activation. Expression of A20 completely blocked CARD domain containing DRIG-Iinduced<br />

IRF-3 Ser396 phosphorylation, homodimerization <strong>and</strong> DNA binding. The level of<br />

A20 inhibition was upstream of the TBK1/IKKe kinases that phosphorylate IRF3 <strong>and</strong> IRF7<br />

but downstream of RIG-I itself, since RIG-I was not a target for the ubiquitin-editing<br />

functions of A20.<br />

To identify the A20 target, a BLAST search was performed that identified an uncharacterized<br />

family of proteins – including the prototypical KIAA1271 – that contains a single CARD-like<br />

domain at the N-terminus. Coexpression of KIAA1271 strongly activated IRF <strong>and</strong> NF-kB<br />

dependent promoters <strong>and</strong> appeared to be the adapter that links RIG-I sensing to downstream<br />

<strong>signaling</strong> through the TBK1/IKKe kinases. F<strong>our</strong> independent groups demonstrated that<br />

MAVS/VISA/IPS-1/CARDIF acts downstream of RIG-I <strong>and</strong> stimulates the expression of<br />

IFNb through the activation of NF-kB <strong>and</strong> IRF-3. MAVS localizes to the mitochondria via a<br />

C-terminal transmembrane domain that is required for its function. Within the context of<br />

hepatitis C virus infection, KIAA1271 appears to be the direct target of the HCV NS3/4A<br />

protease; NS3/4A strongly inhibited KIAA1271-mediated activation of IFNB promoter <strong>and</strong> a<br />

mutated form of KIAA1271, KIAA1271(C508A) which altered the site of NS3/4A proteolytic<br />

cleavage was completely resistant to cleavage. Cleavage of KIAA1271 by NS3/4A disrupts<br />

the C-terminal sequence involved in mitochondrial localization of the adapter. The<br />

characterization of MAVS/KIAA1271 provides the first link between mitochondria <strong>and</strong> the<br />

innate immune response.<br />

How cross-talk between the mitochondrial apoptotic pathways <strong>and</strong> the innate response is<br />

achieved remains unknown. Using vesicular stomatitis virus as a model, we demonstrated that<br />

the intrinsic apoptotic cascade, specifically the BAX-BCL-2-Caspase-9 cascade, was the<br />

primary pathway of VSV-induced apoptosis. Cell death was significantly reduced in BaxBak -/-<br />

murine embryonic fibroblasts (MEFs). Conversely, virus replication was enhanced in the<br />

BaxBak -/- MEFs compared to wild-type cells. Accompanying increased viral replication,<br />

expression of antiviral <strong>and</strong> cytokine genes was also attenuated in infected BaxBak -/- MEFs.<br />

BAX but not BAK was required for IRF-3 phosphorylation <strong>and</strong> DNA binding, thus<br />

substantiating a role for apoptotic <strong>signaling</strong> in the activation of innate immune response.<br />

Therefore, virus-induced apoptosis through a BAX-dependent mitochondrial pathway<br />

represents an early signal for the initiation of IFN antiviral response.<br />

- 51 -


The use of BRET (Bioluminescence Resonance Energy Transfer) for the study of<br />

tyrosine-kinase receptors<br />

Tarik ISSAD<br />

CNRS UMR8104, INSERM U567, Université René Descartes Paris 5, Institut Cochin,<br />

Department of Cell Biology, 22 rue Méchain, 75014 Paris<br />

e.mail: issad@cochin.inserm.fr<br />

During these last years, we have developed an expertise in the use of BRET technology for<br />

the study of protein-protein interaction, particularly in the field of tyrosine kinase receptors<br />

<strong>and</strong> their interaction with regulatory partners. We first demonstrated that the BRET<br />

methodology can be used to monitor lig<strong>and</strong>-induced conformational changes within the<br />

insulin receptor (Boute et al, Mol.Pharm., 2001; Tips, 2002 ; Biochem.Pharm., 2002). We<br />

then used BRET to study, in real time, in living cells, the interaction of the insulin receptor<br />

(IR) with the protein tyrosine phosphatase PTP1B, which is anchored on the cytosolic face of<br />

the endoplasmic reticulum (Boute et al., EMBO Report, 2003). We showed that insulin<br />

rapidly <strong>and</strong> dose-dependently stimulated the interaction of the IR with PTP1B <strong>and</strong> we<br />

demonstrated that internalisation of the IR is necessary for this interaction. In addition, we<br />

demonstrated that PTP1B plays an important role in the regulation of the insulin receptor<br />

immature precursor during its biosynthesis in the endoplasmic reticulum (Boute et al., EMBO<br />

Report, 2003 ; Issad et al., Biochimie 2005). We more recently demonstrated that the BRET<br />

methodology can also be used to monitor lig<strong>and</strong>-induced conformational changes within the<br />

IGF1 Receptor, as well as the interaction of this receptor with PTP1B (Blanquart et al., Mol.<br />

Pharm. 2005).<br />

The receptor-like plasma membrane protein-tyrosine phosphatases PTPalpha <strong>and</strong> PTPespilon<br />

may also play a role in the regulation of the IR. We have shown that in the basal state, the IR<br />

<strong>and</strong> these PTPases are pre-associated at the plasma membrane. Using BRET saturation<br />

experiments, we have shown that insulin does not stimulate the recruitment of these PTPases<br />

to the receptor but rather induced conformational changes within these pre-associated<br />

complexes (Lacasa et al., Mol. Pharm. 2005).<br />

Finally, we also provide evidence that the BRET methodology can be use to monitor the<br />

interaction of the insulin receptor with intracellular adaptors positively or negatively involved<br />

in insulin <strong>signaling</strong>.<br />

Publications<br />

Boute, N., Pernet, K., <strong>and</strong> Issad, T. (2001) The activation state of the insulin receptor<br />

monitored using the Bioluminescence Resonance Energy Transfer methodology Molecular<br />

Pharmacology, 60: 640-645<br />

Tavaré, J.M. <strong>and</strong> Issad, T. (2001) Two-dimensional phosphopeptide mapping of receptor<br />

tyrosine kinases Methods in Molecular Biology 124: 67-85 (Humana Press)<br />

Zilberfarb, V., Siquier, K., Strosberg, A.D., <strong>and</strong> Issad, T. (2001) Effect of dexamethasone on<br />

adipocyte differentiation markers <strong>and</strong> tum<strong>our</strong> necrosis factor alpha expression in human<br />

PAZ6 cells Diabetologia, 44 : 377-386<br />

Grosfeld, A.,. Zilberfarb, V., Turban, S., André, J., Guerre-Millo, M., <strong>and</strong> Issad, T. (2002)<br />

Hypoxia increases leptin expression in human PAZ6 adipose cells Diabetologia 45: 527-530<br />

Lahlou,N., Issad,T., Carel, J-C., Camoin, L., Lebouc,Y., Roger, M., <strong>and</strong> Girard, J. (2002)<br />

Mutations in the human leptin <strong>and</strong> leptin receptor genes as models of serum leptin receptor<br />

regulation Diabetes 51: 1980-1985<br />

- 52 -


Boute, N., Jockers, R. <strong>and</strong> Issad, T. (2002) The use of Resonance Energy Transfer in highthroughput<br />

screening: BRET versus FRET. Trends in Pharmacological Sciences 23: 351-<br />

354<br />

Issad, T., Boute, N. <strong>and</strong> Pernet, K (2002) A homogenous assay to monitor the activity of the<br />

insulin receptor using Bioluminescence Resonance Energy Transfer. Biochemical<br />

Pharmacology 64: 813-817<br />

Guerre-Millo, M., Grosfeld, A., <strong>and</strong> Issad, T. (2002) Stimulatory effect of hypoxia on leptin<br />

release in cultured adipose cells. Obesity Research 10: 856<br />

Issad T., Boute N, Pernet K. (2002) The activity of the insulin receptor assessed by<br />

bioluminescence resonance energy transfer. Ann. N. Y. Acad. Sci. 973: 120-123<br />

Boute N., Boubekeur S., Lacasa D. <strong>and</strong> Issad T. (2003) Dynamics of Interaction between<br />

Insulin Receptor <strong>and</strong> Protein Tyrosine Phosphatase-1B in Living Cells. EMBO reports 4 :<br />

313-319.<br />

Issad T., Boute N., Boubekeur B., Lacasa D. <strong>and</strong> Pernet K. Looking for an insulin pill? Use<br />

the BRET methodology! (2003) Diabetes <strong>and</strong> Metabolism 29: 111-117.<br />

Boute N, Zilberfarb V, Camoin L, Bonnafous S, Le March<strong>and</strong>-Brustel Y, Issad T. The<br />

formation of an intrachain disulfide bond in the leptin protein is necessary for efficient leptin<br />

secretion. Biochimie. 2004;86:351-56.44.<br />

Lacasa D, Boute N <strong>and</strong> Issad T. (2005-a) Interaction of the insulin receptor with the<br />

receptor-like protein tyrosine phosphatases PTPalpha <strong>and</strong> PTPepsilon in living cells. Mol.<br />

Pharmacol. 4:1206-1213.<br />

Issad T, Boute N, Boubekeur S, Lacasa D. (2005) Interaction of PTPB with the insulin<br />

receptor precursor during its biosynthesis in the endoplasmic reticulum. Biochimie. 87:111-<br />

116.<br />

Blanquart C, Boute N, Lacasa D <strong>and</strong> Issad T. (2005-b) Monitoring the activation state of the<br />

Insulin-like Growth Factor-1 Receptor <strong>and</strong> its interaction with PTP1B by using the BRET<br />

methodology. Mol. Pharmacol. Jun 23; [Epub ahead of print]<br />

Issad T. <strong>and</strong> Jockers R. Bioluminescence Resonance Energy Transfer (BRET) to monitor<br />

protein-protein interactions (In Press) Methods in Molecular Biology: Transmembrane<br />

<strong>signaling</strong> protocols (Humana Press)<br />

- 53 -


Protein Kinase C alpha as a potential target in the treatment of acquired antiestrogen<br />

resistance of human breast cancer.<br />

Jan. S. Jepsen 1 , Lisa Frankel 1 , Bo Hansen 2 , Anne E. Lykkesfeldt 1<br />

1 Danish Cancer Society, Str<strong>and</strong>boulevarden 49, DK-2100, Copenhagen, Denmark.<br />

2 Santaris Pharma A/S, Bøge Allé 3, DK-2970, Hørsholm, Denmark.<br />

E-mail: jsj@cancer.dk, lfr@cancer.dk, bh@santaris.com, al@cancer.dk<br />

Initially, antiestrogen therapy is an efficient treatment of hormone-dependent breast cancer.<br />

However, many patients eventually acquire resistance. This presents a serious therapeutic<br />

problem <strong>and</strong> current research aims at elucidating the underlying molecular mechanisms in<br />

order to identify new targets in treatment of patients with resistant tumors.<br />

The Protein Kinase C alpha (PKC alpha) level is increased in several cancer types. However,<br />

limited information is available on the involvement of PKC alpha in antiestrogen resistance.<br />

We screened nine different antiestrogen resistant breast cancer cell lines <strong>and</strong> found that<br />

elevated PKC alpha expression was a general phenomenon. Two of the resistant cell lines<br />

were chosen for further studies: MCF-7/TAM R -1 <strong>and</strong> MCF-7/182 R -6, resistant to tamoxifen<br />

<strong>and</strong> ICI 182,780 (faslodex), respectively. These two cell lines posses highly elevated PKC<br />

alpha protein expression <strong>and</strong> kinase activity levels in comparison to parental MCF-7 cells.<br />

A potent <strong>and</strong> relatively specific PKC alpha inhibitor, Ro-32-0432, resulted in a significant<br />

growth inhibition of these antiestrogen resistant cell lines relative to MCF-7. Moreover,<br />

specific down-regulation of PKC alpha by transient transfection of antisense oligonucleotides<br />

resulted in a 30-40% growth inhibition of TAM R -1 <strong>and</strong> 182 R -6, while MCF-7 remained<br />

unaffected. Additionally, using small hairpin RNA, we generated stable PKC alpha knockdown<br />

cells with almost no PKC alpha protein expression <strong>and</strong> found that PKC alpha knock<br />

down restored the sensitivity of TAM R -1 to tamoxifen. Finally, preliminary results indicate<br />

that MCF-7 clones which overexpress PKC alpha are more resistant to antiestrogen mediated<br />

growth inhibition than parental MCF-7 cells. These results suggest that PKC alpha may be<br />

causally involved in the growth of antiestrogen resistant cells <strong>and</strong> that PKC is a potential<br />

therapeutic target in the treatment of antiestrogen resistant breast cancer.<br />

Recent papers (papers from 2004, 2005)<br />

Frogne T, Jepsen JS, Larsen SS, Fog CK, Brockdorff BL <strong>and</strong> Lykkesfeldt AE. Antiestrogenresistant<br />

human breast cancer cells require activated protein kinase B/Akt for growth. Endocr<br />

Relat Cancer. 2005 12(3): 599-614.<br />

Christensen GL, Jepsen JS, Fog CK <strong>and</strong> Lykkesfeldt AE. Sequential versus combined<br />

treatment of human breast cancer cells with antiestrogens <strong>and</strong> the vitamin D analogue<br />

EB1089 <strong>and</strong> evaluation of predictive markers for vitamin D treatment. Breast Cancer<br />

Research <strong>and</strong> Treatment. 2004 85(1):53-63.<br />

Juncker-Jensen A, Lykkesfeldt AE, Worm J, Ralfkiær U, Espelund U <strong>and</strong> Jepsen JS. Insulinlike<br />

growth factor binding protein 2 is a marker for antiestrogen resistant human breast cancer<br />

cell lines but is not a major growth regulator. Submitted to Growth Hormone & IGF Reseach<br />

Jepsen JS, Pfundheller HM <strong>and</strong> Lykkesfeldt AE: Specific down regulation of p21 (WAF1/CIP1)<br />

<strong>and</strong> the estrogen receptor " in MCF-7 cells by antisense oligonucleotides containing locked<br />

nucleic acid (LNA). Oligonucleotides. 2004 14(2): 147-156.<br />

- 54 -


Jespsen JS, Sorensen MD <strong>and</strong> Wengel J. Locked Nucleic Acid: a potent nucleic acid<br />

analogue in therapeutics <strong>and</strong> biotechnology. Oligonucleotides. 2004 14(2): 130-146.<br />

Vollmer J, Jepsen JS, Uhlmann E, Schetter C, Jurk M, Wader T, Wullner M <strong>and</strong> Krieg AM.<br />

Modulation of antisense or CpG oligodeoxynucleotide-mediated immune stimulation by<br />

locked nucleic acid (LNA). Oligonucleotides. 2004 14(1):23-32.<br />

Jepsen JS <strong>and</strong> Wengel J. LNA-Antisense rivals siRNA for gene silencing.<br />

Current Opinion in Drug Discovery <strong>and</strong> Development. 2004 7(2):188-194.<br />

Jepsen JS. Locked nucleic acid (lna) as a therapeutic tool to knock down gene expression.<br />

Encyclopedia of medical genomics <strong>and</strong> proteomics. Accepted.<br />

Jepsen JS <strong>and</strong> Kauppinen S. Locked nucleic acid (lna) as a tool in biotechnology research<br />

<strong>and</strong> diagnostics. Encyclopedia of medical genomics <strong>and</strong> proteomics. Accepted.<br />

Stein CA, Dias N, Benimetskaya L, Jepsen JS, Lai J <strong>and</strong> Raffo A. LY900003 (Isis 3521) <strong>and</strong><br />

G3139 (Genasense; Oblimersen)- Phosphorothioate Antisense Oligonucleotides With<br />

Pleiotropic Mechanisms of Action. Nucleic Acid Therapeutics in Cancer (2004), pp. 177-198.<br />

Edited by Gerwirtz, A., Humana Press (ISBN: 1-58829-258-4).<br />

Hrdlicka, P.J., Jepsen, J.S., Nielsen, C <strong>and</strong> Wengel, J.: Synthesis <strong>and</strong> biological evaluation of<br />

conformationally restricted <strong>and</strong> nucleobase-modified analogs of the anticancer compound 3'-<br />

C-ethynylcytidine (ECyd). Nucleosides Nucleotides Nucleic Acids. 2005 24(5-7): 397-400.<br />

Hrdlicka PJ, Jepsen JS, Nielsen C <strong>and</strong> Wengel J. Synthesis <strong>and</strong> biological evaluation of<br />

nucleobase-modified analogs of the anticancer compounds 3’-C-ethynyluridine (EUrd) <strong>and</strong><br />

3’-C-ethynylcytidine (ECyd).<br />

Bioorganic & Medicinal Chemistry. 2005 13(4): 1249-1260<br />

Hrdlicka PJ, Andersen NK, Jepsen JS, Hansen FG, Haselmann KF, Nielsen C <strong>and</strong> Wengel J.<br />

Synthesis <strong>and</strong> biological evaluation of branched <strong>and</strong> conformationally restricted analogs of<br />

the anticancer compounds 3’-C-ethynyluridine (EUrd) <strong>and</strong> 3’-C-ethynylcytidine (ECyd).<br />

Bioorganic & Medicinal Chemistry. 2005 13(7): 2597-2621<br />

Kragelund BB, Poulsen K, Andersen KV, Baldursson T, Kroll JB, Neergard TB, Jepsen J,<br />

Roepstorff P, Kristiansen K, Poulsen FM <strong>and</strong> Knudsen J. Conserved residues <strong>and</strong> their role in<br />

the structure, function, <strong>and</strong> stability of acyl-coenzyme A binding protein. Biochemistry. 1999<br />

Feb 23; 38(8):2386-94.<br />

Jepsen JS Locked nucleic acid (LNA) as cancer-therapeutic agent. Dan Med Bull 2004<br />

51:139.<br />

- 55 -


Cell <strong>signaling</strong> by proteinase-activated receptor-2 for interleukin-8 formation in human<br />

lung epithelial cells<br />

Atsufumi Kawabata, Mami Nagataki, Kazumi Moriyuki, Fumiko Sekiguchi, Hiroyuki<br />

Nishikawa*.<br />

Division of Physiology <strong>and</strong> Pathophysiology, School of Pharmaceutical Sciences, Kinki<br />

University, Higashi-Osaka 577-8502, Japan, <strong>and</strong> *Fuso Pharmaceutical Industries Ltd.,<br />

Osaka 536-8523, Japan. E-mail: kawabata@phar.kindai.ac.jp<br />

Proteinase-activated receptor-2 (PAR2) plays a dual role in the respiratory system, being pro-<br />

<strong>and</strong> anti-inflammatory. Stimulation of PAR2 in the airway epithelial cells causes<br />

formation/release of prostagl<strong>and</strong>ins <strong>and</strong> cytokines, leading to bronchodilation <strong>and</strong>/or<br />

inflammation. The present study investigated cell <strong>signaling</strong> pathways responsible for<br />

formation of interleukin-8 (IL-8) following PAR2 stimulation in human lung epithelial cells<br />

(A549), as compared with those for formation of prostagl<strong>and</strong>in E 2 (PGE 2). SLIGRL-NH 2, a<br />

PAR2-activating peptide, but not LSIGRL-NH 2, a scrambled peptide, elicited release of IL-8<br />

as well as PGE 2, which reached a plateau at 18 h <strong>and</strong> 3 h, respectively. PAR2 stimulation also<br />

increased the levels of mRNA for IL-8. Inhibition experiments indicated possible involvement<br />

of MEK-ERK, JNK, Src, protein kinase C <strong>and</strong> EGF receptor tyrosine kinase, but not p38<br />

MAP kinase <strong>and</strong> COX, in PAR2-triggered IL-8 formation, being not necessarily the same as<br />

those for PGE 2 formation. Phosphorylation of JNK, ERK <strong>and</strong> EGF receptors was confirmed<br />

following PAR2 stimulation. Upon activation, PAR2 thus triggers activation of multiple<br />

<strong>signaling</strong> pathways including EGF receptors, JNK <strong>and</strong> MEK-ERK in A549 cells, resulting in<br />

delayed up-regulation <strong>and</strong> release of IL-8 that is pro-inflammatory in the respiratory system.<br />

Recent publications :<br />

1. Kawao, N., Nagataki, M., Nagasawa, K., Kubo, S., Cushing, K., Wada, T., Sekiguchi, F.,<br />

Ichida, S., Hollenberg, M.D., MacNaughton, W.K., Nishikawa, H. <strong>and</strong> Kawabata, A.* (2005)<br />

Signal transduction for proteinase-activated receptor-2-triggered prostagl<strong>and</strong>in E 2 formation<br />

in human lung epithelial cells. J. Pharmacol. Exp. Ther., in press.<br />

2. Nishikawa, H. <strong>and</strong> Kawabata, A.* (2005) PAR-2 as a target for gastric mucosal<br />

protection. Drugs of Future, in press. (Review)<br />

3. Sekiguchi, F., Hasegawa, N., Inoshita, K., Yonezawa, D., Inoi, N., Kanke, T., Saito, N.<br />

<strong>and</strong> Kawabata, A.* (2005) Mechanisms for modulation of mouse gastrointestinal motility by<br />

proteinase-activated receptor (PAR)-1 <strong>and</strong> -2 in vitro. Life Sci., in press.<br />

4. Kanke, T.*, Ishiwata, H., Kabeya, M., Saka, M., Doi, T., Hattori, Y., Kawabata, A. <strong>and</strong><br />

Plevin, R. (2005) Binding of a highly potent protease-activated receptor-2 (PAR2) activating<br />

peptide, [ 3 H]-2-furoyl-LIGRL-NH 2, to human PAR2. Br. J. Pharmacol., 145, 255-263.<br />

5. Kawabata, A.*, Oono, Y., Yonezawa, D., Hiramatsu, K., Inoi, N., Sekiguchi, F., Honjo,<br />

M., Hirofuchi, M., Kanke, T. <strong>and</strong> Ishiwata, H. (2005) 2-furoyl-LIGRL-NH 2, a potent agonist<br />

for proteinase-activated receptor-2, as a gastric mucosal cytoprotective agent. Br. J.<br />

Pharmacol., 144, 212-219.<br />

6. Kawabata, A.* <strong>and</strong> Kawao, N. (2005) Physiology <strong>and</strong> pathophysiology of proteinaseactivated<br />

receptors (PARs): PARs in the respiratory system: cellular <strong>signaling</strong> <strong>and</strong><br />

physiological/pathological roles. J. Pharmacol. Sci., 97, 20-24. (Review)<br />

7. Kanke, T.*, Takizawa, T., Kabeya, M. <strong>and</strong> Kawabata, A. (2005) Physiology <strong>and</strong><br />

pathophysiology of proteinase-activated receptors (PARs): Proteinase-activated receptor-2<br />

(PAR-2) as a potential therapeutic target. J. Pharmacol. Sci., 97, 38-42. (Review)<br />

- 56 -


8. Nishikawa, H., Kawai, K., Tanaka, M., Ohtani, H., Tanaka, S., Kitagawa, C., Nishida,<br />

M., Abe, T., Araki, H. <strong>and</strong> Kawabata, A.* (2005). Protease-activated receptor-2 (PAR-2)related<br />

peptides induce tear secretion in rats: involvement of PAR-2 <strong>and</strong> non-PAR-2<br />

mechanisms. J. Pharmacol. Exp. Ther., 312, 324-331.<br />

9. Kimura, T., Arai, M., Masuda, H. <strong>and</strong> Kawabata, A.* (2004). Impact of a pharmacistimplemented<br />

anemia management in outpatients with end-stage renal disease in Japan. Biol.<br />

Pharm. Bull., 27, 1831-1833<br />

10. Kawabata, A.*, Nakaya, Y., Ishiki, T., Kubo, S., Kuroda, R., Sekiguchi, F., Kawao, N.,<br />

Nishikawa, H. (2004) Receptor-activating peptides for PAR-1 <strong>and</strong> PAR-2 relax rat gastric<br />

artery via multiple mechanisms. Life Sci., 75, 2689-2702.<br />

11. Sekiguchi, F. <strong>and</strong> Kawabata, A.* (2004) Protease-activated receptors (PARs) as<br />

therapeutic targets: development of agonists/antagonists <strong>and</strong> modulation of gastrointestinal<br />

functions. Drug Design Reviews, 1, 287-296. (Review)<br />

12. Kawabata, A.*, Kubo, S., Ishiki, T., Kawao, N., Sekiguchi, F., Kuroda, R., Hollenberg,<br />

M.D., Kanke, T. <strong>and</strong> Saito N. (2004) Proteinase-activated receptor-2-mediated relaxation in<br />

mouse tracheal <strong>and</strong> bronchial smooth muscle: Signal transduction mechanisms <strong>and</strong> distinct<br />

agonist sensitivity. J. Pharmacol. Exp. Ther., 311, 402-410.<br />

13. Kawabata, A.*, Itoh, H., Kawao, N., Kuroda, R., Sekiguchi, F., Masuko, T., Iwata, K.,<br />

Ogawa, A. (2004). Activation of trigeminal nociceptive neurons by parotid PAR-2 activation<br />

in rats. NeuroReport, 15, 1617-1621.<br />

14. Sekiguchi, F., Mita, Y., Kamanaka, Y., Kawao, N., Matsuya, H., Taga, C. <strong>and</strong><br />

Kawabata, A.* (2004). The potent iNOS inhibitor ONO-1714 inhibits nNOS <strong>and</strong> exerts<br />

antinociception in rats. Neurosci. Lett., 365, 111-115.<br />

15. Kawabata, A*, Kanke, T., Yonezawa, D., Ishiki, T., Saka, M., Kabeya, M., Sekiguchi,<br />

F., Kubo, S., Kuroda, R., Iwaki, M., Katsura, K. <strong>and</strong> Plevin, R (2004) Potent <strong>and</strong><br />

metabolically stable agonists for protease-activated receptor-2: Evaluation of activity in<br />

multiple assay systems in vitro <strong>and</strong> in vivo. J. Pharmacol. Exp. Ther. 309, 1098-1107.<br />

16. Kawao, N., Ikeda, H., Kitano, T., Kuroda, R., Sekiguchi, F., Kataoka, K., Kamanaka, Y.<br />

<strong>and</strong> Kawabata, A.* (2004) Modulation of capsaicin-evoked visceral pain <strong>and</strong> referred<br />

hyperalgesia by protease-activated receptors 1 <strong>and</strong> 2. J. Pharmacol. Sci. 61, 683-692.<br />

17. Kawabata, A.*, Kubo, S., Nakaya, Y., Ishiki, T., Kuroda, R., Sekiguchi, F., Kawao, N.<br />

<strong>and</strong> Nishikawa, H. (2004). Distinct roles for protease-activated receptors 1 <strong>and</strong> 2 in vasomotor<br />

modulation in rat superior mesenteric artery. Cardiovasc. Res. 61, 683-692.<br />

18. Kawabata, A.*, Nishikawa, H., Saitoh, H., Nakaya, Y., Hiramatsu, K., Kubo, S.,<br />

Nishida, M., Kawao, N., Kuroda, R., Sekiguchi, F., Kinoshita, M., Kakehi, K., Arizono, N.,<br />

Yamagishi, H. <strong>and</strong> Kawai, K. (2004) A protective role of protease-activated receptor-1 in rat<br />

gastric mucosa. Gastroenterology 126, 208-219.<br />

19. Kawao, N., Hiramatsu, K., Inoi, N., Kuroda, R., Nishikawa, H., Sekiguchi, F. <strong>and</strong><br />

Kawabata, A.* (2003) The PAR-1-activating peptide facilitates pepsinogen secretion in rats.<br />

Peptides 24, 1449-1451.<br />

20. Kamanaka, Y., Kawabata, A.*, Matsuya, H., Taga, C., Sekiguchi, F. <strong>and</strong> Kawao, N.<br />

(2003) Effect of a potent iNOS inhibitor (ONO-1714) on acetaminophen-induced<br />

hepatotoxicity in the rat. Life Sci. 74, 793-802.<br />

21. Kawabata, A.*, Nakaya, Y., Kuroda, R., Wakisaka, M., Masuko, T., Nishikawa, H. <strong>and</strong><br />

Kawai, K. (2003) Involvement of EDHF in the hypotension <strong>and</strong> increased gastric mucosal<br />

blood flow caused by PAR-2 activation in rats. Br. J. Pharmacol. 140, 247-254.<br />

22. Kuroda, R.*, Kawabata, A. (2003) Pain information pathways from the periphery to the<br />

cerebral cortex. Yakugaku Zasshi 123, 533-546 (Review) (in Japanese)<br />

- 57 -


Crosstalk between NF-kB <strong>and</strong> AP-1 in Helicobacter pylori-mediated inflammation in<br />

human gastric epithelial cells<br />

Hyeyoung Kim<br />

Department of Pharmacology <strong>and</strong> Brain Korea 21 Project for Medical Science, Yonsei<br />

University College of Medicine, Seoul 120-752, Korea. Email:<br />

kim626@yumc.yonsei.ac.kr<br />

Previously we have shown that inflammatory enzymes such as cyclooxygenase-2 (COX-2)<br />

<strong>and</strong> inducible nitric oxide syntahse (iNOS) were expressed in H. pylori-infected gastric<br />

epithelial cells. The expression of COX-2 <strong>and</strong> iNOS may be regulated by NF-kB <strong>and</strong> AP-1.<br />

Present study aims to investigate whether H. pylori-induced expressions of COX-2 <strong>and</strong> iNOS<br />

are regulated by NF-kB <strong>and</strong> AP-1 in gastric epithelial AGS cells, <strong>and</strong> whether the<br />

transcriptional regulation of COX-2 or iNOS is inhibited by transfection of antisense<br />

oligodeoxynucleotide (AS ODN) for NF-kB subunit p50 or c-jun dominant negative mutant<br />

gene (TAM-67). As a result, H. pylori induced the expression of COX-2 <strong>and</strong> iNOS via<br />

activation of NF-kB <strong>and</strong> AP-1, which was inhibited in the cells transfected with either p50 AS<br />

ODN or TAM-67. The activated NF-kB complex was p65/p50 hetero- <strong>and</strong> p50/p50 homo-<br />

dimmers while the activated AP-1 complex was a c-jun/c-fos heterodimer in H. pyloriinfected<br />

AGS cells. In conclusion, NF-kB <strong>and</strong> AP-1 share H. pylori-mediated inflammation<br />

<strong>signaling</strong> by inducing COX-2 <strong>and</strong> iNOS expression in gastric epithelial cells. Either<br />

suppression of NF-kB or AP-1 may inhibit H. pylori-induced inflammation in gastric<br />

epithelial cells.<br />

Publication list (2002-2005)<br />

1) NADPH oxidase <strong>and</strong> apoptosis in cerulein-stimulated pancreatic acinar cells. JH Yu, JW<br />

Lim, KH Kim, T Morio, H Kim. Free Rad Biol Med 39: 590-602, 2005<br />

2) NADPH oxidase mediates interleukin-6 expression in cerulein-stimulated pancreatic acinar<br />

cells. JH Yu, JW Lim, H Kim, KH Kim, Int J Biochem Cell Biol 37:1458-1469, 2005<br />

3) Cellular stress - related protein expression in Helicobacter pylori - infected gastric<br />

epithelial AGS cells. JW Lim, H Kim, JM Kim, JS Kim, HC Jung, KH Kim, Int J Biochem<br />

Cell Biol 36:1624-1634, 2004.<br />

4) Oxidative stress - related proteome changes in Helicobacter pylori - infected human gastric<br />

mucosa. HY Baek, JW Lim, H Kim, JM Kim, JS Kim, HC Jung, KH Kim, Biochem J<br />

379:291-299, 2004<br />

5) Helicobacter pylori in a Korean isolate activates mitogen-activated protein kinases, AP-1,<br />

<strong>and</strong> NF-!B <strong>and</strong> induces chemokine expression in gastric epithelial AGS cells. JH Seo, JW<br />

Lim, H Kim, KH Kim, Lab Inv 84:49-62, 2004<br />

6) The Ku antigen-recombination signal-binding protein J! complex binds to the nuclear<br />

factor-!B p50 promoter <strong>and</strong> acts as a positive regulator of p50 expression in human gastric<br />

cancer cells. JW Lim, H Kim, KH Kim, J Biol Chem 279(1):231-237, 2004<br />

7) Role of oxygen free radicals in patients with acute pancreatitis. BK Park, JB Chung, JH<br />

Lee, JH Suh, SW Park, SY Song, H Kim, KH Kim, JK Kang, World J Gastroenterology<br />

9(10):2266-2269, 2003<br />

8) Oxidative stress induces nuclear loss of DNA repair proteins Ku70 <strong>and</strong> Ku80 <strong>and</strong> apoptosis<br />

in pancreatic acinar AR42J cells. JY Song, JW Lim, H Kim, T Morio, KH Kim.. J Biol Chem<br />

278 (38):36676-36687, 2003 Oct.<br />

- 58 -


9) Proteome analysis of rat pancreatic acinar cells: Implication for cerulein - induced acute<br />

pancreatitis, JH Yu, SY Yun, H Kim, KH Kim, Proteomics 3(12):2446-2453, 2003<br />

10) Mass spectrometry <strong>and</strong> t<strong>and</strong>em mass spectrometry analysis of rat pancreatic<br />

mitochondrial ATP synthase: upregulation in pancreatic acinar cells treated with cerulein, JH<br />

Yu, SY Yun, H Kim, KH Kim, Proteomics 3(12):2437-2445, 2003<br />

11) The effect of p38 mitogen-activated protein kinase on mucin gene expression annd<br />

apoptosis in Helicobacter pylori-infected gastric epithelial cells, H Kim, JH Seo, KH Kim,<br />

Ann NY Acad Sci 1010:90-94, 2003<br />

12) Role of NF-!B <strong>and</strong> DNA repair protein Ku on apoptosis in pancreatic acinar cells, JY<br />

Song, JW Lim, H Kim, KH Kim, Ann NY Acad Sci 1010:259-262, 2003<br />

13) Calcium-dependent apoptotic gene expression in cerulein-treated AR42J cells, JH Yu, H<br />

Kim, KH Kim, Ann NY Acad Sci 1010:66-70, 2003<br />

14) Signal transduction of cerulein-induced cytokine expression <strong>and</strong> apoptosis in pancreatic<br />

acinar cells, J Lee, J Seo, H Kim, JB Chung, KH Kim, Ann NY Acad Sci 1010:104-108,<br />

2003<br />

15) NF-kB <strong>and</strong> Bcl-2 in Helicobacter pylori-induced apoptosis in gastric epithelial cells, SH<br />

Chu, JW Lim, KH Kim, H Kim, Ann NY Acad Sci 1010:568-575, 2003<br />

16) Diagnostic significance of antibodies to heat shock proteins, H Kim, Clinica Chemica<br />

Acta, 337:1-10, 2003<br />

17) Cell adhesion - related gene expression by Helicobacter pylori in gastric epithelial AGS<br />

cells, JW Lim, H Kim, KH Kim, Int J Biochem Cell Biol, 35:1284-1296, 2003<br />

18) Role of NF-kB <strong>and</strong> AP-1 on Helicobacter pylori-induced IL-8 expression in AGS cells.<br />

SH Chu, H Kim, JY Seo, JW Lim, N Mukaida, KH Kim, Dig Dis Sci 48(2): 257-265, 2003<br />

19) Expression of Ku70 <strong>and</strong> Ku80 mediated by NF-kB <strong>and</strong> cyclooxygenase-2 are related to<br />

cell proliferation in human gastric cancer cells. JW Lim, H Kim, KH Kim, J Biol Chem<br />

277(48):46093-46100, 2002<br />

20) Transcriptional regulation by thiol compounds in Helicobacter pylori induced IL-8<br />

production in human gastric epithelial cells. JY Seo, H Kim, KH Kim. Ann New York Acad<br />

Sci 973:541-545, 2002<br />

21) Cyclooxygenase-2 expression by transcription factors in Helicobacter pylori-infected<br />

gastric epithelial cells: comparison between HP99 <strong>and</strong> NCTC 11637. JH Seo, H Kim, KH<br />

Kim. Ann New York Acad Sci 973:477-480, 2002<br />

22) Suppression of cerulein-induced cytokine expression by antioxidants in pancreatic acinar<br />

cells. JH Yu, JW Lim, W Namkung, H Kim, KH Kim. Lab Inv 82(10):1359-1368, 2002<br />

23) Effect of mannitol on Helicobacter pylori-induced cyclooxygenase-2 expression in gastric<br />

epithelial AGS cells. H Kim, JY Seo, KH Kim Pharmacology 66(4):182-189, 2002 Dec.<br />

24) Oxidative stress-induced cytokine production in isolated rat pancreatic acinar cells;<br />

Effects of small molecule antioxidants. JY Seo, H Kim, JT Seo, KH Kim, Pharmacology<br />

64(2):63-70, 2002 Feb.<br />

25) Oxidant-sensitive transcription factor <strong>and</strong> cyclooxygenase-2 by Helicobacter pylori<br />

stimulation in human gastric cancer cell lines. H Kim, JW Lim, JY Seo, KH Kim, J<br />

Environmental Pathology, Toxicology <strong>and</strong> Oncology 21:121-129, 2002<br />

- 59 -


Signaling of life <strong>and</strong> death in T cells<br />

Peter H. Krammer<br />

Tumorimmunology Program, German Cancer Research Center, Heidelberg, Germany):<br />

(no abstract provided)<br />

- 60 -


MicroRNA is an anti-apoptotic factor in human brain tumors<br />

Anna M. Krichevsky1, Jennifer A. Chan2, & Kenneth S. Kosik3<br />

1 Department of Neurology, Brigham <strong>and</strong> Women's Hospital, Harvard Medical School;<br />

2 Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical<br />

School, Boston, MA 02115 <strong>and</strong> 3 Neuroscience Research Institute, University of<br />

California Santa Barbara, Santa Barbara, CA 93106, USA<br />

MicroRNAs (miRNAs) are a recently discovered class of small 18-23 nucleotide non-coding<br />

RNA molecules that have been shown to regulate target gene expression in various<br />

organisms. By targeting the mRNA of protein-coding genes for either cleavage or repression<br />

of translation, miRNAs are thought to play critical roles in development, control of growth,<br />

proliferation, <strong>and</strong> cell lineage determination. Expression data <strong>and</strong> location in the human<br />

genome suggested the potential for this new class of regulatory RNAs to influence the<br />

processes of proliferation, differentiation <strong>and</strong> cell death in a human malignancy.<br />

Using <strong>our</strong> homemade oligonucleotide array designed to detect the majority of mammalian<br />

miRNAs identified thus far, we measured the expression levels of miRNAs during normal<br />

brain development, in the process of neurogenesis in vitro <strong>and</strong> in high-grade brain tumors,<br />

glioblastomas. Our study identified a specific miRNA, miR-21, that is markedly elevated in<br />

glioblastomas. We demonstrate that glioblastomas strongly overexpress this specific miRNA<br />

compared to low-grade brain tumors, non-neoplastic fetal <strong>and</strong> adult brain, neuronal <strong>and</strong> glial<br />

cultured cells, <strong>and</strong> stem cell-derived neural precursors, clearly linking this miRNA to<br />

malignancy. Sequence-specific knock-down of miR-21 with modified antisense<br />

oligonucleotides triggers activation of caspases <strong>and</strong> leads to increased apoptotic cell death of<br />

glioblastoma cells in culture, implying a role for this miRNA in gliomagenesis. Wholegenome<br />

expression profiling revealed that multiple proapoptotic genes as well as genes<br />

negatively regulating cell proliferation have been specifically induced after the knock-down<br />

of this miRNA. These findings suggest that the over-expressed miRNA may function as a<br />

micro-oncogene, presumably by blocking expression of key apoptosis-enabling genes.<br />

Moreover, <strong>our</strong> studies not only validate a functional role of miRNA in brain tumors, but also<br />

raise the possibility that miRNAs may serve as novel attractive targets in oncology.<br />

- 61 -


Mechanisms of caspase-independent apoptosis<br />

Guido Kroemer<br />

CNRS-UMR8125, Institut Gustave Roussy, 94805 Villejuif, France<br />

Chemotherapy resistance has often been related to disabled apoptosis with deficient caspase<br />

activation. However, tumor cells tend to die in response to most conventional<br />

chemotherapeutic agents even when caspase activation is inhibited. This caspase-independent<br />

cell death is mediated by a series of metabolic changes, as well as by the mitochondrial<br />

release of caspase-independent death effectors such as AIF. We have recently found that,<br />

even in the absence of any adjuvant, tumor cells dying in response to anthracyclins can elicit<br />

an effective anti-tumor immune response that precludes the growth of inoculated tumors or<br />

leads to the regression of established neoplasia. Caspase inhibition by Z-VAD-fmk or<br />

transfection with the baculovirus inhibitor p35 did not inhibit doxorubicin-induced cell death<br />

(as measured as clonogenic survival), yet suppressed the immunogenicity of dying tumor cells<br />

in a variety of different rodent models of neoplasia. Depletion of DC in vivo curtailed the<br />

immune response against doxorubicin-treated apoptotic tumor cells. Caspase inhibition<br />

suppressed the capacity of doxorubicin-killed cells to be phagocytosed by dendritic cells<br />

(DC), yet had no effect on their capacity to elicit DC maturation. Freshly excised tumors<br />

became immunogenic upon doxorubicin treatment in vitro, <strong>and</strong> intratumoral inoculation of<br />

doxorubicin could trigger the regression of established tumors in immunocompetent mice.<br />

These results delineate a procedure for the generation of cancer vaccines <strong>and</strong> the stimulation<br />

of anti-neoplastic immune responses in vivo. Moreover, they suggest that inhibition of<br />

caspase activation (which leads to a shift from apoptotic to non-apoptotic death modalities)<br />

can abrogate the immunogenic nature of cell death, thus favoring the escape of tumors from<br />

immune surveillance.<br />

- 62 -


Resveratrol inhibits phorbol ester-induced expression of COX-2 through inactivation of<br />

NF-!B <strong>and</strong> AP-1 in mouse skin: role of I!B kinase <strong>and</strong> MAP kinases<br />

Joydeb Kumar Kundu, Young Kee Shin <strong>and</strong> Young-Joon Surh<br />

National Research Laboratory of Molecular Carcinogenesis <strong>and</strong> Chemoprevention,<br />

College of Pharmacy, Seoul National University, Seoul 151-742, South Korea<br />

Resveratrol, an antioxidant <strong>and</strong> anti-inflammatory phytoalexin present in red wine <strong>and</strong> grapes,<br />

was reported to inhibit chemically-induced carcinogenesis in mouse skin. The present study<br />

was aimed to elucidate molecular mechanisms of inhibitory effects of resveratrol on 12-Otetradecanoylphorbol-13-acetate<br />

(TPA)-promoted mouse skin carcinogenesis. Topical<br />

application of resveratrol significantly inhibited TPA-induced COX-2 expression. Resveratrol<br />

diminished the DNA binding of NF-!B by suppressing phosphorylation <strong>and</strong> subsequent<br />

degradation of I!B", <strong>and</strong> resultant nuclear translocation of p65. Resveratrol also blunted<br />

TPA-induced phosphorylation of p65 <strong>and</strong> subsequent interaction of p65 with cyclic AMP<br />

response element binding protein-binding protein. The TPA-induced DNA binding of AP-1<br />

<strong>and</strong> the expression of c-Jun <strong>and</strong> c-Fos were also diminished by resveratrol. Moreover,<br />

pretreatment with resveratrol suppressed the activation of ERK, p38 MAP kinase <strong>and</strong> JNK.<br />

Topical application of TPA caused rapid induction of IKK activity, which was suppressed by<br />

pretreatment with resveratrol or cotreatment with Bay 11-7082, a pharmacological inhibitor of<br />

IKK. Topically applied Bay 11-7082 also abrogated TPA-induced activation of NF-!B <strong>and</strong><br />

AP-1 as well as expression of COX-2, suggesting a regulatory role of IKK in TPA-induced<br />

COX-2 expression in mouse skin in vivo. The TPA-induced phosphorylation of ERK, JNK<br />

<strong>and</strong> p38 MAP kinases was also abrogated by Bay 11-7082 treatment.<br />

List of Publications :<br />

1. Kundu JK, Choi K-Y., <strong>and</strong> Surh Y-J. #-catenin-mediated <strong>signaling</strong>: a novel<br />

molecular target for chemoprevention with anti-inflammatory substances. Biochemica et<br />

Biophysica Acta - Rev. Cancer. 1765 (1): 14-24, 2006.<br />

2. Kundu JK, <strong>and</strong> Surh Y-J. Molecular mechanisms underlying chemoprevention with<br />

resveratrol, Cancer Prev. Res.. 10 (2): 89-98; 2005.<br />

3. Surh Y-J., Kundu JK, Na H-K., <strong>and</strong> Lee J-S. <strong>Redox</strong>-sensitive transcription factors as<br />

prime targets for chemoprevention with anti-inflammatory <strong>and</strong> antioxidative phytochemicals,<br />

J. Nutr. 135 (12): 2993S-3001S; 2005.<br />

4. Prawan A, Kundu JK <strong>and</strong> Surh Y-J. Molecular basis of heme oxygenase-1 induction:<br />

implications for chemoprevention <strong>and</strong> chemoprotection, Antiox. <strong>Redox</strong> Signal. (in press),<br />

2005.<br />

5. Kim SO, Kundu JK, Shin YK, Park J-H., Cho M-H, Kim T-Y. <strong>and</strong> Surh Y-J. [6]-<br />

Gingerol inhibits COX-2 expression by blocking the activation of p38 MAP kinase <strong>and</strong> NF-<br />

!B in phorbol ester-stimulated mouse skin. Oncogene 24(15): 2558-67, 2005.<br />

6. Kundu JK <strong>and</strong> Surh Y-J. Breaking the relay in deregulated cellular signal<br />

transduction as a rationale for chemoprevention with anti-inflammatory phytochemicals,<br />

Mutat. Res. 591 (1-2): 123-146; 2005<br />

7. Chowdhury KK, Saha A, Bachar SC <strong>and</strong> Kundu JK. Analgesic <strong>and</strong> antiinflammatory<br />

activity of Desmodium triflorum DC. J. Biol. Sci., 5 (5) : 581-583; 2005<br />

8. Surh Y-J. <strong>and</strong> Kundu JK. Signal transduction network leading to COX-2 induction: a<br />

road map in search of cancer chemopreventives. Arch. Pharm. Res. 28: 1-15; 2005.<br />

- 63 -


9. Kundu JK, Moss<strong>and</strong>a KA, Na H-K <strong>and</strong> Surh Y-J. Inhibitory effects of extracts of<br />

Sutherl<strong>and</strong>ia frutescens <strong>and</strong> Harpagophytum procumbens on phorbol ester-induced COX-2<br />

expression in mouse skin : AP-1 <strong>and</strong> CREB as potential targets. Cancer Lett. 218: 21-31;<br />

2005<br />

10. Kundu JK <strong>and</strong> Surh Y-J. Molecular basis of chemoprevention by resveratrol : NF-!B<br />

<strong>and</strong> AP-1 as potential targets, Mutat. Res. 255: 65-80; 2004<br />

11. Kundu JK, Chun K-S, Kim SO <strong>and</strong> Surh Y-J. Resveratrol inhibits phorbol esterinduced<br />

cyclooxygenase-2 expression in mouse skin: MAPKs <strong>and</strong> AP-1 as potential<br />

molecular targets. Biofactors 21: 33-39; 2004<br />

12. Kim SO, Chun K-S, Kundu JK <strong>and</strong> Surh Y-J. Inhibitory effects of [6]-gingerol on<br />

PMA-induced COX-2 expression <strong>and</strong> activation of NF-!B <strong>and</strong> p38 MAPK in mouse skin.<br />

Biofactors 21: 27-31; 2004<br />

13. Datta BK, Dutta SK., Khan TH, Kundu JK, Rashid MA, Nahar L <strong>and</strong> Sarker SD.,<br />

Anti-cholinergic, cytotoxic <strong>and</strong> anti-HIV activities of a sesquiterpene <strong>and</strong> a flavone glycoside<br />

from the aerial parts of P. viscosum, Pharmaceutical Biol., 42: 18-23; 2004.<br />

14. Datta BK, Datta SK, Chowdhury MM, Khan TH, Kundu JK, Rashid MA, Nahar L,<br />

Sarker SD. Analgesic, anti-inflammatory <strong>and</strong> CNS depressant activities of sesquiterpenes <strong>and</strong><br />

a flavonoid glycoside from Polygonum viscosum Die Pharmazie, 59: 222-225; 2004<br />

15. Kundu JK, Na H-K, Chun K-S, Kim Y-K, Lee S-J, Lee S-S, <strong>and</strong> Surh Y-J . Inhibition<br />

of phorbol ester-induced COX-2 expression by epigallocatechin gallate in mouse skin <strong>and</strong><br />

cultured human mammary epithelial cells. J Nutr. 133: 3805S-3810S; 2003<br />

16. Das AK, Ahmed F, Bachar SC, Kundu JK <strong>and</strong> Dev S.; Anti-inflammatory effect of<br />

Albizzia lebbeck (Benth.) Bark J. Biol. Sci. 3: 685-687, 2003<br />

17. Shaphiullah M, Bachar SC, Kundu JK, Begum F, Uddin A, Roy SC, Khan MTH.<br />

Antidiarrheal activity of the methanol extract of Ludwigia hyssopifolia Linn Pak. J. Pharm.<br />

Sci 16:7-12; 2003.<br />

18. Datta BK, Datta SK, Rashid M.A, Kundu JK, Hasan CM <strong>and</strong> Sarker SD. Further<br />

Sesquiterpenes from Polygonum viscosum (Polygonaceae), Nat. Prod. Lett. 16: 143-148,<br />

2002<br />

19. Datta BK, Rashid MA, Kundu JK, Rouf ASS, Sarker SD <strong>and</strong> Datta SK. Isolation <strong>and</strong><br />

structure elucidation of viscoazucine, a novel sesquiterpene from Polygonum viscosum.<br />

Pharmazie, 56: 578-9, 2001<br />

20. Kundu JK, Rouf ASS., Hossain MN, Hasan CM <strong>and</strong> Rashid MA. Antitumor activity<br />

of epifriedelanol from Vitis trifolia. Fitoterapia 71: 574-576; 2000<br />

21. Ahmed M, Datta BK, Sadhu SK, Kundu JK <strong>and</strong> Bachar SC. Preliminary biological<br />

studies on stagninol a sesquiterpene isolated from Persicaria stagnina Linn., Pharmazie, 52:<br />

472-75, 1997.<br />

- 64 -


Cross-talk between angiotensin II <strong>and</strong> glucagon receptor <strong>signaling</strong> mediates activation<br />

of mitogen-activated protein kinases ERK 1/2 in rat glomerular mesangial cells.<br />

Xiao C. Li, Oscar A. Carretero, <strong>and</strong> Jia L. Zhuo.<br />

Division of Hypertension <strong>and</strong> Vascular Research, Henry Ford Hospital, Detroit, MI<br />

48202, USA. E-mail: xcli1@hfhs.org<br />

Agonist-induced phosphorylation of MAP kinases extracellular signal-regulated protein<br />

kinases 1 <strong>and</strong> 2 (p-ERK 1/2) plays an important role in the regulation of cell differentiation,<br />

transformation <strong>and</strong> proliferation. We hypothesized that angiotensin II (Ang II), glucagon <strong>and</strong><br />

high glucose (HG) induce ERK 1/2 phosphorylation in cultured rat glomerular mesangial cells<br />

via cross-talk between their <strong>signaling</strong> pathways. Ang II (1 nM), glucagon (1 nM) <strong>and</strong> high<br />

glucose (HG, 25 mM) induced ERK 1/2 phosphorylation (Ang II: 174 ± 16%; glucagon: 214<br />

± 14%; <strong>and</strong> HG: 190 ± 15%; p < 0.01 vs. control), <strong>and</strong> all responses were inhibited by the<br />

AT 1 receptor blocker losartan (Ang II + losartan: 62 ± 21%; glucagon + losartan: 77 ± 14%;<br />

<strong>and</strong> HG ± losartan: 98 ± 10%; p < 0.01 vs. Ang II, glucagon or HG). Pretreatment of<br />

mesangial cells with a phospholipase C (PLC) inhibitor U73122 (10 -6 M), a cAMP-dependent<br />

protein kinase A (PKA) inhibitor H89 (10 -6 M), or a phosphatidylinositol 3-kinase (PI 3kinase)<br />

inhibitor wortmannin (10 -6 M), all markedly attenuated phosphorylation of ERK 1/2<br />

induced by Ang II (Ang II + U73122: 111 ± 13%; Ang II + H89: 86 ± 10%; <strong>and</strong> Ang II +<br />

wortmannin: 114 ± 8%; p < 0.01 vs. Ang II), glucagon (glucagons + U73122: 109 ± 15%;<br />

glucagon + H89: 113 ± 16%; <strong>and</strong> glucagons + wortmannin: 115 ± 9%; p < 0.01 vs. glucagon)<br />

<strong>and</strong> HG (HG + U73122: 90 ±18%; HG + H89: 117 ± 7%; <strong>and</strong> HG + wortmannin: 139 ± 11%;<br />

p < 0.01 vs. HG). These results suggest that AT 1 receptor-activated PLC, cAMP-dependent<br />

PKA <strong>and</strong> PI 3-kinase <strong>signaling</strong> pathways mediate Ang II-, glucagon- <strong>and</strong> hyperglycemiainduced<br />

MAP kinases ERK 1/2 phosphorylation in glomerular mesangial cells. Parts of this<br />

work were supported by a grant from National Institutes of Health (RO1DK067299).<br />

Addendum: A list of recent publications<br />

Widdop, R.E., X.C. Li, <strong>and</strong> B. Jarrott. Regional haemodynamic effects of AT1 angiotensin II<br />

receptor antagonist, CV-11974, in conscious rats. Blood Press. 3 (Suppl 5): 15-20, 1994.<br />

Li, X.C., <strong>and</strong> R.E. Widdop. Regional hemodynamic effects of angiotensin II AT1 receptor<br />

antagonist, CV-11974, in conscious renal hypertensive rats. Hypertension 26: 989-997, 1995.<br />

Li, X.C., <strong>and</strong> R.E. Widdop. Angiotensin type 1 receptor antagonists CV-11974 <strong>and</strong> EXP 3174<br />

cause selective renal vasodilatation in conscious spontaneously hypertensive rats. Clin. Sci.<br />

91: 147-154, 1996.<br />

Widdop, R.E., <strong>and</strong> X.C. Li. A simple versatile method for measuring tail cuff systolic blood<br />

pressure in conscious rats. Clin. Sci. 93: 191-194, 1997.<br />

Li, X.C., P.M. Beart, A.M. James, N.M. Nicole, <strong>and</strong> R.E. Widdop. Type I <strong>and</strong> II metabotropic<br />

receptor agonists <strong>and</strong> antagonists evoke cardiovascular effects after intrathecal administration<br />

in conscious rats. Br. J. Pharmacol. 128: 823-829, 1999.<br />

Paull, J.R.A., X.C. Li, D.B. Sampey, <strong>and</strong> R.E. Widdop. Pharmacodynamic contribution to the<br />

vasodilator effect of chronic AT1 receptor blockade in SHR. Hypertension 37: 91-98, 2001.<br />

Rey, F.E., X.C. Li, O.A. Carretero, J.L. Garvin, <strong>and</strong> P.J. Pagano. Perivascular superoxide<br />

anion contributes to impairment of endothelium-dependent relaxation. Role of gp91phox.<br />

Circulation 106: 2497-2502, 2002.<br />

Li, X.C., <strong>and</strong> R.E. Widdop. AT2 receptor-mediated vasodilatation is unmasked by AT1<br />

receptor blockade in conscious SHR. Br. J. Pharmacol. 142: 821-830, 2004.<br />

- 65 -


Li, X.C., M.S. Karadsheh, P.M. Jenkins, <strong>and</strong> J.A. Stitzel. Genetic correlation between the<br />

free-choice oral consumption of nicotine <strong>and</strong> alcohol in C57BL/6J x C3H/HeJ F2 intercross<br />

mice. Behav. Brain Res. 157: 79-90, 2005.<br />

Li, X.C., M.S. Karadsheh, P.M. Jenkins, <strong>and</strong> J.A. Stitzel. Chromosomal loci that influence<br />

free-choice nicotine consumption in C57BL/6J x C3H/HeJ F2 intercross mice. Brain Res.<br />

(Invited for revision)<br />

Li, X.C., Shao, Y., Ohishi, M., Campbell, D.J., <strong>and</strong> Zhuo, J.L. AT1 receptor-activated<br />

<strong>signaling</strong> mediates angiotensin IV-induced responses in renal microvascular <strong>and</strong> glomerular<br />

mesangial cells. Am. J. Physiol. Renal Physiol. 2005 (in press)<br />

Zhuo, J.L., Li, X.C., Garvin, J.L., Navar, L.G., <strong>and</strong> Carretero, O.A. Intracellular angiotensin II<br />

induces cytosolic Ca2+ mobilization by stimulating intracellular AT1 receptors in proximal<br />

tubule cells. Am. J. Physiol. Renal Physiol. 2005 (in press).<br />

Zhuo, J.L., Carretero, O.A., Peng, H., Li, X.C., Regoli, D., Neugebauer, W., <strong>and</strong> Rhaleb, N-R.<br />

Characterization of N-Acetyl-Seryl-Aspartyl-Lysyl-Proline (Ac-SDKP) receptor binding sites<br />

using 125I-Hpp-Aca-SDKP in rat cardiac fibroblasts. Hypertension 2005 (invited for<br />

revision)<br />

Li, X.C., Carretero, O.A., Navar, L.G., <strong>and</strong> Jia L. Zhuo. AT1A receptor-mediated<br />

accumulation of extracellular angiotensin II in proximal tubule cells: role of cytoskeleton<br />

microtubules <strong>and</strong> tyrosine phosphatase. Am. J. Physiol. Renal Physiol. 2005 (Invited for<br />

revision, F-00405, 2005)<br />

Li, X.C., Carretero, O.A., Shao, Y. <strong>and</strong> Zhuo, J.L. Glucagon receptor-mediated MAP kinase<br />

ERK 1/2 phosphorylation in mesangial cells: role of protein kinase A <strong>and</strong> phospholipase C<br />

<strong>signaling</strong>. Hypertension (2005) (In Press)<br />

- 66 -


Multiple role of histamine H 1 receptor-PKC-MAPK signalling pathway in histaminestimulated<br />

nerve growth factor secretion from glial cells<br />

Metoda Lipnik-Stangelj<br />

Department of Pharmacology <strong>and</strong> Experimental Toxicology, Faculty of Medicine,<br />

University of Ljubljana, Slovenia. E-mail: metoda.lipnik-stangelj@mf.uni-lj.si<br />

Histamine is a potent stimulator of nerve growth factor (NGF) synthesis <strong>and</strong> secretion which<br />

is essential for differentiation, regeneration <strong>and</strong> survival of neurons. The main signalling<br />

pathway, involved in this process, includes histamine-H 1 receptor (H1R)-activated stimulation<br />

of Ca 2+ -dependent protein-kinase C (PKC) <strong>and</strong> mitogen-activated protein-kinase (MAPK).<br />

Several other mediators contribute to the regulation of NGF secretion, <strong>and</strong> there is intense<br />

crosstalk between them. IL-1beta <strong>and</strong> IL-6 also showed strong influence on NGF secretion.<br />

Although interactions between histamine, IL-1beta <strong>and</strong> IL-6 are common in different<br />

physiological <strong>and</strong> pathological responses, they remained unclear in the regulation of NGF<br />

release from glial cells, so far. In the present study, we investigated interactions between<br />

histamine, IL-1beta <strong>and</strong> IL-6 in the regulation of NGF secretion from astroglial cells in<br />

primary culture, <strong>and</strong> the role of H1R-PKC-MAPK signalling pathway in this process.<br />

Histamine, IL-1beta or IL-6, all showed dose- <strong>and</strong> time-dependent stimulation of NGF<br />

secretion from cultured astrocytes. Concomitant treatment of the cells with both, histamine<br />

<strong>and</strong> IL-1beta, significantly enhanced NGF secretion after 24 h<strong>our</strong>s of incubation, in<br />

comparison to the treatment with either histamine or IL-1beta alone. The effect was dosedependent<br />

<strong>and</strong> additive, <strong>and</strong> was effectively blocked by H1R antagonists/inverse agonists<br />

(mepyramine <strong>and</strong> triprolidine), PKC inhibitors (GF 109203X <strong>and</strong> Go 6976 which is Ca 2+<br />

dependent, PKC-alpha isoform selective inhibitor) <strong>and</strong> MAPK kinase inhibitor (PD 98059),<br />

but they have no effect on NGF secretion, stimulated by IL-1beta alone. Simultaneous<br />

treatment of the cells with histamine <strong>and</strong> IL-6 did not show any interaction in the stimulation<br />

of NGF secretion after 24 h<strong>our</strong>s of incubation. On the contrary, significant enhancement of<br />

NGF secretion, in comparison to the treatment with either histamine or IL-6 alone, occurred<br />

after 48 h<strong>our</strong>s of incubation, which indicates long-lasting interactions between histamine <strong>and</strong><br />

IL-6. Using H1R antagonists, PKC inhibitors <strong>and</strong> MAPK kinase inhibitor they were also able<br />

to effectively block the enhancement of NGF secretion after 48 h<strong>our</strong>s of incubation. Our<br />

study suggests that activation of histamine H1R-Ca 2+ -dependent PKC-MAPK signalling<br />

pathway plays a crucial role not only in direct stimulation of NGF secretion by histamine, but<br />

also in indirect stimulation, via different types of interactions between histamine, IL-1beta<br />

<strong>and</strong> IL-6. Thus, H1R could be important therapeutic target in the treatment of certain<br />

neurodegenerative diseases.<br />

1. Lipnik-!tangelj M, "arman-Kr#an M. Histamine <strong>and</strong> IL-6 interaction in the stimulation of<br />

nerve growth factor secretion from cultured astrocytes. Inflamm Res 2005; 54(Suppl 1):S36-7.<br />

2. Lipnik-!tangelj M, "arman-Kr#an M. Histamine-stimulated nerve growth factor secretion<br />

from cultured astrocytes is blocked by protein kinase C inhibitors. Inflamm Res 2004; 53:<br />

S57-8.<br />

3. Lipnik-!tangelj M, "arman-Kr#an M. Activation of histamine H1-receptor enhances<br />

neurotrophic factor secretion from cultured astrocytes. Inflamm Res 2004; 53: 245-52.<br />

4. Lipnik-!tangelj M, "arman-Kr#an M. Pharmacological modulation of NGF secretion from rat<br />

cortical astrocytes in primary culture by histamine H1-H2-<strong>and</strong>-H3-receptor lig<strong>and</strong>s.<br />

Pharmacologist 2002; 44(2 Suppl 1):A110.<br />

5. Lipnik-!tangelj M, Juri$ DM, "arman-Kr#an M. Histamine-induced synthesis <strong>and</strong> secretion<br />

of nerve growth factor from astrocytes. Inflamm Res 1998; 47(Suppl 1):S34-5.<br />

- 67 -


Loss of the Antiproliferative Gene BTG2 in Estrogen Receptor Positive Breast<br />

Carcinoma is Associated with Tumor Grade <strong>and</strong> Overexpression of Cyclin D1 Protein<br />

Hirofumi Kawakubo, Elena Brachtel, Paul Walden <strong>and</strong> Shyamala Maheswaran<br />

Department of Surgical Oncology, Massashusetts General Hospital, Boston, MA 02114<br />

The B-cell Translocation Gene-2 (BTG2) is present in the epithelium of many tissues<br />

including the mammary gl<strong>and</strong> where its expression is regulated during gl<strong>and</strong>ular proliferation<br />

<strong>and</strong> differentiation in pregnancy. Estrogen <strong>and</strong> progestin suppress BTG2 expression<br />

suggesting that these steroids, which stimulate proliferation <strong>and</strong> lobuloalveolar development<br />

of mammary epithelial cells, may down-regulate BTG2 in the mammary gl<strong>and</strong>. Expression<br />

analysis of BTG2 protein in breast cancer revealed the loss of nuclear expression in 46% of<br />

tumors whereas it was readily detectable in the nuclei of adjacent normal gl<strong>and</strong>s. Loss of<br />

BTG2 in estrogen receptor positive breast tumors correlated significantly with increased<br />

histological grade <strong>and</strong> tumor size. Consistent with its ability to inhibit cyclin D1 transcription,<br />

suppression of BTG2 mRNA in the mammary gl<strong>and</strong> during gestation, <strong>and</strong> by estrogen <strong>and</strong><br />

progestin correlated with stimulation of cyclin D1. In estrogen receptor positive breast<br />

carcinomas, loss of nuclear BTG2 expression demonstrated a significant correlation with<br />

cyclin D1 overexpression suggesting that BTG2 may be a factor involved deregulating cyclin<br />

D1 expression in breast cancer. Moreover, cyclin D1 reversed BTG2-mediated inhibition of<br />

breast cancer cell growth indicating a functional antagonism between the two proteins. Thus<br />

coordinated inhibition of BTG2 <strong>and</strong> enhancement of cyclin D1 would constitute a mechanism<br />

to increase estrogen-responsiveness <strong>and</strong> proliferation in breast cancer cells.<br />

1. Kawakubo, H., Carey, J. L., Brachtel, E., Gupta, V., Green, J. E., Walden, P. D., <strong>and</strong> Maheswaran, S. (2004).<br />

Expression of the NF-kappaB-responsive gene BTG2 is aberrantly regulated in breast cancer. Oncogene 23,<br />

8310-8319.<br />

2. Kawakubo, H., Brachtel, E., Kish, J., Muzikansky, A., Gupta, V., Walden, P. D., <strong>and</strong> Maheswaran, S. (2005).<br />

Loss of BTG2 in estrogen receptor positive breast carcinoma is associated with tumor grade <strong>and</strong> overexpression<br />

of cyclin D1 protein. Oncogene.<br />

3. Hoshiya, Y., Gupta, V., Kawakubo, H., Brachtel, E., Carey, J. L., Sasur, L., Scott, A., Donahoe, P. K., <strong>and</strong><br />

Maheswaran, S. (2003a). Mullerian inhibiting substance promotes interferon gamma-induced gene expression<br />

<strong>and</strong> apoptosis in breast cancer cells. J Biol Chem 278, 51703-51712.<br />

4. Gupta, V., Carey, J. L., Kawakubo, H., Muzikansky, A., Green, J. E., Donahoe, P. K., MacLaughlin, D. T.,<br />

<strong>and</strong> Maheswaran, S. (2005). Mullerian inhibiting substance suppresses tumor growth in the C3(1)T antigen<br />

transgenic mouse mammary carcinoma model. Proc Natl Acad Sci U S A 102, 3219-3224.<br />

5. Segev, D. L., Ha, T. U., Tran, T. T., Kenneally, M., Harkin, P., Jung, M., MacLaughlin, D. T.,<br />

Donahoe, P. K., <strong>and</strong> Maheswaran, S. (2000). Mullerian inhibiting substance inhibits breast cancer cell growth<br />

through an NFkappa B-mediated pathway. J Biol Chem 275, 28371-28379.<br />

6. Segev, D. L., Hoshiya, Y., Hoshiya, M., Tran, T. T., Carey, J. L., Stephen, A. E., MacLaughlin, D. T.,<br />

Donahoe, P. K., <strong>and</strong> Maheswaran, S. (2002). Mullerian-inhibiting substance regulates NF-kappa B <strong>signaling</strong> in<br />

the prostate in vitro <strong>and</strong> in vivo. Proc Natl Acad Sci U S A 99, 239-244.<br />

7. Segev, D. L., Hoshiya, Y., Stephen, A. E., Hoshiya, M., Tran, T. T., MacLaughlin, D. T., Donahoe, P. K., <strong>and</strong><br />

Maheswaran, S. (2001). Mullerian inhibiting substance regulates NFkappaB <strong>signaling</strong> <strong>and</strong> growth of mammary<br />

epithelial cells in vivo. J Biol Chem 276, 26799-26806<br />

- 68 -


PI3K Targeting by the Beta-GBP Cytokine. Mitogenic Input <strong>and</strong> Therapeutic Response<br />

Livio Mallucci, <strong>and</strong> Valerie Wells<br />

Cell Signaling Laboratory, Pharmaceutical Sciences Research Division, King's College<br />

London, Franklin Wilkins Building, 150 Stamford Street, London SE1 9NH, UK. Email:<br />

livio.mallucci@kcl.ac.uk<br />

Beta-GBP (beta-galactoside binding protein), a newly identified antiproliferative cytokine<br />

produced by activated CD4 + <strong>and</strong> CD8 + T cells, can enforce programmed cell death in cancer<br />

cells without harming normal cells. We show that the prime target of beta-GBP activated<br />

<strong>signaling</strong> is the p110 catalytic subunit of PI3 kinase. We find that PI3 kinase has a permissive<br />

role in controlling cell cycle <strong>and</strong> survival <strong>signaling</strong> <strong>and</strong> that high mitogenic input, whether<br />

endogenous or enforced, facilitates beta-GBP induced apoptosis via inhibition of PI3 kinase<br />

activity <strong>and</strong> consequent inhibition of Ras-ERK <strong>and</strong> Akt/protein kinase B <strong>signaling</strong>.<br />

References<br />

Mallucci L <strong>and</strong> Wells V (2005) beta-GBP: potential role in cancer therapy. Curr. Opin.<br />

Investig. Drugs 6; 1228-1233.<br />

Ravatn R et al. (2005) Circumventing multidrug resistance in cancer by beta-galactoside<br />

binding protein, an antiproliferative cytokine. Cancer Res. 65; 1631-1634.<br />

Mallucci L et al. (2003) Turning cell cycle controller genes into cancer drugs. A role for an<br />

antiproliferative cytokine (beta-GBP). Biochem. Pharmacol. 66; 1563-1569.<br />

- 69 -


Increased EGF Receptor expression <strong>and</strong> activity is induced by deregulated N-Ras <strong>and</strong><br />

may provide a therapeutic target in NF1 neurofibrosarcoma<br />

Joshua T. Dilworth, Janice M. Kraniak, Michael A. Tainsky, John J. Reiners, Jr., <strong>and</strong><br />

Raymond R. Mattingly<br />

Department of Pharmacology <strong>and</strong> Karmanos Cancer Institute, Wayne State University,<br />

Detroit, Michigan 48201, U.S.A. Email: r.mattingly@wayne.edu<br />

Neurofibromatosis Type 1 (NF1) is the most common inherited cancer predisposition<br />

syndrome. NF1 is caused by functional loss of neurofibromin (Nf1), which compromises Ras<br />

inactivation <strong>and</strong> drives deregulated Ras-dependent <strong>signaling</strong> pathways. We have investigated<br />

a panel of neurofibrosarcoma-derived cell lines <strong>and</strong> have previously demonstrated that two<br />

Nf1-deficient lines have increased basal N-Ras <strong>and</strong> ERK MAPK activation, relative to a<br />

control line that is wild-type for Nf1. We now show that Nf1-deficient cells have an increased<br />

amount <strong>and</strong> activity of the epidermal growth factor receptor (EGFR) <strong>and</strong> demonstrate a<br />

hypersensitive <strong>and</strong> prolonged response to EGF stimulation. Our studies indicate that EGFR<br />

expression is modulated as a result downstream of Ras <strong>signaling</strong>. Maintenance of elevated<br />

EGFR expression depends on serum stimulation <strong>and</strong> the MAPK pathway. Furthermore, using<br />

tetracycline-repressible expression vectors, we show that expression of constitutively active<br />

N- or K-Ras but not H-Ras is sufficient to increase EGFR expression. Inhibition of the EGFR<br />

causes a selective reduction in the proliferation of Nf1-deficient cells. Our research proposes<br />

a mechanism to explain the clinical observation that malignant Schwann cells from NF1<br />

patients overexpress the EGFR. In addition, we provide evidence that supports the EGFR as a<br />

potential pharmacological target in the treatment of this disease.<br />

DAMD170310182<br />

Recent papers ;<br />

S. Beqaj, S. Jakkaraju, R.R. Mattingly, D. Pan & L. Schuger. High RhoA activity maintains<br />

the undifferentiated mesenchymal phenotype, whereas RhoA down-regulation by laminin-2<br />

induces smooth muscle myogenesis. J. Cell Biol. 156: 893-903 (2002)<br />

V.G. Keshamouni, R.R. Mattingly & K.B. Reddy. Mechanism of 17-#-estradiol-induced<br />

ERK1/2 activation in breast cancer cells: A role for HER2 <strong>and</strong> PKC- . J. Biol. Chem. 277:<br />

22558-22565 (2002)<br />

R.R. Mattingly, R.A. Gibbs, R.E. Menard & J.J. Reiners Jr.. Potent suppression of the<br />

proliferation of A10 vascular smooth muscle cells by combined treatment with lovastatin <strong>and</strong><br />

3-allylfarnesol, an inhibitor of protein farnesyltransferase. J. Pharmacol. Exp. Ther. 303: 74 -<br />

81 (2002)<br />

H. Yang, D. Cooley, Q. Ge, R. Andrade & R.R. Mattingly. Phosphorylation of the Ras-<br />

GRF1 exchange factor at Serine-916/898 reveals activation of Ras <strong>signaling</strong> in the cerebral<br />

cortex. J. Biol. Chem. 278: 13278-13285 (2003)<br />

R.E. Menard & R.R. Mattingly. Cell surface receptors activate p21-activated protein kinase<br />

I (PAKI) via multiple Ras <strong>and</strong> PI3-kinase-dependent pathways. Cell. Signal. 15: 1099-1109<br />

(2003)<br />

R.E. Menard & R.R. Mattingly. Gbetagamma subunits stimulate p21-activated kinase1<br />

(PAK1) through activation of PI3-kinase <strong>and</strong> Akt but act independently of Rac1/Cdc42.<br />

FEBS Lett. 556: 187-192 (2004)<br />

- 70 -


D.P. Chopra, R.E. Menard, J. Januszewski & R.R. Mattingly. TNF-alpha mediated<br />

apoptosis in normal human prostate epithelial cells <strong>and</strong> tumor cell lines. Cancer Lett. 203:<br />

145-154 (2004)<br />

D.R. Yingst, K.J. Massey, N.F. Rossi, M.J. Mohanty & R.R. Mattingly. Angiotensin II<br />

directly stimulates activity <strong>and</strong> alters phosphorylation of Na,K-ATPase in rat proximal tubule<br />

with a rapid time c<strong>our</strong>se. Am. J. Physiol. 287: F713-F721 (2004)<br />

J.H. Norum, T. Methi, R.R. Mattingly & F.O. Levy. Endogenous expression <strong>and</strong> PKAdependent<br />

phosphorylation of the guanine nucleotide exchange factor Ras-GRF1 in HEK293<br />

cells. FEBS J. (formerly Eur. J. Biochem.) 272: 2304-2316 (2005)<br />

R.E. Menard & R.R. Mattingly. Active p21-activated protein kinase I (PAKI) rescues<br />

MCF10A breast epithelial cells from undergoing anoikis. Neoplasia 7: 638-645 (2005)<br />

R.R. Mattingly, J.A. Kraniak, J.T. Dilworth, P. Mathieu, B. Bealmear, J.E. Nowak, J.A.<br />

Benjamins, M.A. Tainsky & J.J. Reiners, Jr. The MEK inhibitor PD184352/CI-1040<br />

selectively induces apoptosis in malignant schwannoma cell lines. J. Pharmacol. Exp. Ther. In<br />

press (2005)<br />

S.E. Ziemba, R.R. Mattingly, M.J. McCabe Jr & A.J. Rosenpire. Inorganic mercury inhibits<br />

the activation of LAT in T cell receptor-mediated signal transduction. Toxicol. Sci. In press<br />

(2005)<br />

H. Yang & R.R. Mattingly. The Ras-GRF1 exchange factor coordinates activation of H–Ras<br />

<strong>and</strong> Rac1 to control neuronal morphology. In revision for Mol. Biol. Cell (2005)<br />

- 71 -


Pharmacological inhibitors of DISEASE-RELEVANT CYCLIN-dEPENDENT<br />

PROTEIN KINASES (CDks) <strong>and</strong> GLYCOGEN SYNTHASE KINASE -3 (GSK-3)<br />

Laurent MEIJER<br />

Cell Cycle Laboratory, Station Biologique de Roscoff, C.N.R.S., BP 74, 29682<br />

ROSCOFF cedex, Bretagne, France. E-mail: meijer@sb-roscoff.fr<br />

(http://www.sb-roscoff.fr/CyCell/)<br />

Phosphorylation represents one the most common post-translational mechanism used by cells<br />

to regulate their enzymatic <strong>and</strong> structural proteins. Frequent alteration of protein<br />

phosphorylation in human disease is the reason for an exponentially growing investment in<br />

the optimization <strong>and</strong> therapeutic evaluation of small molecular weight, pharmacological<br />

inhibitors of protein kinases, the enzymes responsible for protein phosphorylation. It is<br />

estimated that nowadays 30-35 % of drug discovery programs in the pharmaceutical industry<br />

target a protein kinase! Currently 55 kinase inhibitors are undergoing clinical evaluation<br />

against diseases such as cancers, inflammation, diabetes, neurodegeneration.<br />

Among the 518 human protein kinases we have selected a few disease-relevant kinases to<br />

screen for small molecular weight inhibitors. Among these, cyclin-dependent kinases (CDKs)<br />

regulate the cell division cycle, apoptosis, transcription, differentiation, nervous system<br />

functions, <strong>and</strong> GSK-3, an essential element of the Wnt <strong>signaling</strong> pathway, is involved in<br />

multiple physiological processes including cell cycle regulation by controlling the levels of<br />

cyclin D1 <strong>and</strong> -catenin, insulin action on glycogen synthesis, axonal outgrowth, neuronal<br />

cell death. Both families of enzymes regulate -amyloid formation <strong>and</strong> tau<br />

hyperphosphorylation, two hallmarks of Alzheimer’s disease. Inhibitors of these kinases have<br />

a strong potential to treat cancers, neurodegenerative diseases (Alzheimer’s, Parkinson’s,<br />

Nieman-Pick type III, stroke, etc…), unicellular parasites. Some of <strong>our</strong> early CDK/GSK-3<br />

inhibitors have reached the pre-clinical <strong>and</strong> clinical stages of pharmaceutical evaluation. For<br />

instance, roscovitine (CYC202, Seliciclib), is currently undergoing phase 2 clinical trials<br />

against leukaemia, lung <strong>and</strong> breast cancers, <strong>and</strong> phase 1 trials against various kidney diseases.<br />

It is undergoing pre-clinical animal evaluation against Alzheimer’s disease stroke <strong>and</strong><br />

Niemann-Pick’s disease type III. Several other families of kinase inhibitors are currently<br />

being developed in the laboratory, a special focus on the bis-indole indirubins will be<br />

provided.<br />

Over a hundred CDK inhibitors [1] <strong>and</strong> about thirty GSK-3 inhibitors [2] have been<br />

identified, among which more than forty have been co-crystallized with CDK2 or CDK5 <strong>and</strong><br />

f<strong>our</strong> with GSK-3. These co-crystal structures are extremely helpful to design further<br />

derivatives with increased potency <strong>and</strong> selectivity. These kinase inhibitors all target the ATPbinding<br />

pocket of the catalytic site of their targets. The actual selectivity of most compounds,<br />

<strong>and</strong> thus the underlying mechanism of their cellular effects, is poorly known. We have<br />

developed affinity chromatography using immobilized inhibitors as a straightforward<br />

approach to identify the actual targets of kinase inhibitors [3,4]. Results show that although<br />

some compounds are quite selective, single target products are very unlikely to be discovered.<br />

This may in fact turn out to be an advantage as cells are unlikely to develop resistance to<br />

multiple target drugs.<br />

[1] Knockaert, M., Greengard, P. <strong>and</strong> Meijer, L., 2002. Pharmacological inhibitors of cyclindependent<br />

kinases. Trends Pharmacol. Sci. 23, 417-425.<br />

- 72 -


[2] Meijer, L., Flajolet, M. <strong>and</strong> Greengard, P., 2004. Pharmacological inhibitors of glycogen<br />

synthase kinase-3. Trends Pharmacol. Sci. 25, 471-480.<br />

[3] Knockaert, M. <strong>and</strong> Meijer, L., 2002. Identifying in vivo targets of cyclin-dependent kinase<br />

inhibitors by affinity chromatography. Biochem. Pharmacol. 64, 819-825.<br />

[4] Bach, S., Knockaert, M., Lozach, O., Reinhardt, J., Baratte, B., Schmitt, S., Coburn, S.P.,<br />

Tang, L., Jiang. T., Liang, D.C., Galons, H., Dierick, J.F., Totzke, F., Schächtele, C., Lerman,<br />

A.S., Carnero, A., Wan, Y., Gray, N. <strong>and</strong> Meijer, L., 2005. Roscovitine targets: protein<br />

kinases <strong>and</strong> pyridoxal kinase. J. Biol. Chem. 280, 31208-31219.<br />

- 73 -


Real-time analysis of Jak/STAT signal transduction in single cells.<br />

Andreas Herrmann 1 , Michael Vogt 1 , Martin Mönnigmann 2 , Bernd Giese 1 , Michael<br />

Sommerauer 1 , Peter C. Heinrich 1 , Gerhard Müller-Newen 1<br />

1 Institut für Biochemie, Universitätsklinikum der RWTH Aachen, Pauwelsstraße 30,<br />

52057 Aachen, Germany. e-mail: mueller-newen@rwth-aachen.de<br />

2 Lehrstuhl für Prozesstechnik, RWTH Aachen, 52056 Aachen, Germany<br />

The Jak/STAT signal transduction pathway plays a central role in acute <strong>and</strong> chronic<br />

inflammation. The pro-inflammatory cytokine interleukin-6 (IL-6) signals through the<br />

cytokine receptor gp130. Upon activation, the receptor-associated tyrosine kinases of the<br />

Janus kinase (Jak) family phosphorylate STAT transcription factors. STAT3 (signal<br />

transducer <strong>and</strong> activator of transcription 3) is preferentially activated in response to IL-6.<br />

Activated STAT3 translocates into the nucleus where it induces target genes that exert<br />

multiple functions in inflammation <strong>and</strong> carcinogenesis.<br />

We have tagged the cytokine IL-6, its receptor gp130 <strong>and</strong> the transcription factor STAT3 with<br />

different fluorescent proteins (yellow fluorescent protein (YFP) or cyan fluorescent protein<br />

(CFP)). These fusion proteins enabled us to study lig<strong>and</strong>-binding <strong>and</strong> receptor dimerization<br />

(Giese et al. (2005) J. Cell. Sci. 118, 5129-40) as well as nuclear translocation of STAT3<br />

(Pranada et al. (2004) J. Biol. Chem. 279, 15114-23) in single cells by the use of confocal<br />

laser-scanning microscopy <strong>and</strong> advanced photo-bleaching techniques. We demonstrated that<br />

non-phosphorylated STAT3 constitutively shuttles between the cytoplasm <strong>and</strong> the nucleus.<br />

Persistent activation of STAT3 is observed in chronic inflammation <strong>and</strong> cancer. To analyze<br />

persistent activated STAT3 we cotransfected double labelled STAT3-CFP-YFP with v-Src<br />

resulting in constitutive tyrosine phosphorylation <strong>and</strong> nuclear accumulation of the fluorescent<br />

transcription factor. By bleaching selectively the YFP moiety of STAT3-CFP-YFP in one<br />

cellular compartment <strong>and</strong> by monitoring the distribution of the CFP <strong>and</strong> YFP fluorescence<br />

over time with high spatial resolution, we show that persistently activated STAT3 shuttles<br />

between cytoplasm <strong>and</strong> nucleus. Computational evaluation of the data by model-based<br />

parameter estimations revealed that activated STAT3 shuttles more rapidly than non-activated<br />

STAT3. We propose that inhibition of nucleocytoplasmic shuttling of persistently activated<br />

STAT proteins is a new target for the treatment of chronic inflammation <strong>and</strong> cancer.<br />

List of recent papers<br />

original papers 2003 -2005 (a selection)<br />

[1] Ancey, C., Küster, A., Haan, S., Herrmann, A., Heinrich, P. C., <strong>and</strong> Müller-Newen, G. (2003)<br />

A fusion protein of the gp130 <strong>and</strong> interleukin-6Ra lig<strong>and</strong>-binding domains acts as a potent interleukin-6<br />

inhibitor.<br />

J. Biol. Chem. 278, 16968-16972<br />

[2] Giese, B., Au-Yeung, C.-K., Herrmann, A., Diefenbach, S., Haan, C., Küster, A., Wortmann, S. B.,<br />

Roderburg, C., Heinrich, P. C., Behrmann, I., <strong>and</strong> Müller-Newen, G. (2003)<br />

Long-term association of the cytokine receptor gp130 <strong>and</strong> the Janus kinase Jak1 revealed by FRAP analysis.<br />

J. Biol. Chem. 278, 39205-39213<br />

[3] Haug, K., Warnstedt, M., Alekov, A. K., S<strong>and</strong>er, T., Ramirez, A., Poser, B., Maljevic, S., Hebeisen, S.,<br />

Kubisch, C., Rebstock, J., Horvath, S., Hallmann, K., Dullinger, J. S., Rau, B., Haverkamp, F., Beyenburg, S.,<br />

Schulz, H., Janz, D., Giese, B., Müller-Newen, G., Propping, P., Elger, C. E., Fahlke, C., Lerche, H., <strong>and</strong> Heils,<br />

A. (2003)<br />

Mutations in CLCN2 encoding a voltage-gated chloride channel are associated with idiopathic generalized<br />

epilepsies.<br />

Nat. Genet. 33, 527-532<br />

- 74 -


[4] Niem<strong>and</strong>, C., Nimmesgern, A., Haan, S., Fischer, P., Schaper, F., Rossaint, R., Heinrich, P. C., <strong>and</strong><br />

Müller-Newen, G. (2003)<br />

Activation of STAT3 by IL-6 <strong>and</strong> IL-10 in primary human macrophages is differentially modulated by SOCS3.<br />

J. Immunol. 170, 3263-3272<br />

[5] de Graaf, K., Hekermann, P., Spelten, O., Herrmann, A., Packman, L. C., Büssow, K., Müller-Newen,<br />

G., <strong>and</strong> Becker, W. (2004)<br />

Characterization of cyclin L2, a novel cyclin with an arginine/serine-rich (R/S) domain: Phosphorylation by<br />

Dyrk1A <strong>and</strong> colocalization with splicing factors.<br />

J. Biol. Chem. 279, 4612-4624<br />

[6] Krüger, T., Benke, D., Eitner, F., Lang, A., Wirtz, M., Hamilton-Williams, E. E., Engel, D., Giese, B.,<br />

Müller-Newen, G., Floege, J., <strong>and</strong> Kurts, C (2004)<br />

Identification <strong>and</strong> functional characterization of dendrititc cells in the healthy murine kidney <strong>and</strong> in experimental<br />

glomerulonephritis.<br />

J. Am. Soc. Nephrol. 15, 613-621<br />

[7] Herrmann, A., Sommer, U., Pranada, A. L., Giese, B., Küster, A., Haan, S., Becker, W., Heinrich, P. C.,<br />

<strong>and</strong> Müller-Newen, G. (2004)<br />

STAT3 is enriched in nuclear bodies.<br />

J. Cell Sci. 117, 339-349<br />

[8] Hebeisen, S., Biela, A., Giese, B., Müller-Newen, G., Hidalgo, P., <strong>and</strong> Fahlke, C. (2004)<br />

The role of the C-terminus in ClC chloride channel function.<br />

J. Biol. Chem. 279, 13140-13147<br />

[9] Behrmann, I., Smyczek, T., Heinrich, P. C., Schmitz-Van De Leur, H., Komyod, W., Giese, B., Müller-<br />

Newen, G., Haan, S., Haan, C. (2004)<br />

Jak subcellular localisation revisited: The exclusive membrane-localisation of endogenous Janus Kinase 1 by<br />

cytokine receptor interaction uncovers the Jak/receptor complex to be equivalent to a receptor tyrosine kinase.<br />

J. Biol. Chem. 279, 34586-35493<br />

[10] Sel<strong>and</strong>er, K. S., Li, L., L., W., Merrell, M., Dahmen, H., Heinrich, P. C., Müller-Newen, G., <strong>and</strong> Harris,<br />

K. W. (2004)<br />

Inhibition of gp130 <strong>signaling</strong> in breast cancer blocks constitutive activation of STAT3 <strong>and</strong> inhibits in vivo<br />

malignancy.<br />

Cancer Res. 64, 6924-6933<br />

[11] Pranada, A. L., Metz, S., Herrmann, A., Heinrich, P. C., <strong>and</strong> Müller-Newen, G. (2004)<br />

Real-time analysis of STAT3 nucleocytoplasmic shuttling.<br />

J. Biol. Chem. 279, 15114-15123<br />

[12] Giese, B., Roderburg, C., Sommerauer, M., Wortmann, S. B., Heinrich, P. C. <strong>and</strong> Müller-Newen, G.<br />

(2005)<br />

Dimerization of the cytokine receptors gp130 <strong>and</strong> LIFR analysed in single cells.<br />

J. Cell. Sci. 118, 5129-5140<br />

recent reviews (a selection)<br />

[R1] Heinrich, P. C., Behrmann, I., Haan, S., Hermanns, H. M., Müller-Newen, G., <strong>and</strong> Schaper, F. (2003)<br />

Principles of interleukin (IL)-6-type cytokine signalling <strong>and</strong> its regulation.<br />

Biochem. J. 374, 1-20<br />

[R2] Müller-Newen, G. (2003)<br />

The cytokine receptor gp130: faithfully promiscuous.<br />

Science STKE 2003, pe40<br />

[R3] Hermanns, H. M., Müller-Newen, G., Heinrich, P. C. <strong>and</strong> Haan, S. (2005)<br />

Bow to you partner for <strong>signaling</strong><br />

Nature Struct. Mol. Biol. 12, 476-478<br />

- 75 -


Transcriptional control of differential glucocorticoid receptor expression: determining<br />

tissue specific expression, <strong>and</strong> protein isoforms.<br />

Jonathan D. Turner 1 <strong>and</strong> Claude P. Muller 1,2<br />

1<br />

Institute of Immunology, Laboratoire National de Santé, 20A rue Auguste Lumière, L-<br />

1011, Luxemb<strong>our</strong>g<br />

2<br />

Department of Immunology, Graduate School of Psychobiology, University of Trier, D-<br />

54290, Germany<br />

Email: jonathan.turner@LNS.ETAT.LU <strong>and</strong> claude.muller@LNS.ETAT.LU<br />

The 5’ UTR of the glucocorticoid receptor plays a key role in determining tissue specific<br />

expression, <strong>and</strong> protein isoforms. Analysis of the 5’ UTR of the hGR has revealed 11 splice<br />

variants of the hGR exon 1, based on 7 exon 1s, 4 of which (1-D to 1-F <strong>and</strong> 1-H) were<br />

previously unknown. All of the exon 1 variants have unique splice donor sites <strong>and</strong> share a<br />

common exon 2 splice acceptor site. Due to an upstream in-frame TGA stop codon the<br />

predicted translation from all splice variants is identical. The f<strong>our</strong> new exon 1s show<br />

remarkable similarity with their rat homologues. Exon 1-D starts <strong>and</strong> finishes 17- <strong>and</strong> 36bp<br />

upstream of the corresponding ends of the rat exon 1 4. Exon 1-E is only 6bp longer than its<br />

homologue 1 5. Exon 1-F contains two short inserts of 11- <strong>and</strong> 6bp when compared to the rat<br />

1 7. 1-H is 18bp longer than the corresponding rat 1 11. In addition to these new exons, we<br />

found that the human exon 1-C occurs as 3 distinct splice variants, covering the region<br />

homologous to the rat exons 1 9 <strong>and</strong> 1 10. All of the alternative hGR exons 1s presented here<br />

were found to be transcribed in human tissue. The human hippocampus expresses mRNA of<br />

all the exon 1 variants, whilst the expression of the other exon 1s seems to be tissue-specific.<br />

While exon 1-D is only in the hippocampus, exons 1-E <strong>and</strong> 1-F are also detected in the<br />

immune system, <strong>and</strong> exon 1-H additionally in the liver, lung <strong>and</strong> smooth muscle. The 5’<br />

region of the hGR is more complex than previously thought, <strong>and</strong> we suggest that each of these<br />

untranslated first exons have a distinct proximal promoter region, providing additional depth<br />

to the mechanisms available for tissue specific expression of the hGR isoforms.<br />

- 76 -


Oncogenic <strong>signaling</strong> by mutant p53<br />

Moshe Oren<br />

Department of Molecular Cell Biology, The Weizmann Institute, Rehovot, Israel<br />

(no abstract provided)<br />

- 77 -


Acute anti-inflammatory properties of statins involve Peroxisome Proliferator-Activated<br />

Receptor-alpha via inhibition of the PKC signalling pathway<br />

Réjane Paumelle 1 , Christophe Blanquart 1 , Olivier Bri<strong>and</strong> 1 , Olivier Barbier 1 , Gaëtane<br />

Woerly 2 Christian Duhem 1 , Frédéric Percevault 1 , Jean-Charles Fruchart 1 , David<br />

Dombrowicz 2 , Corine Glineur 1 <strong>and</strong> Bart Staels 1,3 .<br />

1 Département d’Athérosclerose, U545 Inserm, Institut Pasteur de Lille <strong>and</strong> Faculté de<br />

Pharmacie, Université de Lille II, Lille, France. 2 Inserm U547, Institut Pasteur de Lille,<br />

Lille, France. E-mail: Bart.Staels@pasteur-lille.fr<br />

Statins are inhibitors of 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase used in the<br />

prevention of cardiovascular diseases (CVD). In addition to their cholesterol-lowering<br />

activities, statins exert pleiotropic anti-inflammatory effects, which might contribute to their<br />

beneficial effects not only on CVD, but also on lipid-unrelated immune <strong>and</strong> inflammatory<br />

diseases, such as rheumatoid arthritis, asthma, stroke <strong>and</strong> transplant rejection. However, the<br />

molecular mechanisms involved in these anti-inflammatory properties of statins are<br />

unresolved. The nuclear receptor Peroxisome Proliferator-Activated Receptor (PPAR)-alpha<br />

that is expressed in inflammation regulatory cell types, such as macrophages, exerts potent<br />

anti-inflammatory activities. In this study we present genetic evidence for a role for this<br />

nuclear receptor in mediating the lipid-independent anti-inflammatory activities of statins in<br />

two in vivo models of acute inflammation. Furthermore, the inhibitory effects of statins on<br />

lipopolysaccharide (LPS)-induced inflammatory response genes were abolished in PPARalpha-deficient<br />

macrophages <strong>and</strong> neutrophils, two cell types involved in acute inflammatory<br />

responses. Moreover, we provide a molecular mechanism explaining these observations by<br />

showing that simvastatin inhibited PPAR-alpha phosphorylation by LPS-activated PKCalpha.<br />

A constitutive active form of PKC-alpha inhibited NF-kappaB transrepression by<br />

PPAR-alpha whereas simvastatin enhanced transrepression activity of wild-type PPAR-alpha,<br />

but not of PPAR-alpha mutated in its PKC phosphorylation sites. These data indicate that the<br />

acute anti-inflammatory effect of simvastatin occurs via PPAR-alpha by a mechanism<br />

involving inhibition of PKC-alpha inactivation of PPAR-alpha transrepression activity.<br />

PUBLICATIONS<br />

- Tulasne, D., Paumelle, R., Weidner, M., V<strong>and</strong>enbunder, B. <strong>and</strong> Fafeur, V. The<br />

multisubstrate docking site of the MET receptor is dispensable for MET-mediated RAS<br />

<strong>signaling</strong> <strong>and</strong> cell scattering. 1999. Mol. Biol. Cell. 10, 551-565.<br />

- Paumelle, R., Tulasne, D., Leroy, C., Coll, J., V<strong>and</strong>enbunder, B. <strong>and</strong> Fafeur, V. Sequential<br />

activation of ERK <strong>and</strong> repression of JNK by SF/HGF in MDCK epithelial cells. 2000. Mol.<br />

Biol. Cell. 11, 3751-3763.<br />

- Paumelle, R., Tulasne, D., Kherrouche, Z., Plaza, S., Leroy, C., Reveneau, S.,<br />

V<strong>and</strong>enbunder, B. And Fafeur, V. Hepatocyte growth factor/scatter factor activates the ETS1<br />

transcription factor by a RAS-RAF-MEK-ERK <strong>signaling</strong> pathway. 2002. Oncogene. 21:<br />

2309-2319.<br />

- Blanquart, C., Mans<strong>our</strong>i, R., Paumelle, R., Fruchart, J. C., Staels, B. The protein kinase C<br />

signalling pathway regulates a molecular switch between transactivation <strong>and</strong> transrepression<br />

- 78 -


activity of the Peroxisome Proliferator-Activated Receptor alpha (PPAR{alpha}). 2004. Mol<br />

Endocrinol.18(8):1906-18.<br />

-Teissier, E., Nohara, A., Chinetti, G., Paumelle, R., Cariou, B., Fruchart, J.C., Br<strong>and</strong>es, R.,<br />

Shah, A., Staels, B. Peroxisome proliferator-activated receptor induces NADPH oxydase<br />

activity in macrophages leading to the generation of LDL with PPAR activation properties.<br />

2004. Circ. Res. 95(12):1174-82.<br />

- Paumelle, R., Blanquart, C., Bri<strong>and</strong>, O., Barbier, O, Woerly, G, Duhem, C., Percevault, F.,<br />

Fruchart, J. C., Dombrowicz, D., Glineur, C., <strong>and</strong> Staels, B. Acute anti-inflammatory<br />

properties of statins involve Peroxisome Proliferator-Activated Receptor-alpha via<br />

inhibition of the PKC signalling pathway. In revision in Circ. Res.<br />

- 79 -


Cyclins <strong>and</strong> HDAC/HATs<br />

Richard Pestell<br />

Lombardi Comprehensive Cancer Center, Georgetown University, Washington, USA<br />

(no abstract provided)<br />

- 80 -


Restauration of SHIP-1 activity in human leukemic cells modifies NF-!B activation<br />

pathway <strong>and</strong> cellular survival upon oxidative stress treatment<br />

Geoffrey Gloire*, Edith Charlier*, Souad Rhamouni*, Cédric Volanti*, Alain Chariot*,<br />

Christophe Erneux $ <strong>and</strong> Jacques Piette*<br />

*University of Liège, B-4000 Liège, $ University of Brussels, B-1070 Brussels<br />

Since the discovery that NF-!B 1 could be activated by H 2O 2, several laboratories put a lot of<br />

efforts into dissecting the molecular mechanisms underlying this activation, particularly in<br />

Jurkat leukemic cells. These works led to the observation that NF-!B activation by H 2O 2<br />

involved tyrosine phosphorylation of the inhibitor I!B" through an IKK-independent<br />

mechanism. In the present work, we investigated with more details NF-!B redox regulation<br />

in human leukemic cells. By using different cell lines (CEM, Jurkat <strong>and</strong> Jurkat JR), we clearly<br />

showed that NF-!B activation by H 2O 2 is cell-type dependent: it activates NF-!B through<br />

tyrosine phosphorylation of I!B" in Jurkat cells, but induces an IKK-mediated IkB"<br />

phosphorylation on serines 32 <strong>and</strong> 36 in CEM <strong>and</strong> Jurkat JR cells. We showed that this H 2O 2induced<br />

IKK activation in CEM <strong>and</strong> Jurkat JR cells is mediated by SHIP-1, a lipid<br />

phosphatase which is absent in Jurkat cells. Indeed, the genetic complementation of SHIP-1 in<br />

Jurkat cells made them shifting to an IKK-dependent mechanism upon oxidative stress<br />

stimulation. The phosphatase domain of SHIP-1 is crucial in that process, since the<br />

complementation of Jurkat cells with a mutant of SHIP-1 lacking catalytic activity failed to<br />

mediate IKK activation upon H 2O 2 treatment. We also showed that Jurkat expressing SHIP-1<br />

are much more resistant to H 2O 2-induced apoptosis than the parental cells, suggesting that<br />

SHIP-1 have an important role in protecting leukemic cells from ROS-induced apoptosis.<br />

Overall, <strong>our</strong> study highlight a novel role for SHIP-1 in leukemic cells ROS responses in terms<br />

of signal transduction pathways <strong>and</strong> apoptosis resistance, which can be of interest in<br />

improving ROS-mediated chemotherapies.<br />

- 81 -


Live <strong>and</strong> let die by 73 - mechanisms of degradation <strong>and</strong> chemoresistance<br />

Michael J. Pinkoski<br />

MRC Toxicology Unit<br />

Leicester, United Kingdom<br />

p73 belongs to a family of transcription factors, including p53 <strong>and</strong> p63, that mediates<br />

responses to DNA damage <strong>and</strong> cellular stress by inducing DNA repair, cell cycle arrest <strong>and</strong><br />

apoptosis. p73 is expressed as two isoforms, TA <strong>and</strong> DN, that have opposing biological<br />

effects with DNp73 has a dominant negative/repressive effect on TAp73. Therefore, TA/DN<br />

ratios dictate p73-dependent cellular responses to such insults as genotoxic chemotherapeutic<br />

agents. In fact, DNp73 is associated with reduced survival in hepatocellular carcinoma (HCC)<br />

patients <strong>and</strong> is therefore an indicator of a negative prognosis. Mechanistically, p73 is capable<br />

of engaging both intrinsic <strong>and</strong> extrinsic apoptosis pathways. TAp73 induces transcription of<br />

CD95 <strong>and</strong> increased sensitivity to CD95-induced apoptosis. TAp73 also induces expression of<br />

PUMA, Noxa <strong>and</strong> Scotin to engage death pathways via the mitochondria <strong>and</strong> apoptosome.<br />

Unlike p53, p73 mutations are rarely observed in human cancers, however, p73 activity is<br />

influenced by protein stability. p73 protein degradation is regulated by the E3 ubiquitin ligase,<br />

Itch, whereas stabilization of p73 protein is positively regulated by the nuclear body protein,<br />

PML. Both TA <strong>and</strong> DNp73 proteins are degraded in an Itch dependent manner, but<br />

interestingly, both Itch <strong>and</strong> DNp73 are downregulated following genotoxic stress.<br />

Degradation of DNp73 under these conditions is not mediated by Itch. TAp73, is not<br />

decreased following DNA damage, lending further support to the hypothesis TAp73 acts as a<br />

tum<strong>our</strong> suppressor while DNp73, which can inhibit both TAp73 <strong>and</strong> p53 would represent an<br />

oncogenic activity. Therefore, in addition to DNp73 as a prognostic indicator, these data<br />

suggest that DNp73 <strong>and</strong> Itch are potential targets for cancer therapy.<br />

- 82 -


Hypoxia Signaling. Impact on Tumor metabolism, Cell survival <strong>and</strong> Cell death.<br />

N. M. Mazure. F. Dayan, D. Roux, E. Berra, C. Brahimi-Horn, <strong>and</strong> J. Pouysségur<br />

Institute of Signaling, Developmental Biology <strong>and</strong> Cancer Research, CNRS-UMR 6543,<br />

Centre Antoine Lacassagne, 33 Avenue de Valombrose, 06189 Nice, France<br />

<br />

The function of the Hypoxia-Inducible Factor-1 (HIF-1), the key transcription factor involved<br />

in cellular adaptation to hypoxia is restricted to low oxygen tension (pO 2). As such, this<br />

transcription factor is central in modulating the tumor microenvironment, sensing nutrient<br />

availability <strong>and</strong> controlling anaerobic glycolysis, intracellular pH, <strong>and</strong> cell survival.<br />

Degradation <strong>and</strong> inhibition of the limiting HIF-1" subunit are intimately connected in<br />

normoxia. Hydroxylation of two proline residues by Prolyl Hydroxylase Domain protein 2<br />

(PHD2) earmarks the protein for degradation while hydroxylation of an asparagine residue by<br />

Factor Inhibiting HIF-1 (FIH-1) reduces its transcriptional activity. Indeed, silencing in<br />

normoxia, of either PHD2 or FIH-1, partially induced hypoxic genes, whereas combined<br />

PHD2/FIH-1 silencing generated a full hypoxic gene response. Given the fact that HIF-1"<br />

possesses two transactivation domains (N- <strong>and</strong> C-TAD), we hypothesized on a possible bifunctional<br />

activity of HIF-1a that could be discriminated by FIH-1, a modifier of the C-TAD.<br />

In human cell lines, engineered to overexpress or silence FIH-1, in response to tetracyclin, we<br />

demonstrate by quantitative RT-PCR that a set of hypoxic genes (CAIX, PHD3, PGKI <strong>and</strong><br />

BNIP3) respond differently towards FIH-1 expression. This finding, extended to 30 hypoxiainduced<br />

genes, indicates differential gene expression by the N- <strong>and</strong> C-TAD in response to the<br />

hypoxic gradient.<br />

We propose that the oxygen-sensitive attenuator FIH-1, together with 2 distinct TADs are<br />

central in setting the gene expression repertoire dictated by the cell pO 2.<br />

In the tumor microenvironment context, we are intrigued by the duality of HIF-1 that can<br />

induce either cell survival or cell death. We will discuss <strong>our</strong> unpublished work on the<br />

physiological roles played by the HIF-induced pro-apoptotic gene product BNIP3, autophagy<br />

<strong>and</strong> necrotic cell death.<br />

Ways to exploit this basic knowledge to magnify tumor regression will be presented ?<br />

- 83 -


Alkylating drugs applied in non-cytotoxic doses as a novel compounds targeting<br />

inflammatory signal pathways<br />

Alex<strong>and</strong>er L. Pukhalsky, Galina V. Shmarina, Vladimir A. Alioshkin <strong>and</strong> Alex<br />

Sabelnikov<br />

Research Centre for Medical Genetics, Moscow 115478, Gabrichevsky Institute of<br />

Epidemilogy <strong>and</strong> Microbilogy, Moscow 125212, Russia, East Carolina University,<br />

Greenvill, NC 27858, U.S.A. E-mail: osugariver@medgen.ru<br />

Alkylating drugs (ADs) belonging to the nitrogen mustard family are commonly used as<br />

cytostatic <strong>and</strong> immunosuppressive agents. Our recent in vitro studies demonstrated that in the<br />

case of gradual dose decrease, the number of targets for alkylation in the cell is also reduced<br />

<strong>and</strong> the drug switches from crucial cytostatic to cell growth modifier. At doses of 0.3 mkg/ml<br />

<strong>and</strong> lower, the effects of ADs are no longer associated with DNA damage or altered cytokine<br />

production. Instead, the disruption of signal transduction by the IL-2beta <strong>and</strong>/or TNF-alpha<br />

cell surface receptors is observed. As a result ADs in the doses 100-fold lower than cytostatic<br />

ones are capable to modify lymphocyte activity including the activity of regulatory T cells.<br />

We hypothesized that ADs may have a beneficial effect in the treatment of inflammatory<br />

diseases. Indeed, the application of non-cytotoxic doses of an AD melphalan (MP) reduces<br />

the severity of murine experimental colitis induced by the replacement of drinking water with<br />

5% solution of dextran sulphate sodium (DSS). Daily administration of MP (0.025 mg/kg<br />

body weight) markedly reduced the severity of DSS-colitis as determined by clinical <strong>and</strong><br />

quantitative histological criteria. Moreover, the beneficial effect of MP was also shown in<br />

asthmatic patients. Forty-two patients with moderate <strong>and</strong> severe asthma were enrolled in<br />

r<strong>and</strong>omized placebo-controlled trial: 21 patients were treated with MP (5 daily inhalation of<br />

the drug in the dose of 0.1 mg) <strong>and</strong> 21 subjects were inhaled with placebo. Improvement of<br />

exercise tolerance as well as a marked reduction of exacerbation severity <strong>and</strong> requirement for<br />

inhaled beta2-agonists has been found in MP-treated patients. In 60% of these patients<br />

histological <strong>and</strong> ultrastructural signs of bronchial epithelium regeneration were also revealed.<br />

Thus, ADs at non-cytotoxic concentrations exert beneficial effect both in acute <strong>and</strong> chronic<br />

inflammatory diseases. Such anti-inflammatory activity is thought to be due to blocking of<br />

signal transduction through various cytokine surface receptors. Consequently, different steps<br />

of inflammatory cascade turn up inhibited.<br />

List of publications<br />

1. Toptygina AP, Pulhalsky AL, Alioshkin VA. Immunoglobulin G subclass profile of antmeasles<br />

response in vaccinated children <strong>and</strong> in adults with measles history. Clin Diagn Lab<br />

Immunol, 2005; 12: 845-847.<br />

2. Pukhalsky AL, Shmarina GV, Kapranov NI, Kokarovtseva SN, Pukhalskaya DA,<br />

Kashirskaja NJ Anti-inflammatory <strong>and</strong> immunomodulating effects of clarithromycin in<br />

patients with cystic fibrosis lung disease. Med Inflam 2004; 13:111-117.<br />

3. Pukhalsky AL, Shmarina GV, Bliakher MS, Fedorova IN, Toptygina AP, Fisenko JJ,<br />

Alioshkin VA Cytokine profile after rubella vaccine inoculation: evidence of<br />

immunosuppressive effect of vaccination. Med Inflam 2003, 12(4): 203-207.<br />

4. Sokolov EI, Pukhalsky AL, Tsyplenkova VG, Shevelev VI. Inhalation of ultra-low<br />

doses of alkylating drugs in the treatment of asthma. Pulmonology 2002; 12(3):82-88<br />

(Russian).<br />

- 84 -


5. Sokolov EI, Zykov KA, Pukhalsky AL, Zykov KA, Shmarina GV, Matko LY, Demidov<br />

YI, Kokarovtseva SN, Shevelev VI. Immunomodulating effects of inhaled alkylating drug<br />

melphalam in asthmatic patients. Pulmonology 2002; 12(5):81-86 (Russian).<br />

6. Pukhalsky AL, Shmarina GV. Stimulatory <strong>and</strong> protective effects of alkylating agents<br />

applied in ultra-low concentrations. Pharmacology 2001; 62: 129-132.<br />

7. Shmarina GV, Pukhalsky AL, Kokarovtseva SN, Pukhalskaya DP, Kalashnikova EA,<br />

Kapranov NI, Kashirskaja SJ. Improvement of nutrient absorption may enhance systemic<br />

oxidative stress in cystic fibrosis patients. Med Inflam 2001; 10: 61-67.<br />

8. Shmarina GV, Pukhalsky AL, Kokarovtseva SN, Pukhalskaya DP, Shabalova LA,<br />

Kapranov NI, Kashirskaja SJ. Tumor necrosis factor-"/interleukin-10 balance in normal <strong>and</strong><br />

cystic fibrosis children. Med Inflam 2001; 10: 191-197.<br />

9. Pukhalsky AL, Kapranov NI, Kalashnikova EA, Shmarina GV, Shabalova LA,<br />

Kokarovtseva SN, Pukhalskaya DA, Kasirskaja NJ, Simonova OI. Inflammatory markers in<br />

cystic fibrosis patients with lung Pseudomonas aeruginosa infection. Med Inflam 1999; 8:<br />

159-167.<br />

10. Pukhalsky AL, Shyian SD, Kalashnikova EA, Shmarina GV, Pukhalskaya DA, Bovin<br />

NV Immunomodulating activities of a natural " 1-acid glycoprotein <strong>and</strong> its carbohydrate<br />

chains attached to the protein-free polymer. Med Inflam 1998, 7:115-118.<br />

11. Pukhalsky AL, Vikulova VK, Toptygina AP, Lyashko VN. Genetic heterogeneity of<br />

heat shock protein synthesis <strong>and</strong> the spleen cell sensitivity to antiproliferative effect of<br />

alkylating agents in mice. Bull Exp Biol 1997; 124:561-565.<br />

12. Pukhalsky A, Toptygina A, Khaidukov S. Interleukin-2 receptor # chain as a possible target for low doses<br />

of mafosfamide. Mediators Inflamm 1995; 4:175-180.<br />

13. Pukhalsky AL, Toptygina AP. The comparative study of mechanisms of<br />

antiproliferative effect of mafosfamide <strong>and</strong> cyclosporin A applied in low doses. Bull Exper<br />

Biol 1993; 115 (5): 514-515.<br />

14. Pukhalsky AL, Toptygina AP, Viktorov VV. Immunosuppressive action of cyclophosphamide in mice:<br />

contribution of some factors to determination of strain differences. Int J Immunopharmac 1993; 15: 509-514.<br />

15. Pukhalsky AL, Toptygina AP, Viktorov VV. Pharmacokinetics of alkylating metabolites<br />

of cyclophosphamide in different strains of mice. Int J Immunopharmac 1990; 12:217-223.<br />

16. Yushkov SF, Pukhalsky AL, Rozenberg IB. The effect of sarcolysin <strong>and</strong> endoxan on the<br />

resorption of the tadpoles’ tail muscles in metamorphosis. Pharmacol Toxicol 1970; 6:723-<br />

726 (Russian).<br />

- 85 -


REGULATION OF INFLAMMATION AND GLUCOCORTICOID SIGNALING BY<br />

CURCUMIN AND RESVERATROL<br />

Dr. Irfan Rahman<br />

Department of Environmental Medicine, University of Rochester Medical Center,<br />

Rochester, NY. Email: irfan_rahman@urmc.rochester.edu<br />

Reactive oxygen species (ROS) play a key role in enhancing the inflammation through the<br />

activation NF-kappaB <strong>and</strong> AP-1 transcription factors, <strong>and</strong> nuclear histone acetylation <strong>and</strong><br />

deacetylation (chromatin remodeling) in various inflammatory diseases. We have recently<br />

shown that oxidative stress enhances lung inflammation via expression of pro-inflammatory<br />

mediators through inhibition of histone deacetylase activity <strong>and</strong> NF-kappaB transactivation in<br />

monocytes/macrophages <strong>and</strong> epithelial cells. We also show the antioxidant <strong>and</strong>/or antiinflammatory<br />

effects of dietary polyphenols (curcumin-diferuloylmethane, a principal<br />

component of turmeric) <strong>and</strong> resveratrol (a flavanoid found in red wine), in the control of NFkB<br />

activation, upregulation of glutathione biosynthesis gene via Nrf2 activation, scavenging<br />

effect of ROS by inducing glutathione peroxidase activity, chromatin remodeling <strong>and</strong><br />

subsequently inflammatory gene regulation in macrophages <strong>and</strong> lung epithelial cells. We<br />

further demonstrate that these dietary polyphenols restore glucocorticoid functions (antiinflammatory<br />

property) in response to oxidative stress by upregulation of histone deacetylase<br />

activity. The anti-inflammatory property of curcumin <strong>and</strong> resveratrol may be due to its ability<br />

to induce histone deacetylase activity by reversing the post-translationally modified proteins.<br />

These data provide new information on regulation of inflammatory response by dietary<br />

polyphenols at the molecular level <strong>and</strong> possibly a way forward to inhibit chronic<br />

inflammatory response in various diseases.<br />

- 86 -


HOW ANTITOXIC AND ANTICANCER AGENTS MANEUVER PROGRAMMED<br />

AND UNPROGRAMMED CELL DEATH IN VIVO?<br />

Sidhartha D. Ray<br />

Molecular Toxicology Laboratories, Division of Pharmaceutical Scs,<br />

Arnold & Marie Schwartz College of Pharmacy & Health Sciences, Long Isl<strong>and</strong><br />

University, Brooklyn, NY 11201<br />

Most pro-toxic <strong>and</strong> pro-carcinogenic signals fundamentally share a common platform <strong>and</strong> use<br />

genome-dependent <strong>and</strong> genome-independent events to deregulate the intracellular<br />

microenvironment to ultimately articulate macroscopic changes in vivo. Pro-death signals<br />

tend to perturb the level of oxidative stress (deplete glutathione & induce lipid peroxidation),<br />

stimulate mitochondrial pore formation (release Cyt c), tilt antioxidant balance, jeopardize<br />

energy metabolism (drop ATP production) <strong>and</strong> mishmash ion homeostasis to achieve their<br />

goals. At the molecular level, various cytokines (TNF- ) <strong>and</strong> cell-death controlling elements<br />

(caspases, bcl-XL, p53) dominate in masterminding programmed or unprogrammed cell<br />

death. Using in vivo models, <strong>our</strong> decades of research have shown that select single entity<br />

phytochemicals or a combination of phytochemicals can rescue cells from such chaotic<br />

aftermath <strong>and</strong> propagate antitoxic <strong>and</strong> anticarcinogenic signals in favor cell survival. Using a<br />

carcinogen <strong>and</strong> a non-carcinogen, <strong>our</strong> laboratory has investigated the patterns of programmed<br />

<strong>and</strong> unprogrammed cell death <strong>and</strong> their counteraction by a series of phytochemicals<br />

(curcumin, silymarin, hesperidin, rutin) in the liver in vivo. Regulated pre-exposure (days to<br />

weeks) or the continuous presence (months) of these phytochemicals in vivo carefully<br />

propagate target-specific anti-toxic <strong>and</strong> anti-carcinogenic signals creating a potential sink for<br />

the processes that orchestrate apoptotic/necrotic/apocrotic cell deaths. Although activation or<br />

inhibition of Caspases, select expression of pro <strong>and</strong> anti-apoptotic genes, modulation of DNA<br />

damage/DNA repair processes <strong>and</strong> maintenance of pro- <strong>and</strong> antioxidant balance are the<br />

ultimate key to survival signals, modulation of fundamental biochemical events serve as the<br />

basis in shaping/micromanaging anti-toxic <strong>and</strong>/or anti-carcinogenic outcomes. This talk will<br />

emphasize local <strong>and</strong> global changes (of the parameters described above) at the cellular,<br />

organellar, <strong>and</strong> molecular levels to underst<strong>and</strong> how cytoprotective agents selectively<br />

disseminate anti-toxic <strong>and</strong>/or anti-carcinogenic signals to trigger specific molecular switches<br />

in vivo prior to hepatotoxic or hepatocarcionogenic reactions <strong>and</strong> obliterate lethal<br />

consequences.<br />

- 87 -


Inhibition of histone deacetylase activity as a therapeutic tool in amyotrophic lateral<br />

sclerosis.<br />

Caroline ROUAUX, Irina PANTELEEVA, Frédérique RENE, Jose-Luis GONZALEZ<br />

de AGUILAR, Anne-Laurence BOUTILLIER & Jean-Philippe LOEFFLER<br />

Laboratoire de Signalisation Moléculaire et Neurodégénérescence – INSERM U692 -<br />

Faculté de Médecine - 11, rue Humann - 67085 Strasb<strong>our</strong>g Cedex FRANCE. Tel.: (+33)<br />

390 24 30 88 Fax: (+33) 390 24 30 65.<br />

Motoneuronal death in the spinal cord is a characteristic of Amyotrophic Lateral Sclerosis<br />

(ALS). Our recent data showed a decrease in histone acetylation levels, as well as in protein<br />

levels of the CREB-Binding Protein (CBP), a specific Histone Acetyl Transferase (HAT), in<br />

motoneurons from the lumbar spinal cord of an in vivo mouse model of ALS (SOD1 G86R<br />

mice) when compared to those of wild type animals. The aim of this study was to investigate<br />

whether preventing the loss of acetylation by blocking histone deacetylases with specific<br />

inhibitors (HDACi) could have an impact on motoneuronal death. Therefore, SOD1 G86R<br />

mice were treated with the HDACi Sodium Valproate (VPA) at 250mg/kg/j, IP. VPA<br />

treatment induced a delay in the onset of the symptoms, an increase in motor performances<br />

<strong>and</strong> allowed to significantly reduce motoneuronal death. To study the mechanism of action of<br />

this HDACi, we used a cellular model of motoneurons (the NSC34 neuroblastoma X Spinal<br />

cord cell line) in which oxidative-stress induced-death was mimicked with a pulse of<br />

hydrogen peroxide (H 2O 2). H 2O 2-induced motorneuronal death was accompanied by histone<br />

deacetylation <strong>and</strong> CBP loss. Moreover, this loss resulted from a direct cbp gene repression.<br />

Several HDACi (Trichostatin A: TSA, Sodium Valproate: VPA <strong>and</strong> Sodium Butyrate: NaBu)<br />

were tested. We found that a long-term treatment with each compound had a significant<br />

protective effect on H 2O 2-induced motoneuronal death, reversing histone acetylation decrease<br />

<strong>and</strong> promoting cbp gene expression. Consequently, despite the effects of HDACi on the<br />

neuromuscular junction remain to be elucidated, we believe that their beneficial effects in the<br />

ALS model may result from a protection against motoneurons degeneration.<br />

Rouaux et al., 2003, EMBO 22(24):6537-49<br />

Rouaux et al., 2004, Biochem Pharmacol. 2004, 68(6):1157-64.<br />

Supported by AFM <strong>and</strong> ARS.<br />

- 88 -


Listeria monocytogenes induced Rac1-dependent endothelial histone modification <strong>and</strong><br />

cytokine release<br />

Bernd Schmeck*, Wiebke Beermann*, Vincent van Laak*, Bastian Opitz*, Andreas<br />

Hocke*, Julia Eitel*, Trinad Chakraborty+, Gudula Schmidt#, Holger Barth#, Norbert<br />

Suttorp*, <strong>and</strong> Stefan Hippenstiel*<br />

*) Department of Internal Medicine/Infectious Diseases, Charité – Universitätsmedizin<br />

Berlin, 13353 Berlin, Germany (E-mail: Bernd.Schmeck@charite.de); +) Institute for<br />

Medical Microbiology, Justus-Liebig University of Giessen, 35392 Giessen, Germany; #)<br />

Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-<br />

Ludwig-Universitat Freiburg, 79104 Freiburg, Germany.<br />

Infection of endothelial cells by Listeria monocytogenes is an essential step in the<br />

pathogenesis of listeriosis. Small GTPases of the Rho family act as molecular switches in<br />

signal transduction. We tested the hypothesis that Rho GTPases contribute to the regulation of<br />

cytokine expression in Listeria monocytogenes infection.<br />

L. monocytogenes induced release of distinct CC <strong>and</strong> CXC, as well as Th1 <strong>and</strong> Th2 cytokines<br />

<strong>and</strong> growth factors by endothelial cells whereas non-invasive L. innocua did not induce<br />

cytokine release <strong>and</strong> activated Rac1 as shown by pulldown assay. Cytokine expression was<br />

reduced byinhibition of RhoA, Rac <strong>and</strong> Cdc42 (Clostridium difficile toxin B (TcdB)), <strong>and</strong><br />

Rac1-inhibitor Nsc23766. Activating Rho proteins by Escherichia coli CNF1 increased<br />

Listeria-dependent cytokine expression. We analyzed regulation of IL-8 expression in more<br />

detail: Listeria-induced IL-8 release was reduced by inhibition of RhoA, Rac1 <strong>and</strong> Cdc42<br />

(TcdB) or Rac1while blocking of RhoA/B/C by Clostridium limosum C3 fusion toxin or Rho<br />

kinase by Y27632 had no effect. Activation of RhoA, Rac <strong>and</strong> Cdc42 (CNF1), but not of<br />

RhoA alone (CNF Y), enhanced Listeria-dependent IL-8 release. Next, we analyzed histone<br />

modifications at the gene-promoter of IL-8. Inhibition of RhoA, Rac1 <strong>and</strong> Cdc42 (TcdB) <strong>and</strong><br />

Rac1 (Nsc23766) reduced Listeria-related recruitment of NF-kappaB/p65 <strong>and</strong> RNA<br />

Polymerase II to the IL-8 promoter, as well as Lys-8-acetylation of histone H4 <strong>and</strong> Ser-<br />

10/Lys-14-phosphorylation/acetylation of histone H3 at the IL-8 gene promoter in HUVEC.<br />

In conclusion, Rac1 contributed to epigenetic regulation of L. moncytogenes-induced<br />

cytokine expression by human endothelial cells.<br />

Recent papers:<br />

1: Opitz B, Puschel A, Beermann W, Hocke AC, Forster S, Schmeck B, van Laak V,<br />

Chakraborty T, Suttorp N, Hippenstiel S. Listeria monocytogenes Activated p38 MAPK <strong>and</strong><br />

Induced IL-8 Secretion in a Nucleotide-Binding Oligomerization Domain 1-Dependent<br />

Manner in Endothelial Cells. J Immunol. 2006 Jan 1;176(1):484-90.<br />

2: Brell B, Hippenstiel S, David I, Pries AR, Habazettl H, Schmeck B, Suttorp N,<br />

Temmesfeld-Wollbruck B.<br />

Adrenomedullin treatment abolishes ileal mucosal hypoperfusion induced by Staphylococcus<br />

aureus alpha-toxin--an intravital microscopic study on an isolated rat ileum. Crit Care Med.<br />

2005 Dec;33(12):2810-016.<br />

3: Krull M, Bockstaller P, Wuppermann FN, Klucken AC, Muhling J, Schmeck B, Seybold J,<br />

Walter C, Maass M, Rosseau S, Hegemann JH, Suttorp N, Hippenstiel S. Mechanisms of<br />

Chlamydophila pneumoniae Mediated GM-CSF Release in Human Bronchial Epithelial Cells.<br />

Am J Respir Cell Mol Biol. 2005 Dec 9; [Epub ahead of print]<br />

- 89 -


4: Schmeck B, Huber S, Moog K, Zahlten J, Hocke AC, Opitz B, Hammerschmidt S,<br />

Mitchell TJ, Kracht M, Rosseau S, Suttorp N, Hippenstiel S. Pneumococci induced TLR- <strong>and</strong><br />

Rac1-dependent NF-{kappa}B-recruitment to the IL-8 promoter in lung epithelial cells. Am J<br />

Physiol Lung Cell Mol Physiol. 2005 Nov 18; [Epub ahead of print]<br />

5: Schmeck B, Beermann W, van Laak V, Zahlten J, Opitz B, Witzenrath M, Hocke AC,<br />

Chakraborty T, Kracht M, Rosseau S, Suttorp N, Hippenstiel S. Intracellular bacteria<br />

differentially regulated endothelial cytokine release by MAPK-dependent histone<br />

modification. J Immunol. 2005 Sep 1;175(5):2843-50.<br />

6: N'Guessan PD, Schmeck B, Ayim A, Hocke AC, Brell B, Hammerschmidt S, Rosseau S,<br />

Suttorp N, Hippenstiel S. Streptococcus pneumoniae R6x induced p38 MAPK <strong>and</strong> JNKmediated<br />

caspase-dependent apoptosis in human endothelial cells. Thromb Haemost. 2005<br />

Aug;94(2):295-303. contributed equally<br />

7: Slevogt H, Tiwari KN, Schmeck B, Hocke A, Opitz B, Suttorp N, Seybold J. Adhesion of<br />

Moraxella catarrhalis to human bronchial epithelium characterized by a novel fluorescencebased<br />

assay. Med Microbiol Immunol (Berl). 2005 Jul 30;:1-11 [Epub ahead of print]<br />

8: Brell B, Temmesfeld-Wollbruck B, Altzschner I, Frisch E, Schmeck B, Hocke AC, Suttorp<br />

N, Hippenstiel S.<br />

Adrenomedullin reduces Staphylococcus aureus alpha-toxin-induced rat ileum<br />

microcirculatory damage. Crit Care Med. 2005 Apr;33(4):819-26.<br />

9: Opitz B, Forster S, Hocke AC, Maass M, Schmeck B, Hippenstiel S, Suttorp N, Krull M.<br />

Nod1-mediated endothelial cell activation by Chlamydophila pneumoniae. Circ Res. 2005<br />

Feb 18;96(3):319-26. Epub 2005 Jan 13.<br />

10: Krull M, Kramp J, Petrov T, Klucken AC, Hocke AC, Walter C, Schmeck B, Seybold J,<br />

Maass M, Ludwig S, Kuipers JG, Suttorp N, Hippenstiel S. Differences in cell activation by<br />

Chlamydophila pneumoniae <strong>and</strong> Chlamydia trachomatis infection in human endothelial cells.<br />

Infect Immun. 2004 Nov;72(11):6615-21.<br />

11: Schmeck B, Zahlten J, Moog K, van Laak V, Huber S, Hocke AC, Opitz B, Hoffmann E,<br />

Kracht M, Zerrahn J, Hammerschmidt S, Rosseau S, Suttorp N, Hippenstiel S. Streptococcus<br />

pneumoniae-induced p38 MAPK-dependent phosphorylation of RelA at the interleukin-8<br />

promotor. J Biol Chem. 2004 Dec 17;279(51):53241-7. Epub 2004 Oct 13.<br />

12: Walter C, Zahlten J, Schmeck B, Schaudinn C, Hippenstiel S, Frisch E, Hocke AC,<br />

Pischon N, Kuramitsu HK, Bernimoulin JP, Suttorp N, Krull M. Porphyromonas gingivalis<br />

strain-dependent activation of human endothelial cells. Infect Immun. 2004 Oct;72(10):5910-<br />

8.<br />

13: Schmeck B, Gross R, N'Guessan PD, Hocke AC, Hammerschmidt S, Mitchell TJ,<br />

Rosseau S, Suttorp N, Hippenstiel S. Streptococcus pneumoniae-induced caspase 6dependent<br />

apoptosis in lung epithelium. Infect Immun. 2004 Sep;72(9):4940-7.<br />

- 90 -


Epigenetic Regulation of Stem Cell Maintenance Genes in Prostate Carcinoma<br />

Michèle J. Hoffmann, Mirko Müller, Rainer Engers*, Wolfgang A. Schulz<br />

Urologische Klinik und Institut für Pathologie*, Heinrich-Heine-Universität Düsseldorf,<br />

Germany<br />

E-mail: michele.hoffmann@uni-duesseldorf.de; muellemi@uni-duesseldorf.de;<br />

engers@med.uni-duesseldorf.de ; wolfgang.schulz@uni-duesseldorf.de;<br />

We have previously reported that hypomethylation of LINE-1 retrotransposons is associated<br />

with the progression of prostate carcinoma. As in other cancers, causes <strong>and</strong> consequences of<br />

this alteration are not understood. Most likely, retrotransposon hypomethylation in cancer<br />

cells is related to a global change in chromatin structure that interacts with alterations in DNA<br />

methylation in a mutual fashion, i.e. altered expression of chromatin regulator proteins could<br />

alter DNA methylation <strong>and</strong>/or vice versa. In particular, some genes contributing to the stem<br />

cell character of germ cells <strong>and</strong> early embryonic cells appear to be controlled by DNA<br />

methylation, but also influence chromatin structure <strong>and</strong> DNA methylation in turn, e.g. the<br />

BMI1 polycomb gene <strong>and</strong> CTCFL/BORIS. Altered expression of these genes has been<br />

postulated to occur in many human cancers. We have therefore investigated their expression<br />

<strong>and</strong> relationship to DNA methylation in prostate cancer.<br />

In primary prostate cancer tissues, expression of polycomb EZH2 <strong>and</strong> BMI1 mRNAs was on<br />

average increased <strong>and</strong> decreased, respectively. Expression of CTCFL was increased in a small<br />

number of the cases <strong>and</strong> OCT4 was slightly diminished. No significant association with<br />

overall DNA methylation changes in prostate cancers was observed. However, CTCFL as<br />

well as OCT4 were less methylated in testes than in somatic cells <strong>and</strong> accordingly expressed<br />

much more strongly. Additional experiments using DNA methylation inhibitors showed DNA<br />

methylation to represent the essential mechanism for control of CTCFL expression, whereas<br />

OCT4 down-regulation was more dependent on other factors. No significant changes in DNA<br />

methylation of these two genes were found in prostate cancers, but OCT4 was<br />

hypomethylated in testicular cancers.<br />

Taken together, <strong>our</strong> results argue against a general reactivation of the investigated chromatin<br />

regulators in prostate cancer, with the known exception of EZH2. Since we have investigated<br />

bulk primary tumors, it remains of c<strong>our</strong>se possible that such a reactivation occurs in a small<br />

subset of the tumor cells or in metastases.<br />

Funded by the Deutsche Krebshilfe.<br />

Recent Publications<br />

Schulz WA: Molecular Biology of Human Cancers. An Advanced Student‘s Textbook. Springer, 2005<br />

Maas S, Warskulat U, Steinhoff C, Mueller W, Grimm MO, Schulz WA, Seifert HH (2004) Decreased FAS<br />

expression in advanced stage bladder cancer is not related to p53 status. Urology 63, 392-397<br />

Cronauer MV, Schulz WA, Burchardt T, Ackermann R, Burchardt M (2004) Inhibition of p53 function<br />

diminishes <strong>and</strong>rogen receptor-mediated <strong>signaling</strong> in prostate cancer cell lines. Oncogene 23, 3541-3549.<br />

Betz B, Florl AR, Seifert HH, Dall P, Schulz WA, Niederacher D (2004) DHPLC as a reliable high-throughput<br />

pre-screening method for aberrant promoter methylation in cancer. Hum. Mutat. 23, 612-620<br />

Zhang J, Zotz RB, Li Y, Wang R, Kiel S, Schulz WA, Wen D, Chen Z, Zhang L, Wang S, Gabbert HE, Sarbia<br />

M (2004) Methylenetetrahydrofolate reductase C677T polymorphism <strong>and</strong> predisposition towards esophageal<br />

squamous cell carcinoma in a German Caucasian <strong>and</strong> a northern Chinese population. J. Cancer Res. Clin.<br />

Oncol. 130, 574-580<br />

Florl AR, Steinhoff C, Müller M, Seifert HH, Hader C, Engers R, Ackermann R, Schulz WA (2004) Coordinate<br />

hypermethylation at specific sites in prostate carcinoma precedes LINE-1 hypomethylation. Brit. J. Cancer 91,<br />

985-994<br />

- 91 -


Sant<strong>our</strong>lidis S, Kimura F, Fischer J, Schulz WA (2004) Suppression of clonogenicity by the PCNA-binding<br />

domain of mammalian Dnmt1. Biochem. Cell Biol. 82, 589-596<br />

Raschke S, Balz V, Efferth T, Schulz WA, Florl AR (2005) Homozygous deletions of CDKN2A caused by<br />

alternative mechanisms in different human cancer cell lines. Genes Chromosomes Cancer, 42, 58-67<br />

Thievessen I, Wolter M, Prior A, Seifert HH, Schulz WA (2005) Hedgehog <strong>signaling</strong> in normal urothelial cells<br />

<strong>and</strong> in urothelial carcinoma cell lines. J. Cell. Physiol. 203, 372-377<br />

Hoffmann MJ, Florl AR, Seifert HH, Schulz WA (2005) Multiple mechanisms inactivating CDKN1C in bladder<br />

cancer. Int. J. Cancer 114, 406-413<br />

Kindich R, Florl AR, Jung V, Engers R, Müller M, Schulz WA, Wullich B (2005) Application of a modified<br />

real-time PCR technique for relative gene copy number quantification to the determination of the relationship<br />

between NKX3.1 loss <strong>and</strong> MYC gain in prostate cancer. Clin. Chem. 51, 649-652<br />

Cronauer MV, Schulz WA, Ackermann R, Burchardt M (2005) Effects of WNT/#-catenin pathway activation on<br />

<strong>signaling</strong> through T-cell factor (TCF) <strong>and</strong> <strong>and</strong>rogen receptor in prostate cancer cell lines. Int. J. Oncol. 26,<br />

1003-1040<br />

Rahnenführer J, Beerenwinkel N, Schulz WA, Hartmann C, von Deimling A, Wullich B, Lengauer T (2005)<br />

Estimating cancer survival <strong>and</strong> clincial outcome based on genetic tumor progression scores. Bioinformatics 21,<br />

2438-2446<br />

Wu Q, Hoffmann MJ, Hartmann FH, Schulz WA (2005) Amplification <strong>and</strong> overexpression of the ID4 gene at<br />

6p22.3 in bladder cancer. BMC Mol. Cancer 4, 16 (online)<br />

Sarbia M, Geddert H, Kiel S, K<strong>and</strong>emin Y, Schulz WA, Vossen S, Zotz RD, Willers R, Baldus SE, Schneider<br />

PM, Gabbert HE (2005) Methylenetetrahydrofolate reductase C677T polymorphism <strong>and</strong> risk of adenocarcinoma<br />

of the upper gastrointestinal tract. Sc<strong>and</strong>. J. Gastroenterol. 40, 109-111<br />

Laner T, Schulz WA, Engers R, Müller M, Florl AR (2005) Hypomethylation of the XIST promoter in prostate<br />

cancer. Oncol. Res. 15, 257-264<br />

Kindich R, Florl AR, Kamradt J, Lehmann J, Müller M, Wullich B, Schulz WA (2005) Relationship of NKX3.1<br />

<strong>and</strong> MYC gene copy number ratio <strong>and</strong> DNA hypomethylation to prostate carcinoma stage. Eur. Urol., in press<br />

Wethkamp N, Ramp U, Geddert H, Schulz WA, Florl AR, Suschek CV, Hassan M, Gabbert HE, Mahotka C<br />

(2006) Expression of Death-Associated Protein Kinase during tumor progression of human renal cell<br />

carcinomas: clues for hypermethylation-independent mechanisms of inactivation. Eur. J. Cancer, in press<br />

Schulz WA, Seifert HH (2004) DNA methylation in urological cancers. In: “DNA methylation <strong>and</strong> cancer<br />

therapy” (Szyf M, Hg.), pp.42-58, L<strong>and</strong>es Biosciences<br />

Cronauer MV, Schulz WA, Burchardt T, Anastasiadis AG, De La Taille A, Ackermann R, Burchardt M (2003)<br />

The <strong>and</strong>rogen receptor in hormone-refractory prostate cancer: Relevance of different mechanisms of <strong>and</strong>rogen<br />

receptor <strong>signaling</strong>. Int. J. Oncol. 23, 1095-1102<br />

Grimm MO, Hartmann FH, Schulz WA (2004) Microarrays: Technik und Potenzial beim Prostatakarzinom.<br />

Urologe 43, 653-658<br />

Schulz WA, Müller M (2004) Molekulare Karzinogenese des Prostatakarzinoms. in: Borchers H, Jakse G (eds)<br />

“Die Therapie des Prostatakarzinoms: Die Zukunft hat begonnen”. Akt. Onkol. 122, 39-53.<br />

Hoffmann MJ, Schulz WA (2005) Causes <strong>and</strong> consequences of DNA hypomethylation in human cancer.<br />

Biochem. Cell Biol. 83, 296-321<br />

Schulz WA (2005) Qualified promise: DNA methylation assays for cancer detection <strong>and</strong> classification<br />

(editorial). J. Biomed. Biotechnol. 3, 227-229<br />

Schulz WA, Steinhoff C, Florl AR (2005) Methylation of endogenous human retroelements in health <strong>and</strong><br />

disease. Curr. Topics Microbiol. Immunol., in press<br />

Schulz WA (2006) L1 retrotransposon in human cancers. J. Biomed. Biotechnol., in press<br />

- 92 -


Survey on in vitro cell death induction of multiple myeloma cells upon valproic acid<br />

treatment.<br />

Chantal Schwartz, Valérie Palissot, René N.H.C. Brons <strong>and</strong> Guy Berchem.<br />

Laboratoire d’Hémato-Cancérologie Expérimentale, CRP-Santé, 84, Val Fleury, L-1526<br />

Luxemb<strong>our</strong>g. E-mail :berchem.guy@chl.lu<br />

Multiple myeloma (MM) is, despite the emergence of new treatments in recent years, still a<br />

lethal disease in 2005. Over the last several years, significant insights into the dysregulation<br />

of various signal transduction pathways of MM have emerged <strong>and</strong> have evolved the<br />

development of new agents. Histone deacetylase (HDAC) inhibitors as valproic acid (VPA)<br />

are compounds that inhibit HDAC enzymes that, in conjunction with histone acetylases,<br />

regulate the acetylation state of histones <strong>and</strong>, by extension, the conformational status of<br />

chromatin. VPA is clinically known to be used in treatment of different types of epileptic<br />

diseases. The inhibitory functions of VPA on class I HDAC in combination with its moderate<br />

secondary effects in therapy was recently exploited (Gottlincher M, 2004). VPA has been<br />

described as inducer of apoptosis <strong>and</strong> differenciation in various cancer types <strong>and</strong> leukaemia.<br />

In <strong>our</strong> study, effects of cell death have been studied in cells harvested <strong>and</strong> purified from<br />

routine bone marrow aspirates of several patients with multiple myeloma, as well on the<br />

myeloma cell lines OPM2, RPMI <strong>and</strong> U266 treated with a physiological range of VPA (1-2<br />

mM). Using an XTT based cytotoxicity assay, we observe significant in vitro toxicity already<br />

starting at doses of 1 mM for 24h in the cell lines OPM2 (60% viability) <strong>and</strong> RPMI (80%<br />

viability), as well as on CD138+ selected MM cells from 2 patients. The cell line U266 was<br />

more resistant (90% viability). Incubating in presence of ZVAD, a pan-caspase inhibitor, had<br />

a partial protective effect, but did completely inhibit the cytotoxicity of VPA on OPM2 <strong>and</strong><br />

RPMI, which indicates that cell death through VPA is not only triggerd by the apoptotic<br />

pathway. This was confirmed by flow cytometric analysis with PI/Annexin V staining, where<br />

the cell lines U266 <strong>and</strong> OPM2 did not show apoptosis features, nor in DNA fragmentation<br />

assays. However Western blot analysis of OPM2 <strong>and</strong> RPMI with antibodies against caspase 9<br />

<strong>and</strong> 3 showed an accumulation of their cleaved forms within a time c<strong>our</strong>se of 72h of VPA<br />

treatment, indicating that cell death induced by VPA in MM is not completely independent of<br />

caspase activity.<br />

In summary, these data suggest that VPA has a cytotoxic effect on MM cells, <strong>and</strong> partially but<br />

not primarily induces apoptosis on these cells. Further experiments exploring other possible<br />

mechanisms of cell death will be conducted.<br />

- 93 -


Hypoxia Signal Transduction in Health <strong>and</strong> Disease<br />

Gregg L. Semenza<br />

Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins<br />

University School of Medicine, Baltimore, Maryl<strong>and</strong> 21205 USA. Email:<br />

gsemenza@jhmi.edu<br />

In mammals, delivery of O 2 to cells is regulated by multiple homeostatic mechanisms, which<br />

maintain a balance between O 2 supply <strong>and</strong> dem<strong>and</strong>. O 2 concentrations must be tightly<br />

regulated because of the risk of energy depletion or oxidative damage associated with<br />

inadequate <strong>and</strong> excess O 2, respectively. Hypoxia-inducible factor 1 (HIF-1) is a master<br />

transcriptional regulator of O 2 homeostasis. The half-life <strong>and</strong> transcriptional activity of the<br />

HIF-1 subunit are controlled by O 2-dependent hydroxylation, which provides a<br />

molecular mechanism for transducing changes in oxygenation into changes in gene<br />

expression. HIF-1 controls the expression of genes whose protein products control O 2<br />

delivery through the control of erythropoiesis (erythropoietin) <strong>and</strong> vascularization (vascular<br />

endothelial growth factor, placental growth factor, stromal derived factor 1, platelet-derived<br />

growth factor B, <strong>and</strong> angiopoietin-1<strong>and</strong> -2) growth. In addition to controlling the production<br />

of cytokines that activate vascular endothelial cells, HIF-1 also controls cell-autonomous<br />

responses of endothelial cells <strong>and</strong> bone marrow-derived progenitor cells. In preclinical<br />

studies, administration of an adenovirus encoding a constitutively active form of HIF-<br />

1 promotes both angiogenesis <strong>and</strong> arteriogenesis. A genetic polymorphism in the<br />

human HIF1A gene is associated with absence of coronary collaterals in patients with<br />

ischemic heart disease. HIF-1 overexpression is associated with increased risk of<br />

mortality in many different human cancers <strong>and</strong> HIF-1 contributes to immortalization, genetic<br />

instability, angiogenesis, glycolysis, autocrine growth factor <strong>signaling</strong>, invasion/metastasis,<br />

<strong>and</strong> treatment failure. In the clear cell type of renal cell carcinoma, loss of function of the von<br />

Hippel-Lindau tumor suppressor protein results in dysregulated HIF-1 activity that is<br />

associated with the repression of E-cadherin gene expression, leading to the loss of cell-cell<br />

adhesion that is required for invasion/metastasis. Several novel anti-cancer agents have antiangiogenic<br />

effects that are related to their inhibition of HIF-1 activity. Thus, strategies<br />

designed to inhibit or activate HIF-1 may represent a novel therapies in cancer <strong>and</strong> ischemic<br />

cardiovascular disease, respectively, which are the major causes of mortality in industrialized<br />

societies.<br />

- 94 -


Distinct roles of IRF-3 <strong>and</strong> IRF-7 in the activation of anti-tumor properties of human<br />

macrophages<br />

Mayra Solis 1,* Raphaëlle Romieu-M<strong>our</strong>ez 1,* , Aless<strong>and</strong>ra Nardin 2,* , Véronique Baron-<br />

Bodo 2 , Delphine Goubau 1 , Bernard Massie 3 , Margarita Salcedo 2 <strong>and</strong> John Hiscott 1 .<br />

* R.R-M, M.S. <strong>and</strong> A.N. contributed equally to this work 1 Terry Fox Molecular Oncology<br />

Group, Lady Davis Institute McGill University, Montreal Canada H3T 1E2 2 IDM, Paris<br />

France 3 Biotechnology Research Institute, Montreal Canada H4P 2R2<br />

Type I interferons (IFN-alpha, –beta) exert strong antiviral, antiproliferative <strong>and</strong><br />

antiangiogenic effects <strong>and</strong> are widely used in clinical settings as adjuvants for therapy against<br />

cancer. IFN-beta induction requires the activation of latent transcription factors, including the<br />

interferon regulating factor (IRF)-3, NF-!B <strong>and</strong> ATF-2/c-Jun. Secretion of the newly<br />

produced IFN-beta <strong>and</strong> binding to cognate adjacent type I IFN receptors induces transcription<br />

of the irf7 gene. IRF-7 induces the expression of delayed IFN-alpha genes <strong>and</strong> elicits the full<br />

induction of type I IFN production. When properly activated, macrophages can be<br />

tumoricidal, thus making attractive additions to st<strong>and</strong>ard cancer therapies. Tolerance <strong>and</strong><br />

activity of human autologous IFN-gamma-activated macrophages, (MAK ® cells), have been<br />

assessed in pilot trials in cancer patients. Here, we tested the hypothesis that activation of<br />

IRF-3 <strong>and</strong> IRF-7, with subsequent type I IFN production, may be involved in the acquisition<br />

of new antitumor functions by macrophages. We generated adenoviral vectors for the delivery<br />

of constitutive active mutants of IRF-3 (Ad-F3) or IRF-7 (Ad-F7) into primary human<br />

macrophages. Rapid cell death was observed in Ad-F3- transduced macrophages, whereas<br />

Ad-F7-transduction produced type I IFNs <strong>and</strong> displayed increased expression of genes<br />

encoding TRAIL, IL-12, IL-15 <strong>and</strong> CD80, persisting for at least 72 h<strong>our</strong>s. Expression of<br />

iNOS, TNF-alpha, FasL, IL-1 <strong>and</strong> IL-6 genes was unaltered by Ad-F7 transduction. In<br />

addition, Ad-F3 <strong>and</strong> Ad-F7 transduction appeared to negatively regulate the transcription of<br />

the pro-tumorigenic genes encoding IL-8, VEGF or MMP-2. Ad-F7- expressing macrophages<br />

exerted a cytostatic activity on SK-BR3, MCF-7 <strong>and</strong> COLO-205 cancer cell lines. The latter<br />

cells were previously shown to be insensitive to the action of non-activated macrophages or<br />

MAK cells. In conclusion, transduction of active forms of IRF-3 or IRF-7 appears to<br />

differentially regulate the apoptosis <strong>and</strong> the antitumor properties of primary macrophages,<br />

with IRF-7 active mutant leading to the acquisition of novel anti-tumor effector functions.<br />

- 95 -


New steps toward the use of parvovirus H1 as a therapeutic anti-cancer drug<br />

D.Stéhelin, N.Martin, G.Muharram, A.Bègue, C.Lagrou, A-C.Fl<strong>our</strong>ens A.Roussel,<br />

I.Loison.<br />

CNRS-UMR 8526. Institute of Biology of Lille, 1 rue Calmette-BP 447, 59021 Lille<br />

Cedex. dominique.stehelin@ibl.fr<br />

Recent emphasis (Int J Oncol. 4, 2005, 901; Nature Medicine 8, 2000, 862 ; Nature<br />

Biotechnology 18, 2000, 723) has brought into focus the intriguing property of some viruses<br />

that are able to quite selectively destroy many types of tum<strong>our</strong> cells. Our laboratory has been<br />

studying such a virus, namely parvovirus H1, which opportunistically infects the human<br />

population, but apparently without any known pathology in adults.<br />

Parvovirus H1 exhibits quite remarkable features in cell cultures: 1)It infects preferentially<br />

many types of human tum<strong>our</strong> cells. 2)The cells replicating the virus are rapidly killed (ex vivo<br />

in c.a. 3-5 days), releasing viral progeny. 3)The new virus readily infects the neighb<strong>our</strong>ing<br />

tum<strong>our</strong> cells in a recurrent fashion<br />

In vivo experiments showed that SCID mice injected with human tum<strong>our</strong> cell-lines (i.e.<br />

HeLa) develop tum<strong>our</strong>s <strong>and</strong> rapidly die, unless they are injected with Parvovirus H1, in which<br />

case the tum<strong>our</strong>s regress, then disappear definitely, preventing the animal from dying.<br />

A small Phase1 clinical trial carried on with T.Tursz at the IGR Hospital in Villejuif, in<br />

collaboration with J.Rommelaere (Heidelberg), gave enc<strong>our</strong>aging results: 1)High doses of<br />

parvovirus could be injected, apparently without hampering essential life parameters.<br />

2)Repeated injections could be applied, without observing deleterious high antibody levels.<br />

3)A systemic spread of the virus was observed, where the virus could reach distal metastases<br />

which sustained its replication. 4)The virus disappeared following the last injection.<br />

These promising results enc<strong>our</strong>aged us to further investigate the mechanism by which the<br />

virus is able to selectively destroy tum<strong>our</strong> cells, <strong>and</strong> to decipher why so many different types<br />

of tum<strong>our</strong> cells are efficient targets for parvoviral mediated oncolysis. We will also present<br />

recent achievements concerning the preparation of purified parvovirus grown on cells without<br />

any animal fraction, <strong>and</strong> <strong>our</strong> preliminary predictive test to determine if fresh human tum<strong>our</strong>s<br />

are responsive to parvoviral kelling.<br />

We recently obtained an important research grant that should allow us to progress toward a<br />

potential use of this virus in anticancer therapy.<br />

References:<br />

FAISST, S., D.GUITTARD, A.BENNER, Y.Y.CESBRON, J.R.SCHLEHOFER,<br />

J.ROMMELAERE <strong>and</strong><br />

T.DUPRESSOIR<br />

Dose-dependent regression of HeLa cell-derived tum<strong>our</strong>s in scid mice after parvovirus<br />

H-1 infection<br />

International J<strong>our</strong>nal of Cancer (1998) 75 : 584-589<br />

DUTHEIL, N., F.SHI, T.DUPRESSOIR <strong>and</strong> R.M.LINDEN<br />

Adeno-associated virus site-specifically integrates into a muscle-specific DNA region<br />

Proc. Natl. Acad. Sci. USA (2000) 97 : 4862-4866<br />

- 96 -


<strong>Redox</strong>-Sensitive Transcription Factors as Prime Molecular Targets for<br />

Chemoprevention <strong>and</strong> Cytoprotection<br />

Young-Joon Surh (surh@plaza.snu.ac.kr)<br />

National Research Laboratory of Molecular Carcinogenesis <strong>and</strong> Chemoprevention<br />

College of Pharmacy, Seoul National University, Seoul 151-742, South Korea<br />

There are multiple lines of compelling evidence supporting the association between<br />

inflammatory tissue damage <strong>and</strong> cancer. A new horizon in chemoprevention research is the<br />

recent discovery of molecular links between inflammation <strong>and</strong> cancer. Components of the<br />

cell <strong>signaling</strong> network, especially those converge on redox-sensitive transcription factors<br />

including nuclear factor-kappaB (NF-!B) <strong>and</strong> AP-1 involved in mediating inflammatory<br />

response, have been implicated in carcinogenesis. A wide variety of chemopreventive <strong>and</strong><br />

chemoprotective agents can alter or correct undesired cellular functions caused by abnormal<br />

pro-inflammatory signal transmission mediated by NF-!B <strong>and</strong> AP-1. Modulation of cellular<br />

<strong>signaling</strong> involved in chronic inflammatory response by anti-inflammatory agents hence<br />

provides a rational <strong>and</strong> pragmatic strategy in molecular target-based chemoprevention.<br />

Induction of phase-2 detoxifying or antioxidant enzymes represents an important cellular<br />

defence response to oxidative <strong>and</strong> electrophilic insults. Nrf2 plays a crucial role in regulating<br />

phase-2 detoxifying/antioxidant gene induction. Many chemopreventive <strong>and</strong> chemoprotective<br />

agents have been found to activate this particular redox-sensitive transcription factor, thereby<br />

potentiating cellular antioxidant capacity.<br />

References<br />

1. Surh, Y.-J. (2003) Cancer chemoprevention with dietary phytochemicals. Nature<br />

Reviews Cancer, 3: 768-780.<br />

2. Lim, S.-Y., Jang, J.-H., Na, H.-K., Lu, S., Rahman, I., <strong>and</strong> Surh, Y.-J. (2004) 15-deoxy-<br />

12,14 -prostagl<strong>and</strong>in J2 protects against nitrosative PC12 cell death through up-regulation of<br />

intracellular glutathione synthesis. J. Biol. Chem., 279: 46263-46270.<br />

3. Kim, S.-O., Kundu, J.K., Shin, Y.K., Park, J.-H., Cho, M.-H., Kim, T.-Y., <strong>and</strong> Surh, Y.-J.<br />

(2005) [6]-Gingerol inhibits COX-2 expression by blocking the activation of p38 MAP<br />

kinase. Oncogene, 24, 2558-2567.<br />

- 97 -


Gout-associated uric acid crystals activate the NALP3 inflammasome<br />

Jurg Tschopp<br />

Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerl<strong>and</strong><br />

Development of the acute <strong>and</strong> chronic inflammatory responses known as gout <strong>and</strong> pseudogout<br />

are associated with the deposition of monosodium urate (MSU) or calcium pyrophosphate<br />

dihydrate (CPPD) crystals, respectively, in joints <strong>and</strong> periarticular tissues. Although MSU<br />

crystals were first identified as the etiologic agent of gout in the 18th century1 <strong>and</strong> more<br />

recently as a “danger signal” released from dying cells2, little is known on the molecular<br />

mechanisms underlying MSU- or CPPD-induced inflammation. We will show that MSU <strong>and</strong><br />

CPPD engage the caspase-1 activating NALP3 inflammasome, resulting in the production of<br />

active IL-1b <strong>and</strong> IL-18. Macrophages from mice deficient in various components of the<br />

inflammasome such as Caspase-1, ASC <strong>and</strong> NALP3 are defective in crystal induced IL-1b<br />

activation. Moreover, an impaired neutrophil influx is found in an in vivo model of crystalinduced<br />

peritonitis in inflammasome-deficient mice or mice deficient in the IL-1b receptor<br />

(IL-1R). These findings provide insight into the molecular processes underlying the<br />

inflammatory conditions of gout <strong>and</strong> pseudogout <strong>and</strong> further support a pivotal role of the<br />

inflammasome in several autoinflammatory diseases.<br />

- 98 -


Epigenic control of MHC2TA transcription: varying contributions of DNA <strong>and</strong> histone<br />

modifications in cell-activation <strong>and</strong> interferon-$-mediated induction of gene expression<br />

in cancer cells<br />

Peter J. van den Elsen 1,3 , Marja C.J.A. van Eggermond 1 , Martine J. Jager 2 <strong>and</strong> Tjadine<br />

M. Holling 1<br />

1 Division of Molecular Biology, Department of Immunohematology <strong>and</strong> Blood Transfusion, <strong>and</strong><br />

2 Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherl<strong>and</strong>s.<br />

3 Department of Pathology, VU University Medical Center, Amsterdam, The<br />

Netherl<strong>and</strong>s.<br />

The products of major histocompatibility complex class II (MHC-II) genes encode cell<br />

surface glycoproteins, which play an essential role in the initiation of antigen-specific<br />

immune responses by virtue of their ability to present antigenic peptides to CD4 + T<br />

lymphocytes. Lack of MHC-II molecule expression results in a severe immunodeficiency as<br />

is observed in pediatric patients suffering from the bare lymphocyte syndrome.<br />

Essential for the transcriptional activation of MHC-II genes is the MHC2TA encoded class II<br />

transactivator (CIITA), a co-activator, which interacts with the MHC-enhanceosome bound to<br />

the SXY-promoter module of all MHC-II <strong>and</strong> accessory genes. Lack of MHC-II gene<br />

transcription, therefore, is due to lack of CIITA expression. Constitutive <strong>and</strong> induced<br />

expression of MHC2TA is governed through the employment of several independent<br />

promoters, which are activated following cell type-specific <strong>signaling</strong> pathways (e.g. human T<br />

cells) or in the response to inflammatory cytokines of which interferon-$ (IFN-$) is the most<br />

potent (e.g. fibroblasts <strong>and</strong> epithelial cells).<br />

In human T cell leukemia, the lack of CIITA expression was found to result from epigenetic<br />

modifications at the MHC2TA locus. Besides DNA methylation, we also observed<br />

trimethylation of lysine residue 27 of histone H3 (K27-H3) of the T cell employed MHC2TA<br />

promoter to correlate with lack of MHC2TA transcription.<br />

Interestingly, in uveal melanoma cells different epigenetic phenotypes were observed<br />

involving MHC2TA. In several uveal melanoma cell lines (Mel285 <strong>and</strong> OCM-1) despite<br />

hypermethylation of the uveal melanoma employed MHC2TA promoters, gene transcription<br />

was readily noted following IFN-$ treatment. Interestingly, in these uveal melanoma cells<br />

IFN-$ induction of MHC2TA transcription was associated with a strong increase in<br />

acetylated-histone H3 <strong>and</strong> H4, <strong>and</strong> induction in dimethyl-K4-H3 levels in MHC2TA<br />

chromatin as determined by chromatin immunoprecipitation analyses. In contrast, OMM1.3<br />

cells lacked MHC2TA transcription after IFN-$ stimulation. Surprisingly, the genomic<br />

MHC2TA promoter DNA was unmethylated, which is in contrast to other non IFN-$inducible<br />

cell types. However the lack of MHC2TA transcription correlated with relative high<br />

levels of trimethyl-K27-H3 in MHC2TA promoter chromatin.<br />

Together <strong>our</strong> results indicate varying levels of epigenetic control of MHC2TA transcription in<br />

cancer cells, which may contribute to evasion of host-mediated immunosurveillance.<br />

- 99 -


Recent papers:<br />

1. Holling TM, Van der Stoep N, Quinten E <strong>and</strong> Van den Elsen PJ.<br />

Activated T cells accomplish MHC class II expression through T cell specific occupation of<br />

CIITA promoter III. J. Immunol. 2002; 168: 763-770.<br />

2. Van der Stoep N, Quinten E <strong>and</strong> Van den Elsen PJ.<br />

Transcriptional regulation of the MHC class II transactivator (CIITA) promoter III:<br />

Identification of a novel regulatory region in the 5’-UTR <strong>and</strong> an important role for cAMP<br />

reponsive element binding protein 1 <strong>and</strong> activating transcription factor-1 CIITA promoter III<br />

transcriptional activation in B-lymphocytes. J. Immunol. 2002; 169: 5061-5071.<br />

3. Holling TM, Schooten E, Langerak AW <strong>and</strong> Van den Elsen PJ.<br />

Regulation of MHC class II expression in human T cell malignancies. Blood 2004; 103:<br />

1438-1444.<br />

4. Van der Stoep N, Quinten E, Marcondes Rezende M <strong>and</strong> Van den Elsen PJ.<br />

E47, IRF-4 <strong>and</strong> PU.1 synergize to induce B cell-specific activation of the Class II<br />

Transactivator promoter III (CIITA-PIII). Blood 2004; 104: 2849-2857.<br />

5. Van den Elsen PJ, Holling TM, Kuipers HF <strong>and</strong> Van der Stoep N.<br />

Transcriptional regulation of antigen presentation. Curr. Opin. Immunol. 2004; 16: 67-75.<br />

6. Holling TM, Schooten E <strong>and</strong> Van den Elsen PJ.<br />

Function <strong>and</strong> regulation of MHC class II molecules in T-lymphocytes: of mouse <strong>and</strong> man.<br />

2004; 65: 282-290.<br />

7. Holling TM, Van der Stoep N <strong>and</strong> Van den Elsen PJ.<br />

Epigenetic control of CIITA expression in leukemic T cells. Biochem. Pharmacol. 2004; 68:<br />

1209-1213.<br />

8. Kuipers HF, Biesta PJ, Mommaas AM, Groothuis T, Neefjes J, <strong>and</strong> Van den Elsen PJ.<br />

Statins affect cell surface expression of MHC-II molecules by disrupting cholesterolcontaining<br />

microdomains. Hum. Immunol. 2005; 66: 653-665.<br />

9. Kuipers HF <strong>and</strong> Van den Elsen PJ<br />

Statins <strong>and</strong> control of MHC2TA transcription. Nature Medicine 2005; 11: 365-366.<br />

10. Schooten E, Klous P, Van den Elsen PJ <strong>and</strong> Holling TM.<br />

Lack of MHC class II expression in activated mouse T cells correlates with hypermethylation<br />

at the CIITA-PIII region. Immunogenetics 2005; in press.<br />

- 100 -


Attenuation of MSK1-driven NF-kB gene expression by soy isoflavones does not require<br />

estrogenic activity<br />

Wim V<strong>and</strong>en Berghe, Nathalie Dijsselbloem, Linda Vermeulen, 'Matladi N. Ndlovu,<br />

Elke Boone, Guy Haegeman<br />

Laboratory for Eukaryotic Gene Expression <strong>and</strong> Signal Transduction (LEGEST),<br />

Department of Molecular Biology, Ghent University, K.L. Ledeganckstraat 35, B-9000<br />

Gent, Belgium. Email: w.v<strong>and</strong>enberghe@ugent.be<br />

We have analysed in molecular detail how soy isoflavones (genistein, daidzein, biochanin A)<br />

suppress NF-kB-driven interleukin-(IL)6 expression. In addition to its physiological immune<br />

function as an acute stress cytokine, sustained elevated expression levels of IL6 promote<br />

chronic inflammatory disorders, aging frailty <strong>and</strong> tumorigenesis. Our results in estrogenunresponsive<br />

fibroblasts, mitogen- <strong>and</strong> stress-activated protein kinase (MSK) knock-out cells<br />

<strong>and</strong> estrogen receptor (ER)-deficient breast tumor cells, demonstrate that phytoestrogenic<br />

isoflavones can selectively block nuclear NF-kB transactivation of specific target genes (in<br />

particular IL6), independently of their estrogenic activity. This occurs via attenuation of<br />

mitogen-activated protein/ERK kinase (MEK) <strong>and</strong> extracellular signal-regulated kinase<br />

(ERK) activity, which further downregulates MSK-dependent NF-kB p65 <strong>and</strong> histone H3<br />

phosphorylation. As constitutive NF-kB <strong>and</strong> MSK activity are hallmarks of aggressive<br />

metastatic ER-deficient breast cancer, the MSK <strong>signaling</strong> pathway may become an attractive<br />

target for chemotherapy.<br />

Recent references<br />

Vermeulen, L., De Wilde, G., Van Damme, P., V<strong>and</strong>en Berghe, W. & Haegeman, G.<br />

Transcriptional activation of the NF-!B p65 subunit by mitogen- <strong>and</strong> stress-activated protein<br />

kinase-1 (MSK1). EMBO J<strong>our</strong>nal, 22(6), 1313-1324, 2003.<br />

De Bosscher, K., V<strong>and</strong>en Berghe, W. & Haegeman, G. The interplay between nuclear receptors<br />

<strong>and</strong> NF-!B or AP1: molecular mechanisms for gene repression.<br />

Endocr. Rev., 24(4), 488-522, 2003.<br />

Dijsselbloem, N., V<strong>and</strong>en Berghe, W., De Naeyer, A. en Haegeman, G. Phyto-estrogens:<br />

multi-purpose compounds at the crossroad of hormone replacement, anti-cancer <strong>and</strong> antiinflammatory<br />

therapy? Biochem. Pharm., 68, 1171-1185, 2004.<br />

De Bosscher, K., V<strong>and</strong>en Berghe, W., Beck, I., Lauw, A., Hapgood, J. & Haegeman, G. Antiinflammatory<br />

capacities of a plant-derived, non-steroidal contraceptive compound. Proc Natl<br />

Acad Sci U S A. [Epub ahead of print 21 october] , 2005<br />

De Naeyer, A., V<strong>and</strong>en Berghe, W., Pocock, V., Milligan, S., Haegeman, G., De<br />

Keukeleire, D. : Estrogenic <strong>and</strong> anticarcinogenic properties of kurarinone, a<br />

lav<strong>and</strong>ulyl flavanone from the roots of Sophora flavescens. J<strong>our</strong>nal of Natural Products, 67<br />

(11): 1829-1832, 2004.<br />

De Naeyer, A., V<strong>and</strong>en Berghe, W., De Keukeleire, D. & Haegeman, G. Phytoestrogens in<br />

cancer chemoprevention; characterization <strong>and</strong> beneficial effects of a new flavanone,<br />

- 101 -


kurarinone, a major phytoestrogen constituent of Sophora Flavescens ait. in<br />

“Phytopharmaceuticals in Cancer Chemoprevention” (Modern Nutrition Science) edited by<br />

Debasis Bagchi, USA, CRC Press, 428-448, 2004.<br />

V<strong>and</strong>en Berghe, W., Dijsselbloem, N., De Naeyer, A., Vermeulen, L. en Haegeman, G.<br />

Regulation of NF-kB-driven inflammatory genes by phytoestrogens. In “Oxidative stress,<br />

<strong>Inflammation</strong> <strong>and</strong> Health”, edited by Y-J. Surh,, Marcel Dekker Inc, 61-107, 2005<br />

V<strong>and</strong>en Berghe, W.*, Dijsselbloem, N.*, Vermeulen, L., Ndlovu, M., Boone, E. &<br />

Haegeman, G. Phytoestrogenic soy isoflavones attenuate MSK1-driven NF-kB gene<br />

expression in estrogen receptor deficient breast tumor cells. Cancer Research, in revision<br />

(2005) (*) equally contributed<br />

- 102 -


Talking to chromatin: Silencers Silenced.<br />

Jan Willem Voncken<br />

Div. Molecular Genetics, University of Maastricht, Universiteitssingel 50, 6229 ER,<br />

Maastricht, The Netherl<strong>and</strong>s. Email: w.voncken@molgen.unimaas.nl<br />

Polycomb Group (PcG) proteins modify chromatin structure. Best known for their ability to<br />

maintain transcriptional silencing, PcG proteins are critically involved in many other<br />

important biological processes, ranging from X-chromosome inactivation to stem cell<br />

regeneration. The molecular mechanisms involved in PcG-directed chromatin modification<br />

repression are starting to unravel. Functional regulation of PcG-proteins is poorly understood.<br />

We previously found hat PcG-complexes are phosphorylated <strong>and</strong> dissociate from chromatin<br />

in vivo in a cell cycle-dependent manner. Recent evidence from <strong>our</strong> lab suggests that PcG<br />

proteins are phosphorylated in response to cell signalling independent of cell cycleprogression.<br />

This link suggests a novel mechanism by which PcG-mediated gene silencing<br />

can be dynamically modulated. The implications of <strong>our</strong> findings will be discussed.<br />

- 103 -


A Systems Approach to Protein Kinase Signaling Networks<br />

Michael B. Yaffe M.D., Ph.D.<br />

Howard S. <strong>and</strong> Linda E. Stern Associate Professor<br />

Center for Cancer Research<br />

Depts of Biology <strong>and</strong> Biological Engineering<br />

Associate Member, Broad Institute<br />

Massachusetts Institute of Technology<br />

Cambridge, Massachusetts, USA<br />

Many protein kinases <strong>and</strong> phosphoserine/threonine-binding domains such as 14-3-3<br />

proteins, WW domains, FHA domains, Polo-box domains, <strong>and</strong> BRCT domains function<br />

together within <strong>signaling</strong> networks to control cell cycle progression, the response to DNA<br />

damage, <strong>and</strong> the onset of apoptosis. How signals emerging from these pathways are integrated<br />

<strong>and</strong> processed as a network is unclear. To address this, we have been developing systems<br />

models of <strong>signaling</strong> where kinase activities, protein phosphorylation, binding of substrates to<br />

phosphoserine/threonine binding domains, <strong>and</strong> cellular responses such as cell cycle arrest <strong>and</strong><br />

apoptosis are quantitatively measured at densely sampled points in time, <strong>and</strong> related<br />

mathematically using partial least squares regression <strong>and</strong> principal components analysis.<br />

Using cytokine-induced apoptosis in HT-29 cells as a starting point, we used this approach to<br />

construct a systems model of 7980 intracellular <strong>signaling</strong> events that directly links<br />

measurements to 1440 response outputs associated with apoptosis. The model accurately<br />

predicted multiple time-dependent apoptotic responses induced by a combination of the<br />

death-inducing cytokine tumor necrosis factor (TNF) with the pro-survival factors epidermal<br />

growth factor (EGF) <strong>and</strong> insulin. The model revealed new molecular mechanisms connecting<br />

<strong>signaling</strong> to apoptosis including the role of unsuspected autocrine circuits activated by TGF-"<br />

<strong>and</strong> IL-1", All of the molecular signals could be divided along two primary <strong>signaling</strong> axes<br />

that constitute fundamental dimensions (molecular “basis axes”) within the apoptotic<br />

<strong>signaling</strong> network. Projections of different stimuli along these axes capture the entire<br />

observed apoptotic response, suggesting that cell survival is determined by <strong>signaling</strong> through<br />

this canonical basis set. We are currently applying this methodology to study <strong>signaling</strong> events<br />

that control cell cycle arrest <strong>and</strong> apoptosis after DNA damage.<br />

- 104 -


Ubiquitin ligase Smurf1 controls osteoblast activity <strong>and</strong> bone homeostasis by targeting<br />

MEKK2-JNK pathway<br />

Ying E. Zhang 1 , Motozo Yamashita 1 , Sai-Xia Ying 1 , Cuiling Li 2 , Chuxia Deng 2 , <strong>and</strong><br />

Steven Y. Cheng 1<br />

1 Laboratory of Cellular <strong>and</strong> Molecular Biology, Center for Cancer Research, National<br />

Cancer Institute, National Institutes of Health, Bethesda, MD 20892<br />

2 Genetics of Development <strong>and</strong> Disease Branch, National Institute of Diabetes <strong>and</strong><br />

Digestive <strong>and</strong> Kidney Diseases, National Institutes of Health, Bethesda, MD 20892<br />

Smad ubiquitin regulatory factor (Smurf), a HECT domain ubiquitin ligase, was previously<br />

identified as E3 ligase targeting TGF-beta/BMP pathway through in vitro biochemical<br />

experiments. To investigate the physiological function of Smurf1, we disrupted the mouse<br />

Smurf1 allele through homologous recombination. Smurf1-deficient mice are born normal,<br />

fertile <strong>and</strong> have normal life span. Given the fact that members of the TGF-#eta/BMP<br />

superfamily are crucial inductive signals during embryonic development, the seemingly<br />

normal development of Smurf1 -/- mice <strong>and</strong> the concurrent increase of the Smurf2 transcript<br />

in Smurf1 -/- suggest that the loss of Smurf1 function might have been compensated for, at<br />

least in part, by the elevated Smurf2. Despite of no phenotypic developmental abnormalities<br />

or health problems, Smurf1 -/- mice displayed age-dependent increase of bone mass. The<br />

cause of this increase can be traced to enhanced activities of osteoblasts, which become<br />

sensitized to bone morphogenesis protein (BMP) in the absence of Smurf1. Surprisingly, this<br />

skeletal abnormality is not due to alteration in the Smad-mediated canonical TGF-beta or<br />

BMP <strong>signaling</strong>. Instead, loss of Smurf1 leads to accumulation of phosphorylated MEKK2<br />

<strong>and</strong> activation of the downstream JNK <strong>signaling</strong> cascade. We found that Smurf1 physically<br />

interacts with MEKK2 <strong>and</strong> promotes the ubiquitination <strong>and</strong> turn-over of phosphorylated<br />

MEKK2, which resulted in downregulation of osteoblast activity <strong>and</strong> response to BMP.<br />

These results reveal a novel function of Smurf1 in the regulation of bone homeostasis through<br />

directly targeting MAPK <strong>signaling</strong> pathway, thereby regulating the biological response of the<br />

TGF-beta superfamily <strong>signaling</strong>.<br />

Selected Recent Publications from My Group:<br />

Yu, L., Hebert, M. <strong>and</strong> Zhang, Y. E.: TGF-beta receptor-activated p38 MAP kinase mediates<br />

Smad-independent TGF-beta responses. EMBO J. 21: 3749-3759, 2002.<br />

Ying, S.-X., Zareena J. H., <strong>and</strong> Zhang, Y. E.: Smurf1 facilitates myogenic<br />

differentiation <strong>and</strong> antagonizes the Bone Morphogenetic Protein-2-induced osteoblast<br />

conversion by targeting Smad5 for degradation. J. Biol. Chem. 278: 39029-39036, 2003.<br />

Derynck, R. <strong>and</strong> Zhang, Y. E.: TGF-beta family <strong>signaling</strong>: Smad-dependent <strong>and</strong><br />

Smad independent pathways. Nature 425: 577-584, 2003.<br />

Yamashita, M., Ying, S.-X., Zhang, G.-M., Li, C., Cheng, S. Y., Deng, C.-X., <strong>and</strong><br />

Zhang, Y. E.: Ubiquitin ligase Smurf1 controls osteoblast activity <strong>and</strong> bone homeostasis by<br />

targeting MEKK2 for degradation. Cell 121: 101-113, 2005<br />

- 105 -


Signaling pathways controlling self-tolerance: On the road to systemic autoimmune<br />

diseases<br />

Moncef ZOUALI<br />

INSERM U606, Centre Viggo Petersen, Hôpital Lariboisière,<br />

2, rue Ambroise Paré, 75475 Paris Cedex 10, France,<br />

The immune receptors of lymphocytes are able to sense the nature of bound lig<strong>and</strong>s. Through<br />

coupled <strong>signaling</strong> pathways the generated signals are appropriately delivered to the<br />

intracellular machinery, allowing specific functional responses. A central issue in<br />

contemporary immunology is how the fate of B lymphocytes is determined at the successive<br />

developmental stages <strong>and</strong> how the B cell receptor distinguishes between signals that induce<br />

immune response or tolerance. Experiments with mice expressing transgenes or lacking<br />

signal transduction molecules are providing important clues to the mechanisms that regulate<br />

<strong>signaling</strong> thresholds at different developmental stages. Their relevance for the pathogenesis<br />

of autoimmune diseases in clinical settings is currently the focus of interest, particularly since<br />

in some instances, similar defects have been identified in human autoimmune diseases. The<br />

fact that altered <strong>signaling</strong> has been described in both T cells <strong>and</strong> B cells of autoimmune<br />

patients strengthens the view that the abnormalities observed are relevant to the pathogenesis.<br />

The studies are also revealing novel potential mechanisms of induction of autoimmunity,<br />

which may have a bearing on the underst<strong>and</strong>ing of human diseases.<br />

- 106 -


Workshop presentations<br />

(by alphabetical order)<br />

- 107 -


Efficient DNA <strong>and</strong> siRNA delivery into primary cells <strong>and</strong> cell lines<br />

Amaxa<br />

Gene transfer into primary cells <strong>and</strong> difficult-to-transfect cell lines has up to now been<br />

hampered by inefficient transfection methods. The Nucleofector technology presents a<br />

powerfull tool, which has been successfully used worldwide to transfer DNA plasmids or<br />

siRNA duplexes into primary cells such as neurons, T-cells <strong>and</strong> many cancer cells. Here, we<br />

discuss the latest results <strong>and</strong> applications, that have been obtained by using this method.<br />

Furthermore, we will introduce amaxa’s new Nucleofector 96-well shuttle, which implies all<br />

advantages of the Nucleofector technology in a high throughput manner.<br />

- 108 -


Ambion<br />

MicroRNAs (miRNAs) are small, RNA molecules encoded in the genomes of plants <strong>and</strong><br />

animals. In mammalians, these highly conserved, ~21-mer RNAs regulate the expression of<br />

genes by binding to the 3'-untranslated regions (3'-UTR) of specific mRNAs resulting in<br />

translation inhibition.<br />

In this tsudy, we describe methods to study their expression <strong>and</strong> to investigate their functional<br />

implications.<br />

Expression profile can be easily performed at array scale allowing miRNA global gene<br />

expression studies of different biological processes like cancer, differenciation, viral<br />

infection, etc.<br />

Functional analysis was performed using miRNA Precursor (Pre-miR) molecules that mimic<br />

<strong>and</strong> activate miRNA inhibitory action. Additionally, miRNA Inhibitor (Anti-miR) molecules<br />

that inhibit miRNA action on target mRNAs were used to further confirm the functionality of<br />

certain miRNAs.<br />

Using these tools, we were able to demonstrate the implication of let-7 in the regulation of<br />

Ras protein expression in lung cancer patients.<br />

- 109 -


Molecular Networks in Mammals: Extraction from Literature <strong>and</strong> Pathway Analysis<br />

Ariadne Genomics, Inc.<br />

Using the proprietary high-content linguistics tool MedScan we compiled a comprehensive<br />

database of molecular networks by extracting the information from scientific literature.<br />

MedScan is capable of extracting functional associations between proteins, small molecules,<br />

<strong>and</strong> pathways, recognizes types of regulatory mechanisms involved, effects of regulation <strong>and</strong><br />

experimental conditions.<br />

The resulting database stores 700,000 relationships between mammalian proteins <strong>and</strong><br />

chemicals including facts about protein interactions, promoter binding, molecular<br />

biosynthesis <strong>and</strong> trafficking, <strong>and</strong> cell process regulation. Different approaches towards<br />

reconstructing individual pathways or cascades from this database <strong>and</strong> assigning functional<br />

categories to proteins will be described.<br />

Our visualization software is capable of systematically mining this database for small network<br />

motifs that are robust in regard to the effects induced at the gene expression levels. We have<br />

also developed a Bayesian framework for integration of microarray data <strong>and</strong> binary gene-togene<br />

regulatory relationships. The approach allows the reduction of expression pattern<br />

complexity <strong>and</strong> finds the minimal set of regulatory proteins that are responsible for<br />

differential expression of other genes.<br />

Finding mesoscopic communities in sparse networks<br />

I. Ispolatov, I. Mazo, <strong>and</strong> A. Yuryev<br />

q-bio.MN/0512038, submitted to Phys. Rev. E<br />

Identifying Local Gene Expression Patterns in Biomolecular Networks<br />

A. Y. Sivachenko, A. Yuryev, N. Daraselia, I. Mazo<br />

2005 IEEE Computational Systems Bioinformatics Conference, Aug. 8-11, 2005<br />

Cliques <strong>and</strong> duplication-divergence network growth<br />

Ispolatov, I ; Krapivsky, P L ; Mazo, I ; Yuryev, A<br />

New J.Phys. 7(2005) 145<br />

Binding properties <strong>and</strong> evolution of homodimers in protein-protein interaction networks<br />

Ispolatov, Iaroslav; Yuryev, Anton; Mazo, Ilya; Maslov, Sergei<br />

Nucleic Acids Research 2005 33(11):3629-3635<br />

Extracting Protein Function Information from MEDLINE Using a Full-Sentence Parser<br />

Nikolai Daraselia, Sergei Egorov, Andrey Yazhuk, Svetlana Novichkova, Anton Yuryev, Ilya Mazo<br />

published in Proceeding of the Second European Workshop on Data Mining <strong>and</strong> Text Mining for<br />

Bioinformatics, pp11-18, 2004<br />

A Simple <strong>and</strong> Practical Dictionary-Based Approach for Identification of Proteins in MEDLINE Abstracts<br />

Sergei Egorov PhD, Anton Yuryev PhD, <strong>and</strong> Nikolai Daraselia PhD<br />

J<strong>our</strong>nal of the American Medical Informatics Association 2004<br />

Pathway studio - the analysis <strong>and</strong> navigation of molecular networks<br />

Alex<strong>and</strong>er Nikitin, Sergei Egorov, Nikolai Daraselia <strong>and</strong> Ilya Mazo<br />

BIOINFORMATICS APPLICATIONS NOTE Vol. 19 no. 0 2003, pages 1-3<br />

Extracting human protein interactions from MEDLINE using a full-sentence parser<br />

Nikolai Daraselia, Anton Yuryev, Sergei Egorov, Svetalana Novichkova, Alex<strong>and</strong>er Nikitin <strong>and</strong> Ilya Mazo<br />

BIOINFORMATICS Vol. 19 no. 0 2003, pages 1–8<br />

- 110 -


Organizer: BIOBASE GmbH<br />

Date: January, 26 2006<br />

Time: 3 pm – 4:30 pm<br />

Duration: ca 1 h<strong>our</strong> 30 minutes<br />

Attendees: 110 - 140<br />

Topic: Cutting-edge Biology for Explaining Expression Data<br />

Chair: Alena Kemper, BIOBASE, Wolfenbüttel<br />

Speakers:<br />

Prof. Dr. Edgar Wingender, BIOBASE, Wolfenbüttel<br />

„Making Sense of Transcriptomics <strong>and</strong> Proteomics - New Frontiers of Integrative Biology”<br />

Dr. Hauke Stephan Busch, German Cancer Research Center – DKFZ – TP3, Heidelberg<br />

“CD95-induced Appoptosis: An Example for Large-Scale Modeling of Signal Transduction<br />

Networks”<br />

Dr. Alex<strong>and</strong>er Kel/ Holger Karas, BIOBASE, Wolfenbüttel<br />

„Presentation of the ExPlain System”<br />

Organizer: BIOBASE GmbH<br />

Date: January, 27 2006<br />

Time: 3 pm – 4:30 pm<br />

Duration: ca 1 h<strong>our</strong> 30 minutes<br />

Attendees: 110 - 140<br />

Topic: Cutting-edge Biology for Explaining Expression Data<br />

Chair: Alena Kemper, BIOBASE, Wolfenbüttel<br />

Speakers:<br />

Prof. Dr. Edgar Wingender, BIOBASE, Wolfenbüttel<br />

„Making Sense of Transcriptomics <strong>and</strong> Proteomics - New Frontiers of Integrative Biology”<br />

Prof. Dr. Jürgen Borlak, Fraunhofer Institute of Toxicology <strong>and</strong> Experimental Medicine,<br />

Center for Drug Research <strong>and</strong> Medical Biotechnology, Hannover<br />

„Regulatory Networks of Liver-enriched Transcription Factors in Liver Biology <strong>and</strong> Disease”<br />

Dr. Alex<strong>and</strong>er Kel/ Holger Karas , BIOBASE, Wolfenbüttel<br />

„Presentation of the ExPlain System”<br />

- 111 -


A Novel Fluorescence-based Kinetic Kinase Assay for Use with Recombinant Protein<br />

<strong>and</strong> Cell Lysates<br />

M. Li, J.A. Vrana, M. Surby, X-D Qian, C.J. Clark, M. Schults, B. Imperiali, E.M.<br />

Schaefer<br />

Signal Transduction Division, Invitrogen Corporation, Hopkinton, Massachusetts <strong>and</strong><br />

Department of Chemistry, Massachusetts Institute of Technology<br />

Protein phosphorylation plays a critical role in signal transduction in normal <strong>and</strong> disease<br />

states. Recent advances in the development of specific protein kinase inhibitors illustrates the<br />

potential for a new generation of drugs to treat human cancers. We have adapted a novel<br />

fluorescent peptide substrate-based assay for the rapid, homogeneous <strong>and</strong> sensitive detection<br />

of the activity of a broad range of protein kinases. This assay platform uses the chelationenhanced<br />

fluorophore 8-hydroxy-5-(N,N-dimethylsulfonamido)-2-methylquinoline (referred<br />

to as Sox), which is incorporated into the substrate peptide. A series of specific Ser/Thr <strong>and</strong><br />

Tyr substrate peptides are utilized to measure a range of kinases covering all families of the<br />

kinome. Several key features of this assay include measurement of kinase activity under<br />

optimal kinetics, physiological (mM) ATP, <strong>and</strong> in real time without the use of radioactive<br />

tracer, beads, antibodies or specialized equipment. Importantly, activity increases result in a<br />

corresponding increase in fluorescence. Because the assay can be performed with a wide<br />

range of ATP concentrations it can be used to select for both ATP-competitive <strong>and</strong> ATP-noncompetitive<br />

kinase inhibitors. We have applied this system for the analysis of multiple sample<br />

types including the determination of kinase activity of recombinant proteins <strong>and</strong> using crude<br />

cell lysates in a 96 or 384 well format. This technology, used in conjunction with phosphospecific<br />

antibody based profiling, provides a valuable set of tools for rapidly increasing <strong>our</strong><br />

underst<strong>and</strong>ing of the kinome <strong>and</strong> the nature of <strong>signaling</strong> networks.<br />

- 112 -


Reliable <strong>and</strong> efficient gene Knock-down <strong>and</strong> inducible overexpression in vivo.<br />

Ben Davies#, Kader Thiam*<br />

# Senior Scientific consultant, *Director of Transgenic Technologies<br />

genOway, 181 avenue Jean Jaures, 69362 Lyon cedex 07, France. info@genoway.com<br />

Over the past decades, gene Knock-out <strong>and</strong> overexpression animal models have become<br />

st<strong>and</strong>ard laboratory tools for in vivo functional gene analysis. More recently, attempts have<br />

been made to extend the repertoire of transgenic techniques using RNA interference for gene<br />

Knock-down <strong>and</strong> inducible promoter systems for temporally regulated gene expression,<br />

allowing the compensation <strong>and</strong> developmental problems associated with classical approaches<br />

to be overcome. Despite initial enc<strong>our</strong>aging reports, the application of both these technologies<br />

in vivo has been unreliable. Here we describe two new approaches which allow both<br />

technologies to be robustly <strong>and</strong> consistently applied in a transgenic context.<br />

A major requirement for successful <strong>and</strong> reliable shRNA mediated gene Knock-down in vivo<br />

is a low copy number of integrating constructs. We have established <strong>and</strong> validated a<br />

technology which allows the generation of a high number of independent founder lines with a<br />

pre-screened copy number. Furthermore a second Knock-in based technology has been<br />

designed which takes advantage of ready-to-use validated targeting vectors to introduce a<br />

single copy of the shRNA cassette within a specific permissive genomic site. With both these<br />

technologies, gene knock-down of up to 80% has been reliably demonstrated.<br />

The tetracycline system has been used successfully in cell culture models to achieve<br />

regulated, inducible gene expression. Unfortunately, the application of this technology in vivo<br />

has been plagued by a lack of predictability. The resulting leakiness <strong>and</strong> inconsistency of<br />

expression in the resulting transgenic lines necessitates the generation <strong>and</strong> analysis of many<br />

independent founder lines before a convenient line with the appropriate induction kinetics is<br />

found. We report the development, of a new system which uses a preferential genomic<br />

context to achieve reliable <strong>and</strong> tight regulation of transgene expression. This reliable <strong>and</strong><br />

reproducible inducible systems is reversible <strong>and</strong> is not restricted to preferential tissues.<br />

Through the use of these technologies, the transgenic tool box is now complete to address<br />

gene function in vivo whilst minimizing complicating compensatory <strong>and</strong> developmental<br />

effects. With these methods in place, gene activity can now be reliably <strong>and</strong> exquisitely finetuned,<br />

allowing the most appropriate model for a specific scientific question to be designed.<br />

- 113 -


New tags for transfection <strong>and</strong> protein:protein interaction analysis<br />

T. Schmidt<br />

IBA GmbH, Rudolf-Wissell-Str. 28, D-37079 Göttingen<br />

Magnet Assisted Transfection (MATra) is a new, easy-to-h<strong>and</strong>le, very fast <strong>and</strong> highly<br />

efficient technology to transfect cells in culture. In a first step, nucleic acids are associated<br />

with specific magnetic nanoparticles (MagTag). Exploiting magnetic force the full nucleic<br />

acid dose is then drawn towards <strong>and</strong> delivered into the target cells leading to efficient<br />

transfection without disturbing the membrane architecture, without causing chromosomal<br />

damage or leaving a hole in the cell membrane like other transfection technologies. All types<br />

of nucleic acids from plasmid DNA or siRNA to oligonucleotides can be used with the<br />

MATra approach. Data from a variety of species using cell lines or primary tissue culture<br />

have accumulated including human, monkey, mouse, rat, xenopus, pig, cat or fish.<br />

One-STrEP-tag is an extremely fast <strong>and</strong> efficient method to isolate functional protein<br />

complexes which have formed in the cellular environment after expression of a tagged bait<br />

protein. After disruption of the cells the intact protein complex is separated from the crude<br />

lysate on Strep-Tactin affinity columns. While other systems require tedious optimization<br />

because of high background or two successive purification steps, the One-STrEP system<br />

requires one STEP only. The fast purification under physiological conditions makes even<br />

weakly associated protein complexes amenable to functional studies or mass spectroscopic<br />

identification. To even improve this unsurpassed system, new monoclonal antibodies against<br />

the One-STrEP-tag have been developed. One antibody called Strep-MAB binds nearly<br />

irreversibly to the One-STrEP-tag while the other exhibits easy controllable reversible<br />

binding properties. These complementary <strong>and</strong> Strep-Tactin independent One-STrEP-tag<br />

binding tools open new assay opportunities in protein:protein interaction studies which will be<br />

presented.<br />

- 114 -


Exploring ubiquitin <strong>signaling</strong> with dedicated PIQOR Ubiquitin-PS microarrays<br />

Hartmut Scheel, Corinna B. Scholz, Kay Hofmann<br />

Bioinformatics Group, Miltenyi Biotec GmbH, MACSmolecular Business Unit,<br />

Stöckheimer Weg 1, D-50829 Köln, Germany. E-mail: kay.hofmann@miltenyibiotec.de<br />

The MACSmolecluar business unit of Miltenyi Biotec offers smart products <strong>and</strong> services for<br />

molecular biology research <strong>and</strong> gene expression analysis via the proprietary PIQOR<br />

Microarray platform for topic-defined or custom expression profiling solutions.<br />

Here, we introduce the PIQOR Ubiquitin-PS microarray with a focus on the ubiquitinproteasome<br />

system (UPS). By developing this microarray, we are addressing the growing<br />

need for a deeper underst<strong>and</strong>ing of the UPS <strong>and</strong> a comprehensive monitoring of its<br />

components. It has become clear in the recent years that ubiquitylation plays a key role in<br />

processes like cell cycle progression, protein quality control, DNA repair, stress response,<br />

apoptosis <strong>and</strong> regulation of <strong>signaling</strong>.<br />

The importance of the UPS for signal transduction is not restricted to its well-known role in<br />

the selective degradation of regulatory proteins. Equally important is the role of ubiquitin<br />

modification as a signal by itself. The high complexity of the UPS is underscored by the large<br />

number of components involved in ubiquitin attachment, removal <strong>and</strong> recognition, all similar<br />

to the numbers known for phosphorylation <strong>signaling</strong>. A deregulation of the UPS is associated<br />

with a multitude of diseases, including cancer, catabolic diseases, inflammation <strong>and</strong><br />

neurodegeneration. Therefore, the UPS becomes more <strong>and</strong> more a rich s<strong>our</strong>ce for discoveries<br />

with therapeutic impact <strong>and</strong> ubiquitin research increases steadily.<br />

A hallmark of the UPS is that most proteins involved belong to a limited number of protein<br />

families characterized by common homology domains. The bioinformatics group of Miltenyi<br />

MACSmolecular possesses appropriate tools like the profile technique to mine sequence<br />

databases for proteins with a given homology domain. In a systematic UPS analysis<br />

programme applying this technique, a list of ~1200 genes was generated covering all aspects<br />

of the UPS <strong>and</strong> including many unpublished c<strong>and</strong>idate genes, e.g. approximately 600 E3<br />

ligases were identified. With this catalogue of genes at h<strong>and</strong>, a new microarray was designed,<br />

which allows the systematic detection of mRNA expression changes in the entire UPS. This<br />

new microarray fits nicely into the portfolio of other topic-defined microarrays offered by<br />

Miltenyi Biotec, which already includes microarrays designed for oncology <strong>and</strong> immunology.<br />

- 115 -


Antibody <strong>and</strong> protein delivery in living cells: presentation of a new potent reagent <strong>and</strong><br />

potential applications.<br />

Claire Weill<br />

Polyplus-transfection – Bioparc – BP 90018 Illkirch – FRANCE<br />

In cells, main molecular functions are carried out by proteins. Studying proteins, in terms of<br />

localizations, functions or regulations implies the possibility of transfecting such<br />

biomolecules into cells. Usually, cDNA corresponding to the coding sequence of the protein<br />

is transfected in cells by various methods. However in some cases, the expected protein is<br />

finally not expressed for diverse reasons. The function of a protein may also be elucidated by<br />

an indirect approach: inhibiting RNA coding for the protein, using gene silencing technology<br />

(siRNA). However, the more direct approach consists in introducing the protein of interest<br />

into the cells.<br />

Therefore, we have recently developed a potent reagent, named PULSin, for<br />

cytoplasmic protein delivery. This new protein delivery reagent forms non covalent<br />

complexes with proteins or antibodies. PULSin/protein complexes are internalized via<br />

anionic cell-adhesion receptors present on virtually all cells <strong>and</strong> are released into the<br />

cytoplasm where they disassemble. The protein ends up free, able to diffuse in the cytoplasm<br />

<strong>and</strong> to proceed to its function or target organelle. We will demonstrate that PULSin delivers<br />

efficiently anionic proteins in a large variety of living eukaryotic cells, including primary cells<br />

<strong>and</strong> suspension cells, without any side effect. Moreover, PULSin is also able to transfect<br />

antibodies, allowing users to study proteins or to block their activity in living cells.<br />

Using PULSin opens large areas of interest in various cell biology research domains<br />

like studying lethal proteins or controlling protein level <strong>and</strong> time c<strong>our</strong>se in living cells.<br />

- 116 -


Poster presentations<br />

Poster are classifiied by session<br />

I. Protein Domains<br />

II. Receptor <strong>signaling</strong> <strong>and</strong> G proteins<br />

III. Protein kinase cascades as therapeutic targets<br />

IV. Protein kinase inhibitors: insights into drug design from<br />

structure<br />

V. Phosphatases a key cell <strong>signaling</strong> intermediates<br />

VI. Proteasome degradation pathways<br />

VII. Stem cell specific cell <strong>signaling</strong> mechanisms<br />

VIII. <strong>Inflammation</strong> specific <strong>signaling</strong><br />

IX. Novel compounds targeting inflammatory <strong>signaling</strong> pathways<br />

X. Cell death in cancer<br />

XI. Cell death <strong>and</strong> cardiovascular diseases<br />

XII. Cell death <strong>and</strong> neurodegenerative diseases<br />

XIII. Cell <strong>signaling</strong> pathways leading to regulated chromatin<br />

modifications<br />

XIV. Transcriptional <strong>and</strong> translational control<br />

XV. Reactive oxygen species <strong>and</strong> cell <strong>signaling</strong><br />

XVI. Chemopreventive agents<br />

XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease<br />

<strong>and</strong> then by alphabetical order (first author)<br />

LATE abstracts are at the end of each session<br />

- 117 -


Session I. Protein Domains<br />

- 118 -


Session I : protein domains Poster I, 1<br />

Synthetic corticotropin-like peptide GKVLKKRR, corresponding to the fragment 81-88<br />

of human pro-interleukin-1alpha, is an antagonist of corticotropin receptor<br />

Elena V. Navolotskaya, Vera I. Vanina, Alex<strong>and</strong>er A. Kolobov*, Elena A. Kampe-<br />

Nemm*, Valery M. Lipkin<br />

Branch of Shemyakin <strong>and</strong> Ovchinnikov Institute of Bioorganic Chemistry of the<br />

Russian Academy of Sciences, Science Avenue, 6, 142290 Pushchino, Moscow Region,<br />

Russia, navolots@fibkh.serpukhov.su<br />

*State Research Center for Institute of Highly Pure Bioprepararions, 197110 St.<br />

Petersburg, Russia, kolobov@vs3283.spb.edu<br />

The corticotropin-like octapeptide GKVLKKRR (termed as leucorticotropin), which<br />

corresponds to the amino acid sequence 81-88 in the precursor of human interleukin-1alpha,<br />

was synthesized on automatic peptide synthesizer. Peptide was purified to homogeneity by<br />

reverse-phase HPLC (chromatograph Gilson, France) on Delta Pack C18 column. The<br />

peptide purity was > 99%. Leucorticotropin was labeled with tritium by the high-temperature<br />

solid-state catalytic isotope exchange reaction to specific activity of 20 Ci/mmol. Receptor<br />

binding studies revealed that [3H]leucorticotropin bound with high affinity <strong>and</strong> specificity to<br />

corticotropin receptor on rat adrenal cortex membranes (Kd = 2.2 nM). Leucorticotropin at<br />

concentrations of 0.1 – 1000 n! was found to have no influence on the adenylate cyclase<br />

activity in adrenal cortex membranes, while intranasal injection of leucorticotropin to rats at<br />

doses of 10 - 50 µg/kg was found to inhibit the secretion of 11-oxycorticosteroids from the<br />

adrenals to the bloodstream. Thus, leucorticotropin is an antagonist of corticotrophin<br />

receptor.<br />

- 119 -


Session I : protein domains Poster I, 2<br />

Preparation <strong>and</strong> characterization of recombinant chicken growth hormone (chGH) <strong>and</strong><br />

its putative antagonist chGH G119R mutein<br />

Helena E. Paczoska-Eliasiewicz1, Gili Salomon2, Shay Reicher2, Eugene E.<br />

Gussakowsky3, Anna Hrabia1 <strong>and</strong> Arieh Gertler2<br />

1Department of Animal Physiology, University of Agriculture, Kraków, Pol<strong>and</strong>,<br />

2Faculty of Agricultural, Food <strong>and</strong> Environmental Quality Sciences, The Hebrew<br />

University of Jerusalem, Israel <strong>and</strong> 3Department of Botany, University of Manitoba,<br />

Winnipeg, MB R3T 2N2, Canada.<br />

Synthetic cDNA of chicken GH (chGH) <strong>and</strong> its G119R mutein was synthesized according to<br />

published sequence but optimized for expression in E. coli. The respective cDNAs were<br />

inserted into pMON expression vector <strong>and</strong> transformed into Mon 105 E. coli strain. The<br />

proteins expressed upon induction with nalidixic acid were found almost entirely in the<br />

insoluble inclusion bodies (IBs). The IBs were isolated, the proteins solubilized in 4.5 M urea,<br />

at pH 11.3 in presence of cysteine, refolded <strong>and</strong> purified to homogeneity by anion-exchange<br />

chromatography on Q-Sepharose. The overall yields were 400 to 500 mg from 5 liters of<br />

fermentation. Both proteins were > 98% pure as evidenced by SDS-PAGE <strong>and</strong> contained at<br />

least 95% monomers as documented by gel-filtration chromatography on a SuperdexTM75<br />

column under not denaturing conditions. Circular dichroism analysis revealed that both<br />

proteins have identical secondary structure characteristic of cytokines, namely > 50% of alpha<br />

helix content. Chicken GH was capable of forming a 1:2 complex with recombinant oGHR-<br />

ECD (oGH receptor extracellular domain) though its affinity to ECD as determined by RRA<br />

was 11-fold lower than that of ovine GH (oGH). Correspondingly, its bioactivity, assessed<br />

using PDF-P1 3B9 cells stably transfected with rabbit GHR was 20-25 fold lower, whereas<br />

chGH G119R mutant did not bind to oGHR-ECD <strong>and</strong> was devoid any biological activity in<br />

PDF-P1 3B9 cells. In contrast, in binding experiments carried-out using chicken liver<br />

membranes both oGH <strong>and</strong> chGH showed similar EC50 values in competition with 125I-oGH.<br />

These EC50 <strong>and</strong> were 5-9 fold lower than that of G119R mutein. These results emphasize the<br />

importance of species specificity <strong>and</strong> indicate the possibility of antagonistic activity of chGH<br />

G119R.<br />

- 120 -


Session I : protein domains Poster I, 3<br />

Preparation of leptin antagonists by site-directed mutagenesis of human, ovine, rat <strong>and</strong><br />

mouse leptin's site III: implications on blocking undesired leptin action in vivo.<br />

Gili Salomon1, Leonora Niv-Spector1, Dana Berger1, Isabelle Callebaut2, Jean Djiane3<br />

<strong>and</strong> Arieh Gertler1*<br />

1Faculty of Agricultural, Food <strong>and</strong> Environmental Quality Sciences, The Hebrew<br />

University of Jerusalem, 2LMCP, CNRS UMR7590, Universites Paris 6 & Paris 7,<br />

France, 3Institut National de la Recherche Agronomique, NOPA, France.<br />

As no structural information of the 3D structure of leptin receptor (LEPR) is available the<br />

model of interleukin 6 (IL6) was applied. In that model, a hexameric complex is formed<br />

gradually, first by IL6 molecule which interacts with the IL6R-alpha, then with gp130<br />

forming an inactive trimer which subsequently dimerizes forming an active hexamer, whose<br />

formation is achieved due to interaction of IL6 bound in one trimer (through its site III) with<br />

the immunoglobulin domain (IGD) of gp130 of the other trimer. We have identified the<br />

putative leptin's binding site III by modeling LEPR, on the basis of its alignment with gp130,<br />

<strong>and</strong> fitting leptin on IL6 in the IL6/gp130 complex as leptin's amino acids 39-42 (LDFI),<br />

which are preserved in all leptin species. To verify this hypothesis <strong>and</strong> to test its generality we<br />

have prepared <strong>and</strong> purified to homogeneity human, ovine, rat <strong>and</strong> mouse triple<br />

(L39A/D40A/F41A) <strong>and</strong> quadruple (L39A/D40A/F41A/I42A, human <strong>and</strong> ovine only) leptin<br />

muteins. All six muteins had typical cytokine secondary structure, acted as true antagonists,<br />

namely they interacted with LEPR with affinity similar to the wild type hormone (as<br />

evidenced by SRP <strong>and</strong> RRA), were devoid of biological activity in several leptin-responsive<br />

bioassays <strong>and</strong> specifically inhibited leptin action. Those muteins can be prepared in gram<br />

amounts <strong>and</strong> thus serve as a novel tool of studying leptin function in vitro <strong>and</strong> in vivo. To<br />

prolong their life in circulation some muteins were pegylated using 40 kDa polyethylene<br />

glycol. Although pegylation decreases the affinity, increasing circulation half-life can<br />

recompensate this deficit so pegylated antagonists are expected to be more potent in vivo.<br />

Antagonizing leptin has been suggested as a possible therapy in auto-immune diseases <strong>and</strong><br />

heart failure. Thus leptin antagonists not only offer a novel tool to elucidate the role of leptin<br />

in mammalian physiology <strong>and</strong> pathology but have a potential of becoming a drug.<br />

- 121 -


Session I : protein domains Poster I, 4<br />

A novel Mitogen-Activated Protein Kinase docking site in the N terminus of MEK5alpha<br />

organizes the components of the Extracellular Signal-Regulated Kinase 5 <strong>signaling</strong><br />

pathway<br />

Jan Seyfried, Xin Wang, Giorgi Kharebava, <strong>and</strong> Cathy T<strong>our</strong>nier<br />

Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford<br />

Road, Manchester M13 9PT, UK<br />

The alternative splicing of the mek5 gene gives rise to two isoforms. MEK5beta lacks an<br />

extended N terminus present in MEK5alpha. Comparison of their activities led us to identify a<br />

novel mitogen-activated protein kinase (MAPK) docking site in the N terminus of MEK5 that<br />

is distinct form the consensus motif identified in the other MAPK kinases. It consists of a<br />

cluster of acidic residues at position 61 <strong>and</strong> positions 63-66. The formation of the<br />

MEK5/extracellular signal-regulated kinase 5 (ERK5) complex is critical for MEK5 to<br />

activate ERK5, to increase transcription via MEF2, <strong>and</strong> to enhance cellular survival in<br />

response to osmotic stress. Certain mutations in the ERK5 docking site that prevent<br />

MEK5/ERK5 interaction also abrogate the ability of MEKK2 to bind <strong>and</strong> activate MEK5.<br />

However, the identification of MEK5 alpha mutants with selective binding defect<br />

demonstrates that the MEK5/ERK5 interaction does not rely on the binding of MEK5a to<br />

MEKK2 via their respective PB1 domains. Altogether these results establish that the N<br />

terminus of MEK5a is critical for the specific organization of the components of the ERK5<br />

<strong>signaling</strong> pathway.<br />

- 122 -


Session I : protein domains Poster I, 5<br />

Analysis of transiently overexpressed Src Y527F on phosphoproteome signalling<br />

Jörg von Hagen, Uwe Michelsen, Sven Andrecht, Anja Seiler <strong>and</strong> Rob Hendriks<br />

Merck KGaA, Darmstadt, Germany<br />

Protein phosphorylation <strong>and</strong> dephosphorylation is a major <strong>signaling</strong> method for activating <strong>and</strong><br />

inactivating cell proteins. Here we describe the overexpression of constitutively activated Src<br />

Y527F <strong>and</strong> a novel method to identify target proteins down-stream of Src. Src plays important<br />

roles in cell differentiation, proliferation, <strong>and</strong> survival. For example, stimulation of the<br />

epidermal growth factor receptor, which leads to cell proliferation, results in Src activation.<br />

Src is activated during the G2/M transition. Src plays a role in cell adhesion, cell morphology<br />

<strong>and</strong> motility, <strong>and</strong> bone resorption.<br />

- 123 -


Notes<br />

- 124 -


Notes<br />

Notes<br />

- 125 -


- 126 -


Session II. Receptor <strong>signaling</strong> <strong>and</strong> G proteins<br />

- 127 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 1<br />

Developmental Responses of Chicken Ductus Arteriosus to Oxygen <strong>and</strong> Vasoactive<br />

Mediators<br />

Pia Ågren, Bea Zoer, Lilian Kessels, Jo GR De Mey, Carlos E Blanco <strong>and</strong> Eduardo<br />

Villamor.<br />

Departments of Pediatrics <strong>and</strong> Pharmacology, GROW Research Institute, University of<br />

Maastricht, P.O. Box 5800, 6202 AZ Maastricht, The Netherl<strong>and</strong>s. eiv@paed.azm.nl<br />

Background: At birth, the mammalian ductus arteriosus (DA) undergoes alterations in its<br />

pharmacological responsiveness. This phenomena has not been characterized in the chicken<br />

embryo.<br />

Objective: To characterize DA reactivity in the chicken embryo from the last stage of prenatal<br />

development <strong>and</strong> throughout the perinatal period.<br />

Design/Methods: Isolated DA from 15-d, 19-d non-internally-pipped embryos (total<br />

incubation: 21-d) <strong>and</strong> from 21-d externally-pipped embryos were mounted in a myograph <strong>and</strong><br />

bubbled with 5% O2 /5% CO2/90% N2. The contractile responses induced by O2 (0 to 21%),<br />

KCl, the adrenergic agonist norepinephrine (NE), the "1-adrenoceptor agonist phenylephrine<br />

(Phe), the inhibitor of Kv channels 4-aminopyridine (4-AP), the inhibitor of KATP channels<br />

glibenclamide, the inhibitor of KCa channels tetraethylammonium (TEA), the non-selective<br />

cyclooxygenase (COX) inhibitor indomethacin, the COX-1 inhibitor valeryl salicylate, the<br />

COX-2 inhibitor nimesulide <strong>and</strong> the relaxations (during 62.5 mM KCl-induced contraction)<br />

induced by acetylcholine (ACh), the NO donor sodium nitroprusside (SNP), the adenylate<br />

cyclase activator forskolin, <strong>and</strong> the #-adrenergic receptor agonist isoproterenol were tested.<br />

Results: Contractions induced by O2, KCl, <strong>and</strong> NE increased significantly between 15- <strong>and</strong><br />

21-d. but 4-AP-induced contractions did not change. Glibenclamide <strong>and</strong> TEA did not contract<br />

DA. Indomethacin, valeryl salicylate <strong>and</strong> nimesulide induced a weak contraction that was not<br />

significantly different to the one induced by the vehicle (ethanol). Relaxations induced by<br />

ACh, SNP <strong>and</strong> isoproterenol were markedly reduced in the 21-d embryo. In contrast,<br />

forskolin-induced relaxation was not affected by age. When the DA was divided in two<br />

segments, it was observed that the response to O2 <strong>and</strong> NE was significantly higher in the<br />

pulmonary end <strong>and</strong> the response to KCl <strong>and</strong> SNP was higher in the aortic end.<br />

Conclusions: Transition to ex ovo life is accompanied by dramatic changes in chicken DA<br />

reactivity. These changes are characterized by an increase in the contractile responses <strong>and</strong> a<br />

decrease in relaxation. The reactivity of chicken DA is not homogeneous <strong>and</strong> depends on the<br />

segment studied. Response to oxygen appears to be located in the pulmonary end of the DA.<br />

- 128 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 2<br />

Galpha12 interaction with alphaSNAP induces VE-cadherin localization at endothelial<br />

junctions <strong>and</strong> regulates barrier function<br />

Alex<strong>and</strong>ra V. Andreeva, Mikhail A. Kutuzov, Rita Vaiskunaite, Jasmina Profirovic,<br />

S<strong>and</strong>a Predescu, Asrar B. Malik <strong>and</strong> Tatyana A. Voyno-Yasenetskaya<br />

Department of Pharmacology, College of Medicine, University of Illinois at Chicago,<br />

Chicago, Illinois 60607, U.S.A. Email: a<strong>and</strong>reev@uic.edu<br />

In vascular endothelium, the endothelial-specific cadherin (VE-cadherin) is essential for the<br />

control of permeability <strong>and</strong> angiogenesis. Newly synthesized VE-cadherin is transported to<br />

the plasma membrane <strong>and</strong> forms a homotypic interaction with VE-cadherin from other<br />

endothelial cells, resulting in the assembly of adherens junctions. The involvement of<br />

heterotrimeric G proteins in the regulation of adherens junction function is unclear. Galpha12<br />

has recently been reported to bind directly to the C-terminal region of E-cadherin. In this<br />

study, we identified alphaSNAP, an essential component of the membrane fusion machinery,<br />

as an interactive partner of Galpha12 using yeast two hybrid screening. GST pull-down assays<br />

showed the selective interaction of alphaSNAP with Galpha12 in COS-7 as well as human<br />

umbilical vein endothelial cells (HUVECs). Using domain swapping experiments, we<br />

demonstrated that the N-terminal region of Galpha12 (1-37 aa) was necessary <strong>and</strong> sufficient<br />

for its interaction with alphaSNAP. Galpha13 with its N-terminal extension replaced by that<br />

of Galpha13 acquired the ability to bind to alphaSNAP, whereas Galpha12 with its Nterminus<br />

replaced by that of Galpha13 lost this ability. Using f<strong>our</strong> point mutations of<br />

alphaSNAP, which alter its ability to bind to the SNARE complex, we determined that the<br />

convex rather than concave surface of alphaSNAP was involved in its interaction with<br />

Galpha12. Co-transfection of HUVECs with Galpha12 <strong>and</strong> alphaSNAP stabilized VEcadherin<br />

at the plasma membrane, whereas downregulation of alphaSNAP using siRNA<br />

resulted in the loss of VE-cadherin from the cell surface <strong>and</strong>, when used in conjunction with<br />

Galpha12 overexpression, decreased endothelial barrier function. Our results demonstrate a<br />

direct link between the alpha-subunit of G12 <strong>and</strong> alphaSNAP, <strong>and</strong> suggest a role for this<br />

interaction in regulating the membrane localization of VE-cadherin <strong>and</strong> hence formation of<br />

adherens junctions, control of cell confluence <strong>and</strong> endothelial barrier function.<br />

- 129 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 3<br />

Rap1: a turnabout for the crosstalk between cadherin <strong>and</strong> integrin <strong>signaling</strong><br />

Fiorella Balzac*, Maria Avolio*, Simona Degani*, Floriana Francalanci*, Irina<br />

Kaverina†, Guido Tarone*, J. Victor Small† <strong>and</strong> S. Francesco Retta*<br />

*Department of Genetics, Biology <strong>and</strong> Biochemistry, University of Torino, 10126<br />

Torino, Italy;<br />

†IMBA - Institute of Molecular Biotechnology, Vienna, Austria.<br />

e-mail: francesco.retta@unito.it (S.F. Retta)<br />

The coordinate modulation of cadherin <strong>and</strong> integrin functions plays an essential role in<br />

fundamental physiological <strong>and</strong> pathological processes, including morphogenesis <strong>and</strong> cancer.<br />

However, the molecular mechanisms underlying the functional crosstalk between cadherins<br />

<strong>and</strong> integrins are still elusive.<br />

We show that the small GTPase Rap1, a crucial regulator of the inside-out activation of<br />

integrins, is a target for E-cadherin-mediated outside-in <strong>signaling</strong>. In particular, a strong<br />

activation of Rap1 occurs upon adherens junction disassembly <strong>and</strong> is triggered by E-cadherin<br />

internalization <strong>and</strong> trafficking along the endocytic pathway. In contrast, Rap1 activity is not<br />

influenced by integrin outside-in <strong>signaling</strong>. Moreover, the activation of Rap1 is associated<br />

with <strong>and</strong> controlled by an increased Src kinase activity, <strong>and</strong> is paralleled by the colocalization<br />

of Rap1 <strong>and</strong> E-cadherin at the perinuclear recycling endosome compartment, <strong>and</strong> the<br />

association of Rap1 with a subset of E-cadherin-catenin complexes that does not contain<br />

p120ctn. Finally, the E-cadherin endocytosis-dependent activation of Rap1 is associated with<br />

<strong>and</strong> is required for the formation of integrin-based focal adhesions.<br />

Our findings provide the first evidence of an E-cadherin-modulated endosomal <strong>signaling</strong><br />

pathway involving Rap1, <strong>and</strong> suggest that cadherins may have a novel modulatory role in<br />

integrin adhesive functions by fine-tuning Rap1 activation.<br />

- 130 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 4<br />

CXC receptors <strong>and</strong> chemokines expression in human meningioma: SDF1/CXCR4<br />

<strong>signaling</strong> activates ERK1/2 <strong>and</strong> stimulates meningioma cell proliferation.<br />

Federica Barbieri1, Adriana Bajetto1, Carola Porcile1, Aless<strong>and</strong>ra Pattarozzi1,<br />

Aless<strong>and</strong>ro Massa1, Gianluigi Lunardi1, Gianluigi Zona2, Aless<strong>and</strong>ra Dorcaratto3,<br />

Jean Louis Ravetti3, Renato Spaziante2, Gennaro Schettini1 <strong>and</strong> Tullio Florio1.<br />

1Pharmacology - Dept. of Biology, Oncology <strong>and</strong> Genetics <strong>and</strong> 2Neurosurgery - Dept.<br />

Neuroscience, Ophthalmology <strong>and</strong> Genetics, University of Genova, 3Pathology, San<br />

Martino Hospital – 16132 Genova, Italy. E-mail: federica.barbieri@unige.it<br />

Recent evidence indicates that cancer cells express chemokine (CK) receptors <strong>and</strong> that their<br />

<strong>signaling</strong> is crucial for tumor growth regulation, migration <strong>and</strong> angiogenesis. Here we<br />

analyzed the profiles of expression of CXC CK receptors <strong>and</strong> related CKs in surgical samples<br />

of human meningiomas. RT-PCR was used to identify the presence of mRNA for CXCR1-5<br />

<strong>and</strong> their main lig<strong>and</strong>s (growth-related oncogene, GRO1-2-3/CXCL1-2-3; interleukin 8, IL-<br />

8/CXCL8; monokine-induced g-interferon MIG/CXCL9; $-interferon-inducible-protein-10,<br />

IP-10/CXCL10; stromal cell-derived factor-1, SDF1/CXCL12; B-cell activating chemokine-<br />

1, BCA-1/CXCL13). All the five receptors displayed high percentages of positive cases.<br />

CXCR1 was expressed in 34/37, CXCR2 in 31/35, CXCR3 in 30/36, CXCR4 in 43/55 <strong>and</strong><br />

CXCR5 in 34/36 cases. Conversely, their lig<strong>and</strong>s showed a lower pattern of expression: IL-8<br />

<strong>and</strong> GRO1-3 were detected in 14/35 <strong>and</strong> 15/36 cases, respectively; IP-10 in 15/36, MIG in<br />

10/36, SDF1 in 29/55 <strong>and</strong> BCA-1 in 1/35. The exclusive binding between SDF1 <strong>and</strong> CXCR4<br />

suggests that the SDF1/CXCR4 interaction might play a uniquely biological role. To<br />

investigate the <strong>signaling</strong> mechanisms activated by SDF1 binding to CXCR4 in meningioma,<br />

f<strong>our</strong> short term cultures were obtained from tumor tissues. [3H]-thymidine uptake <strong>and</strong><br />

Western blot were carried out to evaluate the proliferative activity of SDF1 <strong>and</strong> the<br />

involvement of extracellular signal-regulated kinase (ERK1/2) activation. CXCR4 was<br />

functionally coupled as demonstrated by the significant increase of DNA synthesis in primary<br />

cultures which occurred in response to 12.5 nM SDF1. The SDF1-dependent proliferation was<br />

associated with a marked phosphorylation of ERK1/2 <strong>and</strong> was significantly reduced by<br />

PD98059 (a MEK inhibitor), thus linking the SDF1/CXCR4 pathway to meningioma cell<br />

proliferation via ERK1/2 signal transduction. The simultaneous expression of CXCR1-5 <strong>and</strong><br />

their CKs in meningiomas suggests that these receptor-lig<strong>and</strong> pairs could sustain the tumor<br />

proliferation <strong>and</strong>, in particular, the SDF1/CXCR4 axis may be important for this process.<br />

- 131 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 5<br />

Dysregulation of GABAergic Neurotransmission in Mood Disorders<br />

Hendrik Bielau1, Tomasz Gos1, Christian Mawrin2, Kurt Trübner3, Ralf Brisch1,<br />

Gabriela Meyer-Lotz1, Henrik Dobrowolny1, Bruno Baumann1, Hans-Gert Bernstein1,<br />

Bernhard Bogerts1<br />

1Department of Psychiatry, Psychotherapy und Psychosomatic Medicine <strong>and</strong> 2Institute<br />

of Neuropathology; Otto-von-Guericke-University, Leipziger Str. 44, 39120 Magdeburg,<br />

Germany, E-mail: Hendrik.Bielau@Medizin.Uni-Magdeburg.de<br />

3Institute of Legal Medicine, University of Essen, Hufel<strong>and</strong>str. 55, 45122 Essen,<br />

Germany<br />

Alterations of GABAergic neurotransmission are assumed to play a crucial roll in the<br />

pathophysiology of mood disorders. GABA acts via binding to A <strong>and</strong> B receptors, whereas<br />

the B receptor is G-protein-coupled. Glutamatdecarboxylase (GAD) is the key enzyme of<br />

GABA synthesis. The goal of <strong>our</strong> postmortem investigation was the bihemispherical<br />

immunohistochemical staining of GAD neurons in brain structures which are important for<br />

mood regulation. In 20 brains of patients with mood disorders <strong>and</strong> 19 controls (C) an<br />

immunohistochemical staining of GAD-65/67 neurons was performed in dorsolateral<br />

prefrontal cortex, orbitofrontal cortex, anterior cingulate cortex, superior temporal gyrus,<br />

hippocampus <strong>and</strong> medial <strong>and</strong> lateral thalamus with consecutive determination of neuronal<br />

density. In the diagnosis group were 11 patients with bipolar disorder (BD) <strong>and</strong> 9 patients<br />

with major depressive disorder (MDD). The data were tested statistically using ANOVA und<br />

post-hoc Tukey tests. The evaluation revealed significant differences between the groups (C,<br />

BD, MDD) in dorsolateral prefrontal cortex, orbitofrontal cortex, superior temporal gyrus <strong>and</strong><br />

hippocampus. The post-hoc tests demonstrated higher neuronal densities in unipolar patients<br />

compared to bipolar patients <strong>and</strong> controls in dorsolateral prefrontal cortex, superior temporal<br />

gyrus <strong>and</strong> hippocampus. In the orbitofrontal cortex a higher neuronal density was found in<br />

bipolar <strong>and</strong> unipolar patients compared to controls. In the bipolar group dose equivalents of<br />

antidepressants given prior to death correlated positively with the neuronal density in the<br />

hippocampus. In conclusion, the current data on GAD-65/67 point on a dysregulation of the<br />

GABAergic system in mood disorders. Possibly, existing deficits of GABAergic<br />

neurotransmission will be compensated or overcompensated by antidepressants. Moreover,<br />

preliminary data point toward a diminished density of GAD terminals.<br />

- 132 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 6<br />

GEF-H1 is involved in agonist-induced human pulmonary endothelial barrier<br />

dysfunction.<br />

Anna A. Birukova, Alex<strong>and</strong>er D. Verin, Konstantin G. Birukov<br />

Department of Medicine, University of Chicago, Chicago, Illinois 60637, U.S.A. E-mails:<br />

abirukov@medicine.bsd.uchicago.edu, averin@medicine.bsd.uchicago.edu,<br />

kbirukov@medicine.bsd.uchicago.edu,<br />

Endothelial cell (EC) permeability is precisely controlled by cytoskeletal elements (actin<br />

filaments, microtubules, intermediate filaments) <strong>and</strong> cell contact protein complexes (focal<br />

adhesions, adherens junctions, tight junctions). We have recently shown that the edemagenic<br />

agonist thrombin caused partial microtubule (MT) disassembly, which was linked to<br />

activation of small GTPase Rho, Rho-mediated actin remodeling, cell contraction <strong>and</strong><br />

dysfunction of lung EC barrier. GEF-H1 is a MT-associated Rho-specific guanosine<br />

nucleotide (GDP/GTP) exchange factor, which in MT-unbound state stimulates Rho activity.<br />

In this study we tested hypothesis that GEF-H1 may be a key molecule involved in Rho<br />

activation, myosin light chain phosphorylation, actin remodeling <strong>and</strong> EC barrier dysfunction<br />

associated with partial MT disassembly. Our results show that depletion of GEF-H1 or<br />

expression of dominant negative GEF-H1 mutant significantly attenuated permeability<br />

increase, actin stress fiber formation <strong>and</strong> increased MLC <strong>and</strong> MYPT1 phosphorylation<br />

induced by thrombin or MT-depolymerizing agent nocodazole. In contrast, expression of wild<br />

type or activated GEF-H1 mutants dramatically enhanced thrombin <strong>and</strong> nocodazole effects on<br />

stress fiber formation <strong>and</strong> cell retraction. These results show a critical role for the GEF-H1 in<br />

the Rho activation caused by MT disassembly <strong>and</strong> suggest GEF-H1 as a key molecule<br />

involved in crosstalk between MT <strong>and</strong> actin cytoskeleton in agonist-induced Rho-dependent<br />

EC barrier regulation.<br />

- 133 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 7<br />

Role of PAK1 <strong>and</strong> PAK2 in HGF stimulation of cancer cell motility<br />

Michael D. Bright, Anne J. Ridley<br />

Ludwig Institute for Cancer Research (UCL Branch), 91 Riding House Street, London,<br />

W1W 7BS, Engl<strong>and</strong>. E-mail : michael@ludwig.ucl.ac.uk<br />

p21-activated kinases (PAKs) are a family of six serine/threonine protein kinases which signal<br />

downstream of Rac <strong>and</strong> Cdc42. PAKs are divided into two groups with PAK1-3 in group 1<br />

<strong>and</strong> PAK4-6 in Group 2. Each PAK isoform has a C-terminal kinase domain <strong>and</strong> a Cdc42/Rac<br />

interacting binding (CRIB) domain with an N-terminal regulatory region. Group 1 PAKs also<br />

have an autoinhibitory domain (AID) which overlaps the CRIB domain <strong>and</strong> is essential to<br />

their regulation by Rac <strong>and</strong> Cdc42. PAKs are also regulated by phosphorylation at multiple<br />

sites <strong>and</strong> by localization via adapter proteins. PAKs are important in the regulation of<br />

cytoskeletal dynamics <strong>and</strong> are particulary interesting because they are overexpressed in some<br />

cancers <strong>and</strong> have been shown to regulate cancer cell migration <strong>and</strong> invasion.<br />

HGF stimulation induces scattering, enhanced motility <strong>and</strong> morphological changes in several<br />

epithelial cell types. We have studied a variety of human cancer cell lines that respond to<br />

HGF by scattering, <strong>and</strong> found that HGF stimulation of these cells rapidly stimulates<br />

phosphorylation of PAK1 <strong>and</strong> PAK2. In subsequent studies we have focussed on DU145<br />

prostate cancer cells, where HGF induces redistribution of #-catenin from cell:cell junctions<br />

to membrane ruffles as cells detach from each other <strong>and</strong> scatter. Interestingly, the scattering<br />

response of DU145 cells to HGF is dependent on substrate; they scatter when plated on<br />

plastic but not on glass. The effect of siRNA knockdown of PAK1 <strong>and</strong> PAK2 on HGFinduced<br />

scattering <strong>and</strong> motility is being investigated.<br />

- 134 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 8<br />

Expression of chemokine receptor CXCR4 (CD184) in B chronic lymphocytic leukaemia<br />

cells.<br />

Nicolaas H. C. Brons1, Valérie Palissot1, Chantal Schwartz1, Manon Bosseler1,<br />

Bernadette Leners1, Guy J Berchem1,2.<br />

1Laboratoire d'Hémato-Cancérologie Expérimentale, CRP-Santé, 84 Val Fleury, L1526<br />

Luxemb<strong>our</strong>g. E-mail : berchem.guy@chl.lu<br />

2Hémato-Cancérologie, Centre Hospitalier de Luxemb<strong>our</strong>g, 4 rue barblé, L1210<br />

Luxemb<strong>our</strong>g.<br />

Chronic lymphatic leukaemia (CLL) accounts for about 30% of adult leukemias in Western<br />

countries. This lymphoma is very indolent but relentless, with median survivals of almost a<br />

decade. The disease is characterized by the accumulation of a monoclonal population of CD5<br />

positive neoplastic B cells in secondary lymphoid organs, marrow, <strong>and</strong> blood. Although the<br />

slowly proliferating cells are sensitive to chemotherapeutic agents, chemotherapy is almost<br />

never curative <strong>and</strong> relapse inevitably follows. In 1975 Kanti Rai has described a clinical<br />

staging system which until now serves as basis for treatment decisions.<br />

The study was aimed to explore the CD184 also known as the CXCR4 chemokine receptor on<br />

B-lineage chronic lymphocyte leukaemia (B-CLL) cells of various disease stages <strong>and</strong> its<br />

relationship with others receptor expressions. CD 184 is expressed on CLL cells <strong>and</strong> normal<br />

B cells, it is one of the main receptors that mediate B cell entry in a secondary lymphoid<br />

tissues <strong>and</strong> their homing to T cell <strong>and</strong> B cell zones. Marrow stromal cells or nurselike cells<br />

constituvely secrete CXCR12, the lig<strong>and</strong> for CXCR4, thereby attracting <strong>and</strong> rescuing CLL B<br />

cells from apoptosis in a contact dependent fashion. Therefore, CD184 has been mentioned as<br />

a possible prognostic marker in CLL. Flow cytometry was used to detect the CXCR4<br />

expression in patients of different Rai stages The samples were measured on a FACSCanto<br />

(BD) <strong>and</strong> up to 6 markers per tube were analysed by the DivaSoftware (BD). Calibration<br />

curves for the MFE were obtained with Fluorospheres (DakoCytomation, K0110). More than<br />

thirty CD markers have been used to characterized B leukemic cells <strong>and</strong> correlation was<br />

researched with CD184. Of the 44 persons , 11 were healthy donors, 5 had Rai stage 0<br />

disease, 12 had Rai stage I disease, 3 had Rai stage II disease, 4 had Rai stage III disease, 9<br />

had Rai stage IV disease. CXCR4 was consistently expressed on circulating B-CLL with a<br />

mean fluorescence intensity higher than in cells from healthy volunteers. There was a<br />

correlation between CXCR4 expression <strong>and</strong> late stage of the disease. Interestingly,<br />

chemotherapeutic treatments seem to decrease level of this marker down to level of earliest<br />

stages. However, there was no correlation of CD184 level <strong>and</strong> quantitative expressions of<br />

known CD markers tested. Different correlations with previous treatments <strong>and</strong> known<br />

prognostic factors will be described <strong>and</strong> clinical implications will be discussed.<br />

- 135 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 9<br />

Genetically-encoded, FRET-based sensors for monitoring protein kinase <strong>signaling</strong><br />

Justin J. Brumbaugh, Christiane Jost, Carsten Schultz<br />

Gene Expression Program, European Molecular Biology Laboratory (EMBL),<br />

Heidelberg 69117, Germany. Email: brumbaug@embl.de, jost@embl.de,<br />

Schultz@embl.de<br />

Protein phosphorylation <strong>and</strong> dephosphorylation are important forms of posttranslational<br />

regulation <strong>and</strong> a fundamental means for controlling cellular fate <strong>and</strong> function. Deviant kinase<br />

<strong>and</strong> phosphatase activities are responsible for numerous diseases, <strong>and</strong> in particular, cancer.<br />

Due to the far reaching implications of phosphorylation <strong>and</strong> dephosphorylation, it is<br />

imperative to develop tools to closely monitor these dynamic processes in real time.<br />

Previously, we reported the construction of a genetically encoded FRET probe for monitoring<br />

protein kinase C (PKC) activity in living cells (Schleifenbaum et al., J. Am. Chem. Soc.,<br />

2004). To help unravel complex <strong>and</strong> often interdependent, intracellular <strong>signaling</strong> we are<br />

working to adapt this probe to other kinase specificities. To this end we have developed two<br />

novel protein kinase A (PKA) sensors <strong>and</strong>, to the best of <strong>our</strong> knowledge, the first genetically<br />

encoded dual parameter FRET probe that responds independently to PKA <strong>and</strong> PKC<br />

stimulation with differential output. Using an array-based approach <strong>and</strong> automated<br />

microscopy, we have also developed a method using in vivo imaging to screen potential<br />

probes under more comparable conditions <strong>and</strong> with higher throughput. This technology may<br />

lead to more versatile, cell-based assays for kinase activators <strong>and</strong> inhibitors.<br />

- 136 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 10<br />

Stimulation of signal transduction by mono(ADP-ribosyl)ation inhibitors: ecto-ADPRT<br />

as modulators of receptor stimulation?<br />

Claudia Cerella, Maria C. Albertini*, Augusto Accorsi*, Antonio Bergamaschi#, Maria<br />

D'Alessio, Milena De Nicola, Silvia Nuccitelli, Lina Ghibelli<br />

Dipartimento di Biologia, #Cattedra di Medicina del Lavoro, Universita' di Roma Tor<br />

Vergata, Via Ricerca Scientifica 1, 00133, Rome, Italy; *Istituto di Chimica Biologica A.<br />

Fornaini, Universita' di Urbino, Urbino, Italy. E-mail: cerella@uniroma2.it<br />

A set of mono(ADP-ribosyl)ation inhibitors (m-ADPRI) such as 3-aminobenzamide (3-<br />

ABA); nicotinamide (NA); vitamin K1 (K1); vitamin K2 (K2); novobiocin (NVB); miodobenzylguanidine<br />

(mIBG), but not PARP inhibitors (DPQ; low 3-ABA or NA doses)<br />

rapidly <strong>and</strong> specifically mobilise Ca2+ from a thapsigargin-sensitive store in U937 monocytic<br />

<strong>and</strong> Jurkat T lymphocitic cells, as well as their normal counterpart (peripheral blood<br />

monocytes <strong>and</strong> lymphocytes). A capacitative Ca2+ influx ensues. m-ADPRI-induced Ca2+<br />

mobilisation is insensitive to dantrolene, inhibitor of cyclic(ADP-ribose)-sensitive ryanodine<br />

channels. Instead, it is sensitive to U7322 <strong>and</strong> neomycin (neo), inhibitors of phospholipase C<br />

(PLC), the enzyme responsible for production of the endoplasmic reticulum (ER) Ca2+<br />

channels agonist IP3. Accordingly, m-ADPRI stimulate IP3 production. NA-, K2-, NVB- <strong>and</strong><br />

mIBG-induced Ca2+ mobilisation is also sensitive to pertussis toxin (PTX), indicating that<br />

these compounds activate PLC by stimulating G proteins (subtypes Gi). 3-ABA <strong>and</strong> K1 do<br />

not, thus possibly activating PLC by other means (tyrosin kinases?). The direct target(s) of m-<br />

ADPRI, may possibly be ecto-ADPRTs, a set of enzymes with poorly known functions<br />

localised on the external face of plasma membrane. Indeed, competition experiments between<br />

ADPRT substrate (NAD) <strong>and</strong> inhibitor (3-ABA) showed that extracellular NAD dosedependently<br />

inhibited 3-ABA-induced Ca2+ mobilisation. Our current model is that m-<br />

ADPRI may stimulate signal transduction by freeing receptors from a negative modulator<br />

(ADP-ribose) provided by ecto-ADPRTs.<br />

- 137 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 11<br />

Phospholipase C-g1 attenuates growth hormone <strong>signaling</strong> by forming a ternary complex<br />

with Jak2 <strong>and</strong> protein tyrosine phosphatase-1B<br />

Jang Hyun Choi1, Hyeon Soo Kim1, Sun-Hee Kim1, Yun Soo Bae2, H. Moo Kwon3,<br />

Sung Ho Ryu1, <strong>and</strong> Pann-Ghill Suh1*<br />

1 Department of Life Science, Pohang University of Science <strong>and</strong> Technology, Pohang,<br />

Kyungbuk, 790-784, Republic of Korea, E-mail : pgs@postech.ac.kr<br />

2 Divisions of Molecular Life Sciences, Center for Cell Signaling Research, Ewha<br />

Womans University, Seoul 120-750, Republic of Korea<br />

3 Divisions of Nephrology, University of Maryl<strong>and</strong>, Baltimore, Maryl<strong>and</strong> 21201, USA<br />

Growth hormone (GH) binds to its membrane receptor (GHR) <strong>and</strong> regulates many cellular<br />

functions such as proliferation, differentiation <strong>and</strong> chemotaxis. While the activation of GHmediated<br />

<strong>signaling</strong> is well understood, the precise mechanism responsible for its regulation<br />

has not been well assessed. In this study, we demonstrate that PLC-$1 modulates the action of<br />

GH-mediated <strong>signaling</strong> by interacting with tyrosine kinase Jak2 in a GH-dependent manner.<br />

In the absence of PLC-$1 (PLC-$1-/- MEFs), GH-induced JAK2 <strong>and</strong> STAT5<br />

phosphorylations significantly increased <strong>and</strong> the re-expression of PLC-$1 reduced GHinduced<br />

Jak2 activation. Furthermore, GH-induced JAK2 phosphorylation was enhanced by<br />

the specific knock-down of PLC-$1 using siRNA. Interestingly, PLC-$1 physically linked<br />

Jak2 with protein tyrosine phosphatase-1B (PTP-1B), which was implicated in the modulation<br />

of cytokine <strong>signaling</strong> via Jak2. In PLC-$1-/- MEFs, GH-dependent activation of both STAT-5<br />

<strong>and</strong> c-Fos were up-regulated, <strong>and</strong> GH-induced proliferation was potentiated. These results<br />

suggest that PLC-$1 plays a key role in regulation of GH-mediated <strong>signaling</strong> by attenuating<br />

the activation of JAK2.<br />

- 138 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 12<br />

ATP-dependent mechanotransduction by chondrocytes seeded in agarose constructs <strong>and</strong><br />

subjected to dynamic compression<br />

T T Chowdhury <strong>and</strong> M M Knight<br />

Cell <strong>and</strong> Tissue Engineering, Department of Engineering, Queen Mary, University of<br />

London, Mile End Road, London. E1 4NS. UK E-mail : t.t.chowdhury@qmul.ac.uk<br />

Introduction: Mechanical stimulation is essential in maintaining the biochemical properties of<br />

articular cartilage. Both .NO <strong>and</strong> calcium are well known intracellular <strong>signaling</strong> molecules<br />

which play an important role in chondrocyte mechanotransduction. It has been shown that<br />

chondrocytes respond to mechanical stimuli by releasing ATP. Extracellular ATP binds to the<br />

P2Y2 purinoreceptors, which may activate intracellular calcium levels <strong>and</strong> release of .NO,<br />

therefore influencing chondrocyte metabolism. The current study examines whether inhibition<br />

of mechanosensitive ATP by receptor antagonists will influence .NO levels, cell proliferation<br />

<strong>and</strong> proteoglycan synthesis by chondrocytes cultured in agarose constructs <strong>and</strong> subjected to<br />

dynamic compression.<br />

Methods: Bovine chondrocytes were seeded in agarose gel <strong>and</strong> equilibrated in culture for 24<br />

hrs. Using a full characterized cell-straining system, constructs were subjected to 15%<br />

dynamic compression at a frequency of 1Hz for 48 h<strong>our</strong>s in 1 ml of unsupplemented culture<br />

medium or in medium supplemented with 20 µM gadolinium chloride (putative<br />

mechanosensitive ion channel blocker), 100 µM suramin (P2 receptor antagonist) <strong>and</strong> 10<br />

units.ml-1 apyrase (catalyses extracellular ATP). Medium was additionally incubated with 1<br />

µCi.ml-1 [3H]-thymidine <strong>and</strong> 10 µM 35SO4, for the assessment of cell proliferation <strong>and</strong><br />

proteoglycan synthesis, respectively. Nitrite, a stable end product of .NO was measured using<br />

the Griess assay.<br />

Results: The application of dynamic compression resulted in the strain-induced stimulation of<br />

[3H]-thymidine incorporation in unsupplemented constructs (p


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 13<br />

The tyrosine 974 within the LIF-R-chain of the gp130/LIF-R heteromeric receptor<br />

mediates negative regulation of LIF signalling<br />

Thomas Clahsen1, Ute Lehmann1, Claudia Stross1, Heike M. Hermanns1, Rudolf<br />

Volkmer-Engert2, Jens Schneider-Mergener2, Peter C. Heinrich1, Fred Schaper1<br />

1 Department of Biochemistry, RWTH Aachen, Pauwelsstraße 30, D-52074 Aachen,<br />

Germany, E-mail: Thomas.Clahsen@post.rwth-aachen.de<br />

2 Department of Medical Immunology, Universitätsklinikum Charité, Molecular<br />

Libraries <strong>and</strong> Recognition, Ziegelerstrasse 5-9, Berlin D-10117, Germany<br />

Signalling of interleukin-6 <strong>and</strong> interleukin-11 through gp130 homodimeric receptor<br />

complexes has been analysed with respect to initiation <strong>and</strong> termination of signalling in great<br />

detail. Gp130 contains a crucial motif around tyrosine Y759 which mediates negative<br />

regulation through the feedback inhibitor SOCS3 <strong>and</strong> the protein tyrosine phosphatase SHP2.<br />

Signalling of LIF, CNTF, CT-1, CLC or OSM through gp130/LIF-R is believed to be similar<br />

due to the presence of the common signal transducer gp130 within the receptor complexes<br />

utilized, but the difference in the composition of gp130/gp130-homodimers <strong>and</strong> gp130/LIF-Rheterodimers<br />

is likely to be reflected in different signalling. Here, we analysed the<br />

contribution of the LIF-R within the gp130/LIF-R complex to negative regulation mediated<br />

by SHP2 <strong>and</strong> SOCS3. We show that SHP2 contributes to the negative regulation of signalling<br />

through gp130/LIF-R complexes. The inhibitory tyrosine motifs within the cytoplasmic parts<br />

of gp130 <strong>and</strong> the LIF-R act independently. Whereas SHP2 <strong>and</strong> SOCS3 bind directly to the<br />

inhibitory motif of gp130, only SHP2 was found to bind to the corresponding inhibitory<br />

sequence of the LIF-R. This observation was further corroborated by experiments indicating<br />

that mainly gp130 contributes to the inhibition of signalling by SOCS3.<br />

- 140 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 14<br />

Serotonin-induced inhibition of voltage-gated K+ currents <strong>and</strong> contraction in<br />

pulmonary arteries. Signalling via 5-HT(2A) receptors.<br />

Laura Cobeño, Angel Cogolludo, Laura Moreno, Giovanna Frazziano, Tizziana Russo,<br />

Juan Tamargo <strong>and</strong> Francisco Perez-Vizcaino.<br />

Department Pharmacology. School Medicine. Universidad Complutense de Madrid.<br />

28040 Madrid. SPAIN. E-mail: acogolludo@ift.csic.es<br />

Multiple lines of evidence indicate that serotonin (5-HT) <strong>and</strong> voltage-gated potassium (Kv)<br />

channels independently play a central role in the pathogenesis of pulmonary hypertension<br />

(PH). We hypothesized that 5-HT might module the activity of Kv channels, therefore,<br />

establishing a link between these pathogenetic factors in PH. We studied the effects of 5-HT<br />

on Kv currents in rat pulmonary artery smooth muscle cells (PASMC) <strong>and</strong> on contractile<br />

force in isolated pulmonary arteries (PA). 5-HT reduced native Kv currents, depolarized<br />

PASMC <strong>and</strong> caused a contraction of PA. The 5-HT(2A) receptor antagonist ketanserin <strong>and</strong><br />

the 5-HT(1B) receptor antagonist SB224289 inhibited 5-HT-induced contraction by 72 ± 4%<br />

<strong>and</strong> 22 ± 2%, respectively. The contraction induced by 5-HT was also attenuated by the 5-HT<br />

transporter (5-HTT) inhibitor fluoxetine, which also antagonizes 5-HT(2A) receptors, but not<br />

by other 5-HTT inhibitors such as fluvoxamine or citalopram. The inhibitory effects of 5-HT<br />

on Kv currents were prevented by ketanserin <strong>and</strong> fluoxetine, but not by any of the other drugs.<br />

Furthermore, ketanserin inhibited the depolarizing effect induced by 5-HT. 5-HT induced<br />

inhibition of Kv current was insensitive to the PKCzeta pseudosubstrate inhibitor. In contrast,<br />

inhibition of phospholipase C (PLC, U-73122), classic PKCs (Go-6976) or tyrosine kinases<br />

(genistein <strong>and</strong> tyrphostin 23) prevented the effects of 5-HT on Kv currents <strong>and</strong> contractions.<br />

Altogether these results indicate that 5-HT inhibits native Kv currents following the activation<br />

of 5-HT(2A) receptors via PLC, classic PKCs <strong>and</strong> tyrosine kinase. The inhibition of Kv<br />

channels contributes to 5-HT-induced pulmonary vasoconstriction <strong>and</strong> might play a role in<br />

PH.<br />

- 141 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 15<br />

Thromboxane A2-induced inhibition of Kv currents via PKCzeta: Role of reactive<br />

oxygen species <strong>and</strong> Kv1.5 channels<br />

Angel Cogolludo, Laura Moreno, Giovanna Frazziano, Federica Lodi, Laura Cobeño,<br />

Valeria Queirolo, Eduardo Villamor, Juan Tamargo <strong>and</strong> Francisco Perez-Vizcaino.<br />

Department Pharmacology. School Medicine. Universidad Complutense de Madrid.<br />

28040 Madrid. SPAIN. E-mail: acogolludo@ift.csic.es.<br />

Voltage-gated potassium channels (Kv) <strong>and</strong> thromboxane A2 (TXA2) have been involved in<br />

several forms of human <strong>and</strong> experimental pulmonary hypertension. We have reported that the<br />

TXA2 analog U46619, via activation of TP receptors, inhibited Kv currents in rat pulmonary<br />

artery smooth muscle cells (PASMC), increased cytosolic calcium <strong>and</strong> induced a contractile<br />

response in isolated rat <strong>and</strong> piglet pulmonary arteries (PA). These effects were inhibited by<br />

non selective PKC inhibitors <strong>and</strong> by the selective PKCzeta pseudosubstrate inhibitor. Herein,<br />

we have analyzed the role of reactive oxygen species <strong>and</strong> Kv1.5 channels, a major channel<br />

protein contributing to Kv currents in PASMC, in the signalling pathway. U46619 inhibited<br />

Kv currents in native PASMC <strong>and</strong> these effects were strongly inhibited by addition of catalase<br />

to the internal solution in the patch pipette. The membrane permeable hydroperoxide t-butyl<br />

hydroperoxide also inhibited Kv currents. The contractile responses to U46619 in isolated<br />

pulmonary arteries were inhibited by polyethylenglycol-catalase (PEG-catalase, a membrane<br />

permeable form of catalase) <strong>and</strong> the NADPH oxidase inhibitors DPI <strong>and</strong> apocynin. U46619<br />

increased dichlorofluorescein fluorescence, an indicator of intracellular hydrogen peroxide, in<br />

pulmonary arteries <strong>and</strong> this effect was prevented PEG-catalase. U46619 also inhibited Kv<br />

currents in Ltk(-) cells stably expressing hKv1.5 channels. These cells strongly expressed<br />

PKCzeta as measured by Western blot. In conclusion, activation of NADPH oxidase <strong>and</strong> the<br />

subsequent production of hydrogen peroxide are involved in the Kv channel inhibition <strong>and</strong> the<br />

contractile response induced by the TXA2 analog U46619 in rat PA. Kv1.5 channels are<br />

possible targets for the inhibitory effect of TXA2 in native pulmonary arteries.<br />

- 142 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 16<br />

Purification of G protein-coupled receptors <strong>and</strong> associated protein complexes under<br />

native condition<br />

Avais M. Daulat*, Jean-Luc Guillaume*, Pascal Maurice*, Philippe Delagrange$, Ralf<br />

Jockers*#.<br />

* Institut Cochin, Département Biologie Cellulaire. Inserm, U567. CNRS, UMR 8104.<br />

Université Paris Descartes, Faculté de Médecine René Descartes, UMR-S 8104, Paris, F-<br />

75014 France. # Email jockers@cochin.inserm.fr<br />

$ Institut de Recherche SERVIER, Suresnes, France<br />

G Protein-Coupled Receptors (GPCRs) constitute the largest family of membrane receptors<br />

<strong>and</strong> are targeted by about half of the drugs prescribed for human diseases. In contrast to other<br />

protein families the “interactom”, the repertoire of proteins that interact with a given protein,<br />

is only poorly known for GPCRs. Faster progress in the field has been mainly hampered by<br />

the fact that “classical” protein-protein interaction approaches such as yeast two-hybrid <strong>and</strong><br />

affinity purification techniques, are not well-adapted for these multi-transmembrane-spanning<br />

proteins.<br />

Recently, the t<strong>and</strong>em affinity purification (TAP) method has been developed to successfully<br />

purify soluble protein complexes. Here, we adapted this method to the purification of GPCRassociated<br />

protein complexes. This technique relies on a two-step purification protocol of<br />

proteins that associate with the TAP-tagged bait protein in living cells. We established two<br />

HEK 293 clones each stably expressing a distinct C-terminally TAP-tagged GPCR. Among<br />

different detergents tested, digitonin <strong>and</strong> Brij 96 were selected for their ability to solubilize<br />

high amounts of functional receptors. Typical purification yields varied between 20 <strong>and</strong> 30%.<br />

The successful co-purification of receptor-associated proteins was shown by the presence of<br />

heterotrimeric G proteins as determined by western blot <strong>and</strong> mass spectrometric analysis.<br />

Large-scale purifications yielded in coomassie blue stainable protein amounts that were<br />

identified by mass spectrometry.<br />

In conclusion, we established a purification procedure for the isolation of protein complexes<br />

that associated with GPCRs in living cells under basal <strong>and</strong> activated conditions.<br />

- 143 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 17<br />

Functional overlapping between caspase cleavage site <strong>and</strong> CBL binding sequence in the<br />

juxtamembrane domain of the MET receptor.<br />

Julien DEHEUNINCK, Catherine LEROY, Zongling JI, Bénédicte FOVEAU, Vincent<br />

VILLERET, David TULASNE <strong>and</strong> Véronique FAFEUR<br />

CNRS UMR 8117, Institut de Biologie de Lille, Institut Pasteur de Lille, B.P.447, 59021<br />

Lille, FRANCE. E-mail : veronique.fafeur@ibl.fr<br />

Hepatocyte growth factor/ scatter factor (HGF/SF) binds <strong>and</strong> activates the MET tyrosine<br />

kinase receptor to transduce cell survival. Nonetheless, we previously demonstrated that stress<br />

stimuli cause a caspase-dependent cleavage in the MET juxtamembrane domain, which<br />

converts MET into a pro-apoptotic p40 MET fragment. The caspase 3 cleavage occurs at<br />

aspartic acid residue D1000. This residue is juxtaposed to tyrosine Y1001, which is rather<br />

involved in binding CBL upon MET activation by its lig<strong>and</strong>, allowing MET trafficking <strong>and</strong><br />

recycling. We wished to examine the functional relationships between caspase-dependent<br />

cleavage <strong>and</strong> CBL binding at the ESVDY sequence of the juxtamembrane domain of MET.<br />

We first established that the caspase cleavage- <strong>and</strong> CBL binding- sites overlapped, since a<br />

MET D1000N mutant lost the ability both to generate p40 MET <strong>and</strong> to recruit CBL. We then<br />

individually mutated all amino acids of the ESVDY sequence of MET constituting the<br />

caspase <strong>and</strong> CBL sites. While a MET V999A mutant was selectively defective in generating<br />

p40 MET, a MET Y1001F mutant was selectively defective in recruiting CBL, demonstrating<br />

that caspase-dependent cleavage <strong>and</strong> CBL recruitment are dissociable mechanisms. We<br />

further examined the consequence of MET phosphorylation at CBL binding site on the<br />

caspase-dependent cleavage. Using in vitro MET phosphorylation <strong>and</strong> dephosphorylation, we<br />

demonstrated that generation of p40 MET is unfavored when MET is phosphorylated, <strong>and</strong><br />

vice versa. These data were strengthened by modeling studies using the MET peptide<br />

sequence ESVDYR <strong>and</strong> the known structure of caspase 3 or CBL in complex with specific<br />

peptide sequences. These studies show that a phosphorylated Y1001 is not compatible with<br />

specific association of caspase-3 to MET <strong>and</strong> also exclude simultaneous fixation of both<br />

caspase 3 <strong>and</strong> CBL to MET.<br />

In conclusion, we demonstrated that the caspase-dependent cleavage of MET <strong>and</strong> binding of<br />

CBL to the activated MET receptor are dissociable <strong>and</strong> incompatible mechanisms. As a<br />

consequence, we propose that HGF/SF can protect MET against caspase-dependent cleavage<br />

by activating autophosphorylation of the receptor.<br />

- 144 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 18<br />

Identification of PAR1-G protein signalling pathways involved in thrombin-induced<br />

CCL2 release<br />

Xiaoling Deng, Paul F Mercer, Geoffrey J Laurent <strong>and</strong> Rachel C Chambers.<br />

Centre for Respiratory Research, University College London, 5 University Street,<br />

WC1E 6JJ, London, Engl<strong>and</strong>. E-mail: x.deng@ucl.ac.uk<br />

Introduction: In experimental models of lung injury, activation of proteinase-activated<br />

receptor-1 (PAR1) by coagulation proteinases is critical in driving both the inflammatory <strong>and</strong><br />

fibrotic response. PAR1 exerts its pluripotent cellular effects by concomitant activation of<br />

G"i/o, G"q <strong>and</strong> G"12/13. We have previously shown that protection from lung inflammation<br />

<strong>and</strong> fibrosis in PAR1 deficient (PAR1 KO) mice is accompanied by reduced lung levels of the<br />

potent thrombin-inducible protein CCL2 (MCP-1/JE). The aim of this study was to examine<br />

the signalling pathways by which PAR1 activation induces the release of this chemokine by<br />

cultured mouse lung fibroblasts. Methods <strong>and</strong> Results: Wild type mouse lung fibroblasts<br />

(MLFs) were stimulated with thrombin, TFLLR-NH2 <strong>and</strong> RLLFT-NH2 (PAR1 peptide<br />

agonist <strong>and</strong> control peptide respectively), <strong>and</strong> CCL2 protein release into cell culture<br />

supernatants was assessed by ELISA. Both thrombin <strong>and</strong> TFLLR induced CCL2 release by<br />

MLFs in a time- <strong>and</strong> dose-dependent manner. No response was obtained with the control<br />

peptide RLLFT-NH2 or in PAR1 KO fibroblasts. The PAR1 specific antagonist RWJ-58259<br />

(1µM) completely blocked thrombin induced-CCL2 release. In order to investigate potential<br />

signalling pathways involved the effects of pertussis toxin (PTX, G"i/o inhibitor), Ro-318425<br />

(Protein Kinase C (PKC) inhibitor <strong>and</strong> U0126 (MEK1/2 inhibitor) on PAR1 activation<br />

induced-CCL2 release was also assessed. PTX inhibition of G"i/o had no effect on CCL2<br />

release induced by thrombin, whereas Ro-318425 inhibition of PKC reduced CCL2 release by<br />

49±6%, <strong>and</strong> U0126 inhibition of MEK1/2 reduced CCL2 release by 53±8% (all p


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 19<br />

Human Recombinant Vasostatin 1 May Interfere with Cell-Extracellular Matrix<br />

Interactions<br />

Valentina Di Felice1, Francesco Cappello1, Antonella Montalbano1, Nella Ardizzone1,<br />

Claudia Campanella1, Angela De Luca1, Daniela Amelio 2, Bruno Tota2, Angelo Corti3<br />

<strong>and</strong> Giovanni Zummo1<br />

1Human Anatomy Section, Di.Me.S., University of Palermo, Via del Vespro 129, 90127<br />

Palermo, Italia, e-mail: valentina.difelice@unipa.it; 2 Sezione di Fisiologia Organismale,<br />

Dipartimento di Biologia Cellulare, Università della Calabria CAP 87036, Arcavacata di<br />

Rende, Cosenza, Italia, e-mail: tota@unical.it; 3 DIBIT –Department of Oncology, San<br />

Raffaele H Scientific Institute Via Olgettina 58, 20132, Milano, Italia, E.mail<br />

corti.angelo@hsr.it.<br />

Vasostatin 1 (VS-1), the N-terminal fragment derived from the cleavage of Chromogranin A<br />

(CgA), has been shown to exert several biological activities on several tissues <strong>and</strong> organs [1].<br />

Recently, it has been reported that human recombinant VS-1 (STA-CgA 1-78) may alter<br />

myocardial contractility in eel, frog <strong>and</strong> rat hearts [2]. In this study we have explored if STA-<br />

CgA 1-78 can induce intracellular cascades interacting both with adhesion molecules <strong>and</strong>/or<br />

extracellular matrix components (ECM), i.e. involvement of the heat shock protein 90<br />

(HSP90) <strong>and</strong> the endothelial NOS (eNOS), known to be implicated in signal transduction<br />

mechanisms affecting myocardial contractility. We used 3D-cultured adult rat cardiomyocytes<br />

cultivated over fibronectin or fibroblasts or embedded in Matrigel or Collagen type I. Aurion<br />

conjugated VS-1 (Au-STA-CgA1-78) has been used to identify possible sites of interaction of<br />

this molecule with the cell membrane. We found that in <strong>our</strong> 3D-colture, cell-ECM<br />

interactions played a crucial role in the cellular localization of HSP90 as well as in the<br />

expression of eNOS. VS-1 appeared to modulate cell-ECM interactions, thereby remarkably<br />

leading to a different cellular localization of HSP90. Moreover, Au-STA-CgA1-78 was never<br />

detected inside the cell nor overlapping the plasma membrane, but nearby the outer side of the<br />

cardiomyocyte plasmalemma, at a particular distance, typical of integrins. On the whole, these<br />

data suggest that VS-1 does not have a classic receptor on the membrane but that integrins<br />

may represent a non-conventional VS-1 receptor modulating eNOS signalling pathway.<br />

[1] Helle KB (2004) Biol. Res 79 :769-794; [2] Cerra MC, De Iuri L, Angelone T, Corti A,<br />

Tota B (2005) Bas Res Cardiol 100:1-10.<br />

- 146 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 20<br />

Angiotensin receptor signal transduction involving src <strong>and</strong> MAPK in rat nucleus tractus<br />

solitarius<br />

Takayuki Endoh <strong>and</strong> Takashi Suzuki<br />

Department of Physiology, Tokyo Dental College, 1-2-2 Masago, Mihama-ku, Chiba<br />

261-8502, Japan. E-mail : tendoh@tdc.ac.jp<br />

It is recognized that brain contains all the components of the renin-angiotensin systems<br />

(RAS). The nucleus tractus solitarius (NTS) is known to plays a major role in the regulation<br />

of cardiovascular, respiratory, gustatory, hepatic <strong>and</strong> swallowing functions. Voltagedependent<br />

calcium channels (VDCCs) serve as crucial mediators of membrane excitability<br />

<strong>and</strong> Ca2+-dependent functions. The purpose of this study was to investigate the effects of<br />

Angiotensin (Ang) on VDCCs currents (ICa) in the NTS using patch-clamp recording<br />

methods. An application of Ang caused facilitation of L-type ICa. AT1 receptors antagonist,<br />

Losartan antagonized the Ang-induced facilitation of ICa. Intracellular dialysis of the Giprotein<br />

antibody attenuated the Ang-induced facilitation of ICa. Both Src tyrosine kinase<br />

inhibitor <strong>and</strong> mitogen activated protein kinase (MAPK) inhibitor attenuated the Ang-induced<br />

facilitation of ICa. p38 MAPK inhibitor also attenuated the Ang-induced facilitation of ICa.<br />

These results indicate that Ang facilitates L-type VDCCs via Gi-proteins involving Src<br />

tyrosine kinase <strong>and</strong> p38 MAPK kinase mediated by AT1 receptors in NTS.<br />

- 147 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 21<br />

Alzheimer’s Amyloid Precursor Protein in Human Sperm<br />

Margarida Fardilha, S<strong>and</strong>ra I. Vieira, Sara C. T. S. Domingues, Odete A. B. da Cruz e<br />

Silva <strong>and</strong> Edgar F. da Cruz e Silva<br />

Center for Cell Biology, University of Aveiro, 3810-193 Aveiro, Portugal (E-mail:<br />

dacruze@bio.ua.pt)<br />

The Alzheimer’s Amyloid Precursor Protein (APP) is a type I transmembrane glycoprotein<br />

with receptor-like characteristics that originates the Abeta peptide by proteolytic processing.<br />

Abeta is potentially cytotoxic <strong>and</strong> the major component of the cerebral amyloid plaques in<br />

Alzheimer’s Disease (AD) patients. Nonetheless, APP is ubiquitously expressed in<br />

mammalian cells with a broad tissue distribution, <strong>and</strong> Abeta deposition has been reported to<br />

occur in many cells outside the nervous system. Many putative functions have been suggested<br />

for APP, although its precise physiological role remains to be elucidated. As several results<br />

point to a role of chronic inflammation in AD pathogenesis <strong>and</strong> suggest that AD might be a<br />

systemic disorder, the importance of APP function in non-neuronal cells/tissues has gained<br />

further relevance. Previous studies have shown that APLP2 is found in testis <strong>and</strong> sperm. Our<br />

results are consistent with those observations but also point to the presence of APP itself in<br />

human sperm. The present immunocytochemical studies using a variety of antibodies that<br />

either recognize APP-specific epitotes or epitopes shared with other APP family members<br />

reveal quite distinct distributions in human sperm, thus suggesting a putative role for this<br />

important protein in sperm function. We will present the results obtained <strong>and</strong> discuss their<br />

significance for sperm signal transduction mechanisms.<br />

Supported by FCT <strong>and</strong> CBC.<br />

- 148 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 22<br />

Sequential caspase cleavages generate pro-apoptotic factor from MET tyrosine kinase<br />

receptor<br />

Bénédicte FOVEAU*, Catherine LEROY*, Julien DEHEUNINCK, Zongling JI,<br />

Véronique FAFEUR <strong>and</strong> David TULASNE.<br />

*These authors contributed equally to the study<br />

CNRS UMR 8117, Institut de Biologie de Lille, Institut Pasteur de Lille, B.P.447, 59021<br />

Lille, FRANCE.<br />

The receptor of hepatocyte growth factor/scatter factor (HGF/SF), the MET tyrosine kinase<br />

receptor, is known to be essential for normal development <strong>and</strong> cell survival. We previously<br />

showed that stress stimuli cause a cleavage in the MET juxtamembrane domain that converts<br />

the receptor into a pro-apoptotic 40 kDa fragment (p40 MET). We report here an additional<br />

caspase cleavage of MET occurring in its extreme C-terminal region. The cleavage occurs at<br />

aspartic residue 1374 within the DNID sequence of mouse MET. Although this cleavage<br />

removes only the five last amino acids, it has important functional consequences. First, the Cterminal<br />

cleavage favors the juxtamembrane one, leading to efficient generation of the proapoptotic<br />

p40 MET fragment. Second, the p40 MET generated by the two steps cleavage has<br />

enhanced pro-apoptotic activity. These results revealed a tight regulation of MET caspase<br />

cleavages resulting in the reshaping of a survival receptor to a pro-apoptotic factor.<br />

- 149 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 23<br />

Involvement of hydrogen peroxide in the activation of BKCa currents <strong>and</strong> vasodilator<br />

effects of quercetin in rat coronary arteries<br />

Giovanna Frazziano, Angel Cogolludo, Laura Moreno, Ana Briones, Federica Lodi,<br />

Laura Cobeño, Mercedes Salaices, Juan Tamargo, <strong>and</strong> Francisco Perez-Vizcaino.<br />

Department Pharmacology. School Medicine. Universidad Complutense de Madrid.<br />

28040 Madrid. SPAIN. E-mail: frazzygio@jumpy.it<br />

Calcium-activated potassium channels (BKCa) regulate coronary artery tone in vivo, play a<br />

key role in blood pressure regulation <strong>and</strong> have been suggested as novel potential drug targets<br />

in hypertension. The dietary flavonoid quercetin exerts systemic <strong>and</strong> coronary vasodilator<br />

effects in vitro, reduces blood pressure in several rat models of hypertension <strong>and</strong> its<br />

consumption is associated with a lower mortality rate from coronary heart disease in<br />

epidemiological studies. We hypothesized that quercetin might activate BKCa channel in<br />

isolated myocytes from rat coronary arteries <strong>and</strong> that this mechanism might be involved in its<br />

coronary artery relaxant effects. Membrane currents were measured using the whole-cell<br />

configuration of the patch-clamp technique. Quercetin (1-30 microM) increased the outward<br />

currents in the whole range of test potentials <strong>and</strong> increased the frequency of spontaneous<br />

transient outward currents (STOCs) carried by BKCa channels. These effects were abolished<br />

by the selective BKCa blocker iberiotoxin <strong>and</strong> by the hydrogen peroxide scavenger catalase in<br />

the internal pipette solution. The membrane permeable hydroperoxide t-butyl hydroperoxide<br />

also increased outward currents. Quercetin increased dichlorofluorescein fluorescence, a<br />

widely used indicator of intracellular hydrogen peroxide, in coronary arteries with a similar<br />

magnitude <strong>and</strong> time-c<strong>our</strong>se than t-butyl hydroperoxide. Quercetin-induced increase was<br />

prevented by polyethylenglycol-catalase, indicating the role of cytosolic hydrogen peroxide.<br />

The vasodilator effect of low concentrations of quercetin in isolated rat coronary arteries was<br />

inhibited by iberiotoxin. In conclusion, quercetin increased BKCa currents via production of<br />

intracellular hydrogen peroxide. This effect is involved, at least partly, in the coronary<br />

vasodilator effects of quercetin.<br />

- 150 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 24<br />

Role of rafts on µ <strong>and</strong> %-opioid receptor pharmacology in plasma membrane of<br />

mammalian cells.<br />

G.Gaibelet, A.Andre, C.Lebrun, B.Lagane, C.Millot, S.Poupard, A.Saulière, A.Lopez<br />

<strong>and</strong> L.Cezanne<br />

IPBS/CNRS, 205 route de Narbonne, 31062 TOULOUSE cedex, France.<br />

A large body of evidence indicates lateral heterogeneity of lipid <strong>and</strong> protein distribution in the<br />

biological membranes. In this way, lipid assemblies as rafts composed mainly of<br />

sphingomyelin <strong>and</strong> cholesterol have been proposed as lipid-platforms which segregate<br />

membrane components within the cell membrane. In this context these confinements are<br />

expected to be relevant by concentrating interacting molecules in particular regions.<br />

Assuming that lipid rafts contain specific sets of protein including for example Galpha<br />

subunits of heterotrimeric G-protein, we explored the role of rafts in regulating GPCR<br />

functionnality. The RCPGs chosen for <strong>our</strong> studies are the human µ (hMOR) <strong>and</strong> % opioid<br />

receptors (hDOR) expressed in mammalian plasma membrane.<br />

Effect of cholesterol has been evaluated in term of lig<strong>and</strong> binding on both receptors.<br />

Cholesterol depletion affects agonist binding. Surprisingly, for hMOR, this effect is observed<br />

only in presence of Gpp(NH)p. Cholesterol re-complementation restored binding properties in<br />

the two cases, indicating a cholesterol specific effect. Conversely, antagonist binding did not<br />

decrease after cholesterol depletion. These results suggest that a population of high-affinity<br />

receptor state require a rich-cholesterol environment. However, in the case of hMOR,<br />

cholesterol is not sufficient <strong>and</strong> require the presence of G-proteins. In addition, GTP$S<br />

binding experiments, carried out on plasma membrane from cells expressing hMOR, showed<br />

that the coupling efficiency is affected after depletion, suggesting the existence of two distinct<br />

pharmacological behavi<strong>our</strong>. The distribution of receptors in rich <strong>and</strong> poor cholesterol domains<br />

have been investigated. We propose a correlation between the pharmacological properties <strong>and</strong><br />

the distribution of the components of cells membrane expressing hMOR or hDOR.<br />

- 151 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 25<br />

THE SIGNALING GATEWAY MOLECULE PAGES DATABASE<br />

Clare Garvey1, Brenda Riley1, Monica Hoyos-Flight1, Bernd Pulverer1, Barbara<br />

Marte1, Nicole Tindill2, Brian Saunders3, Warren Hedley3, Shankar Subramaniam3,<br />

Timo Hannay1.<br />

1. Nature Publishing Group, 4 Crinan Street, London, N1 9XW. United Kingdom. 2.<br />

Nature Publishing Group, 75 Varick Street, 9th Floor, New York, NY 10013, USA. 3.<br />

San Diego Supercomputer Center, University of California, San Diego, 9500 Gilman<br />

Drive, Mail Code 0505, La Jolla, CA 92093-0505<br />

C.Garvey@nature.com<br />

Signaling@nature.com<br />

Signal transduction is at the core of most biological processes <strong>and</strong> represents a vibrant area of<br />

biological investigation. The Signaling Gateway Molecule Page database is an open access<br />

relational database that aims to provide all relevant published information pertaining to almost<br />

4000 cell <strong>signaling</strong> molecules. The Molecule Pages are authored by experts, anonymously<br />

peer reviewed, <strong>and</strong> published online by Nature Publishing Group. Access to all information<br />

in the Signaling Gateway Molecule Pages is available completely free, making this a res<strong>our</strong>ce<br />

driven by the <strong>signaling</strong> community for the <strong>signaling</strong> community.<br />

Individual Molecule Pages contain both automated data <strong>and</strong> peer reviewed author-entered<br />

data. The automated data component integrates information about the molecule, such as<br />

DNA <strong>and</strong> protein sequence information, structural information, sequence comparisons <strong>and</strong><br />

related sequences, <strong>and</strong> basic biophysical <strong>and</strong> biochemical properties, which are obtained from<br />

external database records. The author-entered component summarizes all significant<br />

published information on a given molecule <strong>and</strong> includes interlinked formalized descriptions<br />

of all its biochemical states.<br />

Over 150 full Molecule Pages have been published so far, 150 are undergoing peer review<br />

<strong>and</strong> 400 are currently being authored. The Signaling Gateway Molecule Page database<br />

integrates <strong>and</strong> summarizes the vast amount of published information on key signal<br />

transduction molecules, linking up not only <strong>signaling</strong> molecules but also <strong>signaling</strong><br />

communities to further accelerate the pace of discovery in cellular <strong>signaling</strong>.<br />

- 152 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 26<br />

Molecular mechanism of CRF induced calcium mobilization in CRF1 <strong>and</strong> CRF2<br />

expressing cells: involvement of different signalling pathways<br />

Eric Gutknecht1-2, Ilse Van der Linden1, Georges Vauquelin2 <strong>and</strong> Frank M.<br />

Dautzenberg1<br />

1CNS Discovery, Johnson & Johnson Research & Development, Turnhoutsweg 30, 2340<br />

Beerse <strong>and</strong> 2MBFA, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussel, Belgium<br />

Corticotropin releasing factor (CRF), the most important mediator of the body’s stress<br />

response activates two receptors (CRF1 <strong>and</strong> CRF2) both belonging to the B-class family of<br />

the GPCRs. CRF1 <strong>and</strong> CRF2 mainly couple to the stimulatory G-protein Galpha s which<br />

activates adenylate cyclase <strong>and</strong> protein kinase A. However, we reported recently that in stably<br />

transfected HEK293 cells, both receptors induce transient calcium (Ca2+) mobilization in the<br />

fluorimetric imaging plate reader (FLIPR) format. Ca2+ mobilization was completely<br />

abolished after depletion of intracellular Ca2+ stores or inhibition of IP3 receptors. In<br />

contrast, a specific inhibitor of the PKA pathway had no effect, whereas blockade of<br />

phospholipase C abolished the Ca2+ mobilization. To better underst<strong>and</strong> the involvement of<br />

the various G-proteins, we incubated cells with pertussis toxin <strong>and</strong> cholera toxin, two well<br />

known inhibitors of Gi <strong>and</strong> Gs proteins. While pertussis toxin partially blocked Ca2+ (40%),<br />

cholera toxin almost completely (80%) inhibited the transient Ca2+ response. To further<br />

assess the contribution of Galpha i in this process <strong>and</strong> the role of beta-gamma subunits, we<br />

transiently transfected the C-terminal part of the G-protein receptor kinase type 2 GRK2 (as a<br />

beta-gamma scavenger) which resulted in a potent reduction of the Ca2+ response. To<br />

elucidate the mechanism of the phospholipase C activation, we transfected different alphasubunits<br />

of the Gq family in CRF1 <strong>and</strong> CRF2(a) expressing cells. While a potentiation of the<br />

Ca2+ signal was observed with Galpha-16, only a weak or no increase in the signal could be<br />

monitored with Galpha o, Galpha olf, Galpha q, or Galpha 11. Thus, in CRF1 <strong>and</strong> CRF2<br />

expressing HEK293 cells, both receptors seems to act through different G-proteins indicating<br />

that several signalling pathways, including Galpha s, are involved in the activation of<br />

phospholipase C.<br />

- 153 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 27<br />

South of Ras: Identification <strong>and</strong> Characterization of Genes Deregulated by Oncogenic<br />

Ras Signaling<br />

Minh-Cam Ha-Thi, Oleg Tchernitsa, Anja Schramme, Balázs Györffy, Anastassia<br />

Malek, Christine Sers <strong>and</strong> Reinhold Schäfer<br />

Laboratory of Molecular Tumor Pathology, Charité Universitätsmedizin Berlin,<br />

Schumannstr. 20/21, D-10117 Berlin, Germany. E-mail: reinhold.schaefer@charite.de<br />

The small GTPase Ras is a molecular <strong>signaling</strong> switch known to play a critical role in tumor<br />

growth <strong>and</strong> development. The oncogenic activity of Ras <strong>signaling</strong> pathways is tightly coupled<br />

with the deregulation of the genetic program. Therefore, it is essential to identify deregulated<br />

target genes <strong>and</strong> to study their contribution to transformed phenotypes. We obtained a<br />

consensus list of genes responding to oncogenic Ras <strong>signaling</strong> in pairs of normal versus Rastransformed<br />

cells of rat, mouse <strong>and</strong> human origin by analyzing differential gene expression<br />

using suppression subtractive hybridization (SSH) <strong>and</strong> by interrogating high-density<br />

microarrays. To systematically study the deregulation of Ras-responsive genes in Rastransformed<br />

cells under various experimental conditions, we established species-specific<br />

customized microarrays representing approx. 300 Ras targets. We used Ras target microarrays<br />

for studying the transcriptional response to conditional Ras activation in a time-resolved<br />

manner <strong>and</strong> for identifying groups of target genes (signal-regulated transcriptional modules)<br />

controlled by specific downstream pathways such as the mitogen-activated kinase (MAPK)<br />

pathway or the phosphatidylinositol-3 kinase (PI3K) pathway in rodent cells. We also<br />

identified 45 genes showing increased transcriptional expression <strong>and</strong> 56 down-regulated<br />

genes in normal (BJELB) <strong>and</strong> H-Ras transformed human embryo fibroblasts (BJELR). To<br />

narrow-down the number of critical target genes, we targeted <strong>signaling</strong> effectors <strong>and</strong><br />

individual Ras-responsive genes by RNA interference. Silencing by siRNA of the<br />

transcriptional regulators Fra-1 <strong>and</strong> HMGA2, frequently up-regulated in Ras-transformed<br />

cells, impairs the proliferative capacity suggesting that these targets are necessary for cellular<br />

transformation. Likewise, knocking-down transformation-sensitive genes in non-transformed<br />

precursor cells can result in neoplastic transformation. Using this approach, a still growing set<br />

of genes capable of negatively affecting cellular transformation <strong>and</strong> also Ras signal<br />

transduction is being established.<br />

- 154 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 28<br />

S100C/A11, an essential element of the signal transduction of TGFb-induced growth<br />

suppression <strong>and</strong> a potential target for therapy against hyper-proliferative human<br />

diseases.<br />

Nam-ho Huh, Eiichi Makino, <strong>and</strong> Masakiyo Sakaguchi.<br />

Department of Cell Biology, Okayama University Graduate School of Medicine,<br />

Dentistry, <strong>and</strong> Pharmaceutical Sciences, Okayama 700-8558, Japan E-mail :<br />

namu@md.okayama-u.ac.jp<br />

TGFb is known to be a potent growth suppressor to epithelial cells, including epidermal<br />

keratinocytes. We have demonstrated that S100C/A11, an EF-h<strong>and</strong>-type Ca++-binding<br />

protein, plays a critical role for the signal transduction. On exposure of normal human<br />

keratinocytes to TGFb, S100C/A11 was specifically phosphorylated at 10Thr by PKCa. The<br />

phosphorylation facilitated the binding of S100C/A11 to nucleolin, resulting in nuclear<br />

translocation of S100C/A11. In nuclei, S100C/A11 liberated Sp1/3 from nucleolin. The<br />

resulting free Sp1/3 transcriptionally activated p21(WAF1), a representative negative<br />

regulator of cell growth. The new pathway involving S100C/A11 was shown to be<br />

indispensable for the growth suppression by TGFb together with the well-known Smadmediated<br />

pathway.<br />

Since the phosphorylated N-terminal domain of S100C/A11 was demonstrated to be<br />

critical for exhibiting the function described above, we introduced into cells a peptide<br />

composed of N-terminal 19 amino acids flanked by HIV-TAT protein transduction domain.<br />

The peptide induced apoptotic cell death in a number of human normal <strong>and</strong> tumor cells. The<br />

induction of apoptotic cell death was apparently independent of p53, p21(WAF1), <strong>and</strong><br />

caspase activity, but treatment with the peptide resulted in partial translocation of apoptosisinducing<br />

factor (AIF) from the cytoplasm to nuclei. These results indicate that the peptide<br />

may lead to the establishment of a new molecular target for the treatment of human cancer<br />

<strong>and</strong> other hyper-proliferative diseases.<br />

References: J Cell Biol 24;163(4):825-35, 2003; J Cell Biol 164(7):979-84, 2004; J Mol<br />

Med 82(9):612-20, 2004; Proc Natl Acad Sci USA. 102(39):13921-6, 2005.<br />

- 155 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 29<br />

Epac activation converts cAMP from a proliferative into a differentiation signal in PC12<br />

cells<br />

Simone Kiermayer, Ricardo M. Biondi, Guido Plotz, Jörg Haupenthal, Stefan Zeuzem,<br />

<strong>and</strong> Albrecht Piiper.<br />

Universitätsklinikum des Saarl<strong>and</strong>es, Department of Internal Medicine II, Kirrberger<br />

Str., Building 41, 66421 Homburg/Saar, Germany. E-mail: inskie@uniklinikumsaarl<strong>and</strong>.de<br />

cAMP plays, amongst others, an important role in the regulation of cell proliferation <strong>and</strong><br />

differentiation. How an elevated intracellular cAMP concentration [cAMP]i induces distinct<br />

biological responses is still unknown. Thus, we investigated the qualitative effects of an<br />

elevated [cAMP]i using cAMP-analoga, which specifically activate the main cAMP effectors,<br />

protein kinase A (PKA) or Epac1 <strong>and</strong> Epac2, exchange factors activating the small GTPases<br />

Rap1 <strong>and</strong> Rap2. As a model we used neuroendocrine PC12 cells, in which elevation of<br />

[cAMP]i activates extracellular signal-regulated kinase (ERK) 1/2 <strong>and</strong> induces low-degree<br />

neurite outgrowth. Our studies revealed that the specific stimulation of PKA triggered<br />

ERK1/2 activation. It was considerably more transient than that, observed upon simultaneous<br />

activation of both PKA <strong>and</strong> Epac. Unexpectedly, the PKA-specific cAMP analog induced cell<br />

proliferation rather than neurite outgrowth. This proliferative <strong>signaling</strong> pathway involved<br />

activation of the epidermal growth factor receptor <strong>and</strong> ERK1/2. Activation of Epac <strong>and</strong> its<br />

down-stream effector Rap1 appeared to extend the duration of PKA-dependent ERK1/2<br />

activation <strong>and</strong> converted cAMP from a proliferative into an anti-proliferative, neurite<br />

outgrowth-promoting signal. Thus, we found strong evidence that the outcome of cAMP<br />

<strong>signaling</strong> can depend on the set of cAMP effectors activated.<br />

- 156 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 30<br />

Expression of the insulin-like growth factor-I receptor in human colorectal cancer <strong>and</strong><br />

associations with Cx26 <strong>and</strong> antiapoptotic marker Bcl-xL.<br />

Mariusz Koda, Stanislaw Sulkowski, Luiza Kanczuga-Koda, Andrzej Wincewicz,<br />

Mariola Sulkowska, Waldemar Famulski, Wojciech Kisielewski.<br />

Department of Pathology, Medical University of Bialystok, Waszyngtona 13, 15-269<br />

Bialystok, Pol<strong>and</strong>. E-mail: sulek@zeus.amb.edu.pl<br />

Insulin-like growth factor (IGF) <strong>and</strong> its receptor (IGF-IR) play an important role in<br />

mitogenesis, apoptosis, growth <strong>and</strong> proliferation of several types of cancers. Overexpression<br />

of IGF-IR in colorectal cancer is associated with increase of cancer cells proliferation <strong>and</strong><br />

migration as well as inhibition of apoptosis. In <strong>our</strong> previous reports we demonstrated<br />

correlations between IGF-IR <strong>and</strong> apoptosis. Moreover we observed relationships between<br />

connexin26 (Cx26) expression <strong>and</strong> apoptotic markers in human colorectal cancer. Recently, it<br />

has been shown that expression of connexins <strong>and</strong> gap junction functions are also regulated by<br />

growth factors including IGF-I. Therefore, in this study we have focused on the relationships<br />

between IGF-IR <strong>and</strong> Cx26 as well as Bcl-xL expression.<br />

A total number of 115 cases of colorectal cancer were examined by immunohistochemistry,<br />

using the avidin-biotin-peroxidase method. Associations among above proteins were assessed<br />

in the entire group of colorectal cancer patients <strong>and</strong> its subgroups depending on lymph node<br />

involvement (N0 <strong>and</strong> N1), histological grade (G2 <strong>and</strong> G3), extent of tumor growth (pT1+pT2<br />

<strong>and</strong> pT3+pT4), histopathologic type (adenocarcinoma <strong>and</strong> mucinous carcinoma), sex, age<br />

(%60 <strong>and</strong> >60)<strong>and</strong> tumor site (colon <strong>and</strong> rectum).<br />

The expression of IGF-IR, Cx26 <strong>and</strong> Bcl-xL was noted in 47%, 56.5% <strong>and</strong> 75.6% of the<br />

tumors, respectively. In the entire group of patients we found the positive correlation between<br />

IGF-IR <strong>and</strong> Cx26 (p


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 31<br />

Expression of insulin <strong>signaling</strong> transmitters <strong>and</strong> glucose transporters at the protein level<br />

in the rat testis.<br />

Kersti Kokk1, 2, Esko Veräjänkorva2, Xiao-Ke Wu2, 4, Helle Tapfer1, Elle Põldoja1,<br />

Helle-Evi Simovart1 <strong>and</strong> Pasi Pöllänen2, 3<br />

1Department of Anatomy, University of Tartu, Tartu, Estonia, E-mail:<br />

Kersti.Kokk@ut.ee<br />

2Department of Anatomy, University of Turku, Turku, Finl<strong>and</strong><br />

3Center for Development/Administration, Kymi Dale (Kymenlaakso) Health Care<br />

District,<br />

FIN-48210 Kotka, Finl<strong>and</strong><br />

4Department of Obstetrics <strong>and</strong> Gynecology, First Affiliated Hospital, Heilongjiang<br />

Traditional Medicine University, Harbin 150040, China.<br />

Insulin Receptor Substrate (IRS) proteins are key mediators in insulin <strong>signaling</strong> from the<br />

insulin receptor <strong>and</strong> play a central role in maintaining basic cellular functions such as growth,<br />

survival <strong>and</strong> metabolism. It takes place through receptor-mediated tyrosine phosphorylation<br />

of the IRS proteins. IRS-1 <strong>and</strong> IRS-2 genes have been considered plausible c<strong>and</strong>idates<br />

involved in the pathogenesis of Type 2 diabetes. A family of glucose transporters (GLUT)<br />

mediates the cellular uptake of glucose. The aim of present study is to demonstrate the<br />

distribution of Insulin Receptor Substrates 1-3 (IRS 1-3), glucose transporters 1-4 (GLUT 1-<br />

4), SIRP1alpha, PKB kinase <strong>and</strong> PI3 kinase in the rat testis to see if signal transduction<br />

mediated by these proteins is active in testicular cells. Wistar rats were used as donors of<br />

testis tissue. Expression of these genes was studied at the protein level using<br />

immunohistochemistry <strong>and</strong> Western blotting. IRS-1, IRS-2, GLUT 1, GLUT 2, GLUT 3 <strong>and</strong><br />

SIRP1alpha were strongly expressed in the different types of cells in all the testes investigated<br />

by immunochemistry. Positive immunoreactions for IRS-3, GLUT 4, PKB kinase <strong>and</strong> PI3<br />

kinase were not found in the rat testis. Immunoblotting experiments demonstrated the<br />

presence of about 26-67 kD reactive with anti- IRS-1, IRS-2, GLUT 1, GLUT 2, GLUT 3,<br />

PKB kinase <strong>and</strong> SIRP 1alpha, but no proteins reactive with IRS-3, GLUT 4 <strong>and</strong> PI3 kinase<br />

antibodies were detected in the rat testis. The present result suggest that proteins like insulin<br />

<strong>and</strong> certain cytokines using IRS-1, IRS-2, GLUT 1, GLUT 2, GLUT 3, PKB kinase <strong>and</strong><br />

SIRP1alpha in their signal transduction can have effects on the epithelial cells of the male<br />

reproductive tract in the rat.<br />

- 158 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 32<br />

Distinct <strong>signaling</strong> <strong>and</strong> actions caused by intracellular lig<strong>and</strong>s (pepducins) for proteinaseactivated<br />

receptor-1 in multiple cells/tissues<br />

Satoko Kubo, Tsuyoshi Ishiki, Ichiko Doe, Fumiko Sekiguchi, Hiroyuki Nishikawa*,<br />

Kenzo Kawai*, Atsufumi Kawabata.<br />

Division of Physiology <strong>and</strong> Pathophysiology, School of Pharmaceutical Sciences, Kinki<br />

University, Higashi-Osaka 577-8502, Japan <strong>and</strong> *Fuso Pharmaceutical Industries Ltd.,<br />

Osaka 536-8523, Japan. E-mail: 0544410001t@kindai.ac.jp<br />

Proteinase-activated receptor-1 (PAR1), a G protein-coupled receptor for thrombin, can be<br />

activated by an N-palmitoylated peptide based on the intracellular loop 3 of human PAR-1,<br />

Pal-RCLSSSAVANRSKKSRALF-amide (P1pal-19), in a C-terminal-dependent manner in<br />

human platelets or PAR1-transfected cells. In contrast, the shorter peptide, Pal-<br />

RCLSSSAVANRS-amide (P1pal-12), could be a PAR1 antagonist. We thus examined the<br />

actions of those peptides in multiple cells/tissues known to express PAR1. P1pal-19, like the<br />

extracellular PAR1 agonist TFLLR-amide (TF), caused Ca2+ signal in human lung epithelial<br />

cells <strong>and</strong> rat gastric mucosal epithelial cells, <strong>and</strong> also delayed prostagl<strong>and</strong>in E2 formation in<br />

the latter cells. P1pal-19 <strong>and</strong> TF produced endothelial NO-dependent vasodilation <strong>and</strong><br />

epithelial prostanoid-dependent bronchodilation. However, P1pal-19 failed to mimic the<br />

contractile activity of TF in the endothelium-denuded blood vessels <strong>and</strong> gastric smooth<br />

muscle. In contrast, P1pal-12 partially inhibited vasorelaxation by TF <strong>and</strong> P1pal-19. In sum,<br />

P1pal-19 mimics the actions of TF in the endothelial <strong>and</strong> epithelial, but not smooth muscle,<br />

cells/tissues, <strong>and</strong> P1pal-12 may act as a PAR-1 antagonist in the vascular endothelium.<br />

- 159 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 33<br />

Small GTPase Ras <strong>and</strong> Rho expression in rat osteoblasts during spaceflight<br />

Yasuhiro Kumei 1 , Sadao Morita 1 , Hitoyata Shimokawa 1 , Kei-ichi Ohya 1 ,<br />

Hisako Katano 2 , Hideo Akiyama 3 , Masahiko Hirano 3<br />

1 Tokyo Medical <strong>and</strong> Dental University, Tokyo, Japan<br />

2 University of Tokyo Medical Science Institute, Tokyo, Japan<br />

3 Toray Research Center, Kamakura, Japan<br />

Rat osteoblasts were cultured for 4 <strong>and</strong> 5 days aboard Space Shuttle <strong>and</strong> solubilized after a 24<br />

hr-treatment with 1",25 dihydroxyvitamin D3. The mRNA levels of Ras <strong>and</strong> Rho were<br />

analyzed by quantitative RT-PCR normalized to glyceraldehyde 3-phosphate dehydrogenase.<br />

The small GTPase Ras <strong>and</strong> Rho are activated by GEF (guanine nucleotide exchange factor)<br />

<strong>and</strong> deactivated by GAP (GTPase-activating factor). Recently we reported that expression of<br />

Ras GEF, Ras-GRF (guanine nucleotide releasing factor) increased to 2- to 5-fold of the<br />

ground controls, during spaceflight. Ras-GRF acts downstream G-protein coupled receptors,<br />

whereas another Ras GEF, SOS (son of sevenless) activates Ras after binding to adaptor<br />

protein Grb2 of receptor tyrosine kinase. The SOS mRNA levels also increased to 3- to 5-fold<br />

during spaceflight, although Ras GAP expression was not altered. Ras activation was<br />

followed by MAP kinase cascades including ERK1/2 expression, <strong>and</strong> FAK (focal adhesion<br />

kinase) increased during spaceflight. Cdc42 is a Rho family protein that responds to<br />

environmental stress <strong>and</strong> induces JNK (Jun-N-terminal kinase). Both Cdc42 GAP <strong>and</strong> GEF<br />

expression was increased 2-fold in the flight cultures, as compared to the ground controls.<br />

Accelerated Cdc42 turnover might result in 2-fold increase of JNK expression. Rho plays<br />

important roles in actin rearrangement. Microgravity may alter physiological change of rat<br />

osteoblasts at least partly via small GTPase.<br />

- 160 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 34<br />

SDF1 controls pituitary cell proliferation through the activation of ERK1/2 <strong>and</strong> the<br />

Ca2+-dependent cytosolic tyrosine kinase, Pyk2<br />

°Aless<strong>and</strong>ro Massa, *Silvia Casagr<strong>and</strong>e, °Adriana Bajetto, °Carola Porcile, °Federica<br />

Barbieri, °Stefano Thellung, °Sara Arena, °Aless<strong>and</strong>ra Pattarozzi, °Monica Gatti,<br />

°Aless<strong>and</strong>ro Corsaro, *Mauro Robello, °Gennaro Schettini, °Tullio Florio.<br />

°Section of Pharmacology, Dept. Oncology Biology <strong>and</strong> Genetics, University of Genova,<br />

Italy. *Unit INFM, Dept. of Physics, University of Genova, Italy .<br />

Stromal cell-Derived Factor 1 (SDF1) is a chemokine of the CXC subfamily involved in the<br />

recruitment of immune cells to sites of inflammation. SDF1 exerts its effects interacting with<br />

CXCR4, a G-protein coupled receptor. CXCR4 is often expressed by tumor cells <strong>and</strong> its<br />

activation causes tumor cell proliferation. To date, no evidence have been provided on the<br />

possible role of SDF1 in pituitary adenoma funcion, although it was reported the expression<br />

of CXCR4 in rat pituitary. In this work, we used GH4C1 cells as a model to study the effects<br />

of SDF1 in pituitary functions. In these cells, SDF1 induced dose-dependent proliferation.<br />

Thus we evaluated the intracellular <strong>signaling</strong> involved in this effect. SDF1 increased cytosolic<br />

[Ca2+] <strong>and</strong> activated Pyk2, ERK1/2 <strong>and</strong> BKCa channels. To correlate these intracellular<br />

effectors with the proliferative activity we inhibited their activity using BAPTA-AM (Ca2+<br />

chelator), PD98059 (MEK inhibitor), salicylate (Pyk2 inhibitor) <strong>and</strong> TEA (K+ channel<br />

blocker). All these compounds reverted SDF1-induced proliferation, suggesting the<br />

involvement of multiple intracellular pathways. To identify a possible cross-talk <strong>and</strong> a<br />

molecular ordering among this pathways, we tested these antagonists on SDF1 dependent<br />

activation of ERK1/2, Pyk2 <strong>and</strong> BKCa channels. We report that: the inhibition of [Ca2+]i<br />

increase or BKCa channels activity did not affect ERK1/2 activation by SDF1; Pyk2<br />

activation was purely Ca2+ dependent, not involving ERK1/2 or BKCa channels; BKCa<br />

channels activity was antagonized by Pyk2 but not by ERK1/2 inhibitors. These data suggest<br />

that SDF1- dependent increase of [Ca2+]i activates Pyk2, that, in turn, regulates BKCa<br />

channel activity. Conversely, ERK1/2 activation is an independent phenomenon. In<br />

conclusion we demonstrate that SDF1 causes proliferation of GH4C1 cells, suggesting that<br />

the activation of CXCR4 may represent a novel regulatory mechanism for pituitary cell<br />

proliferation, that may contribute to pituitary adenoma development.<br />

- 161 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 35<br />

Involvement of Kv1.5 channel endocytosis in serotonin-induced inhibition of voltagegated<br />

K+ currents <strong>and</strong> contraction in pulmonary arteries.<br />

Laura Moreno, Angel Cogolludo, Laura Cobeño, Federica Lodi, Giovanna Frazziano,<br />

Juan Tamargo, <strong>and</strong> Francisco Perez-Vizcaino.<br />

Department Pharmacology. School Medicine. Universidad Complutense de Madrid.<br />

28040 Madrid. SPAIN. E-mail: lmoreno@ift.csic.es<br />

Serotonin (5-HT) <strong>and</strong> voltage-gated K+ (Kv) channels play a central role in the pathogenesis<br />

of pulmonary hypertension (PH). We showed (abstract, this meeting) that 5-HT inhibits Kv<br />

channels in rat pulmonary artery smooth muscle cells (PASMC) establishing a link between<br />

these pathogenetic factors in PH. These effects were mediated by activation of 5-HT(2A)<br />

receptors via PLC, classic PKC <strong>and</strong> tyrosine kinase. Herein we analyzed whether 5-HT<br />

inhibited human Kv1.5 channels stably expressed in Ltk- <strong>and</strong> the possible role of Kv1.5<br />

endocytosis in native PASMC. 5-HT reduced Kv1.5 currents <strong>and</strong> this effect was prevented by<br />

the 5-HT(2A) receptor antagonist ketanserin <strong>and</strong> by the tyrosine kinase inhibitor tyrphostin<br />

23. However, we did not find changes in the Kv1.5 protein phosphorylation in tyrosine<br />

residues using an anti-phosphotyrosine antibody in Kv1.5 immunoprecipitates after treatment<br />

with 5-HT. Inhibition of caveolar disruption (#-cyclodextrin) or inhibition of endocytotic<br />

processes (concanavalin A), prevented the inhibitory effects of 5-HT on Kv currents in native<br />

PASMC. Concanavalin A also inhibited the vasoconstrictor effect of 5-HT in isolated rat<br />

pulmonary arteries. In homogenates from pulmonary arteries, 5-HT(2A) receptors <strong>and</strong><br />

caveolin-1 co-immunoprecipitated with Kv1.5 channels <strong>and</strong> this was increased upon<br />

stimulation with 5-HT. Moreover, confocal microscopy revealed that Kv1.5 channels were<br />

internalized when PASMC were stimulated with 5-HT. In conclusion, activation of 5-HT(2A)<br />

receptors inhibits rat native Kv currents as well as cloned human Kv1.5 currents. 5-HTinduced<br />

inhibition of Kv currents <strong>and</strong> the subsequent vasoconstriction of PA result from the<br />

interaction of 5-HT(2A) receptors with Kv1.5 channels within caveolar membrane<br />

microdomains <strong>and</strong> involve tyrosine phosphorylation <strong>and</strong> endocytotic processes.<br />

- 162 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 36<br />

CLA/PUFA <strong>and</strong> Deoxycholic Acid (DCA)-Induced Signaling in Colorectal Cancer via<br />

the Insulin-Like Growth Factor 1 Receptor (IGF-1R).<br />

Sherif S Morgan, Shawna Comer, <strong>and</strong> Emmanuelle J Meuillet.<br />

The Cancer Biology Graduate Interdisciplinary Program, Nutritional Sciences<br />

Department, <strong>and</strong> Molecular <strong>and</strong> Cellular Biology Department. University of Arizona,<br />

Tucson, Arizona 85721<br />

Colorectal cancer is the second leading cause of cancer-related deaths in the US. Clinical<br />

studies have shown that colorectal cancer patients have higher levels of bile acids (BA) in<br />

colonic lumen, <strong>and</strong> animal studies substantiate the role of BA as tumor promoters. The<br />

molecular mechanism of deoxycholic acid (DCA), which is a hydrophobic cholesterol<br />

derivative remains unclear. Recently, DCA has been shown to perturb the plasma membrane,<br />

which leads to EGFR activation. We sought to investigate a link between the presence of<br />

DCA <strong>and</strong> IGF-IR activity in colorectal cancer cells. Conjugated linoleic acid (CLA) <strong>and</strong> n-3<br />

polyunsaturated fatty acids (PUFA) have been shown to have anti-tumorigenic effects. We<br />

hypothesize that DCA can perturb the plasma membrane, leading to the activation of the IGF-<br />

IR <strong>and</strong> its downstream <strong>signaling</strong> pathways, promoting survival <strong>and</strong> proliferation of colorectal<br />

cancer cells. Conversely, nutritional modulation by increasing intake of PUFA/CLA may<br />

counteract these effects by restoring normal IGF-IR <strong>signaling</strong>. We show that treatment with<br />

DCA leads to activation of IGF-IR <strong>and</strong> ERK-1/2 through the MEK-1 pathway. Counterintuitively,<br />

DCA does not activate AKT; in fact, it counteracts the IGF-induced AKT<br />

activation. DCA appears to preferentially activate ERK, while inhibiting AKT. Treatment<br />

with CLA, on the other h<strong>and</strong>, activates both ERK <strong>and</strong> AKT. Further studies are required to<br />

identify the functional role of activating these pathways, because they may lead to different<br />

cellular responses. These results support the role of DCA as a tumor promoter <strong>and</strong> sheds light<br />

on the potential mechanisms of CLA chemoprevention. These data have important<br />

implications since they support the significant role of nutrition in the prevention <strong>and</strong> treatment<br />

of colorectal cancer.<br />

- 163 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 37<br />

Cellular prion protein <strong>signaling</strong> in serotonergic neuronal cells<br />

Sophie Mouillet-Richard1, Benoît Schneider1, Elodie Pradines1, Mathéa Pietri1,<br />

Myriam Ermonval1, Jacques Grassi2, Vincent Mutel3, Jean-Marie Launay4 & Odile<br />

Kellermann1.<br />

1 Différenciation cellulaire et prions, CNRS UPR1983 94801 Villejuif, France & Institut<br />

Pasteur, Département de biologie cellulaire et Infections, 75724 Paris Cedex 15, France.<br />

mouillet@vjf.cnrs.fr 2 Commissariat à l’Energie Atomique Saclay, DSV-SPI, 91 191 Gif<br />

sur Yvette Cedex, France 3 F. Hoffmann-LaRoche, Basel, Switzerl<strong>and</strong>. 4 Hôpital<br />

Lariboisière 75009 Paris, France.<br />

The cellular prion protein PrPC is the normal counterpart of the scrapie prion protein (PrPSc), itself<br />

the major if not the only component of the infectious agent of transmissible spongiform<br />

encephalopathies (TSE). PrPC is a ubiquitous glycoprotein abundantly expressed in neuronal cells,<br />

which are the targets of TSE pathogenesis. It is attached to the outer membrane leaflet through a GPI<br />

anchor, <strong>and</strong> primarily resides in lipid rafts, which represent subcellular microdomains highly enriched<br />

in cell signalling molecules.<br />

To gain insight into the physiological role of PrPC in relation with the onset, maintenance <strong>and</strong><br />

regulation of neuronal functions, we exploit the properties of the 1C11 neuroectodermal cell line,<br />

endowed with the capacity to convert into fully-functional serotonergic neuronal cells upon<br />

appropriate induction. Within 4 days of differentiation, almost 100% 1C11 cells implement 5-HT<br />

synthesis activity, storage, catabolism, uptake as well as three serotonergic receptors of the 5-HT2B,<br />

5-HT1B/D <strong>and</strong> 5-HT2A subtypes. These G-protein coupled receptors sustain an autoregulation of the<br />

overall serotonergic functions by external 5-HT.<br />

Antibody-mediated ligation of PrPC at the cell surface of 1C11 cells provides a means to mimic the<br />

interaction with a yet-to-be-identified lig<strong>and</strong> <strong>and</strong> to monitor downstream signal transduction events.<br />

While PrPC-associated signalling cascades occur in 1C11 precursor cells as well as other nonneuronal<br />

cell systems, PrPC fulfils some neuronal-specific function in mature 1C115-HT serotonergic<br />

cells. By interacting with the membrane protein caveolin <strong>and</strong> the Fyn tyrosine kinase within a platform<br />

located on neuritic extensions, PrPC mobilizes a PI3kinase-PKC - NADPHoxidase module <strong>and</strong> other<br />

downstream effectors targeting the MAPK kinases ERK1/2.<br />

In addition to recruiting signalling pathways, antibody-mediated ligation of PrPC has impact on the<br />

couplings of the serotonergic receptors present on 1C115-HT serotonergic cells. Concomitant<br />

exposure of 1C115-HT serotonergic cells to 5-HT receptor agonists <strong>and</strong> anti-PrP antibodies<br />

significantly alters the G-protein-mediated couplings associated to all three receptors. PrPC ligation<br />

has diverse outcome depending on the receptor <strong>and</strong> the pathway considered. First, PrP antibodies<br />

selectively augment the PLA2 response imparted by 5-HT2B receptors. Second, PrP ligation fully<br />

abrogates the PLC coupling to 5-HT2A receptors. Third, PrP antibodies reduce the 5-HT1B/D<br />

negative coupling to adenylate cyclase. Because the 5-HT1B/D receptor function is modulated through<br />

antagonising crosstalk that arise from 5-HT2B <strong>and</strong> 5-HT2A receptors, PrPC ligation also impacts on<br />

the functional interactions occurring between the three receptor subtypes.<br />

Hence, PrPC not only mobilizes transduction cascades controlling the cellular redox state <strong>and</strong> ERK1/2<br />

kinases but also interferes with the signalling activity of serotonergic receptors that regulate 5-HT<br />

synthesis, storage <strong>and</strong> uptake. Overall, <strong>our</strong> findings support the notion that PrPC contributes to the<br />

homeostasis of serotonergic neurons. Eventually, they provide a foundation for uncovering the impact<br />

of prion infection on serotonergic neurons.<br />

- 164 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 38<br />

Transformation by oncogenic Ras exhibits JNK-isoform specificity<br />

Christina Nielsen, Jacob Thastrup, Marja Jäättelä <strong>and</strong> Tuula Kallunki<br />

Apoptosis Department, Danish Cancer Society, Str<strong>and</strong>boulevarden 49, 2100<br />

Copenhagen, Denmark. E-mail: tk@cancer.dk<br />

JNK family consists of at least 10 members: JNK1, JNK2 <strong>and</strong> JNK3 isoforms <strong>and</strong> their splice<br />

variants. Active Ras oncogene is expressed in 30-40% of human cancers. Ras, among others,<br />

can activate JNK pathway.<br />

We have recently found out that active Ras cannot transform cells that carry inactivated JNK2<br />

isoform of JNK. Using knockout iMEFs we have observed that JNK2 is essential for Ras<br />

transformation whereas JNK1 is not. Various individual Jnk2-/- iMEFs are severely deficient<br />

in multilayer growth as measured by focus formation <strong>and</strong> soft agar assays. The mouse<br />

tumorigenesis assay further proved that Ras transduced Jnk2-/- iMEFs are clearly less<br />

tumorigenic than the Ras transformed wt <strong>and</strong> Jnk1-/- iMEFs in vivo. We have further verified<br />

<strong>our</strong> findings by down regulating the endogenous JNK2 in wt iMEFs utilizing the short hairpin<br />

(sh) RNA technique. For this we have prepared two JNK2 specific retroviral sh RNA<br />

constructs which both are efficient <strong>and</strong> specific in JNK2 down regulation. Introduction of<br />

either of the JNK2 specific sh RNAs into the wt iMEFs reduced their tumorigenicity clearly.<br />

Novel data strongly suggesting that JNK2 is necessary for Ras induced transformation <strong>and</strong><br />

tumorigenesis will be presented.<br />

- 165 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 39<br />

Identification of suramin as a direct inhibitor of adenylyl cyclase <strong>and</strong> a competitive<br />

inhibitor of [3H]-forskolin binding<br />

Jiri Novotny, Jiri Stöhr, Lenka B<strong>our</strong>ova <strong>and</strong> Petr Svoboda<br />

Department of Biochemistry of Membrane Receptors, Institute of Physiology, Academy<br />

of Sciences, Videnska 1083, Prague 4 <strong>and</strong> Department of Physiology, Faculty of Natural<br />

Sciences, Charles University, Vinicna 7, 120 00 Prague 2, Czech Republic<br />

E-mail: novjiri@biomed.cas.cz<br />

Activity of adenylyl cyclase (AC) is markedly higher in brain cortex from young than from<br />

adult rats. Here we compared the modulatory effects of many different compounds on AC<br />

activity in cerebrocortical preparations from young (12-day-old) <strong>and</strong> adult (90-day-old) rats.<br />

We did not found any substantial difference between the effects of all tested agents <strong>and</strong> AC<br />

activity measured in various conditions was always about two-fold higher in samples from<br />

young animals. Our experiments analysing the presumed modulatory role of suramin revealed<br />

that this pharmacologically important drug may act as a direct inhibitor of AC. The enzyme<br />

activity was diminished to the same extent by suramin in membranes from both tested age<br />

groups. Moreover, suramin was able to reduce AC activity stimulated by Mn2+ or forskolin<br />

in membranes derived from S49 cyc- cells lacking Gs-alpha protein. We also determined<br />

characteristics of [3H]-forskolin binding in cerebrocortical membranes <strong>and</strong> these<br />

measurements allowed us to detect high-affinity, as well as super-high-affinity, [3H]forskolin<br />

binding sites. The binding parameters of these sites differed quite significantly in<br />

samples from both age groups tested. We also assessed the potential effect of suramin <strong>and</strong><br />

results of these analyses suggested that this compound may act as a potent competitive<br />

inhibitor of [3H]-forskolin binding. In summary, <strong>our</strong> present studies extend the existing array<br />

of suramin cellular tagets by identifiying this drug as a potent direct inhibitor of AC <strong>and</strong> [3H]forskolin<br />

binding.<br />

This investigation was supported by Centrum of Neuroscience (LC 554).<br />

- 166 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 40<br />

Oxytocin- <strong>and</strong> vasopressin-induced mitogenic signalling in human small-cell lung<br />

carcinoma<br />

Christel Péqueux1, Marie-Thèrèse Hagelstein2, Jean-Claude Hendrick2 <strong>and</strong> Jean-<br />

Jacques Legros2.<br />

1Tum<strong>our</strong> <strong>and</strong> Development Biology Laboratory, CRCE, Pathology Institute, CHU-B23,<br />

University of Liège, B-4000 Liège, Belgium, e-mail: C.Pequeux@ulg.ac.be;<br />

2Neuroendocrine unit, CIL, Pathology Institute, CHU-B23, University of Liège, B-4000<br />

Liège, Belgium, e-mail: Jean-Jacques.Legros@ulg.ac.be<br />

Neuroendocrine tum<strong>our</strong> cells usually develop very potent autocrine/paracrine signalling<br />

pathways that play a crucial part in the dysregulation of cellular growth. In the study<br />

presented here, we demonstrated, on small cell lung carcinoma (SCLC) cell lines, that the<br />

oxytocin (OT) as well as the vasopressin (VP) genes, normally transcriptionally restricted in<br />

their expression, are activated in SCLC, leading to synthesis <strong>and</strong> secretion of OT <strong>and</strong> VP.<br />

Concomitantly, these tum<strong>our</strong> cells express the various neuropeptide receptors: OTR, V1aR,<br />

V1bR/V3R <strong>and</strong> V2R. We observed that OT, as well as VP, induces a dose dependent <strong>and</strong><br />

time persistent increase of tum<strong>our</strong> cell proliferation. This mitogenic effect of OT was<br />

completely abolished by the OTR antagonist OVTA. We demonstrated that OT- <strong>and</strong> VPinduced<br />

mitogenic effects on SCLC are largely mediated by the OTR <strong>and</strong> the V1aR<br />

respectively <strong>and</strong> that this mitogenic signalling passes through the phosphorylation of ERK1/2<br />

<strong>and</strong> p90RSK in a PLC-, Ca++-, PKC- <strong>and</strong> MEK1/2-dependent pathway. Moreover, we<br />

evidenced that growth of OT- <strong>and</strong> VP-stimulated SCLC could be down regulated by<br />

signalling inhibitors <strong>and</strong> was not a consequence of toxicity. Additionally, we reported that<br />

86% of primary SCLC biopsies analyzed expressed at least the OTR <strong>and</strong> that 71% expressed<br />

the OTR, the V1aR <strong>and</strong> the V2R altogether. Interestingly, in normal human lung, OTR<br />

colocalized with vascular endothelial cells of the lung. Among SCLC plasma samples, the<br />

distribution of elevated OT is 40%, of elevated VP is 43.3% <strong>and</strong> of both elevated hormones is<br />

16.7%. Altogether, these results clearly demonstrate that, in addition to the vasopressinergic<br />

system, OT <strong>and</strong> particularly OTR contribute to give the SCLC tum<strong>our</strong> a survival advantage.<br />

Our data identify pathways by which OT <strong>and</strong> VP induced their mitogenic action on SCLC <strong>and</strong><br />

they highlight that blocking of their receptor signalling represents viable pharmacological<br />

targets.<br />

- 167 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 41<br />

Endoglin modulates Transforming Growth Factor-beta <strong>signaling</strong> pathways in L6E9<br />

myoblasts<br />

Alicia Rodríguez-Barbero, Soraya Velasco, José M. López-Novoa<br />

Instituto Reina Sofía de Investigación Nefrológica. Dpto de Fisiología y Farmacología.<br />

Universidad de Salamanca. Campus Unamuno, Edificio Departamental. Avda. Campo<br />

Charro s/n. 37007. Salamanca. Spain. E-mail: barberoa@usal.es<br />

Endoglin is a transmembrane accessory receptor for transforming growth factor-# (TGF-#).<br />

TGF-# elicits cellular responses via specific type I <strong>and</strong> II serine/threonine kinase receptors<br />

<strong>and</strong> their downstream nuclear effectors, termed Smads. Type I receptors act downstream of<br />

type II receptors <strong>and</strong> determine the signalling specificity within the receptor complex. In most<br />

cells, TGF-# signals via TGF-# type II receptor <strong>and</strong> activin receptor-like kinase (ALK) 5 to<br />

induce Smad2/3 phosphorylation. However, it has recently been shown that in endothelial<br />

cells TGF-# activates two distinct types I receptor pathways, that is, ALK5-inducing Smad2/3<br />

phosphorylation <strong>and</strong> ALK1-promoting Smad1/5 phosphorylation. Endoglin, as well as other<br />

TGF-# <strong>signaling</strong> components, is involved in several pathophysiological processes such as<br />

angiogenesis <strong>and</strong> fibrosis. Mutations in endoglin <strong>and</strong> ALK1 have been linked to the vascular<br />

disorder, hereditary hemorrhagic telangiectasia type 1 (HHT1). However, the function of<br />

endoglin in TGF-#/ALK signalling has remained unclear. To assess the function of endoglin<br />

in TGF-#/ALK signalling, we select a non-endoglin expressed cell type such as L6E9 rat<br />

myoblasts, <strong>and</strong> transfected them with L-endoglin. We found that both, the classic TGF-<br />

#/ALK5 <strong>and</strong> the endothelial specific TGF-#/ALK1 pathways are present <strong>and</strong> functional in<br />

L6E9 myoblasts. TGF-#1 activated both Smad1 y Smad2/3 <strong>and</strong> also induced Smad2/3 <strong>and</strong><br />

Smad4 nuclear translocation without differences between mock <strong>and</strong> endoglin transfected<br />

myoblasts. Nevertheless, endoglin promote Id1 expression <strong>and</strong> reduce PAI-1 activity. Our<br />

results indicate a pivotal role for endoglin in the balance of ALK1 <strong>and</strong> ALK5 signalling. The<br />

progression in <strong>our</strong> underst<strong>and</strong>ing on the functional role of endoglin might have important<br />

relevance in the regulation of pathophysiological processes in which endoglin is involved.<br />

- 168 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 42<br />

Galphaz subunits are essential partners for permanent association of receptor-activated<br />

Galphai subunits to RGS9-2 proteins.<br />

María Rodríguez-Muñoz, David Bermúdez, Elena de la Torre-Madrid, Pilar Sánchez-<br />

Blázquez <strong>and</strong> Javier Garzón.<br />

Laboratorio de Neurofarmacología. Instituto Cajal. Avenida Doctor Arce 37, 28002<br />

Madrid. Spain. Email: mrodriguez@cajal.csic.es<br />

The in vivo administration of an ED80 of morphine induces the transfer of mu-opioid<br />

receptors (MOR) that are associated with Galpha subunits to RGS9-2 proteins (1). The<br />

phosphorylation of specific residues in or near the RGS box <strong>and</strong> the subsequent binding to 14-<br />

3-3 impedes RGS9-2 GAP activity <strong>and</strong> stabilizes this interaction. Thus, by retaining MORactivated<br />

Galphai2 subunits, RGS9-2 proteins play a key role in the onset of opioid tolerance.<br />

Besides the role of RGS9-2 in these processes, morphine also promotes the association of<br />

Galpha subunits with RGS-Z proteins. Since recent studies have shown that Galphaz subunits<br />

are stably associated with RGSZ2, we have examined the contribution of Galphaz to the<br />

sequestering of Galphai subunits by RGS9-2 proteins. Proteins from mouse PAG<br />

synaptosomal membranes were immunoprecipitated with anti-RGS9-2 IgGs <strong>and</strong> analysed by<br />

Western blotting. An anti Phospho-Serine antiserum recognised a 76Kda protein that<br />

corresponded to RGS9-2. Impairing the expression of Galphaz through the subchronic<br />

administration of selective ODNs provoked a 45% reduction in the Ser phosphorylation of<br />

RGS9-2 <strong>and</strong> reduced tolerance to repeated administration of morphine. Conversely, in<br />

Galphai2 knockdown animals <strong>and</strong> mice receiving exogenous Galphaz subunits, the<br />

phosphorylation of RGS9-2 augmented by 36 <strong>and</strong> 42%, respectively. Our data show that<br />

activated GalphazGTP subunits are required for agonist-dependent phosphorylation of RGS9-<br />

2 proteins, which leads to the stabilization <strong>and</strong> sequestering of MOR-activated G-alpha<br />

subunits by RGS9-2 proteins.<br />

(Supported by MEC SAF2003-01121 <strong>and</strong> BMC2002-03228;Instituto de Salud Carlos III<br />

G03/005)<br />

1. Garzón J, Rodríguez-Muñoz M, López-F<strong>and</strong>o A, Sánchez-Blázquez P (2005) J. Biol.<br />

Chem. 280: 8951-8960<br />

- 169 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 43<br />

Conditionnal expression of wild type or mutated Gsalphalpha induced MAPKinases<br />

ERK-1/2, prolactin <strong>and</strong> growth hormone promoters activation in a somato-lactotroph<br />

cell line.<br />

David Romano, Morgane Pertuit, Karine Magalon, Anne Barlier, Ramahefarizo<br />

Rasolonjanahary, Alain Enjalbert et Corinne Gérard.<br />

Interactions Cellulaires Neuroendocriniennes, IFR Jean Roche, Faculté de Médecine<br />

Nord, 13916 Marseille cedex 20, France. E-mail : romano.d@nord.univ-mrs.fr<br />

Mutations of the Gsalphalpha (Gsa) protein, named gsp oncogene, are present in 40% of<br />

human somatotroph adenomas. These mutations induced a constitutive activation of the Gsa<br />

protein <strong>and</strong> an increase of adenylyl cyclase (AC) activity. However its role in tumorigenesis is<br />

not clear because of an overlap of clinical phenotype (hormonal hypersecretion, proliferation)<br />

between tumors bearing or not the gsp oncogene. This overlap might be due to an<br />

overexpression of the wild type (wt) Gsa protein, mimicking the activating mutation, or to the<br />

compensatory mechanisms like the increase of phosphodiesterase activity. To underst<strong>and</strong> the<br />

molecular mechanisms induced by the gsp oncogene, as well as the putative role of<br />

overexpressed wt Gsa, a somato-lactotroph cell line (GH4C1) was stably transfected with wt<br />

Gsalpha or constitutively activated Gsalpha by the R201C mutation, under the control of the<br />

tetracycline operon (GH4C1 Tet-off).<br />

After induction, we observe a similar increase of both wild type (wt) <strong>and</strong> mutated transgene<br />

mRNA levels. Moreover, the time c<strong>our</strong>se of both transgene proteins expression follows their<br />

mRNA synthesis with a maximum after 7 days of induction. However, the wt transgene<br />

protein is more expressed than the mutated one, suggesting a negative post-transcriptionnal<br />

regulation of the gsp oncogene. At a functional level, cAMP level <strong>and</strong> AC activity are<br />

strongly increased after induction of the mutated protein, with a time c<strong>our</strong>se pattern similar to<br />

the expression of the gsp oncogene. However, (i) basal MAPKinases ERK-1/2<br />

phosphorylation, (ii) human prolactin (PRL) <strong>and</strong> growth hormone (GH) promoters activity are<br />

progressively <strong>and</strong> similarly increased by induction of both wt <strong>and</strong> mutated Gsa proteins.<br />

Moreover, using the MAPKinase specific inhibitor U0126, we show that this increase in both<br />

PRL <strong>and</strong> GH promoters activity is MAPKinase-dependent.<br />

So far, these cell lines seem to be interesting models to study the molecular mechanisms<br />

involved in the tumorigenesis of human somatotroph adenomas bearing or not the gsp<br />

oncogene.<br />

- 170 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 44<br />

UVB-induced EGFR-Signaling is mediated via the AhR<br />

Claudia Schäfer1, Ulrike Hübenthal1, Jason Cline1, Melanie Wurm2, Christian Calles1,<br />

Peter Schroeder1, Lars-Oliver Klotz3, Helmut Hanenberg2, Jean Krutmann1, Josef<br />

Abel1 <strong>and</strong> Ellen Fritsche1<br />

Institut für umweltmedizinische Forschung, Auf’m Hennekamp 50; 2Department of<br />

Paediatric Oncology, 3Institute of Biochem. <strong>and</strong> Mol. Biol., University of Düsseldorf,<br />

Universitätsstrasse 1, 40225 Düsseldorf, Germany; e-mail: claudia.schaefer@uniduesseldorf.de<br />

The aryl hydrocarbon receptor (AhR) is a lig<strong>and</strong>-dependend transcription factor that rests in<br />

its inactive form as a multiprotein complex in the cytoplasm of most cells. AhR lig<strong>and</strong>s<br />

include polycyclic aromatic hydrocarbons (PAH) <strong>and</strong> halogenated PAH. Lig<strong>and</strong> binding leads<br />

to dissociation of the interacting proteins, AhR nuclear translocation, heterodimerization with<br />

the AhR nuclear translocator (ARNT) <strong>and</strong> activation of AhR-dependent genes like<br />

cytochrome P450 (CYP) 1A1. CYP1A1 induction by UVB irradiation was recently described<br />

<strong>and</strong> the formation of an endogenous AhR lig<strong>and</strong> by UVB was proposed. Epidermal growth<br />

factor receptor (EGFR)-<strong>signaling</strong> is also initiated by UVB irradiation. Considering that the<br />

AhR lig<strong>and</strong> TCDD causes EGFR-activation in human mammary carcinoma cells, we<br />

investigated whether UVB-initiated EGFR-<strong>signaling</strong> is mediated by activation of the AhR<br />

pathway.<br />

Signaling was studied in the immortalized keratinocyte cell line HaCaT. Irradiation of cells<br />

was carried out with 100 J/m2 UVB. Irradiation led to an AhR-dependent increase in<br />

CYP1A1 <strong>and</strong> COX-2 mRNA as determined by quantitative real-time PCR. Because COX-2<br />

expression can be modulated by EGFR activation, we performed immunocytochemical<br />

analyses of the EGFR. These experiments revealed that clustering <strong>and</strong> internalization of the<br />

EGFR after UVB irradiation are also –at least partially- AhR dependent. Further EGFR<br />

downstream targets which are activated after UVB irradiation are Erk1/2 phosphorylation <strong>and</strong><br />

COX-2 protein expression. We determined by Western blotting that Erk1/2 phosphorylation<br />

<strong>and</strong> COX-2 expression are partially dependent on AhR <strong>signaling</strong>. Involvement of the AhR in<br />

UVB-induced <strong>signaling</strong> was verified by utilization of the AhR antagonist MNF <strong>and</strong> by AhR<br />

gene knockdown. In summary we found that the AhR is involved in UVB-induced activation<br />

of the EGFR <strong>signaling</strong> pathway. Therefore we conclude that the AhR serves as a photosensor<br />

in the cytoplasm of keratinocytes mediating UVB-induced signal transduction.<br />

- 171 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 45<br />

Dual activity of phosphorothioate CpG-oligodeoxynucleotides on HIV: Reactivation of<br />

latent provirus <strong>and</strong> inhibition of productive infection in human T cells<br />

Carsten Scheller*1, Stefan Lamla1, Ulf Dittmer2, Axel Rethwilm1, Eleni Koutsilieri1<br />

Institute of Virology <strong>and</strong> Immunobiology, University of Wuerzburg, Versbacher Strasse<br />

7, 97078 Wuerzburg, Germany<br />

Institute of Virology, UK Essen, Hufel<strong>and</strong>strasse 55, 45122 Essen, Germany<br />

* correspondence to: Carsten Scheller, email: scheller@vim.uni-wuerzburg.de<br />

CpG oligodeoxynucleotides (CpG ODNs) bind to toll-like receptor 9 (TLR-9) <strong>and</strong> trigger<br />

cellular activation in immune cells with antigen-presenting activity, including B-cells <strong>and</strong><br />

dendritic cells. Here we show that treatment of the latently HIV-infected T cell line ACH-2<br />

with the CpG ODNs 2006 or 2040 trigger activation of viral gene expression, demonstrating<br />

that CpG-<strong>signaling</strong> activity can also be found in T cells. In contrast to the stimulating effects<br />

on viral gene transcription in latently-infected cells, CpG ODNs potently suppressed HIV<br />

replication in productively-infected T cells. Inhibition of virus replication was not related to<br />

CpG motifs but was likely caused by the phosphorothioate backbone of the CpG ODNs,<br />

probably by interfering with the viral enzyme reverse transcriptase. These results point to a<br />

novel role of CpG-sequences in the development of immune activation therapies to<br />

accomplish eradication of HIV from its latent T cell reservoir in vivo.<br />

- 172 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 46<br />

5-HT2B <strong>and</strong> alpha1D receptors control redox equilibrium <strong>and</strong> TNF-alpha shedding in<br />

bioaminergic neuronal cells.<br />

Benoit Schneider1, Mathéa Pietri1, Sophie Mouillet-Richard1, Myriam Ermonval1,<br />

Vincent Mutel2, Jean-Marie Launay3 <strong>and</strong> Odile Kellermann1<br />

1Institut André Lwoff-Institut Pasteur, CNRS UPR 1983, Laboratoire Différenciation<br />

Cellulaire et Prions, Villejuif Cedex, France. 2Pharma Research Department, F.<br />

Hoffman-La-Roche Ltd., Base, Switzerl<strong>and</strong>. 3Service de Biochimie, EA3621 Hôpital<br />

Lariboisière, Faculté de Pharmacie, Université apris V, Paris, France. E-mail:<br />

bschneid@vjf.cnrs.fr.<br />

Loss of neuronal homeostasis in neurodegenerative disorders, such as Alzheimer’s,<br />

Parkinson or Prion diseases, may arise from dysregulations of physiological <strong>signaling</strong><br />

pathways due to the accumulation of aggregation-prone proteins. For instance, alterations<br />

with serotonin (5-hydroxytryptamine, 5-HT)- or norepinephrine (NE)- dependent <strong>signaling</strong><br />

pathways have been reported in these pathologies. In addition, oxidative stress is also widely<br />

suspected to be a major cause of neuronal cell dysfunction or death, since post-mortem brain<br />

studies from affected patients exhibit enhanced indices of reactive oxygen species (ROS). A<br />

possible origin of oxidative stress could hence relate to deviation of ROS-coupled <strong>signaling</strong><br />

pathways that normally take part in the control of neuronal functions.<br />

We took advantage of a neuroectodermal progenitor, 1C11, endowed with the capacity<br />

to differentiate into serotonergic or noradrenergic neuronal cells to probe the possible<br />

involvement of 5-HT or NE in regulating the cellular redox state in neurons. Upon induction<br />

by Bt2cAMP, almost 100% of 1C11 cells acquire, within 4 days, a complete serotonergic<br />

phenotype (1C115-HT) including 5-HT synthesis, storage <strong>and</strong> uptake as well as three Gprotein<br />

coupled receptors of the 5-HT1B/1D, 5-HT2B <strong>and</strong> 5-HT2A subtypes. Under<br />

combined addition of Bt2cAMP <strong>and</strong> DMSO, 100% of 1C11 cells convert within 12 days into<br />

fully functional noradrenergic neurons (1C11NE) able to synthesize, store <strong>and</strong> take up NE.<br />

1C11NE cells transduce NE signals through a single alphalpha1D adrenoceptor. Along either<br />

differentiation program, the bioaminergic receptors act as autoreceptors <strong>and</strong> mediate the effect<br />

of 5-HT or NE in the coordination <strong>and</strong>/or onset of all neurotransmitter-associated functions.<br />

By using a panel of specific agonists towards each receptor subclass, we establish that<br />

5-HT2B receptors <strong>and</strong> alphalpha1D adrenoceptors are functionally coupled to ROS synthesis<br />

through NADPH oxidase activation in 1C115-HT <strong>and</strong> 1C11NE cells. The ROS response<br />

imparted by each receptor is restricted to 1C11-derived progenies that have implemented a<br />

complete serotonergic or noradrenergic phenotype. These observations indicate that 5-HT2B<br />

<strong>and</strong> alphalpha1D autoreceptors take part in the control of the cellular redox equilibrium in<br />

neuronal cells. In addition, <strong>our</strong> data identify TACE (TNF-alphalpha Converting Enzyme), a<br />

member of a disintegrin <strong>and</strong> metalloproteinase (ADAM) family, as a downstream target of the<br />

5-HT2B <strong>and</strong> alphalpha1D receptor-NADPH oxidase <strong>signaling</strong> pathway. Upon 5-HT2B or<br />

alphalpha1D receptor stimulation, ROS act as second message signals that fully govern TNFalphalpha<br />

shedding in the surrounding milieu of 1C115-HT <strong>and</strong> 1C11NE cells. TNFalphalpha<br />

may in turn contribute to neuronal homeostasis. Eventually, <strong>our</strong> study may have<br />

implications regarding the origin of oxidative stress as well as up-regulated expression of<br />

proinflammatory cytokines in neurodegenerative disorders, which may relate to the deviation<br />

of normal <strong>signaling</strong> pathways coupled to neurotransmitters.<br />

- 173 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 47<br />

Signaling pathway utilized by HSV-1 to induce NF-kB activation: possible role of<br />

herpesvirus entry receptor A<br />

M. Teresa Sciortino,1 M. Antonietta Medici,1 Francesca Marino-Merlo,1 Daniela<br />

Zaccaria,1 Maria Gioffrè,1 Assunta Venuti,1 A. Pia Camuti,1 S<strong>and</strong>ro Grelli,2 Antonio<br />

Mastino1<br />

1Dept. of Microbiol., Genet. <strong>and</strong> Mol. Sci., Univ. of Messina, Salita Sperone 31, 98166<br />

Messina, Italy. E-mail: antonio.mastino@unime.it. 2Dept. of Exp. Med. <strong>and</strong> Bioch. Sci.,<br />

Univ. of Rome “Tor Vergata”, Rome, Italy.<br />

We have previously demonstrated that wild type herpes simplex 1 (HSV-1), as well as nonreplicating<br />

UV-inactivated HSV-1, promptly activate the nuclear factor kappa B (NF-kB) in<br />

U937 monocytoid cells <strong>and</strong> that glycoprotein D (gD) of HSV-1 is sufficient by itself to exert a<br />

similar effect. We have then investigated the <strong>signaling</strong> pathway utilized by HSV-1 to initiate<br />

NF-kB activation <strong>and</strong>, particularly, whether <strong>our</strong> observation could be related to the capability<br />

of HSV-1-gD to directly stimulate NF-kB through its interaction with the herpesvirus entry<br />

receptor A (HveA). Here we report that: a) exposure to UV-inactivated HSV-1 induced a<br />

MOI-dependent activation of NF-kB in HveA expressing THP-1 monocytoid cells, b)<br />

exposure to soluble gD induced a dose-dependent activation of NF-kB in THP-1 cells that<br />

was inhibited by an antibody able to interfere with gD/HveA interaction, c) cocultivation of<br />

THP-1 cells with an adherent cell line forced to express wild type gD on its surface by stable<br />

transfection led to NF-kB activation, d) cell-to-cell contacts of THP-1 cells with the same<br />

adherent cell line forced to express on its surface, by stable transfection, a mutated form of gD<br />

lacking the capability to interact with HveA, did not activate NF-kB. These results suggest<br />

that HSV-1-gD/HveA interaction is sufficient for triggering a signal transduction pathway<br />

leading to NF-kB activation.<br />

- 174 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 48<br />

Expression of leptin, leptin receptor <strong>and</strong> hypoxia-inducible factor 1 in human<br />

endometrial cancer.<br />

Stanislaw Sulkowski, Mariusz Koda, Andrzej Wincewicz, Luiza Kanczuga-Koda,<br />

Mariola Sulkowska, Boguslaw Musiatowicz.<br />

Department of Pathology, Medical University of Bialystok, Waszyngtona 13, 15-269<br />

Bialystok, Pol<strong>and</strong>. E-mail: sulek@zeus.amb.edu.pl<br />

Recent studies suggested that leptin (Ob) <strong>and</strong> its receptor (ObR) could be involved in the<br />

pathogenesis of various human malignancies, among others in endometrial cancer. Moreover,<br />

hypoxia, which is associated with solid tumors, might stimulate, through hypoxia-inducible<br />

factor 1alpha (HIF-1alpha), expression of Ob <strong>and</strong> ObR. In the present study, we analyzed by<br />

immunohistochemistry the expression of Ob, ObR <strong>and</strong> HIF-1alpha in 60 cases of human<br />

endometrial cancer tissues. Additionally, we assessed correlations among studied proteins as<br />

well as relationships with selected clinicopathological features. Immunoreactivity for Ob,<br />

ObR <strong>and</strong> HIF-1alpha protein was observed in 56.7%, 30.0% <strong>and</strong> 78.3% of endometrial<br />

cancers, respectively. The expression of HIF-1alpha showed a significant positive correlation<br />

with Ob (p


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 49<br />

Activated melanocortin 3 receptor is endocytosed <strong>and</strong> leads to modification of<br />

transcription factor forkhead (Drosophila) homolog (rhabdomyosarcoma) like 1<br />

James M. Wachira, Bindhu Guruswamy, Lawrence C. Uradu <strong>and</strong> Cleo A. Hughes-<br />

Darden<br />

Department of Biology, Morgan State University, 1700 E. Cold Spring Lane, Baltimore<br />

MD 21251, U.S.A. E.mail: jwachir@morgan.edu, binduguru25@yahoo.com,<br />

lawrenceuradu@yahoo.com, chughes@moac.morgan.edu<br />

Melanocortins play a central role in autonomic modulation of metabolism by acting through a<br />

family of highly homologous G-protein coupled receptors. Studies with gene knockout mice<br />

have implicated melanocortin receptors, MC3R <strong>and</strong> MC4R, in the etiology of obesity, insulin<br />

resistance <strong>and</strong> salt-sensitive hypertension. In an attempt to better underst<strong>and</strong> the mechanisms<br />

of function of these receptors, we expressed MC3R <strong>and</strong> MC4R in neuronal cells <strong>and</strong><br />

demonstrated their colocalization to several membrane regions. We now show that in<br />

cultured neuronal cells, MC3R localizes to lipid rafts <strong>and</strong> undergoes endocytic internalization<br />

upon activation by gamma-MSH through a protein kinase sensitive pathway. The appearance<br />

of the internalized receptor in lysosomes suggests that it is subsequently degraded. The<br />

expression of protein kinase A regulatory subunits <strong>and</strong> of c-Jun <strong>and</strong> c-Fos was analyzed by<br />

either immunoblotting or real-time PCR. No discernable changes were observed in the<br />

expression levels of these protein kinase A <strong>and</strong> protein kinase C responsive genes. However,<br />

an observed modification of the protein kinase B responsive FKHRL1 in MC3R expressing<br />

cells suggests an important role for the PI3K/AKT pathway in melanocortin mediated effects.<br />

Immunohistochemical studies showed a robust expression of MC3R protein in brain nuclei<br />

with relevance to cardiovascular function <strong>and</strong> fluid homeostasis thereby suggesting that the<br />

physiological effects of melanocortins on the cardiovascular system arise from effects on the<br />

central nervous system.<br />

Supported by MBRS-Score grant #2SO6-GM051971-08 <strong>and</strong> RCMI grant # G12RR017581.<br />

Lawrence Uradu is supported by NIH/MARC grant #5T34GM007977-21<br />

- 176 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 50<br />

Aberrant activation of Wnt/#-catenin pathway in esophageal squamous cell carcinoma<br />

(ESCC): evidence of FRAT1 overexpression<br />

Yihua Wang1, Shuang Liu1, Wei Zhang1, Hongxia Zhu1, Xiaobo Zhou1, Cuiqi Zhou1,<br />

Guo Zhang1, Lanping Quan1, Jinfeng Bai1, Liyan Xue2, Ning Lu2 <strong>and</strong> Ningzhi Xu1<br />

1Laboratory of Cell <strong>and</strong> Molecular Biology; 2Department of Pathology; Cancer<br />

Institute & Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union<br />

Medical College, Beijing 100021, China. E-mail: xningzhi@public.bta.net.cn<br />

Esophageal squamous cell carcinoma (ESCC) ranks among one of the most frequent cause of<br />

cancer death in the world. Despite recent advances in therapy <strong>and</strong> management, esophageal<br />

cancer remains a killer. Therefore, new therapies based on a better underst<strong>and</strong>ing of the<br />

biology of esophageal cancer are most required. Recently, accumulation of nuclear <strong>and</strong><br />

cytoplasmic #-catenin has been observed in ESCC. However, mechanisms underlying this<br />

phenomenon remain unknown. Frequently rearranged in advanced T-cell lymphomas-1<br />

(FRAT1), strikingly overexpressed in some human esophageal cancer cell lines, is a positive<br />

regulator of the Wnt/#-catenin pathway. Whereas FRAT1 is a regulator of cytoplamsic #catenin,<br />

little is known with regard to the molecular relationship between FRAT1 <strong>and</strong> #catenin<br />

in human cancers, including ESCC.<br />

In this study, FRAT1 cDNA was cloned <strong>and</strong> transfected into human ESCC cells. The effects<br />

of FRAT1 overexpression on cellular growth <strong>and</strong> transcriptional activity of #-catenin/T-cell<br />

factor (TCF) were analyzed. RNA interference (RNAi) was also used to determine the<br />

functions of FRAT1. In situ hybridization <strong>and</strong> immunohistochemical analysis were performed<br />

to identify the level of FRAT1 mRNA expression <strong>and</strong> localization of #-catenin in surgical<br />

specimens of ESCC respectively.<br />

We observe that FRAT1 is overexpressed in ESCC <strong>and</strong> speculate that aberrant activation of<br />

Wnt/#-catenin pathway in esophageal cancer appears to be due to upstream events such as<br />

FRAT1 overexpression. Analysis of freshly resected ESCC specimens showed that FRAT1<br />

was overexpressed in approximately 74% of tumor samples compared to matched normal<br />

ones. We found that overexpression of FRAT1 in ESCC cell line KYSE150 significantly<br />

promoted cell growth, whereas suppression of FRAT1 level by RNAi markedly inhibited<br />

growth of esophageal tumor cells. In addition, FRAT1 overexpression induced the nuclear<br />

accumulation of #-catenin <strong>and</strong> promoted the transcriptional activity of #-catenin/TCF. These<br />

effects were reversed by coexpression of GSK3# or &NTCF4. Furthermore, aberrant<br />

expression of #-catenin was correlated with FRAT1 overexpression in primary human ESCC.<br />

Taken together, <strong>our</strong> data support the novel hypothesis that FRAT1 overexpression is critical<br />

to Wnt/#-catenin pathway activation in ESCC. These findings identify components of Wnt/#catenin<br />

pathway, such as FRAT1 overexpression, as possible therapeutic targets for the<br />

development of novel molecular treatments for human esophageal cancer.<br />

- 177 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 51<br />

PDGFRbetaY857F mutant mediates PDGF-BB-induced signalling <strong>and</strong> chemotaxis,<br />

despite severely reduced kinase activity.<br />

Piotr Wardega, Carl-Henrik Heldin, Johan Lennartsson.<br />

Ludwig Institute for Cancer Research, Uppsala University, Box 595, SE-751 24 Uppsala,<br />

Sweden<br />

Piotr.Wardega(AT)LICR.uu.se,C-H.Heldin(AT)LICR.uu.se,<br />

Johan.Lennartsson(AT)LICR.uu.se<br />

Platelet-derived growth factor receptor (PDGFR) becomes dimerized <strong>and</strong> activated upon<br />

lig<strong>and</strong> binding, which leads to receptor transphosphorylation <strong>and</strong> increases its enzymatic<br />

activity. In the present study, we studied the effects of mutation of tyrosine residue 857 in the<br />

activation loop of PDGFRbeta, to phenylalanine. In agreement with published work, <strong>our</strong><br />

studies show, that PDGFRbetaY857F has much lower kinase activity in vitro in comparison<br />

to PDGFRbetaWT , both as measured as receptor autophosphorylation <strong>and</strong> as phosphorylation<br />

of an exogenous substrate. Interestingly, PDGF-induced tyrosine phosphorylation of<br />

PDGFRbeta, in a cellular context, is not affected by the Tyr857 mutation. There are, however<br />

significant changes in ability of PDGFRbetaY857F to induce signal transduction compared to<br />

the wild-type receptor. For example, we could show that Stat5 signalling differs dramatically<br />

between PDGFRbetaWT <strong>and</strong> PDGFRbetaY857F. We observed almost complete loss of<br />

PDGF-induced Stat5 phosphorylation by the mutant receptor. Surprisingly Stat3, which<br />

belongs to the same group of transcription factors as Stat5, was phosphorylated to the same<br />

extent by mutant <strong>and</strong> wild-type receptor. In addition, we have also found that the Akt <strong>and</strong><br />

JNK signalling pathways are not affected by the PDGFRbetaY857F mutation. Moreover, the<br />

Erk pathway was activated by slower kinetics in the mutant receptor. Interestingly, in spite of<br />

the defect in the PDGFRbetaY857F receptors kinase activity <strong>and</strong> the changes observed in<br />

signal transudction it could still efficiently induce cell chemotaxis. It is known that many<br />

cytoplasmic tyrosine kinases are capable of phosphorylating the PDGFR <strong>and</strong> it is possible that<br />

these may compensate for the lowered enzymatic activity of PDGFRbetaY857F . In summary,<br />

we have demonstrated that the PDGFRbetaY857F has defect kinase activity, but is<br />

nevertheless phosphorylated in the cell, presumably by cytoplasmic kinases <strong>and</strong> can induce<br />

signals sufficient for normal chemotaxis toward PDGF-BB.<br />

- 178 -


Session II : Receptor <strong>signaling</strong> <strong>and</strong> G proteins Poster II, 52<br />

AF6 negatively regulates Rap1-induced cell adhesion<br />

Zhongchun Zhang, Holger Rehmann, Leo S. Price, Jurgen Riedl, <strong>and</strong> Johannes L. Bos<br />

From the Department of Physiological Chemistry <strong>and</strong> Centre of Biomedical Genetics,<br />

University Medical Centre Utrecht, The Netherl<strong>and</strong>s.<br />

Address correspondence to Johannes L. Bos, Department of Physiological Chemistry<br />

<strong>and</strong> Centre of Biomedical Genetics, University Medical Centre Utrecht, Universiteitsweg<br />

100, 3584 CG Utrecht, The Netherl<strong>and</strong>s. Tel. +31302538988; Fax. +31302539035 E-mail:<br />

J.L.Bos@med.uu.nl<br />

Small GTPases of the Ras family are molecular switches, whose functions are guiding<br />

extracellular signals towards cellular responses. Rap, one of the most extensively-studied<br />

members of the family, regulates cell proliferation, cell differentiation, cell death, <strong>and</strong> most<br />

notable cell-matrix <strong>and</strong> cell-cell adhesion.<br />

AF6 is involved in the connection of membrane-associated proteins to the actin cytoskeleton.<br />

It binds to Ras-like small GTPases <strong>and</strong> is suggested to be an effector of both Ras <strong>and</strong> Rap.<br />

In the absence of an immune challenge, T cells circulate the body in a non-adhesive state.<br />

The maintenance of this non-adhesive condition prevents inappropriate immune responses<br />

from occurring. An increase in Rap activation is sufficient to rapidly upregulate T cell<br />

adhesiveness, demonstrating that Rap <strong>signaling</strong> must be tightly controlled in unstimulated T<br />

cells.<br />

In this study, we show that knockdown of AF6 in T lymphocyte by RNAi enhances Rap1induced<br />

integrin-mediated cell adhesion, whereas overexpression of AF6 has the opposite<br />

effect. We conclude that AF6 is a negative regulator of Rap-induced cell adhesion. In<br />

addition, <strong>our</strong> results suggest that endogenous AF6 may function to buffer GTP-Rap in resting<br />

cells, maintaining it in a non-productive complex. Therefore, loss of AF6 function may result<br />

in immunological disorders.<br />

- 179 -


Notes<br />

- 180 -


Notes<br />

Notes<br />

- 181 -


- 182 -


Session III. Protein kinase cascades as therapeutic targets<br />

- 183 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 1<br />

Cyclic nucleotide-dependent inhibition of ecto-nucleotide<br />

pyrophosphatase/phosphodiesterase 1 (E-NPP1) expression.<br />

I. Aerts1, P.P. De Deyn2<strong>and</strong> H. Slegers1<br />

Department of Biomedical Sciences, Cellular Biochemistry1, Neurochemistry <strong>and</strong><br />

behavi<strong>our</strong>2, University of Antwerp, Belgium<br />

Ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (E-NPP1) is a member of a multigen<br />

family which contains NPP1 (PC-1), NPP2 (autotaxine) <strong>and</strong> NPP3 (B10; gp130RB13-6).<br />

These are type II transmembrane metalloenzymes that hydrolyse ATP into AMP <strong>and</strong> PPi or<br />

lysophosphatidylcholine into lysophosphatidic acid. NPP1 is expressed in different tissues<br />

<strong>and</strong> abnormal expression is observed in several pathologies (1).<br />

Glioblastoma multiforme (GBM) is the most aggressive glioma with an expected lifespan of<br />

less than one year. Immunohistochemistry demonstrated that NPP1 expression is not detected<br />

in normal tissue while its expression increased with the gradation of glioma. NPP1 is highly<br />

expressed in reactive astrocytes of GBM.<br />

The rat C6 glioma cell line, an experimental ‘in vitro’ <strong>and</strong> ‘in vivo’ model system for GBM<br />

(2), is used for the study of the expression of NPP1. Cyclic AMP-dependent induction of<br />

differentiation resulted in inhibition of NPP1 expression. The use of specific PKA inhibitors<br />

indicated that this decrease in expression is PKA-dependent. Nitroprusside, a nitric oxide<br />

(NO) donor, inhibited the expression of NPP1 suggesting that NO may be involved in the<br />

latter mechanism. BAY 41-2272, a NO-independent activator of soluble guanylate cyclase<br />

(sGC), attenuated NPP1 expression. The presented data indicated that inhibition of NPP1<br />

expression may be due to NO-induced activation of sGC. In addition dibutyryl cGMP, also<br />

attenuated the expression of NPP1, characterizing that NO/sGC/cGMP as elements of the<br />

signal transduction pathway that modulates the expression of NPP1. The signal transduction<br />

components of the NO-dependent pathway that effects NPP1 expression remain to be<br />

determined.<br />

The elucidation of the mechanism of NPP1 expression may lead to new medications or<br />

therapies for the treatment of patients with GBM.<br />

- 184 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 2<br />

Multiple roles of PI-3’ kinase / Akt <strong>signaling</strong> in human hepatoma cell proliferation <strong>and</strong><br />

drug-induced apoptosis<br />

Catherine alexia 3, Marlène Bras 2, Guillaume Fallot 3, Nathalie Vadrot 3, Fanny<br />

Daniel 3, Malika Lasfer 3, Houda Tamouza 3 & André Groyer 1<br />

1 Inserm U.683 <strong>and</strong> 3 Inserm U.481, Faculté de Médecine Xavier Bichat, 75870 Paris<br />

Cédex 18, France ; 2 Institut Pasteur, Département d'Immunologie, 75724 Paris Cedex<br />

15, France. e-mail: groyer@bichat.inserm.fr<br />

IGF-II <strong>and</strong> type I-IGF receptor (IGF-IR) gene expression is increased in primary liver<br />

tum<strong>our</strong>s <strong>and</strong> transgenic mice over-expressing IGF-II in the liver develop hepatocarcinoma<br />

(HCC) spontaneously, suggesting that alterations of IGF-IR <strong>signaling</strong> in vivo may play a role<br />

in the auto/paracrine control of hepatocarcinogenesis. We have addressed the contribution of<br />

PI-3’K/Akt <strong>signaling</strong> on the proliferation of HepG2 human hepatoma cells <strong>and</strong> on their<br />

protection against doxorubicin-induced apoptosis. Both basal HepG2 cell DNA replication<br />

<strong>and</strong> that stimulated by IGF-IR <strong>signaling</strong> were inhibited by the specific PI-3’K inhibitor<br />

Ly294002 (Ly). In the former case, PI-3’K <strong>signaling</strong> overcame cell cycle arrest in G1 via<br />

increased cyclin D1 protein <strong>and</strong> decreased p130Rb <strong>and</strong> p27kip1 gene expression. Doxorubicin<br />

treatment induced apoptosis in HepG2 cells <strong>and</strong> was concomitant with the proteolytic<br />

cleavage of Akt-1 <strong>and</strong> -2. Drug-induced apoptosis was reversed by IGF-I <strong>and</strong> was (i)<br />

dependent on Akt-1 <strong>and</strong> -2 phosphorylation <strong>and</strong> (ii) accompanied by the inhibition of initiator<br />

caspase-9 activity, suggesting that IGF-IR <strong>signaling</strong> interferes with mitochondria-dependent<br />

apoptosis. Accordingly, Ly enhanced doxorubicin-induced apoptosis <strong>and</strong> suppressed its<br />

reversal by IGF-I. Altogether, the data emphasize the crucial role of PI-3’K/Akt <strong>signaling</strong> (i)<br />

in basal as well as IGF-IR-stimulated HepG2 cell proliferation <strong>and</strong> (ii) in controlling both<br />

doxorubicin-induced apoptosis (e.g., drug-induced cleavage of Akt) <strong>and</strong> its reversal by IGF-I<br />

(protection against apoptosis parallels the extent of Akt phosphorylation). They suggest that<br />

targeting Akt activity or downstream Akt effectors (e.g. GSK3-beta, FOXO transcription<br />

factors) may help define novel therapeutic strategies of increased efficacy in the treatment of<br />

HCC-bearing patients.<br />

- 185 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 3<br />

Grb2 associated binder 1 (Gab1) adaptor/scaffolding protein regulates Erk signal in<br />

human B cells<br />

Adrienn Angyal1, David Medgyesi2, Gabriella Sarmay1<br />

1Department of Immunology, Lor<strong>and</strong> Eotvos University, Budapest 1117, Hungary, email:<br />

a1117@freemail.hu, 2Research Group of the Hungarian Academy of Sciences at<br />

the Department of Immunology, Lor<strong>and</strong> Eotvos University, Budapest 1117, Hungary<br />

Grb2-associated binder 1 (Gab1) is a member of the Gab/dos family of adapter molecules<br />

involved in the signal transduction pathways of a variety of growth factors, cytokines, <strong>and</strong><br />

antigen receptors. In contrast to its positive role upon cytokine <strong>and</strong> growth factor receptor<br />

<strong>signaling</strong>, Gab1 was shown to be a negative regulator of T independent 2 response of<br />

marginal zone B cells in the spleen <strong>and</strong> to participate in a negative feedback loop of T cell<br />

activation. Gab adaptor proteins have several tyrosine residues that are phosphorylated upon<br />

activation of tyrosine kinases <strong>and</strong> consequently bind <strong>signaling</strong> molecules with SH2 domains,<br />

such as SHP-2 tyrosine phosphatase <strong>and</strong> phosphatidyl inositol 3-kinase (PI3-K); <strong>and</strong> also<br />

have pleckstrin homologue (PH) domain, that binds to PIP3 in the cell membrane. Gab1 was<br />

shown to participate in activating ras/MAPK pathway upon EGFR <strong>signaling</strong> that was<br />

mediated by the SHP-2 tyrosine phosphatase <strong>and</strong> the ras regulating protein, rasGAP.<br />

Furthermore, via positive feed back regulation, Gab1 enhances PI3-K activity, resulting in a<br />

high level of phosphatidyl inositol 3,4,5 trisphosphate (PIP3) in the plasma membrane.<br />

The function of Gab1 in B cell <strong>signaling</strong> has not been clarified yet. We explored this question<br />

by using siRNA to temporarily knock down Gab1 in BL41 Burkitt lymphoma cell line. The<br />

transfer of siRNA to the cells was carried out by nucleofector technology (Amaxa). 24 h after<br />

the transfection with Gab1 siRNA the efficiency of silencing <strong>and</strong> its functional consequences<br />

were tested by Western blot analysis. Activation of Erk <strong>and</strong> Akt was followed by using<br />

antibodies specific for the phosphorylated forms of the kinases. Erk activation was almost<br />

completely blocked while Akt phosphorylation was partially reduced in the Gab1 knocked<br />

down samples. These results suggest that in human B cells Gab1 is a major regulator of Erk,<br />

<strong>and</strong> a less potent regulator of Akt/PKB.<br />

- 186 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 4<br />

Mallory Body Keratin Aggresomes Form in Response to Integrin Initiated Cell<br />

Signaling MAP Kinase pathway<br />

Fawzia Bardag-Gorce, Yong Wu, Latoiya Wilson, Jun Li, Li Nan, Barbara A. French<br />

<strong>and</strong> Samuel W. French.<br />

LABioMed at Harbor UCLA Medical Center. Torrance, CA, 90502. USA.<br />

Mallory body formation is observed in the liver cells of alcoholic patients, as well as in other<br />

non-alcoholic diseases. Several factors, including proteasome inhibition, are hypothesized to<br />

be the cause of this phenomenon of MB formation. Feeding mice the drug diethyl-1, 4dihydro-2,<br />

4, 6-trimethyl-3, 5-pyridinedicarboxylate (DDC) for 10 weeks, is a good model to<br />

study Mallory body formation. Recent study showed that the primary culture of hepatocytes<br />

isolated from the liver of these drug primed mice is also a good in vitro model to study this<br />

phenomenon.<br />

The MB formation was in several case associated with tumor formation. In these drugprimed<br />

mice tumor growth was noticed in the liver of mice where the tumor cells showed also<br />

formation of MB. The mitogen–activated protein kinase pathway, a central regulator of<br />

tumor growth <strong>and</strong> cell differentiation, was studied to further examine the array of factors<br />

leading to Mallory body formation. MEK is a central focus of the pathway <strong>and</strong> it is<br />

associated with the up regulation by phosphorylating of the map kinase P44/42 (ERK &).<br />

Western blot analysis exhibited increased phosphorylation of p44/42, which was associated<br />

with an increase of Mallory body formation in liver cells of mice fed DDC. In the DDCprimed<br />

primary culture of hepatocytes there was a marked MBs formation <strong>and</strong> this formation<br />

was significantly reduced when U0126, an inhibitor of MEK1 protein, which reduced<br />

significantly the phosphorylated forms of ERK1/2. Other tumor markers such as AFP, was<br />

examined <strong>and</strong> showed an increase in the protein level when the mice were fed DDC <strong>and</strong><br />

where a significant amount of MBs were formed. This data are consistent with the conclusion<br />

that Mallory body formation is associated with preneoplastic state of the cell.<br />

- 187 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 5<br />

Sanguinarine inhibits VEGF-induced Akt phosphorilation<br />

Giuseppina Basini, Sujen E. Santini, Simona Bussolati, Francesca Grasselli<br />

Dipartimento di Produzioni Animali, Biotecnologie Veterinarie, Qualità e Sicurezza<br />

degli Alimenti, Sezione di Fisiologia Veterinaria, Università di Parma, 43100 Parma,<br />

Italy. E-mail: basini@unipr.it<br />

Angiogenesis, the process by which new blood vessels develop from pre-existing ones, plays<br />

an essential role during embryogenesis, adult vascular remodelling <strong>and</strong> in several pathological<br />

disorders including cancers, ischemic cardiovascular disease, diabetic retinopathy, wound<br />

healing <strong>and</strong> inflammation. Unraveling the pro- <strong>and</strong> anti-angiogenetic mechanisms would offer<br />

therapeutic options to ameliorate or perhaps even cure disorders that are now leading causes<br />

of mortality. Research conducted over the past 10 years has demontrated that VEGF is a<br />

major regulator of angiogenesis by interacting with cellular receptors <strong>and</strong> communicating<br />

with the nucleus through a network of intracellular <strong>signaling</strong> pathways. Sanguinarine (SA), an<br />

alkaloid isolated from Sanguinaria Canadensis, is known for its antiangiogenetic effects by<br />

suppressing basal <strong>and</strong> VEGF-induced new vessel growth. The present study was aimed to<br />

evaluate possible effect of SA on Akt phosphorylation, a critical event in VEGF-mediated<br />

angiogenesis. Briefly, 2'105 immortalized porcine aortic endothelial cell (AOC) generously<br />

provided by José Yelamos (Hospital Universitario Virgen de la Arrixaca, El Palmar, 30120<br />

Murcia, Spain) were seeded in 1 ml M199 <strong>and</strong> treated with: 1) VEGF (100 ng/ml); 2) SA (300<br />

nM); 3) VEGF+SA at room temperature. After a 10 min incubation, Akt activity was<br />

evaluated by Akt/PKB Kinase Activity Assay Kit (Stressgene, Victoria, Canada). VEGF<br />

treatment significantly (p


Session III : Protein kinase cascades as therapeutic targets Poster III, 6<br />

Constitutively active cytoplasmic c-Jun N-terminal kinase 1 is a dominant regulator of<br />

dendritic architecture: role of microtubule-associated protein 2 as an effector.<br />

Benny Bjorkblom, Nina Ostman, Vesa Hongisto, Vladislav Komarovski, Jan-Jonas<br />

Filen, Tuula Nyman, Tuula Kallunki, Michael J. C<strong>our</strong>tney, Eleanor T. Coffey.<br />

Turku Centre for Biotechnology, Åbo Akademi <strong>and</strong> Turku University, Turku FIN-<br />

20521, Finl<strong>and</strong>. Email:ecoffey@btk.fi<br />

Normal functioning of the nervous system requires precise regulation of dendritic shape <strong>and</strong><br />

synaptic connectivity. Here, we report a severe impairment of dendritic structures in the<br />

cerebellum <strong>and</strong> motor cortex of c-Jun N-terminal kinase 1 (JNK1)-deficient mice. Using an<br />

unbiased screen for c<strong>and</strong>idate mediators, we identify the dendrite-specific high-molecularweight<br />

microtubule-associated protein 2 (MAP2) as a JNK substrate in the brain. We<br />

subsequently show that MAP2 is phosphorylated by JNK in intact cells <strong>and</strong> that MAP2<br />

proline-rich domain phosphorylation is decreased in JNK1-/- brain. We developed<br />

compartment-targeted JNK inhibitors to define whether a functional relationship exists<br />

between the physiologically active, cytosolic pool of JNK <strong>and</strong> dendritic architecture. Using<br />

these, we demonstrate that cytosolic, but not nuclear, JNK determines dendritic length <strong>and</strong><br />

arbor complexity in cultured neurons. Moreover, we confirm that MAP2-dependent process<br />

elongation is enhanced after activation of JNK. Using JNK1-/- neurons, we reveal a dominant<br />

role for JNK1 over ERK in regulating dendritic arborization, whereas ERK only regulates<br />

dendrite shape under conditions in which JNK activity is low (JNK1-/- neurons). These<br />

results reveal a novel antagonism between JNK <strong>and</strong> ERK, potentially providing a mechanism<br />

for fine-tuning the dendritic arbor. Together, these data suggest that JNK phosphorylation of<br />

MAP2 plays an important role in defining dendritic architecture in the brain.<br />

- 189 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 7<br />

Proliferation of human melanoma cells related to high expression of PKA regulatory<br />

subunit 1A protein<br />

Sara Bondioni, Erika Peverelli , Monica Rodolfo1 ,Stefano Ferrero 2, Silvano Bosari 2,<br />

Paolo Beck-Peccoz Anna Spada, Andrea Lania <strong>and</strong> Giovanna Mantovani.<br />

Institute of Endocrine Sciences, University of Milan, Ospedale Maggiore IRCCS,<br />

1Istituto Nazionale dei Tumori <strong>and</strong> 2Pathology Unit, Department of Medicine, Surgery<br />

<strong>and</strong> Dentistry, A.O. San Paolo <strong>and</strong> Ospedale Maggiore IRCCS; Milan, Italy.<br />

The two regulatory subunits (R1 <strong>and</strong> R2) of PKA are differentially expressed in several<br />

cancer cell lines <strong>and</strong> studies indicate distinct roles for these subunits in growth control. The<br />

aim of this study was to evaluate the expression of the different PKA regulatory subunits<br />

(R1A, R2A, R2B) in human melanomas, as well as the effects of selective subunit activation<br />

on cell proliferation. Immunohystochemistry demonstrated high expression levels of the three<br />

PKA regulatory subunits studied in all melanoma samples. On the contrary, in normal<br />

melanocytes R1A was absent or expressed at very low levels, suggesting a higher R1/R2 ratio<br />

in tumoral tissue compared to normal one. The effect of R1/R2 ratio on proliferation was<br />

assessed in 6 human melanoma cell lines that show the same PKA regulatory subunits<br />

expression pattern of melanoma samples. The R2-selective cAMP analogue 8-Cl-cAMP dosedependently<br />

inhibited cell proliferation (60% inhibition at 5 M 8-Cl-cAMP) through the<br />

induction of apoptosis. Inhibition of cell proliferation was also obtained by R1A silencing<br />

through siRNA technique. Our data indicate that a high R1/R2 ratio promotes proliferation in<br />

human melanoma cells.<br />

- 190 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 8<br />

Effects of pituitary adenylate cyclase-activating polypeptide (PACAP) on ERK1/2<br />

(p44/p42 Mitogen-Activated Protein Kinases) cascade in pancreatic beta-cells<br />

Christophe Broca, Safia Costes, Nathalie Linck, Joel Bockaert, Stephane Dalle <strong>and</strong><br />

Gyslaine Bertr<strong>and</strong>.<br />

Equipe Avenir INSERM; IGF; INSERM U661; CNRS UMR5203; 141 rue de la<br />

Cardonille, 34094 Montpellier cedex 5, France. E-mail : christophe.broca@igf.cnrs.fr<br />

PACAP, neuropeptide belonging to the vasoactive intestinal peptide (VIP)/glucagon/secretin<br />

family <strong>and</strong> interacting with both specific (PAC1) <strong>and</strong> VIP-shared (VPAC) receptors, plays an<br />

important role in pancreatic beta-cell function. The ERK1/2 cascade, which modulates cell<br />

proliferation <strong>and</strong> gene transcription but also cytoplasmic processes such as insulin granules<br />

exocytosis, could thus mediate part of PACAP pancreatic effects. So, the aim of <strong>our</strong> study<br />

was to investigate in pancreatic beta-cells (INS-1E <strong>and</strong> rat islets) the relationships between<br />

PACAP <strong>and</strong> ERK1/2 activation (measured by western blotting of doubly phosphorylated<br />

activated forms of these kinases) as well as the <strong>signaling</strong> cascade hereby activated.<br />

First, in the absence of glucose, PACAP (100 nM) transiently (15 min) stimulated ERK1/2<br />

phosphorylation whereas in the presence of a stimulating glucose concentration (8.3 mM),<br />

PACAP induced a sustained <strong>and</strong> long lasting (( 3 h) stimulation of ERK1/2 phosphorylation<br />

following a biphasic pattern. The activation of ERK1/2 cascade by PACAP was clearly<br />

glucose-dependent (0-20 mM), <strong>and</strong> was totally abolished by the MEK (ERK1/2 kinases)<br />

inhibitor PD98059 (20 µM). Moreover, PACAP-induced ERK1/2 phosphorylation was<br />

clearly sensitive to the inhibition of the protein kinase A (PKA) by 10 µM H89 (-40%), the<br />

inhibition of L-type calcium channel by 2 µM nifedipin (-65%), but also sensitive to the<br />

inhibition of the phosphatidylinositol 3-kinase (PI3K) by 100 nM wortmannin or 10 µM<br />

LY294002 (-40% <strong>and</strong> -50% respectively). Finally, compared to PACAP <strong>and</strong> forskolin, we<br />

demonstrated that VIP exerts only minor effects on ERK1/2 cascade either in the absence or<br />

the presence of glucose.<br />

We thus concluded that PACAP exerts a sustained <strong>and</strong> prolonged effect on the pancreatic<br />

beta-cell ERK1/2 cascade, mainly through the PAC1 receptor, <strong>and</strong> also through the<br />

stimulation of both calcium-, PKA- <strong>and</strong> PI3K-cascade. The coupling of PAC1 receptors to<br />

ERK1/2 could play an important role in the insulinotropic function as well as in the survival<br />

of pancreatic beta-cells, <strong>and</strong> thus be relevant in diabetes physiopathology.<br />

- 191 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 9<br />

Reduced Bcr-Abl function leads to erythroid differentiation of K562 cells via downregulation<br />

of ERK<br />

A. Brózik*, Cs. Heged!s**, N.P. Casey***, A. Bors*, M. Geiszt****, K. Német* <strong>and</strong> M.<br />

Magócsi*<br />

* National Medical Centre, Dept. of Molecular <strong>and</strong> Cell Biology, Budapest, Hungary<br />

** Eötvös Loránd Univ. Fact. of Science<br />

*** Univ. of Tasmania, Division of Medicine, Australia<br />

**** Semmelweis. Univ. Dept. of Physiology, Budapest, Hungary<br />

E-mail: magocsi@biomembrane.hu<br />

The chimeric bcr-abl gene encodes a constitutively active tyrosine kinase that leads to<br />

abnormal transduction of growth <strong>and</strong> survival signals. Selective inhibition of this tyrosine<br />

kinase with Gleevec reduces the autophosphorylation of Bcr-Abl protein <strong>and</strong> subsequently the<br />

activity of ERK1/2 MAP kinases in K562 cells. According to <strong>our</strong> earlier observation that<br />

down-regulation of ERK1/2 activities is able to induce differentiation of various erythroid cell<br />

lines, we show here that inhibition of the MEK/ERK signalling pathway by the specific MEK<br />

inhibitor UO126 is sufficient to induce hemoglobin expression in both mRNA <strong>and</strong> protein<br />

levels - similarly to the effect of Gleevec treatment. Gleevec strongly reduces Bcr-Abl,<br />

STAT-5, <strong>and</strong> ERK phosphorylation, decreases Bcl-xl level <strong>and</strong> induces caspase-3 dependent<br />

apoptosis. In contrary, UO126 reduces only ERK activity <strong>and</strong> does not induce cell death.<br />

For studying the effect of reduced Bcr-Abl function on erythroid differentiation at the level of<br />

bcr-abl transcript we applied siRNA approach. Based on <strong>our</strong> FISH studies, K562 cells carry<br />

more than 10 copies of the fusion bcr-abl gene. We used retroviral vector with GFP reporter<br />

<strong>and</strong> H1 promoter driven DNA templates for siRNA mediated degradation of bcr-abl mRNA.<br />

Despite the high (> 90%) transduction efficiency we could detect only a transient decrease in<br />

Bcr-Abl protein <strong>and</strong> in phosphorylated ERK1/2 levels. This transient change in Bcr-Abl<br />

signalling was sufficient to induce hemoglobin mRNA expression at 96-144 h<strong>our</strong>s after<br />

transduction without significant cell death. These results suggest that reduced Bcr-Abl<br />

function can influence the differentiation blockade in chronic myeloid leukemia cells via the<br />

ERK pathway.<br />

- 192 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 10<br />

Protein Kinase B (c-akt): a molecular switch in regulation of lineage choice decisions<br />

during myelopoiesis<br />

Mir<strong>and</strong>a Buitenhuis, Liesbeth Verhagen, Christian Geest, Hanneke van Deutekom <strong>and</strong><br />

Paul J. Coffer.<br />

Molecular Immunology Lab, Dept. of Immunology, University Medical Center Utrecht,<br />

Lundlaan 6, 3584 EA Utrecht, The Netherl<strong>and</strong>s.<br />

Phosphatidylinositol 3-kinase (PI3K) has been reported to play a critical role in<br />

proliferation <strong>and</strong> survival of a variety of cell types; however, a role in regulating blood cell<br />

production is largely unknown. We have utilized an ex-vivo differentiation system, using<br />

human umbilical cord blood derived CD34+ hematopoietic progenitor cells, to investigate the<br />

role of PI3K <strong>and</strong> its downstream effectors in myeloid differentiation.<br />

Inhibition of PI3K, with the pharmacological inhibitor LY294002, completely blocked<br />

proliferation <strong>and</strong> differentiation during granulopoiesis. To pin-point the molecular<br />

mechanisms responsible for this effect, we utilized specific pharmacological inhibitors of<br />

PKB/c-akt or mTOR, both down-stream effectors of PI3K. Inhibition of PKB blocked<br />

progenitor proliferation without affecting cell survival. Interestingly, inhibition of PKB,<br />

abrogated neutrophil maturation, but conversely dramatically enhanced terminal<br />

differentiation of eosinophils. Retroviral transductions were performed to ectopically express<br />

a constitutively active PKB (myrPKB) in CD34+ cells. Transduction of cells with myrPKB<br />

resulted in enhanced neutrophil differentiation <strong>and</strong> monocyte development compared to cells<br />

transduced with empty vector alone. Conversely, ectopic expression of myrPKB resulted in an<br />

almost complete block in eosinophil differentiation, surprisingly demonstrating neutrophil<br />

differentiation even in the presence of IL-5. In order to investigate the role of PKB in<br />

regulation of hematopoiesis in vivo, #2-microglobulin-/-NOD/SCID mice were transplanted<br />

with CD34+ cells ectopically expressing active myrPKB or dominant-negative PKBcaax.<br />

Transplantation of mice with CD34+ cells ectopically expressing myrPKB resulted in<br />

enhanced neutrophil <strong>and</strong> monocyte development, whereas ectopic expression of PKBcaax<br />

induced eosinophil development in vivo. To investigate which downstream signal<br />

transduction pathways could be involved in PKB mediated regulation of myelopoiesis,<br />

hematopoietic progenitors were either retrovirally transduced with myrPKB or treated with<br />

pharmacological inhibitors, <strong>and</strong> western blot analysis was performed. Preliminary data<br />

indicate that phosphorylation of C/EBPalpha, a transcription factor known to play an<br />

important role in regulation of myelopoiesis, is inhibited upon PKB activation in<br />

hematopoietic progenitors.<br />

Taken together, these results demonstrate that PKB activity plays a critical role in the<br />

regulation of cell fate choices during myeloid lineage commitment, most likely via regulation<br />

of phosphorylation of C/EBPalpha. High PKB activity promotes neutrophil differentiation<br />

<strong>and</strong> monocyte development, while reduction of PKB activity is required to induce eosinophil<br />

differentiation.<br />

- 193 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 11<br />

Anaphylactic shock critically depends on PI3K <strong>and</strong> eNOS-derived NO<br />

Anje Cauwels1, Ben Janssen2 <strong>and</strong> Peter Brouckaert1<br />

1Department for Molecular Biomedical Research, Ghent University <strong>and</strong> VIB,<br />

Technologiepark 927, B-9052 Ghent, Belgium; 2Department of Pharmacology <strong>and</strong><br />

Toxicology, University of Maastricht, Maastricht 6200 MD, The Netherl<strong>and</strong>s<br />

Anaphylaxis is an acute, severe <strong>and</strong> life-threatening systemic allergic reaction that occurs<br />

after the reexposure to a specific antigen. Allergens include insect stings, medications,<br />

antibiotics, anesthetics, contrast materials, certain foods, latex exposure or even exercise. A<br />

serious anaphylactic reaction is associated with severe hypotension <strong>and</strong> requires immediate<br />

parenteral treatment with epinephrine <strong>and</strong> aggressive volume resuscitation. However,<br />

anaphylactic cardiovascular collapse can be resistant to epinephrine <strong>and</strong> intravenous fluids.<br />

Platelet activating factor (PAF) is implicated in the cardiovascular dysfunctions occurring in<br />

various shock syndromes, including anaphylaxis. It is generally accepted that the excessive<br />

production of the vasodilator nitric oxide (NO) causes inflammatory hypotension <strong>and</strong> shock.<br />

NO may be produced by the constitutive NO synthases eNOS (NOS3) or nNOS (NOS1), or<br />

by the transcriptionally regulated inducible iNOS (NOS2). At this time, iNOS is regarded to<br />

be the major producer of shock-inducing NO. Nevertheless, the contribution of NO to PAFinduced<br />

or anaphylactic shock is still ambiguous. We studied PAF <strong>and</strong> anaphylactic shock in<br />

conscious mice. Surprisingly, hyperacute PAF or anaphylactic shock <strong>and</strong> mortality entirely<br />

depended on NO, produced not by the inducible iNOS but by the constitutive eNOS, rapidly<br />

activated via the (alternative) phosphatidylinositol-3-kinase (PI3K) pathway. In contrast to the<br />

unsubstantiated axioma that only excessive iNOS-derived NO underlies cardiovascular<br />

collapse in shock, <strong>our</strong> data support the surprising concept that eNOS-derived NO is the<br />

principal vasodilator in PAF-induced or anaphylactic shock <strong>and</strong> define eNOS <strong>and</strong>/or PI3K as<br />

new possible targets for treating anaphylaxis.<br />

- 194 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 12<br />

Small GTPases Ras modulate transforming growth factor-beta 1-induced extracellular<br />

matrix síntesis.<br />

Begoña Cenador, Isabel Fuentes-Calvo, Eugenio Santos1, José M. López-Novoa <strong>and</strong><br />

Carlos Martínez-Salgado2.<br />

Departamento de Fisiología y Farmacología, Universidad de Salamanca, Avda. Campo<br />

Charro s/n, 37007 Salamanca, 1Centro de Investigación del Cáncer, Campus Miguel de<br />

Unamuno s/n, 37007 Salamanca, <strong>and</strong> 2Unidad de Investigación, Hospital Universitario<br />

de Salamanca, Paseo San Vicente 58-182, 37007 Salamanca.<br />

Ras proteins are small GTPases with prooncogenic effect that act as transducers of<br />

extracellular signals that regulate cell survival, growth <strong>and</strong> differentiation. Transforming<br />

growth factor beta 1 (TGF-beta 1) has a relevant role in the origin <strong>and</strong> maintenance of<br />

glomerulosclerosis (GSC) <strong>and</strong> tubule-interstitial fibrosis (TIF). TGF-beta <strong>and</strong> Ras signalling<br />

pathways are close related: TGF-beta 1 overcomes Ras mitogenic effects <strong>and</strong> Ras counteracts<br />

TGF-beta signalling. TIF is associated to increases in Ras, <strong>and</strong> with the activation of its<br />

signalling pathways Erk/MAPK (mitogen activated protein kinases) <strong>and</strong> phosphatidylinositol-<br />

3-kinase (PI3K)/Akt in a renal fibrosis model. We have studied the role of Ras, Erk/MAPK<br />

<strong>and</strong> Akt in TGF-beta 1 mediated extracellular matrix (ECM) synthesis, using embrionary<br />

fibroblasts from knockout (KO) mice for Sos 1 (sos 1-/-), a guanidine nucleotide exchange<br />

factor that is the main Ras activating protein. Fibronectin <strong>and</strong> collagen type I expression, as<br />

well as total collagen synthesis, is much bigger in sos 1-/- fibroblasts than in control sos 1+/+<br />

fibroblasts, both in basal conditions <strong>and</strong> after TGF-beta 1 treatment. Ras activation is higher<br />

in sos 1-/- fibroblasts than in controls. Phosphorylated Akt expression is also bigger in sos 1-/-<br />

fibroblasts than in controls, but there are no differences in phosphorylated Erk expression.<br />

These studies show that Ras may down-regulate TGF-beta 1-induced ECM synthesis, <strong>and</strong> the<br />

Akt signalling pathway participates in the increase in ECM synthesis in sos 1-/- fibroblasts.<br />

- 195 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 13<br />

Yeast mitochondrial biogenesis is regulated via the cAMP protein kinase tpk3p<br />

Cyrille Chevztoff, Michel Rigoulet <strong>and</strong> Anne Devin<br />

IBGC du CNRS, 1 rue Camille Saint Saëns, 33077 BORDEAUX cedex, France. e-mail :<br />

anne.devin@ibgc.u-bordeaux2.fr<br />

In most organisms, the cellular mitochondrial content is modulated by environmental stimuli<br />

<strong>and</strong>/or in response to energy dem<strong>and</strong> changes. Several cAMP targets <strong>and</strong> transcription factors<br />

have been shown to be involved in the up-modulation of mitochondrial biogenesis when<br />

energy dem<strong>and</strong> increases. In the yeast Saccharomyces cerevisiae, the Ras/cAMP/protein<br />

kinase pathway is involved in many physiological adaptations of cells upon environmental<br />

changes. Among the adaptation processes occurring during the transition between exponential<br />

growth to stationary phase, the down-modulation of the cytochrome content <strong>and</strong> of the<br />

respiratory activity of yeast cells plays a role in the maintenance of a high growth yield. The<br />

question is therefore raised as to the role of the Ras/cAMP-protein kinase A pathway in this<br />

adaptative process. We have previously shown that mutant strains exhibiting an increased or<br />

decreased activity of this pathway have a coordinated change in mitochondrial enzyme<br />

content. The question thus arose as to whether a specific kinase might be involved in this<br />

process. We show that out of the three cAMP protein kinases in yeast, tpk3p is the one<br />

involved in mitochondrial biogenesis. Indeed, when grown on non-fermentescible carbon<br />

s<strong>our</strong>ce TPK3- cells have a significantly decreased amount of mitochondria. Respiratory rates,<br />

enzymatic equipment as well as mitochondrial DNA amount are decreased in this strain.<br />

Mitochondria isolated from the TPK3- strain display distinct energetic features when<br />

compared to the wild type, pointing out a role of Tpk3p in the biogenesis of the mitochondrial<br />

respiratory chain. Moreover, this <strong>signaling</strong> pathway clearly involves reactive oxygen species,<br />

as shown by the full reversal of the decreased amount of mitochondria in the TPK3- strain by<br />

anti-oxidant. The activity of transcription factors involved in mitochondrial biogenesis is<br />

decreased in the TPK3- strain <strong>and</strong> restored by anti-oxidant addition, thus indicating that<br />

mitochondria to nucleus <strong>signaling</strong> might go through reactive oxygen species. This study<br />

points out a crucial role of reactive oxygen species generated in a tpk3p deficient strain for the<br />

adjustment of mitochondrial biogenesis to cellular energy dem<strong>and</strong>.<br />

- 196 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 14<br />

MAPK pathways influence the release of .NO <strong>and</strong> PGE2 in IL-1# stimulated<br />

chondrocyte/agarose constructs subjected to dynamic compression<br />

T T Chowdhury, K Elliot*, D M Salter*, D L Bader <strong>and</strong> D A Lee<br />

Queen Mary, University of London, Mile End Road, London. E1 4NS. UK<br />

*Edinburgh University Medical School, Edinburgh. EH16 4TJ<br />

E-mail : t.t.chowdhury@qmul.ac.uk<br />

Introduction: Nitric oxide (.NO) <strong>and</strong> prostagl<strong>and</strong>in E2 (PGE2) are catabolic mediators<br />

induced by IL-1# <strong>and</strong> are therefore potentially important pharmacological targets in<br />

osteoarthritis (OA). In chondrocytes, the application of mechanical load has been shown to<br />

counteract the IL-1# induced .NO <strong>and</strong> PGE2 release. Furthermore, the cytokine can activate<br />

all three members of the MAPK family. However, the role of the MAPK subtypes in the<br />

release of .NO <strong>and</strong> PGE2 in chondrocytes is complex <strong>and</strong> may primarily involve<br />

phosphorylation of p38 or JNK <strong>and</strong> activation of NF!B <strong>and</strong> AP-1. The current study<br />

examines whether inhibition of these pathways will abolish the IL-1# induced .NO <strong>and</strong> PGE2<br />

release in mechanically stimulated chondrocyte/agarose constructs.<br />

Methods: Bovine chondrocytes seeded in agarose gel were cultured under free-swelling<br />

conditions, for 48 hrs in medium containing 0 or 10 ng.ml-1 IL-1#, supplemented with 0 to<br />

1000 ng.ml-1 SB203580 (inhibitor of p38 MAPK) or 0 to 50 nM JNK II (inhibitor of JNK-1,<br />

2, 3), or 0 to 1000 ng.ml-1 PDTC (inhibits NFkB activation) or 0 to 1000 ng.ml-1 curcumin<br />

(inhibitor of AP-1). For the mechanical loading experiments, all constructs were subjected to<br />

15 % dynamic compression (1 Hz) for 48 hrs under the following conditions: 0 or 10 ng.ml-1<br />

IL-1#, +/- 10 ng.ml-1 SB203580 or 5 nM JNK II or 10 ng.ml-1 PDTC or 10 ng.ml-1<br />

curcumin.<br />

Results: For constructs cultured under free-swelling conditions, SB203580 <strong>and</strong> JNK II<br />

resulted in a dose-dependent inhibition of nitrite <strong>and</strong> PGE2 release in IL-1# stimulated<br />

constructs. PDTC <strong>and</strong> curcumin reversed the IL-1# induced nitrite release whereas PGE2<br />

levels were only partially inhibited. For the mechanical loading experiments, dynamic<br />

compression could counteract the IL-1# induced nitrite release. Compression-induced<br />

inhibition of nitrite release was abolished by SB203580, PDTC <strong>and</strong> curcumin <strong>and</strong> partially<br />

reduced by JNK II. The application of dynamic compression resulted in the strain-induced<br />

inhibition of PGE2 release in IL-1# stimulated constructs. This effect was abolished by<br />

SB203580 <strong>and</strong> JNK II <strong>and</strong> partially reduced by PDTC <strong>and</strong> curcumin.<br />

Discussion: These results suggest the involvement of MAPKs <strong>and</strong> the transcription factors,<br />

AP-1 <strong>and</strong> NFkB in the IL-1# induced release of .NO <strong>and</strong> PGE2. Further work will determine<br />

the sequential activation of the signalling cascades associated with mechanical load <strong>and</strong> IL-<br />

1#. Moreover, the temporal organization of the multiple pathways will provide important<br />

information for the pharmacological <strong>and</strong> biophysical treatment of degenerative joint disease<br />

as observed in OA.<br />

Acknowledgements:<br />

This work was supported by a project grant from the Wellcome Trust, project number,<br />

073972<br />

- 197 -


Session III : Protein kinase cascades as therapeutic targets Poster III,<br />

The carcinogen Lindane disrupts autophagy at the maturation step by triggering the<br />

MAPK ERK pathway<br />

Elisabeth A. Corcelle1, Marielle Nebout1, Soumeya Bekri2, Nils Gauthier3, Paul<br />

Hofman4, Philippe Poujeol5, Patrick Fénichel1 <strong>and</strong> Baharia Mograbi1<br />

1INSERM, U670, IFR 50, Faculté de Médecine, Avenue de Valombrose, F-06107 Nice<br />

Cedex 02, France, 2Groupe Appareil Digestif et Environnement (EA3234), Faculté de<br />

Médecine, Bd Gambetta F-76183 Rouen cedex, 3INSERM, U627, IFR50, F-06107 Nice,<br />

4INSERM, E0215/Laboratoire de Pathologie Clinique et Expérimentale, Hôpital<br />

Pasteur, Nice <strong>and</strong> 5CNRS UMR 6548, Faculté des Sciences, Nice. E-mail :<br />

mograbi@unice.fr<br />

Macroautophagy (hereafter referred to as autophagy) has emerged as a key tumor suppressor<br />

pathway. During this process, the cytosolic constituents are sequestered into autophagosomes,<br />

which subsequently fuse with lysosomes to become autolysosomes where their contents are<br />

finally degraded. Although a reduced autophagy has been demonstrated in human tumors or<br />

in response to oncogenes <strong>and</strong> carcinogens, the underlying mechanism(s) remain(s) unknown.<br />

Here, we show that widely used carcinogen Lindane promotes vacuolation of Sertoli cells. By<br />

electron <strong>and</strong> immunofluorescent microscopy analyses, we demonstrated that these structures<br />

are acid autolysosomes, containing cellular debris, <strong>and</strong> labeled by LC3, Rab7 <strong>and</strong> LAMP1,<br />

markers of autophagosomes, late endosomes <strong>and</strong> lysosomes respectively. Such Lindaneinduced<br />

vacuolation results from significant delay in autophagy degradation, in relation with a<br />

decline of the lysosomal activity of Aryl Sulfatase A. At molecular level, we show that this<br />

defect in autolysosomal maturation is independent of mTOR <strong>and</strong> p38 inhibitions. Rather, the<br />

activation of the mitogen-activated protein kinase (MAPK)/ERK pathway is required for<br />

Lindane to disrupt the autophagic pathway. Most importantly, we provide the first evidence<br />

that sustained activation of ERK pathway is sufficient to commit cell to autophagic<br />

vacuolation. Taken together, these findings strongly support that the aberrant sustained<br />

activation of ERK by the carcinogen Lindane disrupts the maturation of autophagosomes into<br />

functional autolysosomes. Our findings therefore suggest the possibility that high constitutive<br />

ERK activity found in all cancers may provide a malignant advantage by impeding the tumor<br />

suppressive function of autophagy.<br />

- 198 -


Session III : Protein kinase cascades as therapeutic targets Poster III,<br />

DJ-1, a negative regulator of PTEN, is widely expressed in ovarian carcinoma<br />

Ben Davidson1, Vivi Ann Flørenes1, Martina Skrede1, Reuven Reich2<br />

Department of Pathology1, the National Hospital-Norwegian Radium Hospital,<br />

University of Oslo, Montebello N-0310 Oslo, Norway<br />

2 Department of Pharmacology <strong>and</strong> Experimental Therapeutics, School of Pharmacy,<br />

Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel<br />

E-mail: ben.davidson@radiumhospitalet.no; vivi.ann.florenes@radiumhospitalet.no;<br />

martina@skrede.name; reich@cc.huji.ac.il<br />

DJ-1 has recently been shown to be a negative regulator of PTEN in Drosophila <strong>and</strong> in human<br />

lung <strong>and</strong> breast carcinoma. The objective of this study was to analyze DJ-1 mRNA expression<br />

in primary <strong>and</strong> metastatic ovarian carcinoma. Seventy-two peritoneal <strong>and</strong> pleural effusions,<br />

43 primary tumors <strong>and</strong> 14 solid metastases were analyzed for DJ-1 mRNA expression using<br />

RT-PCR. p-AKT <strong>and</strong> PTEN protein expression was analyzed in 70 effusions using Western<br />

blotting. DJ-1 mRNA was frequently expressed at all anatomic sites (65/72 effusions, 35/43<br />

primary carcinomas <strong>and</strong> 11/14 metastases), with similar expression at these sites (p>0.05,<br />

Kruskal-Wallis test). In effusions, expression was higher in peritoneal compared to pleural<br />

specimens (p=0.047, Mann-Whitney test) <strong>and</strong> in post-chemotherapy compared to primary<br />

diagnosis specimens (p=0.012, Mann-Whitney test). Western blotting showed p-AKT<br />

expression in 80% of effusions, with loss of PTEN in 90% of specimens. Patients with postchemotherapy<br />

effusions expressing higher DJ-1 levels had shorter progression-free survival in<br />

univariate analysis (p=0.027). Our data show that DJ-1 expression is frequent in ovarian<br />

carcinoma, that its expression is associated with aggressive clinical c<strong>our</strong>se, <strong>and</strong> that it is<br />

associated with AKT phosphorylation <strong>and</strong> silencing of PTEN in metastatic cells in effusions.<br />

DJ-1 expression may therefore mediate enhanced AKT <strong>signaling</strong> in this cancer type, in<br />

addition to AKT2 amplification.<br />

- 199 -


Session III : Protein kinase cascades as therapeutic targets Poster III,<br />

The mitogen-activated protein kinases (MAPK) p38 <strong>and</strong> JNK are markers of tumor<br />

progression in breast carcinoma<br />

Ben Davidson1, Sophya Konstantinovsky2, Lilach Kleinberg1, Mai TP. Nguyen1, Assia<br />

Bassarova1, Gunnar Kvalheim3, Jahn M. Nesl<strong>and</strong>1, Reuven Reich2<br />

Departments of Pathology1 <strong>and</strong> Oncology3, the National Hospital-Norwegian Radium<br />

Hospital, University of Oslo, Montebello N-0310 Oslo, Norway<br />

2 Department of Pharmacology <strong>and</strong> Experimental Therapeutics, School of Pharmacy,<br />

Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel<br />

E-mail: ben.davidson@radiumhospitalet.no; sophya@md.huji.ac.il;<br />

lilachkl@odont.uio.no; assia.bassarova@radiumhospitalet.no;<br />

gunnar.kvalheim@klinmed.uio.no; j.m.nesl<strong>and</strong>@medisin.uio.no; reich@cc.huji.ac.il<br />

The objective of this study was to investigate the activation of mitogen-activated protein<br />

kinases (MAPK) in breast carcinoma effusions, <strong>and</strong> to analyze its relationship to anatomic site<br />

<strong>and</strong> clinical parameters. Activated MAPK (p-ERK, p-JNK <strong>and</strong> p-p38) expression was studied<br />

in 42 effusions <strong>and</strong> 51 corresponding solid tumors (23 primary, 28 metastases) using<br />

immunohistochemistry. Hormone receptor <strong>and</strong> HER2 status, proliferation (Ki-67) <strong>and</strong><br />

apoptosis (p85-PARP fragment) were assessed. MAPK levels, activity <strong>and</strong> activation ratio<br />

(phospho/pan-MAPK ratio) were analyzed in 19 effusions using Western blotting (WB).<br />

Nuclear expression of p-p38 <strong>and</strong> p-JNK was significantly higher in effusions compared with<br />

both primary tumors (p


Session III : Protein kinase cascades as therapeutic targets Poster III, 15<br />

A cross-talk between the Wnt <strong>and</strong> the adhesion-dependent <strong>signaling</strong> pathways governs<br />

the chemosensitivity of acute myeloid leukemia<br />

Fabienne De Toni , Loïc Ysebaert, Stéphane Manenti <strong>and</strong> Claire Racaud Sultan.<br />

Centre de Physiopathologie de Toulouse Purpan, IFR 30, INSERM U563, Département<br />

“Oncogenèse et signalisation dans les cellules hématopoïétiques”, CHU PURPAN, 3<br />

place du Dr Baylac 31059 TOULOUSE Cedex 03, FRANCE. E-mail :<br />

fadetoni@toulouse.inserm.fr<br />

Relapses following chemotherapy are a major hindrance to patients' survival in acute myeloid<br />

leukemia (AML). To investigate the role of the hematopoietic niche in the chemoresistance of<br />

leukemic cells, we examined two pathways: one mediated by adhesion molecules/integrins,<br />

<strong>and</strong> the other by soluble factors of the morphogen Wnt pathway. In <strong>our</strong> study, both the<br />

adhesion of leukemic blasts to fibronectin <strong>and</strong> the addition of Wnt antagonists induced,<br />

independently, resistance of AML cells to daunorubicin in a cell survival assay. Using<br />

pharmacological inhibitors <strong>and</strong> siRNA, we showed that both resistance pathways required the<br />

activity of the Glycogene Synthase Kinase 3b (GSK3b). Moreover, the AML cell protection<br />

downstream of GSK3b was mediated by NF-kB. A link between the adhesion <strong>and</strong> the Wnt<br />

pathway was found, as adhesion of U937 on human osteoblasts, a component of the<br />

hematopoietic niche, triggered the secretion of the Wnt antagonist sFRP-1 <strong>and</strong> supported<br />

resistance to daunorubicin. The osteoblast-conditioned medium could also confer<br />

chemoresistance to U937 cells cultured in suspension, <strong>and</strong> this cell protective effect was<br />

abrogated after depletion of sFRP-1. In the context of this potential double in vivo resistance,<br />

modulators of the common signal GSK3b <strong>and</strong> of its target NF-kB could represent important<br />

novel therapeutic tools.<br />

F. De Toni et al, Oncogene 2006 in press<br />

- 201 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 16<br />

Repeated exposures of human skin fibroblasts to UVB at subcytotoxic level trigger<br />

premature senescence through the TGF-ß1 <strong>signaling</strong> pathway.<br />

Florence Debacq-Chainiauxa, Céline Borlona, Thierry Pascala, François Eliaersa,<br />

Noëlle Ninannea, Françoise de Longuevilleb, José Remaclea <strong>and</strong> Olivier Toussainta<br />

a Laboratory of Biochemistry <strong>and</strong> Cellular Biology, Department of Biology, University<br />

of Namur (FUNDP), Rue de Bruxelles 61, 5000 Namur, Belgium<br />

b Eppendorf Array Technologies, Rue du Séminaire 12, 5000 Namur, Belgium<br />

e-mail: florence.chainiaux@fundp.ac.be<br />

Premature senescence of human diploid fibroblasts (HDFs) can be induced by exposures to a<br />

variety of oxidative stress <strong>and</strong> DNA damaging agents. In this study we developed a model of<br />

UVB-induced premature senescence of skin HDFs. After a series of 10 subcytotoxic (nonproapoptotic)<br />

exposures to UVB at 250 mJ/cm2, the so-called biomarkers of senescence were<br />

markedly expressed : growth arrest, senescence associated ß-galactosidase activity,<br />

senescence-associated gene overexpression, deletion in mitochondrial DNA. Using a low<br />

density DNA microarray to study gene expression profiles of 240 senescence- <strong>and</strong> stressrelated<br />

genes, a set of 44 genes were found as differentially expressed in this model among<br />

which Transforming Growth Factor-ß1 (TGF-ß1). Neutralizing antibodies against TGF-ß1 or<br />

its receptor II (TßRII) sharply attenuated the senescence-associated features, suggesting a role<br />

for TGF-ß1 in UVB-induced premature senescence. Both the latent <strong>and</strong> active forms of TGFß1<br />

were increased with time after the last UVB-stress. This model represents a proof-ofconcept<br />

as alternative in vitro model in photoaging research for screening potential antiphotoaging<br />

compounds.<br />

- 202 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 17<br />

Deguelin controls angiogenic <strong>and</strong> invasive properties of endothelial cells by modulating<br />

the Akt/IkappaB kinase pathway cells.<br />

Raffaella Dell’Eva1, Claudia Ambrosini 1, Douglas M. Noonan2, Adriana Albini,1 <strong>and</strong><br />

Nicoletta Ferrari1<br />

1Experimental Oncology A Laboratory, National Cancer Research Institute (IST), L.go<br />

R. Benzi 10, 16132 Genova, Italy. 2Università degli Studi dell'Insubria, Varese, Italy. Email:<br />

nicoletta.ferrari@istge.it, raffaella.delleva@istge.it<br />

Tumor growth relies on angiogenesis a process controlled by multiple growth factors<br />

generating a multitude of signals that require careful coordination to guarantee a proper<br />

vascular network. Here we evaluate the potential of deguelin, a natural plant productisolated<br />

from Mondulea seriacea (Leguminosae) effective on both premalignant <strong>and</strong> malignant HBE<br />

cells, <strong>and</strong> investigate its mechanisms of action. Deguelin causes extensive biological<br />

modifications of endothelial cells by decreasing their growth rate while protecting them from<br />

apoptosis, repressing their ability to migrate <strong>and</strong> invade but not affecting their capacity to<br />

organize into capillary like structures. Inhibition of invasion by deguelin was associated with<br />

decreased metalloprotease MMP- 2 activity. In vivo, deguelin blocks angiogenesis in the<br />

Matrigel plug assay. Basal levels of phosphorylated Akt (pAkt) were reduced after brief<br />

exposure to deguelin (1h) <strong>and</strong> inhibition of phosphorylation of IkB by TNFalpha in the<br />

presence of deguelin correlates well with the ability of deguelin to decrease activation of NFkB<br />

in the nucleus. Our data suggest that inhibition of NF-kB by deguelin may be an effective<br />

strategy to control angiogenesis. Our results justify further studies to evaluate the utility of<br />

deguelin as a chemopreventive agent.<br />

- 203 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 18<br />

Different ability to up-regulate FasL expression may confer differential response of<br />

glioma cells to CB2-selective <strong>and</strong> non-selective cannabinoids<br />

Aleks<strong>and</strong>ra Ellert-Miklaszewska 1, Bozena Kaminska 2, Liliana Konarska 1:<br />

1Dept. of Biochemistry <strong>and</strong> Clinical Chemistry, Medical University, Banacha 1 str., 02-<br />

097 Warsaw, Pol<strong>and</strong>, Email: aellert@nencki.gov.pl, 2Lab. of Transcription Regulation,<br />

Nencki Institute, Pasteura 3 str., 02-093 Warsaw, Pol<strong>and</strong><br />

Cannabinoids induce apoptosis of glioma cells in vitro <strong>and</strong> tumor regression in vivo. Despite<br />

a growing interest, the molecular mechanisms of their pro-apoptotic action, induced by<br />

stimulation of either of receptor type, CB1 <strong>and</strong>/or CB2, still remain elusive. We have<br />

observed that synthetic cannabinoids: WIN-55,212-2 (non-selective CB1/CB2 agonist) <strong>and</strong><br />

JWH133 (CB2-selective), affect growth <strong>and</strong> viability of glioma cells. However, the<br />

morphological changes <strong>and</strong> the time c<strong>our</strong>se of apoptotic cell death were different, depending<br />

on the drug added. Thus, the aim of the present study was to identify pathways that<br />

distinguish cannabinoid actions triggered by CB1/CB2 <strong>and</strong> selective CB2 stimulation in C6<br />

glioma cells. We found that both compounds induce similar pattern of changes in MAPK<br />

activation (p42/44 Erk, p38, JNK). However, the levels of phosphorylated (active) protein<br />

kinase B (PKB/Akt) decreased significantly during incubation with WIN55,212-2, while it<br />

was less pronounced after exposure to JWH133. The increase of the active form of Forkhead<br />

transcription factor (FKHR), one of Akt downstream targets, <strong>and</strong> its translocation to the<br />

nucleus occur after WIN55,212-2 but not JWH133 addition. Since FKHR has been implicated<br />

in inducing the expression of genes critical for cell death, such as FasL gene, we studied the<br />

role of Akt/FKHR pathway in regulation of FasL expression during WIN55,212-2- <strong>and</strong><br />

JWH133-induced apoptosis. RT-PCR analysis revealed the up-regulation of fasL mRNA level<br />

only after WIN55,212-2 treatment, whereas it did not occur after JWH133, unless the cells<br />

were pre-treated with LY294002, phosphatidylinositol-3 kinase (PI3K) inhibitor. The extent<br />

of FasL expression correlated with the degree of PI3K/Akt inhibition, Forkhead activation<br />

<strong>and</strong> apoptosis induction, suggesting that the observed difference in Akt inhibition might be<br />

responsible for the diversified effects of the two cannabinoids. Our findings help to<br />

underst<strong>and</strong> differences in anti-tumor efficiency of cannabinoid receptor agonists, which in<br />

turn might be of clinical relevance in glioma treatment. Supported by the grant No. 06/2002<br />

from the Polish Pharmacy <strong>and</strong> Medicine Development Foundation by Polpharma S.A.<br />

- 204 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 19<br />

The ERK5 pathway promotes cell survival by down-regulating FasL expression<br />

following osmotic stress stimulation<br />

Katherine G Finegan1, Xin Wang1, Andrew C. Robinson1, Lynette Knowles1, Roya<br />

Khosravi-Far2, Katherine A. Hinchliffe1, Raymond P. Boot-H<strong>and</strong>ford3 <strong>and</strong> Cathy<br />

T<strong>our</strong>nier1<br />

Faculty of Life Sciences1 <strong>and</strong> Wellcome Trust Center for Cell Matrix3, University of<br />

Manchester, Michael Smith Building, Oxford Road Manchester M130PT, UK;<br />

Deparment of Pathology2, Harvard Medical School, Boston MA 02215, USA<br />

Extracellular signal-regulated kinase 5 (ERK5) is a mitogen-activated protein kinase (MAPK)<br />

that is activated by dual phosphorylation via a unique MAPK/ERK kinase, MEK5. The<br />

analysis of the targeted deletion of the erk5 <strong>and</strong> mek5 genes has demonstrated a physiological<br />

significance in their role in promoting cell survival. We have identified how the ERK5<br />

signalling pathway regulates this process using as <strong>our</strong> models fibroblasts deficient in either<br />

ERK5 or MEK5 expression. We found that ERK5 was required for mediating the survival of<br />

fibroblasts under basal conditions <strong>and</strong> in response to sorbitol treatment. Increased FasL<br />

expression acted in a positive feedback loop to enhance apoptosis of ERK5- or MEK5deficient<br />

cells under conditions of osmotic stress. Compared to wild type cells, erk5-/- <strong>and</strong><br />

mek5-/- fibroblasts treated with sorbitol displayed a reduced Protein Kinase B (PKB) activity<br />

associated with increased Foxo3a activity. Based on these results we conclude that the ERK5<br />

signalling pathway promotes cell survival by down-regulating FasL expression via a<br />

mechanism that implicates PKB-dependent inhibition of Foxo3a downstream of<br />

Phosphatidylinositol 3-Kinase.<br />

- 205 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 20<br />

Constitutive active EGFR <strong>and</strong> Erk1/2 is promising targets for treatment of antiestrogen<br />

resistant breast cancer cell lines<br />

Thomas Frogne, Jan S. Jepsen <strong>and</strong> Anne E. Lykkesfeldt<br />

Department of Tumor Endocrinology, Institute of Cancer Biology, Danish Cancer<br />

Society Str<strong>and</strong>boulevarden 49, DK-2100 Copenhagen, Denmark E-mail: tfr@cancer.dk<br />

Breast cancer patients with advanced disease often benefit from endocrine therapy. However,<br />

after an initial response, most patients will develop resistance towards treatment. In <strong>our</strong><br />

department, we have established a large panel of antiestrogen resistant cell lines, hereby<br />

mimicking antiestrogen resistant tumor growth. We have previously shown that these cell<br />

lines are dependent on activated Protein Kinase B (PKB/Akt) to maintain growth in the<br />

presence of antiestrogen <strong>and</strong> we are currently investigating the importance of the ErbB<br />

receptor family. When we compare the parental <strong>and</strong> resistant cell lines under basal growth<br />

conditions we see an upregulation of the Epidermal Growth Factor Receptor (EGFR) protein<br />

<strong>and</strong> an increase in activity of the prominent downstream targets Extracellular Regulated<br />

Kinase 1 & 2 (Erk1/2), in <strong>our</strong> antiestrogen resistant cells. Furthermore, the resistant cell lines<br />

possessed increased activation of ErbB2 <strong>and</strong> had lost protein expression of ErbB4. To asses<br />

the importance of EGFR <strong>and</strong> Erk1/2 in growth of the resistant cell lines we used the small<br />

molecule inhibitors ZD1839 <strong>and</strong> U0126, which inhibits activation of EGFR <strong>and</strong> Erk1/2,<br />

respectively. These experiments showed a significantly stronger growth inhibition of the<br />

antiestrogen resistant cell lines when compared to parental cells. Further studies with these<br />

inhibitors showed that activation of EGFR <strong>and</strong> Erk1/2 is not only required for growth of the<br />

antiestogen resistant cell lines but also for the development of antiestrogen resistance.<br />

Based on these observations, we conclude that constitutive activation of EGFR <strong>and</strong> Erk1/2 is<br />

required for both development <strong>and</strong> growth of antiestrogen resistant cell lines.<br />

We are currently evaluating expression of activated Akt <strong>and</strong> Erk1/2 in clinical material in<br />

order to evaluate whether antiestrogen resistant tumors overexpress active Akt <strong>and</strong>/or Erk1/2<br />

<strong>and</strong> whether these two kinases may be a target for treatment of antiestrogen resistant breast<br />

cancer.<br />

- 206 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 21<br />

H- <strong>and</strong> N-Ras isoforms modulate transforming growth factor-beta 1-induced<br />

proliferation <strong>and</strong> extracellular matrix síntesis.<br />

Isabel Fuentes-Calvo, Begoña Cenador, Alej<strong>and</strong>ro Esteller, Eugenio Santos1, José M.<br />

López-Novoa <strong>and</strong> Carlos Martínez-Salgado2.<br />

Departamento de Fisiología y Farmacología, Universidad de Salamanca, Avda. Campo<br />

Charro s/n, 37007 Salamanca, 1Centro de Investigación del Cáncer, Campus Miguel de<br />

Unamuno s/n, 37007 Salamanca, <strong>and</strong> 2Unidad de Investigación, Hospital Universitario<br />

de Salamanca, Paseo San Vicente 58-182, 37007 Salamanca.<br />

Transforming growth factor beta 1 (TGF-beta 1) has a relevant role in the origin <strong>and</strong><br />

maintenance of glomerulosclerosis (GSC) <strong>and</strong> tubule-interstitial fibrosis (TIF). Ras proteins<br />

are small GTPases with prooncogenic effect that act as transducers of extracellular signals<br />

that regulate cell survival, growth <strong>and</strong> differentiation. TGF-beta <strong>and</strong> Ras signalling pathways<br />

are close related: TGF-ß1 overcomes Ras mitogenic effects <strong>and</strong> Ras counteracts TGF-beta<br />

signalling. TIF is associated to increases in Ras, Erk <strong>and</strong> Akt activation in a renal fibrosis<br />

model. We study the role of N- <strong>and</strong> H-Ras isoforms, <strong>and</strong> the involvement of the Ras effectors<br />

Erk <strong>and</strong> Akt, in TGF-ß1-mediated ECM synthesis <strong>and</strong> proliferation, using embrionary<br />

fibroblasts from double knockout (KO) mice for H- <strong>and</strong> N-Ras (H-ras-/-/N-ras-/-) isoforms<br />

<strong>and</strong> from heterozygote mice (H-ras+/-/N-ras+/-). ECM synthesis is increased in basal<br />

conditions in H-ras-/-/N-ras-/- fibroblasts, this increase being bigger after stimulation with<br />

TGF-beta 1. TGF-beta 1-induced fibroblast proliferation is smaller in H-ras-/-/N-ras-/- than in<br />

H-ras+/-/N-ras+/- fibroblasts. Erk activation is decreased in H-ras-/-/N-ras-/- fibroblasts;<br />

inhibition of Erk activation reduces fibroblast proliferation. Akt activation is higher in double<br />

KO fibroblasts than in heterozygotes; inhibition of Akt activation also inhibits ECM<br />

synthesis. We can suggest that H- <strong>and</strong> N-ras isoforms down-regulate ECM synthesis, <strong>and</strong><br />

mediate proliferation, in part through MEK/Erk activation. PI3K-Akt pathway activation may<br />

be involved in the increase in ECM synthesis observed in the absence of H- <strong>and</strong> N-Ras.<br />

- 207 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 22<br />

The possible role of PI3-kinase in melanoma cell migration.<br />

Dorota Gil1, Joanna Duli"ska-Litewka1, Anna Pituch Noworolska2 <strong>and</strong> Piotr Laidler1<br />

1Departament of Medical Biochemistry, Kopernika 7, 31-034 Kraków 2Department of<br />

Clinical Immunology, Polish-American Children's Hospital, Jagielonian University<br />

Medical College, e-mail: dgil@poczta.fm<br />

Integrins are heterodimeric ("/# subunits) cell surface glycoproteins that bind to ECM<br />

proteins <strong>and</strong> activate members of intracellular signalizing pathways cascades that can<br />

stimulate or regulate motility <strong>and</strong> invasivnes, cell growth, <strong>and</strong> survival. Cancer cells very<br />

often switch the types of extracellular matrix receptors (integrins) they express, fav<strong>our</strong>ing<br />

ones that transmit progrowth signals. The expression of "3#1 integrin has been indicated as<br />

possibly associated with invasive phenotype not only trough formation of a new cell-matrix<br />

contact sites but also by regulating the expression of matrix-degrading enzymes, MMP-2 or<br />

MMP-9 metalloproteinases. "3#1 integrin is a laminin (LN) receptor <strong>and</strong> recent studies<br />

showed that laminin–5 <strong>and</strong> "5–containing laminins, such as laminin–10, <strong>and</strong> –11, are<br />

preferred lig<strong>and</strong>s for "3#1 integrin. Interaction of "3#1 integrin with LN–5 may provide<br />

evidences to how it transduces signals that affect cell behavior.<br />

METHODS: Studies were carried out on human melanoma metastatic cell lines: A 375 (from<br />

solid tumor) <strong>and</strong> 1205Lu (from mouse lung). Cells were culture on plastic dishes alone or<br />

covered with LN-5. Expression of integrins was studied by flow cytometry, secretion of<br />

MMP-2 <strong>and</strong> MMP-9 by zymography, <strong>and</strong> the invasive potential by using conventional<br />

Boyden chambers. Expression of cell <strong>signaling</strong> proteins was analyzed using Western Blot.<br />

RESULTS: LN–5 stimulated up regulation of MMPs activity in both cell lines. Signaling<br />

pathway necessary for migration of melanoma cells likely involved PI3-kinase activity as the<br />

addition of the PI3-K specific inhibitor, LY294002, decreased the level of active MMP-2 in<br />

A 375 <strong>and</strong> LU1205 cells. Western blot analyses of whole cell lysates confirmed the inhibition<br />

of PI3-kinase activity after addition of LY294002, as determined with anti-phospho–AKT<br />

(Ser 473) antibodies <strong>and</strong> observed increase in the level of phospho–AKT in the cells cultured<br />

on LN–5.<br />

CONCLUSION: Activation of PI3-kinase in response to ECM signals carried through<br />

specific integrin receptor is likely an important event involved in metastasis of melanoma.<br />

Underst<strong>and</strong>ing the intracellular <strong>signaling</strong> pathways in response to ECM proteins may be<br />

useful in the development of tools for selective blocking of some <strong>signaling</strong> pathways.<br />

This work is suported by the KBN: Jagiellonian University Medical College (W)/256/P/L)<br />

<strong>and</strong> FP5 EU project “European Searchable Tumor Cell Lines Data Bank” – ESTDAB – NAS:<br />

QLRI – CT-2001-01325.<br />

- 208 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 23<br />

CK2 modulates AKT kinase activity: implication in cell survival <strong>and</strong> death<br />

Barbara Guerra<br />

Dep. of Biochemistry & Molecular Biology, University of Southern Denmark, DK-5230<br />

Odense, Denmark. E-mail: bag@bmb.sdu.dk<br />

Protein kinase CK2 is a Ser/Thr kinase ubiquitously distributed <strong>and</strong> highly conserved in all<br />

eukaryotic organisms so far investigated. CK2 is implicated in many cellular functions e.g.<br />

cell growth, cell cycle progression, transformation <strong>and</strong> cell survival. Mounting evidence has<br />

indicated that CK2 is also implicated in cancer. For instance, a potent biological synergy in<br />

the development of primitive multiclonal lymphoid leukemias has been observed in bitransgenic<br />

mice overexpressing CK2alpha-catalytic subunit <strong>and</strong> c-Myc. Accelerated<br />

lymphomagenesis has been reported in mice overexpressing CK2 <strong>and</strong> deficient in the<br />

expression of p53. These <strong>and</strong> other data have indicated that the inappropriate expression of<br />

CK2, in cooperation with other proteins, might augment the oncogenic potential of cells. In<br />

line with these observations, the phosphatidylinositol 3-kinase (PI3K)/AKT signal<br />

transduction pathway has also been shown to be central in many intracellular processes such<br />

as proliferation, angiogenesis, motility <strong>and</strong> cell survival. The fact that a search for substrates<br />

of AKT has led to the identification of several components of the apoptotic machinery, <strong>and</strong><br />

that the PI3K pathway is negatively regulated by PTEN, a dual-specificity lipid <strong>and</strong> protein<br />

phosphatase considered to be a tum<strong>our</strong> suppressor gene product, gave, indeed, rise to the idea<br />

that AKT is a key element in the regulation of cell survival. Recently, we were able to show<br />

for the first time that the specific inhibition of the PI3K-AKT <strong>signaling</strong> pathway in<br />

combination with the depletion of either CK2 subunits by antisense oligodeoxynucleotides<br />

leads to an enhanced drug-induced apoptotic response. In vitro as well as in vivo studies have<br />

revealed that the individual CK2 subunits interact with AKT enhancing AKT kinase activity<br />

<strong>and</strong> that the complex formation is not modulated by the phosphorylation status of AKT. These<br />

data identifies a novel molecular mechanism that leads to modulation of AKT activation<br />

raising the possibility that CK2 <strong>and</strong> AKT might be implicated in common pathways that<br />

control cell proliferation <strong>and</strong> survival. Moreover, they underline the importance of developing<br />

efficient strategies to directly inhibit AKT <strong>and</strong> CK2 as valuable therapeutic approach in<br />

cancer therapy.<br />

- 209 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 24<br />

Animal models <strong>and</strong> drug design for therapy of Met-triggered cancers<br />

Hessameh Hassani1, Nicolas Pietrancosta1, Bernard Maigret2, Jean-Louis Kraus1,<br />

Flavio Maina1 & Rosanna Dono1<br />

1IBDML, Campus de Luminy-Case 907, 13288 Marseille Cedex 09 France - 2UMR<br />

CNRS/UHP 7565, Université Henri Poincare, Nancy 1, BP 239, 54506 V<strong>and</strong>oeuvre-les-<br />

Nancy Cedex, France. E-mails: dono@ibdm.univ-mrs.fr<br />

Hyperactivity of the receptor tyrosine kinase (RTK) Met is crucial for development of<br />

invasive/metastatic phenotype of neoplastic cells. Met-expressing cells acquire invasive<br />

phenotype during progression of breast cancer, <strong>and</strong> autocrine activation by its lig<strong>and</strong> HGF is<br />

responsible for spontaneous metastasis to the lung. Mutations or amplifications of Met human<br />

gene have been found in different types of cancer cells <strong>and</strong> Met is a prognostic marker for<br />

different carcinomas. To date, small-molecule drugs can selectively <strong>and</strong> efficiently inhibit<br />

RTK activity both in vitro <strong>and</strong> in vivo <strong>and</strong> act as anticancer drugs. So far, there are no<br />

pharmaceutical compounds able to interfere with Met activity in human cancers. We have<br />

undertaken a multi-disciplinary approach, which combines cell biology, mouse genetics,<br />

chemistry <strong>and</strong> non-invasive monitoring of primary tum<strong>our</strong>s <strong>and</strong> metastasis, to develop<br />

chemical compounds that specifically inhibit Met-triggered oncogenic events both in vitro<br />

<strong>and</strong> in vivo. By performing biological <strong>and</strong> biochemical studies, we identify two compounds<br />

that block Met-induced cell scattering, receptor trans-phosphorylation <strong>and</strong> intracellular<br />

signalling in culture cells. We are currently performing chemical modifications in order to<br />

optimize their efficacy <strong>and</strong> specificity. Computer-assisted molecular modelling has allowed<br />

us to underst<strong>and</strong> the mechanism of action of the novel Met inhibitors <strong>and</strong> maximize the<br />

chance of predicting the most efficient chemical changes. This approach has led to the<br />

generation of a Met inhibitor one log more active than the previous ones. The characterization<br />

of this novel Met inhibitor is ongoing. We are also generating mouse models of Met-mediated<br />

tumor <strong>and</strong> metastasis suitable to study activity of Met inhibitor in vivo. By taking advantage<br />

of Cre-mediated recombination, transgenic mice will over-express Met in a temporal <strong>and</strong><br />

spatial regulated manner together with the Luciferase reporter to allow non-invasive<br />

monitoring of tum<strong>our</strong>s <strong>and</strong> metastasis.<br />

- 210 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 25<br />

Novel Involvement of the Immunomodulator AS101 in IL-10 Signal Transduction, Via<br />

the Tyrosine Kinase Fer<br />

Rami Hayun, Sali Shpungin, Hana Malovani, Michael Albeck, Uri Nir <strong>and</strong> Benjamin<br />

Sredni<br />

Safdié Institute for AIDS <strong>and</strong> Immunology Research, Faculty of Life Sciences, Bar-Ilan<br />

University, Ramat-Gan, Israel. E-mail: srednib@mail.biu.ac.il<br />

Department of Chemistry, Bar-Ilan University, Ramat-Gan, Israel.<br />

Faculty of Life Sciences, Bar Ilan University, Ramat Gan, Israel. E-mail:<br />

nir@mail.biu.ac.il<br />

Interleukin-10 (IL-10) plays a major role in proliferation of many tumor cells <strong>and</strong> activates<br />

the transcription factor Stat3 by tyrosine phosphorylation. The immunomodulator ammonium<br />

trichloro(dioxoethylene-o,o') tellurate (AS101) sensitizes several tumors to chemotherapy, by<br />

inhibiting the tumor IL-10 autocrine loop. The tyrosine kinase Fer, is essential for the<br />

proliferation of some malignant cell-lines <strong>and</strong> its connection to Stat3 activation<br />

was demonstrated in a few cases. This study examined the role of AS101 in IL-10 signal<br />

transduction <strong>and</strong> the correlation between Fer <strong>and</strong> Stat3, in human peripheral blood<br />

mononuclear cells (PBMC). We showed that Fer was associated with Stat3 in PBMC <strong>and</strong> in<br />

the RAW 264.7 cell line. Recombinant IL-10 increased the tyrosine phosphorylation of Stat3,<br />

up regulated the levels of Fer, <strong>and</strong> increased the association of Fer with phosphorylated Stat3<br />

(pYStat3). The immunomodulator AS101 as well as IL-10 receptor antagonist, Inhibited IL-<br />

10 secretion, reduced the phosphorylation of Stat3, decreased the levels of Fer protein <strong>and</strong><br />

decreased the association between Fer <strong>and</strong> pYStat3. All AS101 activities were totally<br />

abrogated by exogenous addition of recombinant IL-10. These results indicate that AS101<br />

plays as a negative regulator in IL-10 signal transduction, <strong>and</strong> able to dephosphorylate the<br />

proto-oncogene Stat3, <strong>and</strong> modulates the ability of Fer to associate with pYStat3. Therefore,<br />

anti-IL-10 treatment using AS101 may be effective in certain malignancies <strong>and</strong> other<br />

pathologies where IL-10 secretion is elevated <strong>and</strong> Stat3 is continuously phosphorylated.<br />

- 211 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 26<br />

Different <strong>signaling</strong> pathways mediates the strain-induced CREB activation in cardiac<br />

fibroblasts<br />

Britta E. Husse, Gerrit Isenberg<br />

Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, D-<br />

06097 Halle/S., Germany. E-mail: britta.husse@medizin.uni-halle.de<br />

The transcription factor CREB (cAMP response element binding protein) mediates the<br />

mechanical strain-induced gene expression in the heart. We investigated which <strong>signaling</strong><br />

pathways are involved in the CREB activation by strain using cultured ventricular fibroblasts<br />

from adult rat hearts. The analysis of phospho-CREB by immunocytochemistry showed a<br />

nearly complete CREB activation (93.2±6.1%) by cyclical mechanical strain (1 Hz, 5%<br />

elongation, 15 min) which did not distinguish from the whole number of CREB-positive<br />

fibroblasts (87.8±9.6%). The strain-induced CREB activation was partially reduced by PKA<br />

inhibition (19.9±11.2%), by PKC inhibition (31.2±11.2%) but not by CaMK II inhibition. The<br />

inhibition of several members of the MAPK cascade revealed a reduction of strain-induced<br />

CREB phosphorylation by 24.4±5.8% (Src-inhibition), by 27.7±5.8% (Raf1-inhibition) <strong>and</strong><br />

by 23.8±8.6% (MEK-inhibition). The PI3-K inhibition reduced strain-caused phospho-CREB<br />

by 19.3±5.4%. The inhibition of p38, the stress-sensitive pathway, decreased the straininduced<br />

CREB phosphorylation by 21.5±12.8%. The time-dependent CREB activation by<br />

cAMP stimulation with 10µM forskolin was transient with a peak after 5 min; from<br />

18.5±6.3% to 42.9±15.9% phospho-CREB positive cells. The PKC-induced CREB activation<br />

by 500 nM PMA was sustained beginning after 10 min; from 12.1±5.0% to 54.3±10.4%<br />

phospho-CREB positive cells. Our results suggest that the complete strain-induced CREB<br />

phosphorylation includes several <strong>signaling</strong> pathways. We discuss this wide-ranging<br />

possibilities of CREB activation as safety for CREB-mediated gene expression in the heart.<br />

- 212 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 27<br />

Signal transduction pathway of CNP-dependent inhibition of upregulation of PAI-1<br />

expression by TNF-alpha in endothelial cells<br />

Hanna Jerczynska, Czeslaw Cierniewski, Zofia Pawlowska<br />

Department of Molecular & Medical Biophysics, Medical University in Lodz, Pol<strong>and</strong>. Email:<br />

pawlow@zdn.am.lodz.pl<br />

Plasminogen activator inhibitor type 1 (PAI-1) plays a fundamental role in regulation of the<br />

fibrinolytic system. PAI-1 synthesis is upregulated by multiple factors, but only few are<br />

known to be able to decrease PAI-1 expression. In <strong>our</strong> previous study we found that C-type<br />

natriuretic peptide (CNP), which modulates salt <strong>and</strong> water balance as well as vascular tone,<br />

was an effective inhibitor of PAI-1 synthesis <strong>and</strong> release from endothelial cells, it also reveals<br />

stronger inhibitory activity for TNF-alpha - stimulated cells. The <strong>signaling</strong> pathways required<br />

for this effect have not been elucidated. It was reported that NF-kB is involved in TNFregulated<br />

PAI-1 production. We have evidenced that the activation of MAPK kinase cascade<br />

is a necessary step in induction of PAI-1 expression by TNF in endothelial cells. In this study,<br />

the effects of signal transduction inhibitors on TNF-induced PAI-1 expression in the presence<br />

of CNP were examined. We used PD 98059, an ERK1/2 inhibitor, LY 294002, a PI3K/Akt<br />

inhibitor, <strong>and</strong> 8-bromo-cGMP, a soluble analog of cGMP to characterize the signal<br />

transduction pathway involved in CNP action in human endothelial cells. Involvement of NFkB<br />

activation in this process was also investigated.<br />

Our results imply that CNP inhibition of TNF-activated PAI-1 gene expression takes place via<br />

regulation of signal transduction pathways involving PI3K/Akt <strong>and</strong> cGMP-dependent protein<br />

kinase, possibly by inhibiting NF-kB-dependent pathways. The activation of ERK seems not<br />

to be implicated in this process. Thus, <strong>our</strong> results explain at least in part the mechanism<br />

involved in PAI-1 expression inhibition by CNP in TNF-stimulated endothelial cells.<br />

Supported by grants: QLK3-CT-2002-30326 (5th EU FP) <strong>and</strong> Medical University of Lodz,<br />

Pol<strong>and</strong>.<br />

- 213 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 28<br />

PI3K$ - A Novel Target for Rheumatoid Arthritis. Closing the loop - From genetic<br />

validation <strong>and</strong> rationale drug design to therapeutic proof of concept.<br />

Ji H. 1, Rückle T. 1, Camps M. 1, Rintelen F. 1, Ardissone A. 2, Gillieron C. 1, Bertschy-<br />

Meier D. 1, Cirillo R. 2, Hirsch E. 3, Wymann MP. 4, Vanhaesebroeck B. 5, Schwarz<br />

M.K. 1 <strong>and</strong> Rommel C. 1*<br />

1Serono Pharmaceutical Research Institute, Serono International S.A., 14, Chemin des<br />

Aulx, 1228 Plan-les-Ouates, Geneva, Switzerl<strong>and</strong> 2LCG-RBM, Serono<br />

International S.A., Via Ribes 1, 10010 Colleretto Giacosa, Italy. 3Department of<br />

Genetics, Biology <strong>and</strong> Biochemistry, University of Torino, Via Santena 5bis, 10126<br />

Torino, Italy. 4Department Clinical & Biological Sciences, Institute of Biochemistry &<br />

Genetics, Centre of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel,<br />

Switzerl<strong>and</strong>. 5 Ludwig Institute for Cancer Research, 91 Riding House Street, London,<br />

W1W 7BS, UK.<br />

*christian.rommel@serono.com<br />

Small molecule inhibitors targeting class I PI3K isoforms have vast potential use for the<br />

treatment of inflammatory <strong>and</strong> autoimmune disorders as well as cancer <strong>and</strong> cardiovascular<br />

diseases. However, the lack of specificity, isoform selectivity <strong>and</strong> poor biopharmaceutical<br />

profile of PI3K inhibitors has so far hampered rigorous disease-oriented <strong>and</strong><br />

pharmacologically relevant target validation. PI3K$ plays a crucial role in mediating<br />

leukocyte chemotaxis as well as mast cell degranulation, mainly in response to G-protein<br />

coupled receptor activation <strong>and</strong> hence represents a high value target for autoimmunity <strong>and</strong><br />

inflammation.<br />

In <strong>our</strong> laboratories we have studied PI3K$ deficient mice in various murine models of<br />

rheumatoid arthritis (RA). We show that these mice are largely protected in these models,<br />

which feature the effector phase of RA, resulting from impaired neutrophil migration. These<br />

findings further validate PI3K$ as a therapeutic target.<br />

Exploiting the interface of Molecular Medicine, Medicinal Chemistry <strong>and</strong> Experimental<br />

Pharmacology, we have developed potent <strong>and</strong> selective small molecule PI3K$ inhibitors, that<br />

upon oral treatment suppress the progression of joint inflammation <strong>and</strong> cartilage damage in<br />

murine models of RA, reproducing the protective effects exhibited by PI3K$ deficient mice.<br />

SAR of two generations of inhibitors, supported by X-ray crystallography <strong>and</strong> structure based<br />

design, has led to the identification of the lead compound AS-605240 (Ki = 7.8 nM), active in<br />

animal models of cell recruitment, mast cell activation <strong>and</strong> inflammation.<br />

Our results identify selective PI3K$ inhibitors as potential therapeutics for the treatment of<br />

RA.<br />

- 214 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 29<br />

Multiple roles for beta-arrestins in FSH signalling : a 5 S/T cluster in the C-terminus of<br />

the receptor is important for beta-arrestin recruitment, desensitization <strong>and</strong><br />

internalization but not for beta-arrestin-mediated Erk activation.<br />

E. Kara, P. Crepieux, C. Gauthier, N. Martinat, V. Piketty, F. Guillou, E. Reiter<br />

UMR INRA-CNRS-Univ. T<strong>our</strong>s-Haras Nationaux, PRC, Equipe " Mécanismes d’action<br />

des Gonadotropines ", 37380 Nouzilly France<br />

The follicle-stimulating hormone receptor (FSH-R) is a seven transmembrane spanning<br />

receptor that couples to G"s upon agonist stimulation. #-arrestins (#-arr) are rapidly recruited<br />

to the FSH-activated receptor <strong>and</strong> play key roles in the desensitization <strong>and</strong> internalization<br />

processes. In addition, #-arr are able to activate the ERK pathway through a G proteinindependent<br />

mechanism.<br />

We have identified a cluster of five Ser/Thr residues in the c-terminus of the FSH-R (T638,<br />

T640, S641, S642 <strong>and</strong> T644) which is important for #-arr recruitment. When these residues<br />

were mutated into Alanines (5A), the FSH-induced recruitment of #-arr 1 <strong>and</strong> #-arr 2 was<br />

reduced by 90% as assessed by co-immunoprecipitation with the FLAG-receptor. As<br />

expected, cAMP accumulation by the 5A mutant was enhanced when compared to the wildtype<br />

receptor. Consistantly, internalization of the 5A mutant was reduced when compared to<br />

the wild-type control. In striking contrast however, the ability of the 5A mutant to activate<br />

ERK via the #-arr pathway was actually increased when compared to the wild-type receptor.<br />

Together, these results suggest that at least two functionally distinct pools of #-arr are<br />

recruited to the activated FSH-R : 1) the predominant part interacts with the Ser638-Thr644<br />

cluster, involved in both desensitization <strong>and</strong> internalization <strong>and</strong> exerts a negative role on the<br />

ERK activation by both Gs <strong>and</strong> #-arr-dependent mechanisms, 2) a smaller proportion of the<br />

#-arr does not interact with the Ser/Thr cluster <strong>and</strong> is required for ERK activation through a<br />

#-arr-dependent pathway.<br />

- 215 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 30<br />

Design <strong>and</strong> functional activity of phosphopeptides with potential immunomodulating<br />

capacity, based on the sequence of Grb2 associated binder 1 (Gab1)<br />

Akos Kertesz1, Balazs Takacs1, Gyorgyi Varadi2, Gabor K. Toth2, Gabriella Sarmay1<br />

1Department of Immunology, Lor<strong>and</strong> Eotvos University, Budapest 1117, Hungary, email:<br />

sarmayg@cerberus.elte.hu <strong>and</strong> 2Department of Medical Chemistry, University of<br />

Szeged, Szeged 6720, Hungary, e-mail: tgabor@mdche.szote.u-szeged.hu<br />

Grb2-associated binder 1 (Gab1) is a scaffolding/adaptor protein involved in the signal<br />

transduction pathways of growth factors, cytokines, <strong>and</strong> antigen receptors. Gab adaptor<br />

proteins have several tyrosine residues which are phosphorylated upon lig<strong>and</strong> mediated<br />

tyrosine kinase activation <strong>and</strong> bind <strong>signaling</strong> molecules with SH2 domains, such as SHP-2<br />

tyrosine phosphatase, <strong>and</strong> phosphatidyl inositol 3-kinase (PI 3-K). We hypothesized that<br />

phosphopeptides corresponding to motifs of Gab1 may interfere with the corresponding<br />

<strong>signaling</strong> molecules in B cells, thus modulating the cell activation. Our main goal is to design<br />

cell membrane permeable phosphopeptides based on the sequence of Gab1 that may regulate<br />

the B-cell response.<br />

Phosphopeptides representing Gab1’s tyrosine phosphorylated motifs were synthesized <strong>and</strong><br />

tested. Using peptide pull down assay, we identified SHP-2, Lyn <strong>and</strong> PLCg binding to the<br />

same motif, 621GD-LD633. Furthermore, the 621GD-LD633 phosphopeptide bound <strong>and</strong><br />

powerfully activated SHP-2, while the PI3-K binding 442EL-PN453 <strong>and</strong> 467IQ-GP478<br />

peptides less efficiently activated the phosphatase. In order to test the role of the 621-633<br />

motif on B-cell <strong>signaling</strong>, the phosphopeptide was coupled to a membrane permeable<br />

transporting peptide, octanoyl-R8 (OR8). Confocal laser scanning microscopy analysis of<br />

cells exposed to the BODIPY dye labeled peptide showed that the peptide is taken up by B<br />

cells <strong>and</strong> it is localized mostly in lysosome. Next, B cells were treated with membrane<br />

permeable phosphopeptide, OR8(GD-LD) then the alteration of the intracellular free [Ca2+]<br />

<strong>and</strong> tyrosine phosphorylation of intracellular proteins were tested. OR8(GD-LD) transiently<br />

raised the level of [Ca 2+] in B-cells, <strong>and</strong> selectively modified the phosphorylation of certain<br />

intracellular proteins.<br />

Conclusion: cell membrane permeable phosphopeptides designed based on the sequence of<br />

Gab adaptor protein may selectively modulate intracellular <strong>signaling</strong> pathways thus may be a<br />

s<strong>our</strong>ce of further drug development.<br />

- 216 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 31<br />

LRRK1 protein kinase activity is stimulated upon binding of GTP to its Roc domain<br />

Daniel Korr, Luisella Toschi, Peter Donner, Hans-Dieter Pohlenz, Bertram Weiss, <strong>and</strong><br />

Bertolt Kreft<br />

Research Laboratories, Schering AG, Muellerstr.178, 13342 Berlin, Germany,<br />

E-mail: bertolt.kreft@schering.de<br />

Human leucine-rich repeat kinase 1 (LRRK1) is a multi-domain protein of unknown function<br />

belonging to the ROCO family of complex proteins. Here, we report the molecular<br />

characterization of human LRRK1 <strong>and</strong> show, for the first time, that LRRK1 is both a<br />

functional protein kinase <strong>and</strong> a GDP/GTP-binding protein. Binding of GTP to LRRK1 is<br />

specific, requires the GTPase-like Roc domain, <strong>and</strong> leads to a stimulation of LRRK1 kinase<br />

activity. LRRK1 is the first example of a GTP-regulated protein kinase harboring both the<br />

kinase effector domain <strong>and</strong> the GTP-binding regulatory domain. Hence, we propose a model<br />

in which LRRK1 cycles between a GTP-bound active <strong>and</strong> a GDP-bound inactive state.<br />

Moreover, we mutated LRRK1 to mimic mutations previously identified in LRRK2/dardarin,<br />

the only human paralogue of LRRK1, that have been linked to autosomal-dominant<br />

parkinsonism. We demonstrate that three of f<strong>our</strong> mutations analyzed significantly<br />

downregulate LRRK1 kinase activity. Ultimately, the results presented for LRRK1 may<br />

contribute to the elucidation of LRRK2’s role in the pathogenesis of Parkinson’s disease.<br />

- 217 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 32<br />

Activation of p38 MAP kinase in diosgenin-induced apoptosis in human rheumatoid<br />

synovial cells with increased COX-2 expression<br />

Bertr<strong>and</strong> Liagre1, David Y. Léger1, Pascale Vergne-Salle2, Philippe Bertin2, Richard<br />

Trèves2 <strong>and</strong> Jean-L. Beneytout1.<br />

1Laboratoire de Biochimie, UPRES EA 1085, Faculté de Pharmacie, Limoges, France.<br />

E-mail: bertr<strong>and</strong>.liagre@unilim.fr; 2Service de Rhumatologie et Thérapeutique, CHRU<br />

Dupuytren, Limoges, France.<br />

Alterations in the apoptosis of synovial cells have been described in resident synoviocytes <strong>and</strong><br />

associated with the pathogenesis of RA. The role of cyclooxygenase-2 (COX-2) <strong>and</strong><br />

prostagl<strong>and</strong>ins in synoviocyte death is still unknown. Recently, we showed that diosgenin, a<br />

plant steroid, induced apoptosis in human RA FLS with COX-2 overexpression (Liagre B, et<br />

al. (2004) Arthritis Res. Ther. 6:R373-R383). In a new work, we studied the kinase (Akt <strong>and</strong><br />

MAPK) signalling pathway after diosgenin-induced apoptosis in the human RA FLS.<br />

Particular attention was paid to the modulation of COX-2 expression <strong>and</strong> activity in RA<br />

synoviocyte viability. Our results showed that 40 µM diosgenin caused an inhibition of Akt<br />

phosphorylation. Moreover, after diosgenin treatment, p38 MAP kinase activation was<br />

increased in contrast with JNK or ERK activation. Phosphorylation of p38 was correlated<br />

with an increase of COX-2 expression <strong>and</strong> activity. However, diosgenin inhibited nuclear<br />

factor-kB (NF-kB) in apoptotic conditions in human RA FLS. In order to clarify if activation<br />

of p38 was directly associated with diosgenin-induced RA FLS apoptosis, we used a selective<br />

inhibitor of p38 (SB 203580) to verify DNA fragmentation after diosgenin treatment. We also<br />

studied in the same conditions the level of COX-2 expression <strong>and</strong> prostagl<strong>and</strong>in E2 (PGE2)<br />

production. Pre-treatment with SB 203580 before diosgenin treatment reduced the production<br />

of mononucleosomes <strong>and</strong> oligonucleosomes in comparison with diosgenin alone.<br />

Furthermore, this inhibition of diosgenin-induced apoptosis was correlated with an inhibition<br />

of COX-2 expression <strong>and</strong> a decrease of PGE2 synthesis.<br />

In conclusion, we showed for the first time that diosgenin-induced apoptosis in human RA<br />

FLS is associated with an increase of p38 activity <strong>and</strong> a decrease of Akt phosphorylation <strong>and</strong><br />

NF-kB activation. Overexpression of COX-2 is consecutive at the activation of p38 after<br />

diosgenin treatment. Future studies should evaluate whether diosgenin could be used as a<br />

therapeutic agent for RA in association with a COX-2 inhibitor.<br />

This work was supported by La Société Française de Rhumatologie.<br />

- 218 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 33<br />

Protein kinase C-eta as a possible therapeutic target in breast cancer<br />

Etta Livneh, Galia Oberkobitz, Noa Rotem, Galia Karp, Adva Maissel <strong>and</strong> Galit Shahaf.<br />

Department of Microbiology <strong>and</strong> Immunology, Ben Gurion University, Beer Sheva<br />

84105, Israel, e-mail:etta@bgumail.bgu.ac.il<br />

Protein kinase C (PKC) is involved in several major signal transduction pathways that control<br />

gene expression, cell growth <strong>and</strong> differentiation. However, these kinases were not thoroughly<br />

explored for their chemotherapeutic values. Here we have explored PKCeta as a possible<br />

target for chemotherapy in breast cancer. PKCeta was recently suggested as a therapeutic<br />

target in glioblastoma multiforme. PKCeta, expressed primarily in epithelial cells, appears as<br />

a c<strong>and</strong>idate regulator of mammary gl<strong>and</strong> proliferation or differentiation, as its expression is<br />

specifically up-regulated in the mammary gl<strong>and</strong> during the transit from resting to pregnant<br />

state. This prompt us to examine the hormonal regulation of its expression by estrogen <strong>and</strong><br />

progesterone as their levels are also up-regulated during pregnancy. We show that estradiol<br />

gradually increased the expression of PKCeta in the estrogen-responsive lines MCF-7 <strong>and</strong><br />

T47D, but not in the estrogen non-responsive line MDA-MB 231. In contrast to the observed<br />

up-regulation of PKCeta, the PKCdelta isoform was down-regulated while PKCalpha<br />

expression was unaltered, demonstrating different response to estradiol stimulation.<br />

Moreover, the presence of progesterone, involved in the differentiation of the mammary<br />

gl<strong>and</strong>, reduced the estrogen- induced PLCeta expression in a time-dependent manner.<br />

Interestingly, <strong>our</strong> recent studies suggest also a role in apoptosis for PKCeta. Here we have<br />

investigated its role in cell death induced by the DNA damaging agents UV irradiation <strong>and</strong><br />

the anti-cancer drug– Camptothecin (CPT). Our studies showed that the inducible expression<br />

of PKCeta in MCF-7 cells provided partial resistance against cell death, which was<br />

accompanied by increased cell survival <strong>and</strong> PARP cleavage. Furthermore, we show that JNK<br />

activity required for apoptosis in MCF-7 cells was inhibited by the inducible expression of<br />

PKCeta. Thus, <strong>our</strong> studies suggest that PKCeta has an important role in the hormonal<br />

regulation, cell cycle control <strong>and</strong> resistance to DNA damage in breast cancer cells. These<br />

properties could make PKCeta a target for chemotherapeutic intervention in breast cancer.<br />

- 219 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 34<br />

Spatial regulation of PKCeta: C1b domain <strong>and</strong> the pseudosubstrate containing fragment<br />

target PKCeta to the Golgi <strong>and</strong> the Nuclear Envelope<br />

Adva Maissel, Mairav Marom, Marat Shtutman, Galit Shahaf <strong>and</strong> Etta Livneh<br />

Department of Microbiology <strong>and</strong> Immunology, Faculty of Health Sciences <strong>and</strong> the<br />

Cancer Research Center, Ben Gurion University, Beer Sheva 84105, Israel<br />

Protein kinase C (PKC) represents a family of serin/threonine kinases, playing a central role<br />

in the regulation of cell growth, differentiation <strong>and</strong> transformation. These enzymes differ in<br />

their primary structure, biochemical properties, tissue distribution <strong>and</strong> sub-cellular<br />

localization. The specific cellular functions of PKC isoforms is largely controlled by their<br />

localization. PKCeta, a member of the novel subfamily, is expressed predominantly in<br />

epithelial tissues. However, not much is known with respect to its mechanism of activation<br />

<strong>and</strong> regulation. Our recent studies suggest its role in cell cycle control. In this work we show<br />

that PKCeta is localized at the Golgi apparatus, ER <strong>and</strong> the nuclear envelope. Furthermore,<br />

using GFP-fusion proteins of the different functional domains of PKCeta we deciphered the<br />

specific structural domains of the protein responsible for its apparent localization. We show<br />

that the cysteine-rich repeat C1b is responsible for its Golgi localization, while for its<br />

presence at the ER/nuclear envelope the pseudosubstrate containing fragment coupled to the<br />

C1 domain is required. In response to short-term activation by PMA we show translocation of<br />

PKCeta to the plasma membrane <strong>and</strong> the nuclear envelope. Moreover, we demonstrate that<br />

the C1b is sufficient for its translocation to the plasma membrane. Using an antibody specific<br />

for phosphorylated S675 (the hydrophobic site described as one of the priming sites of PKC<br />

enzymes), we show that, PKCeta is hyperphosphorylated on the hydrophobic site under these<br />

conditions of short-term PMA treatment. Interestingly, accumulation of PKCeta at the nuclear<br />

envelope as well as hyperphosphorylation on the hydrophobic site also occurred in response<br />

to serum-starvation. It should be noted that interaction of PKCeta with the cyclin E/Cdk2<br />

complex at the perinuclear region was recently reported by us in response to serum-starvation.<br />

Thus, <strong>our</strong> studies demonstrate translocation of PKCeta to the nuclear envelope, <strong>and</strong> suggest<br />

that the spatial regulation of PKCeta could be important for its cellular functions including<br />

effects on cell cycle control <strong>and</strong> involvement in tumor promotion.<br />

- 220 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 35<br />

Dlk/ZIP Kinase is involved in Mitosis <strong>and</strong> Cytokinesis<br />

Steve M<strong>and</strong>erscheid, Gerd L<strong>and</strong>sberg, Dominik Brinckmann <strong>and</strong> Karl Heinz<br />

Scheidtmann<br />

Institute of Genetics, University of Bonn, Roemerstr. 164 D-53117 Bonn, Germany. Email:<br />

stevem<strong>and</strong>er@web.de<br />

Dlk/ZIP Kinase is a Serin /Threonin specific Kinase of the proapoptotic DAP Kinase family.<br />

Dlk/ZIP Kinase does not induce apoptosis per se, rather it seems to be involved in regulation<br />

of transcription, contractile processes <strong>and</strong> mitosis, particularly cytokinesis. During mitosis<br />

Dlk/ZIP Kinase associates with centrosomes, centromeres <strong>and</strong> the contractile ring. In order to<br />

elucidate the biological role of these interactions <strong>and</strong> to map the interaction domains of<br />

Dlk/ZIP Kinase with these structures we established cell lines stably expressing N- <strong>and</strong> Cterminal<br />

truncation mutants at a low level. Mutant &N1 consisting of the extracatalytic<br />

domain colocalized with centrosomes <strong>and</strong> the contractile ring but not with<br />

centromere/kinetochore structures. Constitutive expression of this mutant did not reveal any<br />

phenotype indicating that it did not act in a dominant negative manner, at least not at the low<br />

expression level.<br />

On the other h<strong>and</strong>, &C2NLS lacking the C-terminal 110 amino acid residues including the<br />

leucine zipper, but containing a heterologous NLS did not associate with centrosomes or the<br />

contractile ring, though it was found associated with chromatin. In contrast to &N1-expressing<br />

cells, the cell line expressing &C2NLS exhibited a multinucleation phenotype. Interestingly,<br />

the &C2NLS cell line appeared heterogeneous. The majority of cells expressed the GFP<br />

fusion construct at a low level whereas about a quarter of the cells exhibited strong<br />

overexpression, mainly in the nucleus. Overexpression strongly correlated with generation of<br />

large round or deformed nuclei. Multinucleated cells were observed too.<br />

Cells were additionally analysed for phosphorylation of histone H3, expression <strong>and</strong><br />

localization of chromosomal passenger proteins (aurora B, INCENP, <strong>and</strong> survivin). H3<br />

phosphorylation of Serin 10 <strong>and</strong> Threonin 11 was normal, the chromosomal passenger<br />

showed the known distribution pattern. Our data show for the first time that Dlk might be<br />

involved in mitotic processes, prior to cytokinesis.<br />

Acknowledgement : This work is supported by DFGGrant Sche 246/16 <strong>and</strong> “b<strong>our</strong>se de<br />

formation-recherche” BFR 03/021<br />

- 221 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 36<br />

The JNK/NF-kB balance controls the life span of retinoic acid-treated APL cells.<br />

Julie Mathieu1, Stéphane Giraudier2, Michel Lanotte1 <strong>and</strong> Françoise Besançon1<br />

1INSERM U685, Centre Hayem, Hôpital St Louis, 1 avenue Claude Vellefaux, 75475<br />

Paris, France; 2 INSERM U362, Institut Gustave Roussy, 39 rue Camille-Desmoulins,<br />

94805 Villejuif, France.<br />

E-mail : Julie.mathieu@stlouis.inserm.fr;Francoise.besancon@stlouis.inserm.fr<br />

All-trans retinoic acid (ATRA) significantly improves the survival of patients with acute<br />

promyelocytic leukemia (APL) by inducing granulocytic differentiation of leukemia cells.<br />

Since an activation of the transcription factor NF-kappa B occurs during ATRA-induced<br />

maturation of APL cells, a mechanistic link between these two processes was investigated.<br />

Using an in vitro model for APL, we report that ectopic overexpression of a repressor of NFkappa<br />

B activation did not affect granulocytic differentiation. Importantly, NF-kappa B<br />

inhibition markedly resulted in a decreased viability of the differentiated cells which<br />

correlated with increased apoptosis. Apoptosis was accompanied by a sustained activation of<br />

the c-Jun N-terminal kinase (JNK). Inhibition of JNK by the specific inhibitor SP600125 or<br />

by transfection of a dominant-negative mutant of JNK1 reduced the percentage of apoptotic<br />

cells, thus showing that JNK activation constitutes a death signal. Furthermore, impairment of<br />

NF-kappa B activation resulted in increased levels of reactive oxygen species (ROS) upon<br />

ATRA treatment. ROS accumulation was suppressed by the antioxidant butylated<br />

hydroxyanisol, which also abolished ATRA-induced JNK activation <strong>and</strong> apoptosis.<br />

Altogether, <strong>our</strong> results demonstrate an anti-apoptotic effect of NF-kappa B activation during<br />

ATRA-induced differentiation of NB4 cells <strong>and</strong> identify repression of ROS-mediated JNK<br />

activation as a mechanism for this effect. Our observations also suggest that NF-kappa B<br />

<strong>signaling</strong> may contribute to an accumulation of mature APL cells <strong>and</strong> participate in the<br />

development of ATRA syndrome.<br />

- 222 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 37<br />

Determination of human epidermal growth factor receptors downstream <strong>signaling</strong><br />

phosphoproteins in breast cancer using suspension beads protein array.<br />

Jean-Louis Merlin, Elisabeth Longavenne, Fadila Chergui, Carole Ramacci, Marie<br />

Rouyer, Sanae Bouali, Pascal Genin, Agnès Leroux.<br />

Centre Alexis Vautrin, EA3452 Université Henri Poincaré, Nancy, France<br />

jl.merlin@nancy.fnclcc.fr<br />

Human epidermal growth factor receptors (HER) have major therapeutic implications in<br />

breast cancer. Receptor-directed monoclonal antibodies as well as tyrosine kinase inhibitors<br />

are designed to block HER downstream <strong>signaling</strong> <strong>and</strong> achieve proliferation control <strong>and</strong><br />

apoptosis induction. In this study, we validate the use of multiplex phosphoproteins singlestep<br />

analysis using suspension beads protein array to explore the functionality of HER<br />

downstream <strong>signaling</strong> in two human breast cancer cell lines, MDA-MB231 <strong>and</strong> MDA-<br />

MB468 : bearing different status of PTEN expression : MDA-MB468 are PTEN-null cell<br />

lines.<br />

The kinetics of expression of phosphorylated key-proteins of EGFR downstream <strong>signaling</strong><br />

(EGFR, AKT, p70S6 kinase, ERK1/2, GSK3 <strong>and</strong> p38MAPK) was measured by multiplex<br />

analysis using suspension beads protein array in EGF treated-cells. EGF exposure led to<br />

elevated expression of phospho-EGFR in both cell lines. Variations of expression of HER<br />

downstream <strong>signaling</strong> phosphoproteins were detected among the cell lines. EGF exposure led<br />

to increased phospho-AKT expression in PTEN expressing cells within the 5 minutes<br />

following exposure to EGF. No variation in phospho-AKT expression was observed in the<br />

PTEN-null cell line.<br />

In breast cancer specimens, great variations of phosphoproteins expression were observed<br />

showing that before treatment initiation HER downstream <strong>signaling</strong> functionality can differ<br />

from patient to patient. Such variations could probably be implicated <strong>and</strong> explain differences<br />

in clinical response to anti-HER drugs.<br />

All suspension beads protein array results, achieved in cell lines or breast cancer specimens,<br />

were compared <strong>and</strong> found fully consistent with western blot.<br />

This study demonstrates the great interest of suspension beads protein array for single-step<br />

phosphoproteins multiplex analysis. Suspension beads protein array can be used with sizelimited<br />

tumor specimens <strong>and</strong> yield accurate determination of phosphorylation rates. These<br />

results validate the use of multiplexed phosphoproteins analysis using suspension beads<br />

protein array to assess functionality of HER dowstream <strong>signaling</strong> as predictive <strong>and</strong>/or<br />

surrogate marker for clinical response to anti-HER drugs.<br />

- 223 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 38<br />

Phosphorylation by p42/44 MAPK regulates the activity <strong>and</strong> localization of human<br />

hypoxia inducible factor HIF-1alpha.<br />

Ilias Mylonis1, Georgia Chachami1,2, Martina Samiotaki3, George Panayotou3,<br />

Efrosyni Paraskeva2, Eleni Georgatsou1, Sophia Bonanou1 <strong>and</strong> George Simos1<br />

1Laboratory of Biochemistry <strong>and</strong> 2Laboratory of Physiology, Department of Medicine,<br />

University of Thessaly, Larissa, Greece.<br />

3Protein Chemistry Laboratory, B.S.R.C. «Alex<strong>and</strong>er Fleming», Vari, Greece<br />

E-mail: simos@med.uth.gr<br />

Hypoxia inducible factor 1 (HIF-1) is a heterodimeric transcriptional activator that controls<br />

the expression of most of the genes induced by hypoxic conditions. The molecular<br />

mechanisms that regulate the expression <strong>and</strong> activity of its inducible subunit HIF-1alpha,<br />

involve several post-translational modifications including hydroxylation, acetylation <strong>and</strong><br />

phosphorylation. In order to study the role of HIF-1alpha phosphorylation we have used<br />

human recombinant HIF-1alpha as a substrate in kinase assays with cell extracts. Our data<br />

show that at least two different nuclear protein kinases can interact with <strong>and</strong> modify HIF-<br />

1alpha <strong>and</strong> one of them was identified as the p42/p44 MAPK. Analysis of phosphorylated<br />

HIF-1alpha by mass spectroscopy revealed two serine residues as possible MAPK<br />

phosphorylation sites in its carboxy-terminal part. Site-directed mutagenesis of these two<br />

residues significantly reduced the phosphorylation of HIF-1alpha by either cell extracts or<br />

purified MAPK, showing that they represent major in vitro modification sites. When the<br />

mutant forms of HIF-1alpha were introduced into HeLa cells, they exhibited much lower<br />

transcriptional activity than the wild-type. Furthermore, the GFP-tagged HIF-1alpha mutants<br />

were excluded from the nucleus in contrast to the predominant nuclear localization of the<br />

wild-type form. These data suggest that phosphorylation of the identified sites by MAPK is<br />

required for nuclear accumulation of HIF-1alpha <strong>and</strong> subsequent activation of the hypoxia<br />

target genes.<br />

- 224 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 39<br />

Doxorubicin-induced MAP kinase activation in hepatocyte cultures is independent of<br />

oxidant damage<br />

Rosaura Navarro, Rosa Martínez, Idoia Busnadiego, M. Begoña Ruiz-Larrea <strong>and</strong> José<br />

Ignacio Ruiz-Sanz<br />

Department of Physiology, Medicine <strong>and</strong> Dentistry School, University of the Basque<br />

Country, 48080-Bilbao, Spain. E-mail: mbego.ruizlarrea@ehu.es<br />

Doxorubicin (DOX) is a potent anticancer drug, whose clinical use is limited due to its<br />

toxicity. DOX cytotoxic effects have been associated with reactive oxygen species (ROS)<br />

generated during the drug metabolism. ROS induce <strong>signaling</strong> cascades leading to changes in<br />

the phosphorylation status of target proteins, which are keys for cell survival or apoptosis.<br />

The mitogen-activated protein kinase (MAPK) cascades are routes activated in response to<br />

oxidative stress. In this work the effects of DOX on cytotoxicity, indicators of oxidative stress<br />

(malondialdehyde –MDA- <strong>and</strong> GSH), <strong>and</strong> the phosphorylation status of extracellular signalregulated<br />

kinases (ERKs), c-Jun N-terminal kinases (JNKs) <strong>and</strong> p38 kinases were analyzed in<br />

primary cultures of rat hepatocytes. DOX (1- 50 µM) did not modify LDH release into the<br />

medium during the incubation time up to 6 h. The levels of MDA, determined by HPLC, <strong>and</strong><br />

the intracellular GSH were constant during the treatment with the drug. GSH levels from<br />

mitochondria extracted by Percoll gradient from cultured hepatocytes were not modified by<br />

DOX, thus excluding its depletion or any impaired mitochondrial uptake. Characterization of<br />

proteins by Western blot analysis revealed that DOX increased phosphorylation of p38<br />

kinases, JNK1/2 <strong>and</strong> ERK1/2. In conclusion, DOX triggers activation of ERK, JNK <strong>and</strong> p38<br />

kinases in primary hepatocyte cultures independently of oxidant damage.<br />

This work was supported by the Basque Government (Research Project <strong>and</strong> Predoctoral<br />

Training Grant to R.N.) <strong>and</strong> the University of the Basque Country (UPV00081.327-E-<br />

15294/2003).<br />

- 225 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 40<br />

Superoxide anions are involved in doxorubicin-induced ERK activation in hepatocyte<br />

cultures<br />

Rosaura Navarro, Idoia Busnadiego, M. Begoña Ruiz-Larrea <strong>and</strong> José Ignacio Ruiz-<br />

Sanz<br />

Department of Physiology, Medicine <strong>and</strong> Dentistry School, University of the Basque<br />

Country, 48080-Bilbao, Spain. E-mail: joseignacio.ruizs@ehu.es<br />

Doxorubicin (DOX), an antineoplastic agent widely used for the treatment of cancer, belongs<br />

to the anthracycline family of antitumor antibiotics. DOX may undergo one-electron<br />

reduction to the corresponding semiquinone free radical by flavin-containing reductases.<br />

Under aerobic conditions, the semiquinone radical reacts rapidly with oxygen to generate<br />

superoxide anion, undergoing redox-cycling. At moderate concentrations, ROS play an<br />

important role as regulatory mediators in <strong>signaling</strong> processes. We have shown that DOX<br />

increased phosphorylation of enzymes comprising MAP kinase cascades in primary<br />

hepatocyte cultures, <strong>and</strong> that this action was independent of oxidant damage. In particular,<br />

extracellular signal-regulated kinase (ERK) was phosphorylated by the drug treatment. In this<br />

work, we have determined the possible involvement of particular free radicals in DOXinduced<br />

ERK phosphorylation in hepatocyte cultures by using specific free radical<br />

scavangers. The levels of ERK phosphorylation were measured by Western blot analysis with<br />

an anti-Thr202/Tyr204-phosphorylated 44/42 MAPK antibody. Deferoxamine (iron chelator),<br />

catalase (hydrogen peroxide removing enzyme) or alpha-tocopherol (peroxyl-radical<br />

scavenger) did not affect DOX-increased ERK phosphorylation levels. However, the cellpermeable<br />

superoxide dismutase mimetic MnTBAP, <strong>and</strong> the flavin-containing enzyme<br />

inhibitor diphenyleneiodonium, reverted DOX-induced effects. These results suggest that<br />

superoxide anions, probably generated by DOX metabolism, are involved in the effects of the<br />

anthracycline on the MAP kinase cascade activation.<br />

This work was supported by the Basque Government (Research Project <strong>and</strong> Predoctoral<br />

Training Grant to R.N.) <strong>and</strong> the University of the Basque Country (UPV00081.327-E-<br />

15294/2003).<br />

- 226 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 41<br />

Doxorubicin transitorily triggers Akt/PKB activation in primary cultures of rat<br />

hepatocytes<br />

Rosaura Navarro, Idoia Busnadiego, M. Luisa Hernández, M. Begoña Ruiz-Larrea <strong>and</strong><br />

José Ignacio Ruiz-Sanz<br />

Department of Physiology, Medicine <strong>and</strong> Dentistry School, University of the Basque<br />

Country, 48080-Bilbao, Spain. E-mail: joseignacio.ruizs@ehu.es<br />

The serine-threonine protein kinase Akt/PKB (protein kinase B) has received much interest in<br />

recent years because it is a key mediator of cell proliferation <strong>and</strong> seems to play an important<br />

role in tumorigenesis <strong>and</strong> resistance to chemotherapeutic drugs. The kinase suppresses<br />

chemotherapy-triggered apoptosis through interaction with critical molecules that regulate or<br />

execute apoptosis. Although most reports have shown that chemotherapy decreases Akt<br />

activity, the potent anticancer drug doxorubicin can increase Akt activity, depending on the<br />

cell type. Thus, in several cancer cell lines, the drug triggers a transient phosphorylation <strong>and</strong><br />

activation of Akt at early time points, prior to the detection of apoptosis. In this work, we<br />

have determined in primary cultures of rat hepatocytes the effects of doxorubicin on the<br />

phosphorylation status of Akt. The levels of Akt phosphorylation were measured by Western<br />

blot analysis with an anti-Ser473-phosphorylated Akt antibody. Doxorubicin (10 µM) had a<br />

biphasic behavi<strong>our</strong>, increasing (60%) Akt phosphorylation at 30 min <strong>and</strong> decreasing it (40%)<br />

at 1h. At later times, the Akt phosphorylation status remained similar to control cells (without<br />

doxorubicin). The phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin prevented Akt<br />

activation. The profile of the phsophorylation levels of Akt with doxorubicin was completely<br />

different from that found for the pro-apoptotic JNK, which increased in a dose- <strong>and</strong> timedependent<br />

manner.<br />

This work was supported by the Basque Government (Research Project <strong>and</strong> Predoctoral<br />

Training Grant to R.N.) <strong>and</strong> the University of the Basque Country (UPV00081.327-E-<br />

15294/2003).<br />

- 227 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 42<br />

Phosphoinositide 3-kinase/Akt pathway as a potential therapeutic target in patients with<br />

high risk myelodysplastic syndromes<br />

Maria Nyåkern1, Carlo Finelli2, Pier Luigi Tazzari3, Costanza Bosi2, Matilde Yung<br />

Follo1, Lucio Cocco1, Alberto M. Martelli1<br />

1Department of Anatomy, Cell Signalling Laboratory, Università di Bologna, 40126<br />

Bologna, Italy;2Institute of Hematology <strong>and</strong> 3Blood Bank, Policlinico S.Orsola-<br />

Malpighi, Università di Bologna, 40138 Bologna, Italy. E-Mail:<br />

alberto.martelli@unibo.it<br />

The serine/threonine kinase Akt, a downstream effector of phosphatidylinositol 3-kinase<br />

(PI3K), is known to play an important role in anti-apoptotic <strong>signaling</strong> <strong>and</strong> has been implicated<br />

in the aggressiveness of a number of different human cancers including acute myeloid<br />

leukemia (AML). Myelodysplastic syndromes are hematopoietic stem cell disorders<br />

characterized by ineffective hematopoiesis, leading to blood cytopenias, <strong>and</strong> by a high risk of<br />

progression to overt AML. The progression of MDS to AML is thought to be associated with<br />

abrogation of apoptotic control mechanisms. However, little is known about the signal<br />

transduction pathways which may be involved in enhanced survival of MDS cells. In this<br />

report, we have performed immunocytochemical <strong>and</strong> flow cytometric analysis, to evaluate the<br />

levels of activated Akt in bone marrow or peripheral blood mononuclear cells from patients<br />

diagnosed with MDS. We observed high levels of Ser473 phosphorylated Akt (p-Akt)<br />

staining in 90 % of the cases (n=22) diagnosed as high risk MDS, whereas mononuclear cells<br />

from normal bone marrow or low risk MDS patients showed low or absent Ser473 p-Akt<br />

staining. Furthermore, all high risk MDS patients also demonstrated high expression of the<br />

Class I PI3K p110& catalytic subunit <strong>and</strong> a decreased expression of PTEN. Pharmacological<br />

inhibitors of the PI3K/Akt pathway (SH-5, perifosine) caused apoptotic cell death of bone<br />

marrow mononuclear cells isolated from high risk MDS patients. Taken together, <strong>our</strong> results<br />

suggest that Akt activation might be one of the factors contributing to the decreased apoptosis<br />

rate observed in patients with high risk MDS. PI3K/Akt inhibition may constitute a novel<br />

therapeutic strategy if apoptosis induction of MDS mononuclear cells is the goal.<br />

- 228 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 43<br />

Expression of HPV E5 protein induces expression of EP4 prostanoid receptor through<br />

PKA- <strong>and</strong> CREB-dependent pathway in C33A cervical cancer cells.<br />

Jung Min OH <strong>and</strong> Yong-Sung JUHNN<br />

Department of Biochemistry <strong>and</strong> Molecular Biology, Cancer Research Institute, Seoul<br />

National University College of Medicine, Seoul 110-799, Korea<br />

HPV (Human Papilloma Virus) infection is the major cause of cervical cancer development.<br />

The role of HPV E6 <strong>and</strong> E7 protein in the carcinogenesis has been extensively studied,<br />

however, the role of E5 protein in cervical carcinogenesis is not understood clearly. The<br />

prostagl<strong>and</strong>in <strong>signaling</strong> pathway mediated by cyclooxygenase-2(COX-2) is activated in<br />

cervical cancer cells <strong>and</strong> many other types of cancer cells, therefore, this study aimed to<br />

analyze the effect of E5 protein on prostagl<strong>and</strong>in <strong>signaling</strong> pathway.<br />

Stable expression of E5 protein caused an increase in COX-2 protein <strong>and</strong> EP4 prostanoid<br />

receptor in C33A cervical cancer cells. Stable expressing COX-2 protein also increased<br />

mRNA expression of EP4 <strong>and</strong> prostagl<strong>and</strong>in E synthase <strong>and</strong> secretion of prostagl<strong>and</strong>in E2<br />

(PGE2), major product of COX-2 in inflammatory process, in C33A cervical cancer cells.<br />

Treatment of E5 expressing cells with NS-398, a COX-2 inhibitor, decreased the expression<br />

of EP4, <strong>and</strong> furthermore, treatment of HeLa cells that expresses high level of COX-2 with<br />

NS-398 also resulted in decrease in EP4 mRNA expression <strong>and</strong> PGE2 secretion. Treatment<br />

with H89, a PKA inhibitor or decoy CRE reduced expression of EP4 mRNA in COX-2<br />

expressing cells. However, expression of COX-2 in HaCaT cells, immortalized human<br />

keratinocytes did not induce expression of EP4.<br />

Because the EP4 protein level did not increase in parallel with EP4 mRNA following E5<br />

overexpression, the degradation rate of EP4 protein was analyzed. The EP4 protein in COX-2<br />

expressing cells was found to degrade faster in the presence of cycloheximide than that of<br />

vector-transfected control cells. The degradation of EP4 protein was blocked by treatment<br />

with MG-132, a proteasome inhibitor.<br />

From this result, we conclude that HPV E5 protein induces EP4 mRNA through COX-2-,<br />

PKA-, <strong>and</strong> CREB-dependent pathway in cervical cancer cells, not in HaCaT cells.<br />

- 229 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 44<br />

Differential modulation of AMPK <strong>signaling</strong> pathways by low or high levels of exogenous<br />

reactive oxygen species in colon cancer cells<br />

In-Ja Park, Jin-Taek Hwang 1 , Young Min Kim 2 , Joohun Ha 1 <strong>and</strong> Ock Jin Park<br />

Department of Food <strong>and</strong> Nutrition, Hannam University, 133 Ojeong-dong Daedeok-gu,<br />

Daejeon 306-791, Korea, E-mail: ojpark@hannam.ac.kr<br />

1 Department of Biochemistry <strong>and</strong> Molecular Biology, Medical Research Center for<br />

Bioreaction to Reactive Oxygen Species, Kyung Hee University College of Medicine,<br />

Seoul 130-791, Korea<br />

2 Department of Biological Sciences, Hannam University, Daejeon 306-791, Korea<br />

ROS have been regarded as undesirable metabolic byproducts often linked to macromolecule<br />

damage, aging or degenerative diseases. However, ROS emerge as important intracellular<br />

<strong>signaling</strong> molecules, which act as mediators or second messengers at nontoxic concentrations<br />

through receptor-transducing pathways. In cancer cell system, ROS exert a paradoxical effect.<br />

ROS can promote tumor growth by transforming normal cells through activation of<br />

transcription factors or inhibition of tumor suppressor genes, whereas the elevated levels<br />

would inhibit tumor cells through the stimulation of proapoptotic-signals. The excess ROS<br />

generate cell cycle arrest <strong>and</strong> apoptotic cell death or even necrosis in severe cases. Therefore,<br />

the maintenance of ROS homeostasis is extremely important to cell <strong>signaling</strong> <strong>and</strong> the<br />

regulation of cell death.<br />

The present study was undertaken to examine the effect of low <strong>and</strong> high concentrations of<br />

H 2O 2 on cancer cell proliferation <strong>and</strong> apoptosis, <strong>and</strong> AMPK <strong>signaling</strong> pathways in HT-29<br />

human colon cancer cells. Non-toxic dose of H 2O 2 (10 uM) induced cancer cell proliferation,<br />

whereas the toxic level of 1000 uM H 2O 2 induced apoptosis. The stimulation of cell<br />

proliferation was accompanied with an increase in cyclooxygenase-2 (COX-2), <strong>and</strong> apoptosis<br />

induced by high-dose H 2O 2 was correlated with the activation of AMPK <strong>and</strong> negatively<br />

correlated with COX-2 expression. These results suggest that ROS at non-toxic levels can<br />

stimulate cancer cell growth by regulating AMP-activated protein kinase (AMPK) <strong>and</strong>/or<br />

COX-2, <strong>and</strong> the abundant exogenous ROS linked to the growth inhibition through modulating<br />

AMPK <strong>signaling</strong> pathways.<br />

- 230 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 45<br />

Identification of <strong>signaling</strong> pathway leading to increase of mitochondrial cytochrome-c<br />

oxidase (COX) <strong>and</strong> mitofusin-2 (Mfn-2) proteins during insulin-mediated myogenesis in<br />

vitro<br />

Patrycja Pawlikowska1, Barbara Gajkowska2, Micha# M. Godlewski1, Arkadiusz<br />

Orzechowski1*<br />

1*Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw<br />

Agricultural University, Now<strong>our</strong>synowska 159, 02-776 Warsaw, Pol<strong>and</strong>, e-mail:<br />

orzechowski@alpha.sggw.waw.pl, 2Department of Cell Ultrastructure MRC, Polish<br />

Academy of Sciences, Warsaw, Pol<strong>and</strong>, *corresponding author.<br />

Mitochondrial fusion mediated by Mfn-2 has been recently shown to play a crucial role in the<br />

embryonic development, antiapoptosis through DNA protection against free radicals-induced<br />

lesions <strong>and</strong> maintaining unchanged mtDNA pool. The purpose of this study was to examine<br />

the role of mitochondria in insulin-dependent myogenesis in vitro. Two differently encoded<br />

(nuclear <strong>and</strong> mitochondrial) subunits of cytochrome-c oxidase (COX), myogenin <strong>and</strong> Mfn-2<br />

expressions were determined by Western blots. Our studies with LY294002 confirmed<br />

assumption that insulin stimulates myogenesis though PI3-K-dependent <strong>signaling</strong> pathway.<br />

Similarly, if MAPKK (MEK) was inhibited by PD98059 insulin-mediated myogenesis from<br />

C2C12 muscle cells was accelerated. Additionally, immunoblots revealed that insulin raised<br />

the expression of subunit I <strong>and</strong> IV of COX in L6 muscle cells. Apparently, insulin-dependent<br />

induction of COX I was mediated by PI3-K, since blockade of insulin action with LY294002<br />

resulted in decreased level of mitochondrial but not nuclear encoded subunit of COX.<br />

Moreover, electron <strong>and</strong> confocal microscopy showed that insulin activates formation of a<br />

network of elongated, tubular mitochondria. Increased mitochondrial length <strong>and</strong><br />

interconnectivity was not observed upon PI3-K inhibition with LY294002. Subsequently, we<br />

decided to study transmembrane GTPase – Mfn-2 which seems to be essential for<br />

mitochondrial fusion. We observed that insulin induces Mfn-2 protein in L6 myoblasts.<br />

Moreover, we found that inhibition of MAPKK-dependent <strong>signaling</strong> pathway additionally<br />

elevated both Mfn-2 <strong>and</strong> myogenin protein levels. Although correlation does not prove<br />

causation, we speculate that Mfn-2 participates in insulin-mediated muscle differentiation. We<br />

conclude that insulin-dependent myogenesis is associated with increased level of COX I <strong>and</strong><br />

Mfn-2 as well as augments fusion of mitochondria. The above-mentioned effects are inhibited<br />

by LY294002, whereas inhibition of MAPKK with PD98059 led to additional amplification<br />

of this phenomenon.<br />

- 231 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 46<br />

Tyrosine nitration of MEK <strong>and</strong> ERK induces their autophosphorylation <strong>and</strong> activation<br />

in vivo <strong>and</strong> in vitro<br />

Elena Pinzar*, Maria Garrido°, Daniela Urrecheaga°*, Guillaume Bonnot*, Patrick<br />

Levy* <strong>and</strong> Serge P. Bottari *#<br />

*Laboratoire HP2, UFR de Médecine, Grenoble Universités, Domaine de la Merci,<br />

38700 La Tronche, France, °Facultad de Farmacia, Universidad Central de Venezuela,<br />

Caracas, Venezuela, #Département de Biologie Intégrée, CHU de Grenoble, 38043<br />

Grenoble, France. E-mail: Serge.Bottari@ujf-grenoble.fr<br />

Angiotensin II (Ang II) has been shown to activate MAPK pathways <strong>and</strong> to induce production<br />

of reactive oxygen <strong>and</strong> nitrogen species. Nitric oxide (NO) <strong>and</strong> superoxide (O2-.) as well as<br />

peroxinitrite (ONOO-) have been previously reported to stimulate MAPK in a few cell types.<br />

We therefore investigated whether ERK could be nitrated in rat vascular smooth muscle cells<br />

(VSMC) upon exposure to Ang II. Stimulation of cells with 10 nM Ang II induced a<br />

prominant <strong>and</strong> sustained nitration of ERK1/2 as revealed by immunoprecipitation <strong>and</strong><br />

immunoblotting. ERK nitration was accompanied by activation of the enzyme <strong>and</strong> showed a<br />

kinetic profile different than that of phosphorylation. Both Ang II-induced ERK nitration <strong>and</strong><br />

phosphorylation were mediated by the AT1 receptor. The ONOO- donor 3morpholinosydnonimine<br />

hydrocloride (SIN-1) also stimulated nitration <strong>and</strong> activation of<br />

recombinant unactive ERK <strong>and</strong> MEK. Nitration <strong>and</strong> phosphorylation of ERK1/2 by Ang II<br />

was significantly inhibited by the NAD(P)Hoxidase inhibitors 4-(2-aminoethyl)<br />

benzenesulfonyl fluoride (AEBSF) <strong>and</strong> diphenylene iodonium (DPI). Moreover, the selective<br />

inducible nitric-oxide synthase (iNOS) inhibitor 1400W but not the endothelial NOS inhibitor<br />

L-NAME, completely inhibited nitration <strong>and</strong> activation of ERK. The ONOO- <strong>and</strong> O2-.<br />

scavengers, respectively ebselen <strong>and</strong> myricetin, drastically blunted Ang II-stimulated nitration<br />

<strong>and</strong> activation of ERK. Conversely, the MEK inhibitor U0126, did not affect ERK nitration,<br />

but completely blocked ERK phosphorylation <strong>and</strong> activation. Taken together these data<br />

indicate that Ang II can activate ERK1/2 both through nitration via reactive nitrogen speciessensitive<br />

pathways <strong>and</strong> by phosphorylation through the Ras-Raf-MEK pathway.<br />

Interestingly, ERK nitration appears to be a prerequisite for its activating phosphorylation by<br />

MEK in response to Ang II in VSMC.<br />

- 232 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 47<br />

HSP27 SCAFFOLDS MK2 TO THE AKT SIGNAL COMPLEX<br />

Madhavi J. Rane<br />

Department of Medicine, Molecular Signaling Group, University of Louisville,<br />

Louisville, KY 40202, U.S.A. E-mail:mrane@louisville.edu<br />

The phosphatidylinositol 3-kinase (PI3K)/Akt (protein kinase B, PKB) <strong>signaling</strong> pathway<br />

plays a critical role in cell growth, proliferation, <strong>and</strong> cell survival. Up regulation of this<br />

pathway has been demonstrated in various carcinomas. Thus, inhibiting Akt activation <strong>and</strong><br />

promoting cell death provides a strategy for targeted cancer therapy. We have shown<br />

previously that Akt/Hsp27 interaction regulates neutrophil apoptosis <strong>and</strong> that MK2 acts as<br />

PDK2 for Akt. The current study tested the hypothesis that Hsp27 regulates Akt activation<br />

<strong>and</strong> promotes cell survival by scaffolding MK2 to the Akt signal complex. Here we show that<br />

loss of Akt/Hsp27 interaction by anti-Hsp27 antibody treatment in neutrophils or by<br />

transfection of Hsp27 siRNA in HK-11 cells resulted in induction of cellular apoptosis.<br />

Transfection of myristoylated Akt (AktCA) in HK-11 cells induced Akt Ser473<br />

phosphorylation, activation, <strong>and</strong> Hsp27 Ser82 phosphorylation. Co-transfection of AktCA<br />

<strong>and</strong> Hsp27siRNA in HK-11 cells specifically silenced Hsp27 expression, without altering<br />

expression of Akt. Silencing Hsp27 expression resulted in the loss of Akt/MK2 interaction,<br />

loss of Akt Ser473 phosphorylation, activation, Hsp27 Ser82 phosphorylation, <strong>and</strong> induced<br />

HK-11 cell death. Deletion mutagenesis studies identified acidic linker region (117-128) on<br />

Akt as an Hsp27 binding region. Deletion of amino acids 117-128 on Akt resulted in loss of<br />

its interaction with Hsp27 <strong>and</strong> MK2, but not Hsp90 as demonstrated by immunoprecipitation<br />

<strong>and</strong> GST pull-down studies. Co-transfection studies in HEK-293 cells demonstrated that<br />

constitutively active MK2 (MK2EE) phosphorylated Aktwt on Ser473 but failed to<br />

phosphorylate Akt&117-128 mutant. In conclusion, Hsp27 scaffolds MK2 to Akt signal<br />

complex <strong>and</strong> promotes Akt activation <strong>and</strong> cell survival. Veterans Administration Grant <strong>and</strong><br />

AHA 0335278N<br />

- 233 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 48<br />

Transforming Growth Factor-#1 Induces Cyclooxygenase-2 Gene Expression In Human<br />

Mesangial Cells: Role of MAPK And PI3K pathways<br />

Alicia Rodríguez-Barbero, Fern<strong>and</strong>o Dorado, Atanasio P<strong>and</strong>iella*, José M. López-<br />

Novoa.<br />

Instituto Reina Sofía de Investigación Nefrológica. Dpto de Fisiología y Farmacología.<br />

*Centro de Investigaciones Biológicas. (CISC). Universidad de Salamanca. Campus<br />

Unamuno, Edificio Departamental. Avda. Campo Charro s/n. 37007. Spain. E-mail:<br />

barberoa@usal.es<br />

Transforming growth factor-# (TGF-#) plays a fundamental role in the progression of renal<br />

diseases. Mesangial cells play a major role in physiological <strong>and</strong> pathophysiological renal<br />

processes. Accumulating evidence has suggested that eicosanoids derived from<br />

cyclooxygenase 2 (COX-2) participate in a number of pathological processes in immunemediated<br />

renal diseases. Mesangial cells express receptors for TGF-# <strong>and</strong> COX-2 expression<br />

can be induced in mesangial cells. However, there are no studies on the possible role of TGF-<br />

# on COX-2 expression in human mesangial cells (HMC). The aims of this study were to<br />

assess whether TGF-#1 stimulates COX-2 expression in primary HMC, <strong>and</strong> to determine the<br />

routes involved in TGF-#1-induced COX-2 expression <strong>and</strong> PGE2 synthesis. TGF-#1 induced<br />

COX-2 promoter activity, COX-2 mRNA <strong>and</strong> protein expression in HMC. COX-2 induction<br />

was accompanied by increased PGE2 synthesis. To establish the role that ERK1/2 <strong>and</strong> p38<br />

MAPK as well as PI3K/Akt play in TGF-#1-dependent COX-2 expression, HMC were pretreated<br />

with inhibitors of these pathways. Inhibition of ERK1/2 by the MEK inhibitors,<br />

PD98059 or U0126 <strong>and</strong> inhibition of p38 MAPK activity by SB203580 notably decreased the<br />

TGF-#1-dependent COX-2 protein induction <strong>and</strong> PGE2 synthesis in HMC. In addition, pretreatment<br />

of HMC with the PI3K inhibitor LY294002 attenuated TGF-#1-induced COX-2<br />

expression <strong>and</strong> PGE2 synthesis. These results demonstrate that TGF-#1 regulates COX-2<br />

expression in HMC through the activation of ERK1/2, p38 MAPK, <strong>and</strong> PI3K. These data can<br />

help to elucidate the molecular mechanisms underlying the regulation of COX-2 <strong>and</strong> open up<br />

specific strategies for the treatment of glomerular disease.<br />

- 234 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 49<br />

Yeast as a Tool for the Study of the Mammalian PI3K-PTEN-Akt Pathway<br />

Isabel Rodríguez-Escudero,1 Françoise M. Roelants,2 César Nombela,1 Jeremy<br />

Thorner,2 María Molina1 <strong>and</strong> Víctor J. Cid1<br />

1Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense<br />

de Madrid, Pza. de Ramón y Cajal s/n, 28040 Madrid, Spain. E-mail:<br />

vicjcid@farm.ucm.es<br />

2Department of Molecular <strong>and</strong> Cell Biology, Division of Biochemistry <strong>and</strong> Molecular<br />

Biology, University of California, Berkeley, California 94720 USA.<br />

Generation of phosphatidylinositol (3,4,5)-trisphosphate (PIP3) by class I<br />

phosphatidylinositol 3-kinase (PI3K) is at the head of crucial <strong>signaling</strong> pathways in metazoa,<br />

like those involved in the control of cell proliferation <strong>and</strong> apoptosis. Therefore, this lipid<br />

kinase, its antagonist the phosphatidylinositol 3-phosphatase (PTEN), <strong>and</strong> its main<br />

downstream effector protein kinase B (PKB/c-Akt) are key therapeutic targets. With the aim<br />

of facilitating genetic, molecular <strong>and</strong> pharmacological analyses on these proteins, we have<br />

expressed mammalian PI3K (p110alpha), PTEN <strong>and</strong> c-Akt in the model organism<br />

Saccharomyces cerevisiae. Expression of mammalian PI3K dramatically inhibited yeast cell<br />

growth, an effect that depended on conversion of intracellular phosphatidylinositol (4,5)bisphosphate<br />

(PIP2) to PIP3, a lipid that does not naturally exist in S. cerevisiae. All cellular<br />

effects were fully reverted by co-expression of catalytically-active human PTEN, <strong>and</strong><br />

ameliorated by a PI3K inhibitor (LY294002). This system allows an easy platform for in vivo<br />

pharmacological studies <strong>and</strong> screens on these proteins. We have studied the effects of PIP2<br />

depletion on the yeast cytoskeleton <strong>and</strong> <strong>signaling</strong>. Interestingly, conversion of PIP2 to PIP3<br />

causes activation of a yeast MAPK pathway, suggesting a role of phosphoinositides upstream<br />

MAPK <strong>signaling</strong> in yeast. We were able to reproduce PIP3-dependent re-localization to the<br />

plasma membrane <strong>and</strong> phosphorylation of heterologous c-Akt in yeast. Endogenous yeast<br />

protein kinases are responsible for phosphorylation of heterologous c-Akt, suggesting that<br />

Akt-activating kinases are conserved through phylogeny. Thus, yeast might also provide a<br />

new tool for the discovery of novel Akt activators.<br />

- 235 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 50<br />

Competition effects shape the response sensitivity <strong>and</strong> kinetics of phosphorylation cycles<br />

in cell <strong>signaling</strong><br />

Carlos Salazar <strong>and</strong> Thomas Höfer<br />

Theoretical Biophysics, Institute of Biology, Humboldt University Berlin, Invalidenstr.<br />

42, 10115 Berlin, Germany. E-mail: carlos.salazar@rz.hu-berlin.de<br />

Activation-inactivation cycles of <strong>signaling</strong> proteins <strong>and</strong> transcription factors catalyzed by<br />

kinases <strong>and</strong> phosphatases are a core component of cellular signal transduction. We present a<br />

concise kinetic description of phosphorylation cycles that starts by considering the elementary<br />

steps of enzyme-target binding <strong>and</strong> catalysis. A rapid-equilibrium approximation for protein<br />

interactions is used to reduce the set of parameters. Generally no explicit rate laws exist for<br />

the kinase <strong>and</strong> phosphatase; linear or Michaelis-Menten rate equations can be obtained in<br />

special cases. Key parameters that determine system behavior are the concentrations of kinase<br />

<strong>and</strong> phosphatase relative to the target protein <strong>and</strong> the affinities of the two enzymes for the<br />

different phosphorylation states of the target. By scanning the space of these parameters, we<br />

obtain a phase diagram that shows the existence of graded, ultrasensitive <strong>and</strong> bistable<br />

behaviors as well as a previously undescribed biphasic response. Two kinds of competition<br />

effect turn out to shape the behavior: (1) the degree of product inhibition of each enzyme, <strong>and</strong><br />

(2) the competition between kinase <strong>and</strong> phosphatase to bind the target protein, as determined<br />

by their relative target affinities <strong>and</strong> concentrations. Like the steady-state response curve, the<br />

response time was found to be strongly dependent on the kinetic design of the<br />

phosphorylation cycles, <strong>and</strong> the order in which (de)phosphorylation of multiple residues<br />

proceed. Sequential mechanisms of phosphate processing are characterized by a higher degree<br />

of competition between phosphorylation <strong>and</strong> dephosphorylation, which results in sharper<br />

response curves <strong>and</strong> slower response times than in r<strong>and</strong>om mechanisms. Taken together, <strong>our</strong><br />

kinetic analysis allows us to identify key parameters that should be given priority in<br />

experimental measurements <strong>and</strong> to arrive at experimentally testable predictions concerning<br />

both the steady-state response <strong>and</strong> the kinetics of phosphorylation cycles <strong>and</strong> cascades.<br />

- 236 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 51<br />

JNK1 isoform is required for M-CSF <strong>and</strong> LPS induced MKP1 (DUSP1) expression<br />

Ester Sánchez-Tilló, Mónica Comalada, Consol Farrera, Jordi Xaus, Annabel F.<br />

Valledor, Carme Caelles, Jorge Lloberas <strong>and</strong> Antonio Celada<br />

Macrophage Biology Group, Institute of Biomedical Research, Barcelona Science Park,<br />

University of Barcelona, Spain. Phone 34934037165; Fax 34934034747; acelada@ub.edu<br />

Macrophages proliferate in the presence of M-CSF in a process that depends on early (5 min)<br />

<strong>and</strong> short ERK activation. The addition of LPS arrests proliferation <strong>and</strong> prolongs ERK<br />

activation, as well as p38 <strong>and</strong> JNK, which are required for an inflammatory response. The<br />

decision to proliferate or to become activated is correlated with the extent <strong>and</strong> duration of<br />

MAPK regulation, which is controlled by mitogen kinase phosphatases (MKPs). In <strong>our</strong><br />

model, the phosphatase responsible for ERK deactivation is MKP1 (DUSP1), which is<br />

induced by M-CSF or LPS with different time-c<strong>our</strong>se <strong>and</strong> independent of ERK activation. We<br />

observed that MKP1 induction by both M-CSF <strong>and</strong> LPS is transcriptionally dependent on<br />

JNK activity while other MAPKs are not involved. The inhibition of MKP1 by JNK caused a<br />

slight elongation of ERK-1/2 <strong>and</strong> p38 activation, as well as inhibition of M-CSF-dependent<br />

proliferation. Using macrophages from PKC' knock-out mice, we observed that although M-<br />

CSF <strong>and</strong> LPS activate the same pathway to induce MKP1, only crosstalk between PKC' <strong>and</strong><br />

JNK is present in LPS-stimulated macrophages. The two JNK genes, jnk1 <strong>and</strong> jnk2 are<br />

constitutively expressed in macrophages, but only the JNK1 isoform is involved in MKP1<br />

induction by M-CSF or LPS, as determined using single knock-out mice. Moreover, JNK1 is<br />

required for pro-inflammatory cytokine biosynthesis (TNF-", IL-1# <strong>and</strong> IL-6) <strong>and</strong> NO<br />

production triggered by activation of LPS <strong>and</strong> TNF-". The induction of these cytokines is<br />

independent of MKP1 expression, as determined in MKP1 knock-out mice. Our results show<br />

that stimuli that induce MAPK activation also lead to the induction of attenuators, which<br />

provides a negative feedback mechanism through which to control the activation of other<br />

MAPKs. JNK could be a novel therapeutic target to regulate MKP1 expression in tum<strong>our</strong>s<br />

<strong>and</strong> in chronic inflammatory responses.<br />

EN.REFLIST<br />

- 237 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 52<br />

MAPKinase gene expression – as determined by microarray analysis - distinguishes<br />

uncomplicated from complicated reconstitution after major surgical trauma<br />

E. Marion Schneider1, Manfred Weiss2, Weidong Du1, Fabian Kriebel1, Gerhard<br />

Leder3, Klaus Buttenschön3, Ulrich C. Liener3, Uwe Bernd Brückner3<br />

fabiankriebel@web.de<br />

1Sektion Experimentelle Anaesthesiologie; 2Abteilung Klinische Anaesthesiologie,<br />

3Abteilung Chirurgie, Universitaetsklinikum Ulm, Steinhoevelstrasse 9; 89075 Ulm,<br />

Germany<br />

Objective: A prospective pilot study in patients with major surgical trauma at an university<br />

hospital was set up to identify <strong>signaling</strong> pathways by microarray expression analysis, which<br />

may be distinct in individual patients <strong>and</strong> may be indicative for complicated versus uneventful<br />

reconstitution post trauma.<br />

Methods: RNA was prepared from peripheral blood drawn into PAXgene tubes from three<br />

patients before <strong>and</strong> exactly 24h after trauma. These RNA were then transcribed into cDNA,<br />

Cy3- or Cy5-labeled, <strong>and</strong> hybridized onto a cDNA microchip, red vs. green fluorescence was<br />

quantified using a microarray scanner. Patient (patients before <strong>and</strong> post trauma) versus control<br />

(healthy donors) blood cell gene expressions were compared using fluorescence intensities<br />

<strong>and</strong> appropriate normalizations.<br />

Results: In addition to a number of rather non-specific stress response genes, activated in all<br />

patients, we found a remarkable number of differences in gene expression patterns of<br />

individual patients. Some of the differing genes were associated with uncomplicated<br />

convalescence such as up-regulation of both the ERK5 pathway (MAPK7, mitogen-activated<br />

protein kinase-7) <strong>and</strong> transcription factors which stimulate hematopoiesis <strong>and</strong> tissue<br />

remodeling (MEF2, myocyte enhancer factor-2, BMP-2, bone morphogenic protein-2,<br />

TNFRSF11a (TNF-R superfamily, syn. RANK), <strong>and</strong> RUNX-1, runt related transcription<br />

factor-1). Chemokine genes active in stem cell recruitment from the bone marrow as well as<br />

dendritic cell <strong>and</strong> NK maturation (SCYA14 (HCC-1, hemofiltrate cc chemokine )), were<br />

increased as well as activators of the lymphoid compartment (TNFRSF7 (CD27), CD3zeta<br />

<strong>and</strong> perforin (PRF1)). In contrast, all these transcripts were down-regulated in complicated<br />

reconstitution <strong>and</strong> later development of septic shock. Moreover, p38 kinase (MAPK14), S100<br />

molecules <strong>and</strong> members of the lipoxygenase pathway were associated with a more eventful<br />

outcome. In none of the patients, we found c-Jun-stress activated factor (JNK) pathway to be<br />

markedly activated, also p105 (Rel) or the “NFkB inducible kinase” (NIK, MAP3K14) were<br />

not found to be up-regulated post surgical trauma.<br />

Conclusions: Microarray expression studies are a promising tool for screening <strong>and</strong> then<br />

selecting differentially regulated genes in favorable as compared to complicated reconstitution<br />

post trauma. Gene up-regulation patterns may emphasize previously unrecognized molecules<br />

<strong>and</strong> <strong>signaling</strong> pathways enc<strong>our</strong>aging the appropriate protein <strong>and</strong> functional assays in an<br />

individual patient but also in larger patient cohorts.<br />

- 238 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 53<br />

Modulation of MPF <strong>and</strong> MAPK pathways activities by physiological pHi changes.<br />

Chantal Sellier1, Anne F Antoine1, Stéphane Flament2, Arlette Lescuyer1, Jean-Pierre<br />

Vilain1 <strong>and</strong> Jean-François Bodart1.<br />

1 - Laboratoire de Régulation des signaux de division, EA 1033, IFR 118, UTSL, SN3, F-<br />

59655 Villeneuve d’Ascq cedex, France. E-mail : chantal_sellier@yahoo.fr ; Anne-<br />

Frederique.Antoine@univ-lille1.fr ; Arlette.Lescuyer@univ-lille1.fr ; Jean-<br />

Pierre.Vilain@univ-lille1.fr ; Jean-François.Bodart@univ-lille1.fr<br />

2 - Faculté des Sciences, EA 3442, UHP, Bvd des Aiguillettes, BP 239, F-54506<br />

V<strong>and</strong>oeuvre Lès Nancy cedex, France. E-mail : stephane.flament@scbiol.uhp-nancy.fr<br />

Intracellular pH (pHi) plays a key role in mitogenic signal transduction, development <strong>and</strong><br />

maintenance of transformed phenotypes. Nevertheless, cell cycle mechanisms sensitive to pHi<br />

remain to be determined. Analogous to G2/M transition, Xenopus oocyte maturation is<br />

characterized by germinal vesicle breakdown (GVBD), MPF (Cyclin B/Cdk1) <strong>and</strong> Mos-<br />

MAPK pathways activation. These events are accompanied by a transient alkalization. We<br />

determined that an acidifying compound, NH4Cl, delayed progesterone-induced GVBD in a<br />

dose-dependent manner. Cyclin B phosphorylation, Cdk1 Y-15 dephosphorylation as well as<br />

p39Mos accumulation, MAPK <strong>and</strong> p90Rsk phosphorylations induced by progesterone were<br />

delayed by acidification. The delay induced by NH4Cl was prevented by injection of MOPS<br />

buffer pH 7.7. In contrast, alkalyzing treatment such as Tris buffer pH 9 injections,<br />

accelerated GVBD, MPF <strong>and</strong> MAPK activation. Strickingly, we observed that NH4Cl<br />

strongly inhibited thiophosphorylated active MAPK-induced GVBD <strong>and</strong> MPF activation<br />

while it had no or little effect on the MPF autoamplification loop <strong>and</strong> GVBD triggered by egg<br />

cytoplasm injection. Nevertheless, Tris pH 9 did not have any effects on egg cytoplasm- or<br />

active MAPK-induced GVBD. Thus, dynamic of early events driving MAPK <strong>and</strong> MPF<br />

activation induced by progesterone may be negatively or positively regulated by pHi changes<br />

whereas only MAPK-induced GVBD pathway was inhibited by acidification. Finally, using<br />

electrophysiological approch, we observed that NH4Cl induced an activation of Ca2+activated<br />

Cl current suggesting an elevation of intracellular Ca2+ concentration. Therefore,<br />

effects of pHi on early meiosis dynamic might be related to subsequent change in Ca2+<br />

homeostasis.<br />

- 239 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 54<br />

Protective effect of a peptide derived from the endogenous PKC inhibitor PKI55 on the<br />

neurosecretory function in ischemic brain slices<br />

Rita Selvatici1, Sofia Falzarano1, Lara Franceschetti1, Remo Guerrini2, Silvia Marino3<br />

<strong>and</strong> Anna Siniscalchi3<br />

1Department of Experimental <strong>and</strong> Diagnostic Medicine, Section of Medical Genetics;<br />

2Department of Pharmaceutical Sciences, 3Department of Clinical <strong>and</strong> Experimental<br />

Medicine, Section of Pharmacology, University of Ferrara, Italy. E-mail: svr@unife.it<br />

We have recently identified the PKI55 protein, coding for 55 amino acids, that is normally<br />

poorly translated in vivo <strong>and</strong> acts as a specific modulator of either cPKC-" <strong>and</strong> nPKC-&<br />

isozymes. PKI55 remains relatively inactive until PKC attains an active conformation <strong>and</strong><br />

reaches a critical concentration in the cell; it is not modified by the enzyme but irreversibly<br />

associates with its target, thus behaving as a suicidal inhibitor. The inhibition <strong>and</strong> degradation<br />

of over-activated PKC isozymes may prevent unfav<strong>our</strong>able changes in cellular phenotypes,<br />

resulting from PKC overexpression (Selvatici, J Mol Evol 2003 57:131). In the present work<br />

we compared the in vitro biochemical activity of the PKI55 protein, of its 39-amino acids Nterminal<br />

fragment (G39) <strong>and</strong> of its 16 amino acids C-terminal fragment (G16) on specific<br />

PKC recombinant isozymes, by measuring the initial rate of phosphate incorporation from<br />

32P-ATP into saturating amounts of histone IIIS. PKI55 concentration-dependently inhibited<br />

PKC activity, provided that calcium ions were present in the assay medium (IC50=6µM). The<br />

inhibitory activity was retained by G16 (IC50=50µM), but not by G39. The active fragment<br />

G16 was tested in an in vitro model of brain ischemia: superfused guinea pig cerebral cortex<br />

slices, continuously electrically (10 Hz) stimulated, were exposed to 20 min of oxygenglucose<br />

deprivation (OGD, Badini, Neurochem Int 1997 31:817), <strong>and</strong> then reperfused for 1<br />

h<strong>our</strong> (REP). PKC activity was increased to 244±36% at the end of OGD, while acetylcholine<br />

(ACh) release, taken as an index of the neurosecretory function, was reduced to 35±2% of the<br />

controls. Following REP, a reduction in PKC activity to 54±7% was displayed, indicating a<br />

down regulation of previously activated PKC (Selvatici, J Neurosci Res 2003 71:64); ACh<br />

release only partially recovered (REP=69±6% of the controls), suggesting persistence of<br />

neuronal suffering. PKC activation during OGD was attenuated by 50µM G16 <strong>and</strong> the<br />

consequent down regulation following REP was prevented. Moreover, ACh release fully<br />

recovered to normal values (REP+G16=91±4% of the controls). These data suggest a<br />

neuroprotective action for G16, the active fragment of the endogenous PKC inhibitor PKI55.<br />

- 240 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 55<br />

Effects of chemical ischemia in cerebral cortex slices. Focus on mitogen activated<br />

protein kinase cascade.<br />

Anna Siniscalchi1, Sabrina Cavallini1, Silvia Marino1 Sofia Falzarano2, Lara<br />

Franceschetti2,<br />

<strong>and</strong> Rita Selvatici2<br />

1Department of Clinical <strong>and</strong> Experimental Medicine, Section of Pharmacology,<br />

2Department of Experimental <strong>and</strong> Diagnostic Medicine, Section of Medical Genetics;<br />

University of Ferrara, Italy. E-mail: snn@unife.it<br />

Mitochondrial toxins could model, in vitro, the energy failure that occurs during transient<br />

brain ischemia in vivo, in alternative to either oxygen <strong>and</strong> glucose deprivation, or to high<br />

glutamate treatment. In <strong>our</strong> laboratory, sodium azide (NaN3), combined with the glycolysis<br />

blocker, 2-deoxyglucose (2-DOG), has been used to induce chemical ischemia (CI) in rat<br />

cerebral cortex slices (Cavallini, Neurochem Int 2005, 47:482), <strong>and</strong> the involvement of<br />

glutamate overflow, calcium overload <strong>and</strong> nitric oxide efflux in its mechanism of action has<br />

been shown. In the present work, the downstream events following CI have been addressed:<br />

superfused rat cerebral cortex slices, continuously electrically (5 Hz) stimulated, were treated<br />

with 10 mM NaN3 plus 2 mM 2-DOG for 5 min, then reperfused with normal medium for<br />

one h<strong>our</strong> (REP). For Ras/MAP kinases analysis by Western blotting, membranes were<br />

incubated with rabbit polyclonal antibodies against p21Ras, ERK1/2 (p44/42), phospho-<br />

ERK1/2, p38, phospho-p38, SAPK/JNK <strong>and</strong> phospho-SAPK/JNK. Densitometric analysis of<br />

autoradiographic b<strong>and</strong>s was performed with a Bio-Rad densitometer. The level of p21 Ras<br />

was increased by 40 % immediately after CI, <strong>and</strong> did not recover to control values following<br />

REP. The total levels of both ERK1 <strong>and</strong> ERK2 were reduced by CI <strong>and</strong> partially recovered to<br />

control values in REP; their phosphorylation degree (phosphorylated to total protein level<br />

ratio, about 50 % in the controls) did not significantly change either under CI or under REP<br />

conditions. The phosphorylation degree of MAPK p38, very low in control slices (17%), was<br />

not modified by either CI or REP. The activation of SAPK/JNK was full in the controls <strong>and</strong><br />

was reduced to 70% after CI <strong>and</strong> to 64% following REP. All these effects were prevented by<br />

the NMDA receptor antagonist MK801, 10 µM, suggesting the involvement of glutamate.<br />

The present findings show that CI reduces the MAPK levels, possibly because of acute energy<br />

depletion, although the p21Ras protein is increased. The decrease in SAPK/JNK, observed<br />

under CI/REP conditions, may lead to necrotic neuronal death, since its activation has been<br />

related to both apoptosis <strong>and</strong> neuroprotection mechanisms. These results could be of interest<br />

in view of preventive treatments of ischemia/reperfusion-induced brain damages.<br />

- 241 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 56<br />

Hantavirus infection of lung microvascular endothelial cells activates the extracellular<br />

regulated kinase 1 /2 (ERK1/2) <strong>and</strong> phosphoinositol3-kinase (PI3K) pathways leading to<br />

proinflammatory cytokine/chemokine production.<br />

Christina F. Spiropoulou, Karen L. Hutchinson, Cesar Albarino, Thomas G. Ksiazek<br />

<strong>and</strong> Pierre E. Rollin<br />

Special Pathogens Branch, Division of Viral <strong>and</strong> Rickettsial Diseases, Centers for<br />

Disease Control <strong>and</strong> Prevention, Atlanta, GA 30333, USA. E-mail:ccs8@cdc.gov<br />

Hantavirus pulmonary syndrome (HPS), is a severe respiratory illness with approximately<br />

40% mortality which is caused by New World hantaviruses. In humans, the primary target<br />

organ for virus infection is the lung, <strong>and</strong> severe disease symptoms have been attributed to<br />

massive lung endothelial cell capillary leakage. Immunopathology studies have shown the<br />

presence of monocytic <strong>and</strong> lymphocytic cell infiltrates in the microvasculature of patients but<br />

no evidence of endothelial cell disruption. Hantavirus infection of primary microvascular<br />

endothelial cells in vitro also does not cause cell disruption but induces the upregulation of a<br />

number of proinflammatory chemokines. These observations have led to the hypothesis that<br />

host immune responses play a crucial role in the pathogenesis of hantavirus diseases. This<br />

study was designed to examine pathways participating in the induction of proinflammatory<br />

chemokines by hantavirus infection. Hantavirus entry <strong>and</strong> replication activates the<br />

extracellular regulated kinase 1 /2 (ERK1/2) <strong>and</strong> phosphoinositol3-kinase (PI3K) pathways.<br />

By using specific pharmacological inhibitors we have shown that both pathways contribute to<br />

the induction of proinflammatory cytokines/chemokines by hantaviruses. However, we<br />

observed a partial inhibition using inhibitors specific for the ERK1/2 pathway whereas total<br />

inhibition of virus induced cytokines <strong>and</strong> chemokines was seen with LY294002, an inhibitor<br />

of PI3K. Studies leading to discovery of possible pharmacotherapeutic interventions for HPS<br />

are important given the lack of effective hantavirus vaccines.<br />

- 242 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 57<br />

Extracellular pH changes activate the p38-MAPK signalling pathway in the vertebrate<br />

heart<br />

Konstantina Stathopoulou, Catherine Gaitanaki <strong>and</strong> Isidoros Beis<br />

Department of Animal <strong>and</strong> Human Physiology, School of Biology, Faculty of Sciences,<br />

University of Athens, Panepistimioupolis, Athens, 157 84, GREECE. E-mail:<br />

kstath@biol.uoa.gr, cgaitan@biol.uoa.gr, ibeis@biol.uoa.gr<br />

The acid-base balance is one of the most important physiological parameters that affect the<br />

proper cellular function. In the case of heart, ischemia is closely related to acidosis, whereas<br />

growth factors <strong>and</strong> peptide hormones induce alkalinization. In the present study we<br />

investigated the effect of extracellular pH changes on the activation of the p38-MAPK<br />

signalling pathway in the isolated perfused amphibian heart. Alkalosis (pH 8.5 or 9.5)<br />

maximally activated p38-MAPK within 2 min (4.17- <strong>and</strong> 3.20-fold, compared to control<br />

values, respectively) <strong>and</strong> this effect was reversible since the kinase phosphorylation levels<br />

decreased upon heart reperfusion with normal Tris-Tyrode’s buffer. On the contrary, acidosis<br />

activated p38-MAPK moderately, but persistently (1.65-fold, at 1 min <strong>and</strong> 1.91-fold, at 60<br />

min). Na+/H+ exchanger inhibitors amiloride <strong>and</strong> HOE642 (a kind gift from Aventis Pharma<br />

Deutschl<strong>and</strong> GmbH) abolished p38-MAPK phosphorylation induced by alkalosis, whereas the<br />

Na+/K+-ATPase inhibitor ouabain partially attenuated this activation, results indicating that<br />

this signalling pathway depends on these cation exchangers. What is more, alkalosis (pH 8.5)<br />

induced a significant MAPKAPK2 (2.59-fold, 2 min) <strong>and</strong> HSP27 phosphorylation (5.33-fold,<br />

2 min) in a p38-MAPK-dependent manner, as demonstrated with experiments using 1 µM<br />

SB203580. Furthermore, the phosphorylated forms of p38-MAPK <strong>and</strong> HSP27 were<br />

immunohistochemically detected in the perinuclear region <strong>and</strong> dispersedly in the cytoplasm of<br />

ventricular cells during alkalosis. All the above data suggest that the p38-MAPK signalling<br />

pathway is activated by extracellular pH changes <strong>and</strong> in the case of alkalosis this activation<br />

seems to function protectively in the amphibian heart.<br />

Acknowledgements: This study was funded by the Special Research Account of the<br />

University of Athens <strong>and</strong> the Pythagoras I grant (70/3/7399). Ms Stathopoulou is a recipient<br />

of a State Scholarships Foundation fellowship.<br />

- 243 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 58<br />

The MEKK1-JNK Pathway Regulates Mouse Eyelid Morphogenesis<br />

Atsushi Takatori <strong>and</strong> Ying Xia<br />

Department of Environmental Health, University of Cincinnati Medical Center,<br />

Cincinnati, Ohio 45267 U.S.A<br />

MEK kinase 1 (MEKK1) is a mitogen-activated protein kinase kinase kinase, known as an<br />

upstream regulator for the c-Jun NH2-terminal kinases (JNKs). Knocking out Mekk1 in mice<br />

results in eye-open at birth (EOB) phenotype that leads to severe eye pathologies. There are<br />

two Jnk isoforms, Jnk1 <strong>and</strong> Jnk2, that are ubiquitously expressed in various tissues. This<br />

study is aimed at underst<strong>and</strong>ing the distinct role for JNK1 <strong>and</strong> JNK2 in transmitting the<br />

MEKK1 signals during eyelid morphogenesis. Mekk1-/-, Jnk1-/- <strong>and</strong> Jnk2-/- mice were<br />

backcrossed to C57BL/6 background <strong>and</strong> intercrosses were carried out to generate Mekk1+/-<br />

Jnk1-/-<strong>and</strong> Mekk1+/-Jnk2-/- mice. We find that the Mekk1+/-Jnk1-/-, but not Mekk1+/-<br />

Jnk2-/-, mice displayed EOB similar to the Mekk1-/- mice. Unlike the Mekk1+/-Jnk2-/-<br />

fetuses, which showed a thin layer of eyelid epithelium covering the ocular surface at<br />

embryonic day 16.5, the Mekk1+/-Jnk1-/- fetuses lacked eyelid closure <strong>and</strong> had fully exposed<br />

cornea. Immunohistochemistry studies showed similar levels of JNK phosphorylation, but a<br />

much reduced c-Jun phosphorylation <strong>and</strong> plasminogen activator inhibitor-1 (PAI-1)<br />

expression in the developing eyelid epithelium of the Mekk1+/-Jnk1-/-, compared to that of<br />

the wild type <strong>and</strong> the Mekk1+/-Jnk2-/- fetuses. Considering the crucial role of PAI-1 in<br />

epithelial cell migration, we studied the keratinocyte migration using an in vitro wound<br />

healing assay. Our results indicated that the Mekk1+/-Jnk1-/- keratinocytes had slower<br />

wound closure than the wild type <strong>and</strong> Mekk1+/-Jnk2-/- keratinocytes. Taken together, <strong>our</strong><br />

results show that in the absence of JNK1, one functional Mekk1 allele is insufficient to cause<br />

c-Jun phosphorylation <strong>and</strong> epithelial cell movement, responsible for defective eyelid<br />

morphogenesis of the Mekk1+/-Jnk1-/- fetuses. JNK1 appears to be more critical than JNK2<br />

in transmitting the MEKK1 signals to c-Jun phosphorylation <strong>and</strong> target gene expression<br />

during mouse embryonic eyelid development.<br />

Support: NIH EY 15227<br />

- 244 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 59<br />

Erucylphospho-N,N,N-trimethylpropylammonium (ErPC3) acts through redistribution<br />

of protein kinases within lipid rafts <strong>and</strong> signal transduction changes in leukemic cells<br />

Iana Tsoneva1, Spiro M. Konstantinov2, Hansjörg Eibl3, Martin R. Berger4<br />

1,4Inst. of Biophysics, BAS, Acad. G. Bonchev St., bl 21, 1113 Sofia, Bulgaria e-mail:<br />

Itsoneva@obzor.bio21.bas.bg<br />

2,4Lab for Experimental Chemotherapy, Dept. of Pharmacology, Medical University of<br />

Sofia, 2 Dunav St., 1000 Sofia, Bulgaria; e-mail: Pgen@TU-Sofia.bg<br />

4 !Unit of Toxicology <strong>and</strong> Chemotherapy, German Cancer Research Center, INF 280,<br />

69120 Heidelberg, Germany; e-mail: M.Berger@DKFZ.de<br />

3Max Planck Inst. of Biophysical Chemistry, Am Faßberg, 37077, Göttingen, Germany<br />

ErPC3 belongs to the group of alkylphosphocholines which are membrane targeting<br />

antineoplastic agents <strong>and</strong> act as signal transduction modulators. Our hypothesis was that<br />

ErPC3 can modulate the protein content of lipid rafts. The non receptor tyrosine kinase c-Abl<br />

is widely expressed in malignant hematopoietic cells <strong>and</strong> its oncogenic variant Bcr-Abl has<br />

enough leukemogenic potential for the development of acute lymphoblastic <strong>and</strong> chronic<br />

myeloid leukemia. Therefore, the panel of cell lines included two groups: cells with bcr-abl<br />

rearrangement (K-562 <strong>and</strong> LAMA-84) <strong>and</strong> cells with c-Abl only (HL-60 <strong>and</strong> SKW-3). Whole<br />

cell lysates were analyzed by Western blotting. Lipid rafts were isolated from membranes by<br />

sucrose gradient centrifugation <strong>and</strong> analyzed for cAbl (p160) <strong>and</strong> BCR-ABL (p210). Whole<br />

cells mounted by cytospin were stained for the ganglioside GM1 content by cholera toxin B<br />

subunit conjugated to FITC. ErPC3 elicited changes in Rb protein status that included<br />

increased expression <strong>and</strong> fragmentation. Furthermore, the alkylphosphocholine caused time<br />

dependently increased association of cAbl <strong>and</strong> BCR-ABL proteins to lipid rafts. This process<br />

was accompanied by reorganisation of lipid rafts containing ganglioside GM1. When<br />

comparing ErPC3 with cytosine arabinoside, the antimetabolite did not induce significant<br />

changes in the cAbl content of the lipid rafts. The mechanism of action of ErPC3 is related to<br />

its effects on lipid rafts as well as on Rb activation <strong>and</strong> induction. A deeper underst<strong>and</strong>ing of<br />

molecular mechanisms especially those related to modulation of signal molecule content in<br />

membrane lipid rafts will contribute to better evaluation <strong>and</strong> selection of anticancer lysolipids<br />

such as ErPC3.<br />

- 245 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 60<br />

Molecular mechanisms of the deleterious action of ceramide on insulin signalling:<br />

involvement of caveolae<br />

Sophie Turban1, Xavier Lelièpvre2, Soazig Le Lay3, Olivier B<strong>our</strong>ron2, Harinder S.<br />

Hundal1 & Eric Hajduch2.<br />

1Department of Molecular Physiology, Dundee, UK, e-mail: s.turban@dundee.ac.uk,<br />

2Inserm U671, Paris, France, 3Max Planck Institute, Dresden, Germany<br />

Several studies have revealed that in peripheral tissues, insulin resistance might be associated<br />

with intracellular formation of ceramide, the main secondary messenger of the sphingomyelin<br />

transmembrane signalling pathway. We have recently shown that ceramide inhibited insulininduced<br />

activation of protein kinase B (PKB), a key protein in insulin signalling, by<br />

promoting its association <strong>and</strong> phosphorylation by protein kinase Cz (PKCz).<br />

Caveolae are small invaginations of the plasma membrane that are enriched in cholesterol <strong>and</strong><br />

sphingolipids like ceramide. Interestingly, many signalling proteins such as PKCz have been<br />

found in those microdomains. The purpose of the study was to investigate the mechanisms<br />

involved in the negative effect of ceramide on the insulin pathway <strong>and</strong> to determinate whether<br />

caveolae would be the place where the lipid inhibits PKB via the activation of PKCz. We<br />

show that ceramide induces the recruitment of PKCz in the caveolin rich domain. Moreover<br />

the concomitant recruitment of PKB is observed in those compartments. We report that<br />

cholesterol depletion <strong>and</strong> destruction of caveolae structures in 3T3-L1 adipocytes, L6 muscle<br />

cells <strong>and</strong> isolated human adipocytes with methyl-beta-cyclodextrin prevents the deleterious<br />

effect of ceramide on the insulin-induced translocation <strong>and</strong> activation of PKB in the plasma<br />

membrane. In addition we demonstrate that in isolated adipocytes from caveolin-1 knock-out<br />

mice, ceramide did not alter the insulin-induced phosphorylation of PKB. All together these<br />

findings indicate that in insulin sensitive tissues, ceramide inhibit PKB via PKCz within<br />

caveolae.<br />

- 246 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 61<br />

Involvement of NF!B activating pathways in DLBCL pathogenesis<br />

Joost C. van Galen1, Bauke Ylstra1, Chris J.L.M. Meijer1, Jettie J.F. Muris1, Cindy<br />

P.E, Giroth1, Gert J. Ossenkoppele2, Joost J. Oudejans1.<br />

Departments of Pathology1 <strong>and</strong> Hematology2, VU university medical center, De<br />

Boelelaan 1117, 1081 HV Amsterdam, the Netherl<strong>and</strong>s. E-mail: j.vangalen@vumc.nl.<br />

Intrinsic resistance to apoptosis of lymphoma cells is a probable mechanism causing<br />

chemotherapy resistance <strong>and</strong> eventual fatal outcome in patients with diffuse large B cell<br />

lymphomas (DLBCL). Microarray studies have demonstrated that DLBCL can be classified<br />

in activated B-cell (ABC) like or germinal center B-cell (GCB) like DLBCL. Clinical<br />

outcome in ABC like DLBCL is worse than for GCB like DLBCL.<br />

Poor outcome in ABC like DLBCL is explained by constitutive activation of the NF!B<br />

pathway resulting in high expression levels of many apoptosis inhibitory genes. The<br />

mechanism behind this NF!B activation however is unknown. In this study we determined<br />

expression profiles of possible pathways leading to NF!B activation in these DLBCL.<br />

We investigated whether an ABC like phenotype correlates with expression of genes involved<br />

in NFkB activation.<br />

Using dual channel oligo arrays we performed genome-wide microarray analysis in a group of<br />

46 primary nodal DLBCL to determine GCB versus ABC like phenotype. Next we performed<br />

hierarchical cluster analysis of only genes coding for intracellular signal transduction factors<br />

involved in NF!B activation. Both profiles were compared to each other.<br />

We found that in ABC like DLBCL expression of NFkB target genes correlated with the<br />

expression of genes upstream from NFkB activation that are implicated in B-cell receptor<br />

(BCR) <strong>signaling</strong>. No correlation was observed with activation of pathways downstream of the<br />

TNF receptor family.<br />

We conclude that NF!B activation in ABC DLBCL is correlated with constitutive BCR<br />

receptor <strong>signaling</strong>. Thus interfering with the function or expression of BCR target genes<br />

might provide new possibilities to restore apoptosis sensitivity in ABC like DLBCL cells <strong>and</strong><br />

thus improve clinical outcome in these patients.<br />

- 247 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 62<br />

Reciprocal Relationship Between RhoC GTPase <strong>and</strong> Caveolin-1 Leads to Altered<br />

MAPK Signaling During Pancreatic Cancer Cell Migration <strong>and</strong> Invasion.<br />

Cynthia M. van Golen <strong>and</strong> Kenneth L. van Golen.<br />

Department of Internal Medicine, Division of Hematology/Oncology, The University of<br />

Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109 email:<br />

kgolen@umich.edu<br />

BACKGROUND: In the current study we investigated the role of caveolin-1 (cav-1) in<br />

pancreatic adenocarcinoma (PC) cell migration <strong>and</strong> invasion; initial steps in metastasis. Cav-<br />

1 is the major structural protein in caveolae; small *-shaped invaginations within the plasma<br />

membrane. Caveolae are involved in signal transduction, wherein cav-1 acts as a scaffolding<br />

protein to organize multiple molecular complexes regulating a variety of cellular events.<br />

Recent evidence suggests a role for cav-1 in promoting cancer cell migration, invasion <strong>and</strong><br />

metastasis; however, the molecular mechanisms have not been described. The small<br />

monomeric GTPases are among several molecules which associate with cav-1. Classically,<br />

the Rho GTPases control actin cytoskeletal reorganization during cell migration <strong>and</strong> invasion.<br />

RhoC GTPase is overexpressed in aggressive cancers that metastasize <strong>and</strong> is the predominant<br />

GTPase in PC. Like several GTPases, RhoC contains a putative cav-1 binding motif.<br />

RESULTS: Analysis of 10 PC cell lines revealed high levels of cav-1 expression in lines<br />

derived from primary tumors <strong>and</strong> low expression in those derived from metastases.<br />

Comparison of the BxPC-3 (derived from a primary tumor) <strong>and</strong> HPAF-II (derived from a<br />

metastasis) demonstrates a reciprocal relationship between cav-1 expression <strong>and</strong> p42/p44 Erk<br />

activation with PC cell migration, invasion, RhoC GTPase <strong>and</strong> p38 MAPK activation.<br />

Furthermore, inhibition of RhoC or p38 activity in HPAF-II cells leads to partial restoration<br />

of cav-1 expression. CONCLUSIONS: Cav-1 expression inhibits RhoC GTPase activation<br />

<strong>and</strong> subsequent activation of the p38 MAPK pathway in primary PC cells thus restricting<br />

migration <strong>and</strong> invasion. In contrast, loss of cav-1 expression leads to RhoC-mediated<br />

migration <strong>and</strong> invasion in metastatic PC cells.<br />

- 248 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 63<br />

Functional Genomics of Coronavirus-Host Interactions using a High-Throughput<br />

siRNA Strategy<br />

Hélène Verheije1, Matthijs Raaben1, Lucas Pelkmans2, Eberhard Krausz3,4, Eugenio<br />

Fava3,4, Peter J.M. Rottier1, Cornelis A.M. de Haan1<br />

1Virology Division, Department of Infectious Diseases & Immunology, Faculty of<br />

Veterinary Medicine, Utrecht University, The Netherl<strong>and</strong>s; 2EHT Zurich, Institute for<br />

Molecular Systems Biology, Zürich, Switzerl<strong>and</strong>; 3Max Planck Institute for Molecular<br />

Cell Biology <strong>and</strong> Genetics, Dresden, Germany; 4MPI-CBG High-Throughput<br />

Technology Development Studio (HT-TDS), Dresden, Germany. E-mail:<br />

h.verheije@vet.uu.nl<br />

Viruses replicate within cells by using the cellular machinery to their own advantage. In<br />

general much is known about the pathogens, however, only little is known about the<br />

contributions of the host. Our research aims to analyse coronavirus-host interactions by using<br />

a functional genomics screen, based on a recently developed high throughput genome-wide<br />

RNAi strategy (Pelkmans et al. 2005). The screen is designed to identify genes whose direct<br />

loss-of-function phenotypes result in altered viral entry <strong>and</strong> replication. As a proof of<br />

principle 54 individual kinases, known to be involved in endocytosis, were analyzed for their<br />

role in coronavirus entry. The extensively characterized mouse hepatitis virus (MHV), a close<br />

relative of the severe acute respiratory syndrome (SARS) virus, was used for the initial<br />

screen. HeLa cells were transfected with the different siRNAs <strong>and</strong> 72h after transfection the<br />

cells were infected with MHV at a low multiplicity of infection. MHV infection was<br />

monitored by analyzing reporter gene expression at an early time point post infection.<br />

Downregulation of several kinases resulted in inhibition of infection, suggesting a role for<br />

these kinases in MHV internalization. Also an increase in infection efficiency was found for<br />

some kinases. In a follow-up study, a genome-wide kinase <strong>and</strong> phosphatase siRNA library,<br />

targeting 750 kinases <strong>and</strong> 180 phosphatases, will be used to identify more host genes whose<br />

loss-of-function results in impaired entry <strong>and</strong> replication. This might provide interesting leads<br />

for the development of anti-coronaviral agents.<br />

- 249 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 64<br />

PDGFRbetaY857F mutant mediates PDGF-BB-induced signalling <strong>and</strong> chemotaxis,<br />

despite severely reduced kinase activity.<br />

Piotr Wardega, Carl-Henrik Heldin, Johan Lennartsson.<br />

Ludwig Institute for Cancer Research, Uppsala University, Box 595, SE-751 24 Uppsala,<br />

Sweden<br />

Piotr.Wardega@LICR.uu.se , C-H.Heldin@LICR.uu.se,<br />

Johan.Lennartsson@LICR.uu.se<br />

Platelet-derived growth factor receptor (PDGFR) becomes dimerized <strong>and</strong> activated upon<br />

lig<strong>and</strong> binding, which leads to receptor transphosphorylation <strong>and</strong> increases its enzymatic<br />

activity. In the present study, we studied the effects of mutation of tyrosine residue 857 in the<br />

activation loop of PDGFRbeta, to phenylalanine. In agreement with published work, <strong>our</strong><br />

studies show, that PDGFRbetaY857F has much lower kinase activity in vitro in comparison<br />

to PDGFRbetaWT , both as measured as receptor autophosphorylation <strong>and</strong> as phosphorylation<br />

of an exogenous substrate. Interestingly, PDGF-induced tyrosine phosphorylation of<br />

PDGFRbeta, in a cellular context, is not affected by the Tyr857 mutation. There are, however<br />

significant changes in ability of PDGFRbetaY857F to induce signal transduction compared to<br />

the wild-type receptor. For example, we could show that Stat5 signalling differs dramatically<br />

between PDGFRbetaWT <strong>and</strong> PDGFRbetaY857F. We observed almost complete loss of<br />

PDGF-induced Stat5 phosphorylation by the mutant receptor. Surprisingly Stat3, which<br />

belongs to the same group of transcription factors as Stat5, was phosphorylated to the same<br />

extent by mutant <strong>and</strong> wild-type receptor. In addition, we have also found that the Akt <strong>and</strong><br />

JNK signalling pathways are not affected by the PDGFRbetaY857F mutation. Moreover, the<br />

Erk pathway was activated by slower kinetics in the mutant receptor. Interestingly, in spite of<br />

the defect in the PDGFRbetaY857F receptors kinase activity <strong>and</strong> the changes observed in<br />

signal transudction it could still efficiently induce cell chemotaxis. It is known that many<br />

cytoplasmic tyrosine kinases are capable of phosphorylating the PDGFR <strong>and</strong> it is possible that<br />

these may compensate for the lowered enzymatic activity of PDGFRbetaY857F . In summary,<br />

we have demonstrated that the PDGFRbetaY857F has defect kinase activity, but is<br />

nevertheless phosphorylated in the cell, presumably by cytoplasmic kinases <strong>and</strong> can induce<br />

signals sufficient for normal chemotaxis toward PDGF-BB.<br />

- 250 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 65<br />

Leukocyte mitogen activated protein kinase cascade activity associated with the LRRK2<br />

G2019S mutation <strong>and</strong> Parkinson's disease<br />

Linda R. White1, Sylvia N. Kvam2, Mathias Toft1,3, Matthew Farrer3, Jan O. Aasly2<br />

1Department of Neuroscience, Norwegian University of Science <strong>and</strong> Technology,<br />

2Department of Neurology, University Hospital of Trondheim, N-7006 Trondheim,<br />

Norway, <strong>and</strong> 3Department of Neuroscience, Mayo Clinic College of Medicine,<br />

Jacksonville, Florida, USA. E-mail: linda.white@ntnu.no<br />

Several pathogenic mutations in the leucine-rich repeat kinase 2 (LRRK2, Park8) gene have<br />

now been identified in connection with familial <strong>and</strong> sporadic parkinsonism. Of these, the<br />

substitution 6055G>A is the hitherto most common cause of genetically-related Parkinson's<br />

disease (PD) to be found, accounting for over 2 % of PD patients in Norway (Aasly et al.,<br />

Ann Neurol 2005;57:762-5). The mutation is autosomal dominant, the clinical features<br />

including asymmetric resting tremor, bradykinesia <strong>and</strong> rigidity, with a good response to<br />

levodopa treatment. It is thus indistinguishable from idiopathic PD. LRRK2 is a large,<br />

multidomain protein, <strong>and</strong> the 6055G>A mutation results in a single amino acid substitution<br />

(G2019S) in the mitogen-activated protein kinase kinase kinase (MAPKKK) domain, at the<br />

start of the kinase activation site (a highly-conserved region). This is therefore expected to<br />

affect enzyme activity. Since PD develops in the later decades of life, it is likely that changes<br />

in MAPK cascades attributable to the mutation might be identifiable prior to disease<br />

development, <strong>and</strong> in tissues other than the brain's dopaminergic system. We have therefore<br />

isolated leukocytes from the blood of Norwegian patients bearing the Park8 G2019S mutation<br />

<strong>and</strong> diagnosed as suffering from PD, as well as from patients identified as mutation carriers,<br />

but who have not yet developed any movement disorder, <strong>and</strong> from relatives identified as not<br />

carrying the mutation. These groups have been compared with a positive control group<br />

(patients with typical PD of no known cause, genetic or otherwise (idiopathic PD)), <strong>and</strong> age<br />

<strong>and</strong> sex-matched healthy controls. The phosphorylation of proteins central to MAPKmodulated<br />

signal transduction has been assayed in these samples by protein microarray <strong>and</strong><br />

ELISA, <strong>and</strong> highly significant differences between the groups identified.<br />

- 251 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 66<br />

Inhibition of gap junction intercellular communication by capsaicin in rat liver<br />

epithelial cells are mediated by phosphorylation of Cx43 <strong>and</strong> activation of ERK1/2<br />

through activation of NADPH oxidase<br />

Ji Hyuck Yim, Ki Won Lee, Jung Yeon Kwon, <strong>and</strong> Hyong Joo Lee*<br />

Department of Food Biotechnology, School of Agricultural Biotechnology, <strong>and</strong><br />

Center for Agricultural Biomaterials, Seoul National University, Seoul 151-742,<br />

Republic of Korea<br />

Many studies have focused on the antimutagenic or anticarcinogenic activities of capsaicin<br />

(trans-8-methyl-N-vanillyl-6-nonenamide) which is a major pungent ingredient in red pepper.<br />

Recently, several studies have shown that anticarcinogenic effects of capsaicin may be partly<br />

involved in the induction of apoptosis in several cancer cell lines. A recent study suggests that<br />

the mechanism that makes antiproliferative activity of capsaicin in a human hepatocarcinoma<br />

cells is due to their ability to produce hydrogen peroxide (H2O2) via activation of NADPH<br />

oxidase. However, it can be argued that such inappropriate H2O2 generation may also have a<br />

deleterious effect on nonmalignant cells as well. The present study investigated whether<br />

capsaicin generates H2O2 at the intracellular levels <strong>and</strong>, if so, whether the formation of H2O2<br />

by capsaicin induces carcinogenicity in normal rat liver epithelial cells. Capsaicin at the<br />

concentration of 50µM completely inhibited gap junction intercellular communication (GJIC;<br />

a carcinogenic phenomenon) in WB-F344 normal rat liver epithelial cells (WB cells) in a<br />

dose-dependant manner. The inhibition of GJIC by capsaicin was mediated by<br />

hyperphosphorylation of connexin 43 (Cx43) <strong>and</strong> activation of extracellular-regulated kinase<br />

1/2 (ERK1/2) as demonstrated using pharmacological inhibitors of ERK1/2. Pretreatment of<br />

WB cells with catalase <strong>and</strong> inhibitors of NADPH oxidase significantly reversed capsaicininduced<br />

inhibition of GJIC <strong>and</strong> hyperphosphorylation of Cx43, suggesting that ROS may<br />

mediate the inhibition of GJIC induced by capsaicin. These results indicate that the<br />

antiproliferative effects of capsaicin on cancer cells are partially due to their prooxidant<br />

actions, thereby indicating their potential toxicity <strong>and</strong> carcinogenicity.<br />

- 252 -


Session III : Protein kinase cascades as therapeutic targets Poster III, 67<br />

Renal Akt kinase <strong>signaling</strong> in Zucker diabetic fatty rats (ZDF).<br />

Jana Zdychova1, Jessie N. Lindsley2, Sharon Anderson2, Radko Komers1,2.<br />

1Diabetes Center, Institute for Clinical <strong>and</strong> Experimental Medicine, 140 21 Prague 4,<br />

Czech Republic; 2Division of Nephrology <strong>and</strong> Hypertension, Oregon Health&Science<br />

University, 97239 Portl<strong>and</strong>, OR, USA. E-mail: rako@medicon.cz<br />

Impaired insulin <strong>signaling</strong> via PI-3-kinase (PI3K) <strong>and</strong> Akt in skeletal muscle, liver <strong>and</strong> fat has<br />

been implicated in the pathophysiology of insulin resistance (IR) including Type 2 diabetes<br />

(DM2). Kidney is also a target of insulin actions. However, unlike the skeletal muscle, renal<br />

Akt <strong>signaling</strong> in DM2 has been so far less studied. To address this issue, renal cortical activity<br />

<strong>and</strong> protein expression of Akt, its down-stream effector mTOR, <strong>and</strong> Akt antagonist PTEN<br />

were studied in 4 <strong>and</strong> 12-week old ZDF (ZDF4 <strong>and</strong> ZDF12), a model of DM2, <strong>and</strong> in agematched<br />

controls (Zucker lean, ZL4 <strong>and</strong> ZL12). Akt <strong>and</strong> mTOR activities were also studied in<br />

separate groups of 12 week old animals after a short-term treatment with PI3K inhibitor<br />

wortmannin (W, 100 µg/kg bwt) or vehicle (DMSO). Akt activity (in vitro kinase assay) <strong>and</strong><br />

expression of active Akt (P-Akt) were increased in ZDF12 as compared to ZL12 (p


Notes<br />

- 254 -


Notes<br />

Notes<br />

- 255 -


- 256 -


Session IV. Protein kinase inhibitors: insights into drug<br />

design from structure<br />

- 257 -


IV. Protein kinase inhibitors: insights into drug design from structure Poster IV, 1<br />

The mechanism of anti-proliferation by epidermal growth factor receptor tyrosine<br />

kinase inhibitor ZD1839 in human oral squamous cell carcinoma.<br />

Nam K. Jeon, Jin Kim <strong>and</strong> Eun J. Lee<br />

Department of Oral Pathology, Oral Cancer Research Institute, BK21 project for<br />

Medical Science, Yonsei University College of Dentistry, Seoul, 120752, KOREA Email:e16lee@yumc.yonsei.ac.kr<br />

Activation of the epidermal growth factor receptor (EGFR) <strong>and</strong> downstream <strong>signaling</strong><br />

pathways have been implicated in causing resistance to EGFR-targeted therapy in solid<br />

tumors, including the head <strong>and</strong> neck tumors. To investigate the mechanism of<br />

antiproliferation to EGFR inhibition in oral cancer, we compared EGFR tyrosine kinase<br />

inhibitor (Gefitinib, Iressa, ZD1839) with respect to its inhibitory effects on three kinases<br />

situated downstream of EGFR: MAPK, Akt, <strong>and</strong> glycogen synthase kinase-3ß (GSK-3ß). We<br />

have demonstrated that ZD1839 induces growth arrest in oral cancer cell lines by independent<br />

of EGFR-mediated <strong>signaling</strong>. Cell cycle kinetic analysis demonstrated that ZD1839 induces a<br />

delay in cell cycle progression <strong>and</strong> a G1 arrest; this was associated with increased expression<br />

of both p27KIP1 <strong>and</strong> p21CIP/WAF1 proteins. We found that the resistance to the<br />

antiproliferative effects of gefitinib, in vitro was associated with uncoupling between EGFR<br />

<strong>and</strong> MAPK inhibition, <strong>and</strong> that GSK-3ß activation <strong>and</strong> degradation of its target cyclin D1<br />

were indicators of a high cell sensitivity to gefitinib. In conclusion, <strong>our</strong> data show that the<br />

uncoupling of EGFR with mitogenic pathways can cause resistance to EGFR inhibition in<br />

oral cancer. (This study was supported by the BK21 project for Medical Science, <strong>and</strong> by a<br />

grant of the national Cancer control R&D Program 2003(No. 0320230), Ministry of Health &<br />

Welfare, Republic of Korea.)<br />

- 258 -


IV. Protein kinase inhibitors: insights into drug design from structure Poster IV, 2<br />

Three dimensional structures of Pim-1 inhibitor complexes give new insights for the<br />

development of target specific kinase inhibitors<br />

Stefan Knapp, A.N. Bullock, J. É. Debreczeni, O. Y. Fedorov, B. D. Marsden. E.<br />

Meggers<br />

University of Oxford, Structural Genomics Consortium (SGC), Botnar Research Center,<br />

Oxford OX3 7LD, UK. E-mail: stefan.knapp@sgc.ox.ac.uk<br />

The Structural Genomics Consortium (SGC) is a not-for-profit organization that aims to<br />

determine the three dimensional structures of proteins of medical relevance, <strong>and</strong> place them in<br />

the public domain without restriction. Due to the important role in current drug development<br />

we solve 12 human kinases so far <strong>and</strong> selected human Pim-1 kinase for further follow up<br />

studies.<br />

The kinase Pim-1 plays a pivotal role in cytokine <strong>signaling</strong> <strong>and</strong> is implicated in the<br />

development of a number of tumors. The three dimensional structure of Pim-1 is<br />

characterized by unique structural features within the ATP binding pocket which makes it<br />

particularly important to determine how inhibitors bind to this kinase. We determined 6 high<br />

resolution crystal structures of Pim-1 in complex with a potent inhibitors of the<br />

bisindolylmaleimide class (BIM-1), a imidazo [1,2-b]pyridazine, three novel ruthenium half<br />

s<strong>and</strong>wich organometallic complexes as well as a complex with a high affinity substrate<br />

peptide.<br />

The determined structural data revealed a number of rather unexpected features of Pim-1<br />

inhibitor interaction. For example the imidazo[1,2-b]pyridazine inhibitor formed no hydrogen<br />

bond with the hinge region of Pim-1 as is usually observed for this inhibitor class <strong>and</strong> other<br />

kinase inhibitors. Instead the imidazo[1,2-b]pyridazine forms a bifurcated hydrogen bond<br />

with the active site lysine K67 <strong>and</strong> a water molecule that is coordinated by E89 <strong>and</strong> D186.<br />

Structures of ruthenium half s<strong>and</strong>wich organometallic complexes which inhibit Pim-1 with<br />

low pico molar affinity revealed that all three inhibitors fit tightly into the binding pocket with<br />

a shape complementarity that would not be possible to achieve with a conventional organic<br />

compound. The presented data suggest a number of strategies for the further development of<br />

potent <strong>and</strong> selective Pim-1 inhibitors which may find applications for the treatment of Pim-1<br />

dependent cancer types.<br />

- 259 -


IV. Protein kinase inhibitors: insights into drug design from structure Poster IV, 3<br />

The Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor ZD1839 (‘Iressa)<br />

Suppresses Proliferation <strong>and</strong> Invasion of Human Oral Squamous Carcinoma Cells via<br />

MMP, uPAR Dependent Mechanism<br />

Eun J. Lee, Jin H. Whang, Nam K. Jeon, Young E. Kwak <strong>and</strong> Jin Kim<br />

Department of Oral Pathology, Oral Cancer Research Institute, BK21 project for<br />

Medical Science, Yonsei University College of Dentistry, Seoul, 120-752, KOREA<br />

E-mail:e16lee@yumc.yonsei.ac.kr<br />

Oral squamous cell carcinomas (OSCCs) are characterized by a marked propensity for local<br />

invasion <strong>and</strong> dissemination to cervical lymph nodes. Overexpression of the epidermal growth<br />

factor receptor (EGFR) <strong>and</strong> high levels of certain matrix metalloproteinases (MMP) have<br />

been implicated in the development of squamous-cell carcinoma of oral cancer. This study<br />

attempted to determine the mechanisms underlying the effects of ZD1839 on the cellular<br />

level, <strong>and</strong> to characterize the effects of ZD1839 with regard to human OSCC cell growth <strong>and</strong><br />

invasion/migration. Cell cycle kinetic analysis demonstrated that ZD1839 induces a delay in<br />

cell cycle progression <strong>and</strong> a G1 arrest, which was associated with the up-regulation of cyclindependent<br />

kinase inhibitors (CDKI) p27KIP1 <strong>and</strong> p21CIP1/WAF1. The up-regulation of<br />

CDKI may be mediated by a p53-independent <strong>and</strong> hnRNPC1/C2-dependent pathway. In<br />

addition, 100nM ZD1839 demonstrated that both MMP-2 <strong>and</strong> MMP-9 enzyme activity were<br />

decreased by ~25-30%. The current in vitro study indicates that the inhibition of proliferation<br />

<strong>and</strong> invasion/migration in OSCC cell lines by ZD1839 results in an anticancer effect via<br />

multiple cellular <strong>and</strong> molecular mechanisms, <strong>and</strong> suggests that ZD1839 may be useful in<br />

inhibiting <strong>and</strong> /or preventing metastasis. (This study was supported by the BK21 project for<br />

Medical Science, <strong>and</strong> by a grant of the national Cancer control R&D Program (02-PJ1-PG3-<br />

20801-0015) <strong>and</strong> 2003(No. 0320230), Ministry of Health & Welfare, Republic of Korea.)<br />

- 260 -


IV. Protein kinase inhibitors: insights into drug design from structure Poster IV, 4<br />

Aberrant methylation of p15INK4B <strong>and</strong> p14ARF <strong>and</strong> deletion of p16INK4A are<br />

frequent events in liver fluke related cholangiocarcinoma<br />

Temduang Limpaiboon1,4, Preeda Prakrankamanant1, Inthira Tussakhon1, Patcharee<br />

Chinnasri1, Patcharee Jearanaikoon1,4, Chawalit Pairojkul2,4, Banchob Sripa2,4,<br />

Vajarabhongsa Bhudhisawasdi3,4<br />

1Department of Clinical Chemistry, Centre for Research <strong>and</strong> Development of Medical<br />

Diagnostic Laboratories, Faculty of Associated Medical Sciences, 2Department of<br />

Pathology, 3Department of Surgery, 4Liver Fluke <strong>and</strong> Cholangiocarcinoma Research<br />

Center, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thail<strong>and</strong>.<br />

E-mail:temduang@kku.acth<br />

Cholangiocarcinoma (CCA) is the highest incidence cancer in Northeast Thail<strong>and</strong>. CCA is<br />

caused by liver fluke (Opisthorchis viverrrini) infection resulting in both genetic <strong>and</strong><br />

epigenetic alterations. The INK4 locus located on chromosome 9p21 consisting of three<br />

genes, p14ARF, p16INK4A <strong>and</strong> p15INK4B, which are involved in cell cycle regulation <strong>and</strong><br />

reported to be inactivated in many human cancers through genetic <strong>and</strong> epigenetic events. To<br />

underst<strong>and</strong> mechanisms of inactivation of these three genes in CCA, we analyzed loss of<br />

heterozygosity (LOH) at chromosomal region 9p21 using three polymorphic microsatellite<br />

markers; D9S1752, D9S171 <strong>and</strong> D9S161 <strong>and</strong> determined promoter hypermethylation of<br />

p14ARF, p16INK4A <strong>and</strong> p15INK4B using methylation-specific PCR in 79 CCA. LOH was<br />

found at least one or more loci in 32 of 74 informative cases (43.2%) in which high frequency<br />

was found in D9S1752 (34.9%) <strong>and</strong> less frequency was observed in D9S171 (27.6%).<br />

Aberrant methylation was present predominantly in p15INK4B (50.6%) <strong>and</strong> p14ARF (41.2%)<br />

<strong>and</strong> less frequently in p16INK4A (25.3%). Our results suggest that promoter<br />

hypermethylation is a predominant inactivation mechanism of p15INK4B <strong>and</strong> p14ARF,<br />

whereas, deletion is a major event for inactivation of p16INK4A during CCA development.<br />

This finding may lead to a new approach of CCA treatment.<br />

- 261 -


IV. Protein kinase inhibitors: insights into drug design from structure Poster IV, 5<br />

Exploring the chemotherapeutic benefit of combining Gemcitabine (Gemzar) <strong>and</strong><br />

Rapamycin<br />

Ofer Merimsky1,2, Yaara Gorzalczany2 <strong>and</strong> Ronit Sagi-Eisenberg2<br />

1Department of Oncology, The Tel-Aviv S<strong>our</strong>asky Medical Center, Sackler School of<br />

Medicine, Tel Aviv University, Tel Aviv 69978 Israel <strong>and</strong> 2Dept. of Cell <strong>and</strong><br />

Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978<br />

Israel<br />

Two major obstacles encountered while treating cancer patients are achieving the right<br />

balance between the effectiveness of the drug <strong>and</strong> its cytotoxic side effects as well as<br />

overcoming mechanisms of existing or acquired resistance. The emerging solution appears to<br />

constitute a combination of a “traditional” cytotoxic drug with a “<strong>signaling</strong>” drug.<br />

Gemcitabine (Gemzar), is an antimetabolite drug that inhibits DNA synthesis <strong>and</strong> displays<br />

some activity as a single agent in treating pancreatic <strong>and</strong> sarcoma cancer. However,<br />

gemcitabine treatment is often associated with acquired resistance. Rapamycin is a selective<br />

inhibitor of mTOR, a phosphatidylinositol-3-kinase (PI3K)-related kinase that controls cell<br />

proliferation, cell survival <strong>and</strong> adhesion-independent survival <strong>and</strong> migration. Combining<br />

Gemzar <strong>and</strong> rapamycin might thus provide a therapeutic advantage. Here we report a patient<br />

who underwent resection of abdominal leiomyosarcoma at the time of renal transplantation.<br />

While on rapamycin for rejection prevention, no sarcoma re-growth was evident. On<br />

interruption of rapamycin, after spontaneous kidney rejection, the sarcoma relapsed, <strong>and</strong><br />

metastasized. Re-introduction of rapamycin <strong>and</strong> co-administration of gemcitabine yielded a<br />

prolonged remission <strong>and</strong> symptoms alleviation. To define the molecular impact of this drug<br />

combination, we have used SK-LMS cells, a human leiomyosarcoma cell line as a model.<br />

Indeed, we show that both gemcitabine <strong>and</strong> rapamycin inhibit cell proliferation, only<br />

rapamycin alone or when combined with gemcitabine reduce significantly the amount of<br />

phosphorylated ERK as well as a the amount of bcl-2. In view of the fact that gemcitabineinduced<br />

cell death is determined by bcl-2 content, <strong>our</strong> results provide a strong basis for a<br />

novel regimen based on Gemzar/rapamycin combination in cancer treatment.<br />

- 262 -


Notes<br />

- 263 -


Notes<br />

Notes<br />

- 264 -


- 265 -


Session V. Phosphatases as key cell <strong>signaling</strong> intermediates<br />

- 266 -


V. Phosphatases as key cell <strong>signaling</strong> intermediates Poster V, 1<br />

SHP-1 tyrosine phosphatase in human erythrocytes.<br />

Bragadin M.1, Clari G.2, Ion Popa F.2, Bordin L.2<br />

1 Dept. of Environmental Sciences, Ca’ Foscari University of Venice, Calle Larga<br />

S.Marta Dorsoduro 2137, Venice, Italy; e-mail: bragadin@unive.it<br />

2 Dept. of Biological Chemistry, University of Padua, Viale G. Colombo 3, 35121 Padua,<br />

Italy; e-mail: giulio.clari@unipd.it ; luciana.bordin@unipd.it<br />

SHP-1 is a SH2-domain containing protein Tyr-phosphatase expressed in hematopoietic cell<br />

lines, which is hypothesized to play a largely negative role in signal transduction. In human<br />

erytrocytes, the phospho-Tyr level of proteins, mainly transmembrane b<strong>and</strong> 3 protein, is<br />

closely controlled by the antithetic activity of Tyr-protein kinases <strong>and</strong> phosphatases, resulting<br />

in a dephosphorylated state. Only after particular stimuli, as with oxidizing agents, diamide or<br />

pervanadate, or thiol alkylating compound, N-ethyl maleimide (NEM), Tyr-phosphorylation<br />

of b<strong>and</strong> 3 can be triggered, inhibiting Tyr-phosphatase action <strong>and</strong> inducing erytrocyte<br />

membrane reorganization.<br />

We demonstrate that, in human red blood cells, SHP-1 is present in membranes from resting<br />

cells, but in 5 percent of the protein amount. Interstingly, this amount increases up to<br />

threefold following NEM treatement of intact cells, whereas diamide <strong>and</strong> pervanadate do not<br />

alter the normal protein location. In addition, SHP-1 translocation from cytosol to membrane<br />

is not affected by b<strong>and</strong> 3 P-Tyr level, since it is not mediated by the SH2-P-tyr recruiment<br />

mechanism, <strong>and</strong> localizes into the cytoskeletal compartment. B<strong>and</strong> 3 is the target of SHP-1,<br />

which dephosphorylates tyrosines located in acidic sequences such as Tyr 8,21 <strong>and</strong> 904.<br />

All together, these findings support the idea that, in human erytrocytes, the normal level of<br />

Tyr-phosphorylation of membrane protein, mainly b<strong>and</strong> 3, must be down regulated. We<br />

hypothesize that the simultaneous presence of both SHP-2 <strong>and</strong> SHP-1 ensures b<strong>and</strong> 3<br />

dephosphorylation in different conditions: SHP-2, through interaction of its SH2 domain/s to<br />

P-Tyr protein, is regulated by the b<strong>and</strong> 3 Tyr-phosphorylation level; SHP-1 may be involved<br />

by simple membrane rearrangement.<br />

- 267 -


V. Phosphatases as key cell <strong>signaling</strong> intermediates Poster V, 2<br />

Novel Cdc25 inhibitors: activity in MCF-7 cells <strong>and</strong> potentiation of cytotoxic drugs<br />

Souhayla El Maâdidi, Laurent Brault <strong>and</strong> Denyse Bagrel<br />

Laboratoire d’Ingénierie Moléculaire et Biochimie Pharmacologique, UFR SciFA,<br />

Université Paul Verlaine-Metz, Campus Bridoux, rue du Général Delestraint, 57070<br />

Metz, France. E-mail: bagrel@univ-metz.fr<br />

The Cdc25s dual-specificity phosphatases are key regulators of cell cycle progression which<br />

dephosphorylate <strong>and</strong> activate cycline dependent kinases. Three homologs of Cdc25<br />

phosphatases exist in humans: Cdc25A, Cdc25B, <strong>and</strong> Cdc25C. Cdc25B <strong>and</strong> Cdc25C regulate<br />

the G2/M cell cycle transition while Cdc25A is responsible for regulating the G1/S transition.<br />

The overexpression of Cdc25A <strong>and</strong> B is frequently associated with a wide variety of cancers.<br />

Taken into account there regulatory role, the aim of this study is a development of new Cdc25<br />

inhibitors. In <strong>our</strong> laboratory, novel maleic anhydride derivatives have been synthetised<br />

(named 6a to 6j) <strong>and</strong> tested in vitro using pNPP as substrate on the three recombinant GST-<br />

Cdc25A, GST-Cdc25B <strong>and</strong> GST-Cdc25C . These inhibitors differ in the size of their fatty<br />

acid chain, the most active of them having the longest chain. The in vitro inhibitory activity<br />

was confirmed by testing those compounds on two breast cancer cell lines: MCF-7 <strong>and</strong> its<br />

counterpart resistant to vincristine Vcr-R. F<strong>our</strong> compounds were chosen: 6a presenting the<br />

shortest fatty acid chain (6 carbons) <strong>and</strong> inactive in vitro, 6e having a mid-long chain (11<br />

carbons) showing an intermediate activity, <strong>and</strong> the 2 most potent inhibitors in vitro (6i <strong>and</strong> 6j)<br />

with the longest side chain (17 carbons). These two compounds induced a cell cycle blockage<br />

in G0/G1 phase <strong>and</strong> a decrease of cell proportions in S <strong>and</strong> G2/M phases after 24 <strong>and</strong> 48h<br />

treatments. Moreover, apoptosis was triggered within 48h-treatment, without oxidative burst.<br />

We also showed that 6i <strong>and</strong> 6j are able to potentiate the effects of cisplatin <strong>and</strong> adriamycin, at<br />

least by an additional effect. Thus, we can conclude that maleic anhydride-derivatives may<br />

mediate apoptosis through a cell cycle blockage via inhibition of the phosphatases Cdc25 <strong>and</strong><br />

may present a potential interest in therapeutic strategies against cancer.<br />

- 268 -


V. Phosphatases as key cell <strong>signaling</strong> intermediates Poster V, 3<br />

Cell adhesion to the extracellular matrix increases the proliferation of acute myeloid<br />

leukemia (AML) cells <strong>and</strong> up-regulates the CDC25A phosphatase.<br />

Anne Fern<strong>and</strong>ez-Vidal, Loïc Ysebaert, Christine Didier, Stéphane Manenti.<br />

Centre de physiopathologie Toulouse-Purpan, INSERM U563-IFR30, Département<br />

« Oncogénèse et signalisation dans les cellules hématopoïétiques », CHU Purpan, 3 place<br />

du Dr Baylac 31024 Toulouse Cedex 3, France. E-mail : fervidal@toulouse.inserm.fr.<br />

Microenvironmental stimuli are key factors in normal <strong>and</strong> leukemic hematopoietic<br />

differenciation. In this contexte, we investigated the influence of the major extracellular<br />

matrix component, fibronectin, on the proliferative state of acute myeloid leukemia <strong>and</strong><br />

normal hematopoietic cells. Although fibronectin inhibited the proliferation rate of immature<br />

normal CD34+ cells, it stimulated the proliferation of immature leukemic cells at similar<br />

differentiation state (KG1a cell line). These data suggest that hematopoietic cells shift from a<br />

negative to a positive proliferative response to cell adhesion during the leukemogenic<br />

transformation process. As a general feature, we found that cell adhesion to fibronectin matrix<br />

increases the proliferation of various cell lines. Importantly, we identified the dual specificity<br />

phosphatase Cdc25A as a key effector of this effect. Indeed, adhesion to fibronectin of<br />

leukemic cells dramatically up-regulates the cellular level of this protein, in good correlation<br />

with the proliferative response. By contrast, Cdc25A protein down-regulation was observed in<br />

normal CD34+ cells, nicely fitting with the inhibition of the proliferation observed in these<br />

cells. Furthermore, siRNA- <strong>and</strong> pharmacological inhibitor-based experiments strongly suggest<br />

that Cdc25A regulation in this context is indeed involved in the adhesion –dependent<br />

proliferation process. Altogether, these data demonstrate for the first time an integrindependent<br />

Cdc25A protein regulation, making of this cell cycle effector an important link<br />

between extracellular stimuli <strong>and</strong> the cell cycle machinery. This also make of this phosphatase<br />

<strong>and</strong> of its molecular regulators, a potential therapeutic target in the acute myeloid leukemia<br />

pathologies.<br />

- 269 -


V. Phosphatases as key cell <strong>signaling</strong> intermediates Poster V, 4<br />

The Vaccinia virus VH1-related (VHR) dual-specific protein phosphatase is upregulated<br />

in cervix cancer cells lines.<br />

Rachel Henkens, Philippe Delvenne, Jacques Boniver, Tomas Mustelin, Michel<br />

Moutschen <strong>and</strong> Souad Rahmouni.<br />

Department of Pathology, Liège University, Liège, Belgium.<br />

Protein tyrosine phosphatases (PTPases) regulate a variety of important processes in cells<br />

including cell cycle progression <strong>and</strong> several are involved in cancer. The 21-kDa Vaccinia<br />

virus VH1-related (VHR) dual-specific protein phosphatase plays a critical role in cell cycle<br />

progression <strong>and</strong> is itself regulated during its progression. Using RNA interference, it has been<br />

demonstrate that cells lacking VHR arrest in G1 <strong>and</strong> G2 phases of the cell cycle <strong>and</strong> show<br />

signs of beginning of cell senescence. We report here for the first time that VHR PTPase is<br />

upregulated in several cervix cancer cell lines. Using immunohistochemistry <strong>and</strong><br />

immunoblotting techniques, we show that this dual-specific phosphatase is upregulated in<br />

HPV positive cells lines such as Hela S3, CaSki, SiHa as well as in HPV negative cervix<br />

cancer cell lines such as C33 <strong>and</strong> HT3 compared to normal keratinocytes (NK). In addition to<br />

the upregulation of this protein, we report that VHR has a cytoplasmic localization in NK<br />

while its localized in the cytoplasm <strong>and</strong> in the nucleus of the cancer cell lines investigated.<br />

The semi-quantitative RT-PCR analysis shows no difference between the mRNA levels in the<br />

different cell lines compared to NK. Finally, we report that the VHR upregulation observed is<br />

at least partially due to the half life differences between the normal keratynocytes (+/- 7h) <strong>and</strong><br />

the different cervix cancer cell lines (>12h).<br />

- 270 -


V. Phosphatases as key cell <strong>signaling</strong> intermediates Poster V, 5<br />

Regulation of apoptosis signal-regulating kinase 1 (ASK1) by polyamine levels via<br />

protein phosphatase 5<br />

Mikhail A. Kutuzov, Alex<strong>and</strong>ra V. Andreeva <strong>and</strong> Tatyana A. Voyno-Yasenetskaya.<br />

Department of Pharmacology, College of Medicine, University of Illinois at Chicago,<br />

Chicago, Illinois 60607, U.S.A. Email: kutuzov@uic.edu<br />

Protein phosphatase 5 (PP5) is a member of the PPP family of protein Ser/Thr phosphatases,<br />

conserved from protists to plants <strong>and</strong> animals. Recent years have seen increasing evidence for<br />

PP5 involvement in a variety of <strong>signaling</strong> pathways, suggesting that PP5 is multifunctional<br />

phosphatase, similarly to more extensively characterized PPP phosphatases PP1 <strong>and</strong> PP2A. A<br />

number of proteins have been identified that interact with PP5 <strong>and</strong> either regulate its activity,<br />

or are PP5 substrates, or play a role of scaffold. PP5 may also be regulated, at least in vitro,<br />

by low molecular weight compounds, such as polyunsaturated fatty acids <strong>and</strong> fatty acyl-CoA<br />

esters. We found that PP5 is strongly inhibited by micromolar concentrations of a natural<br />

polyamine spermine. This inhibition was observed both with p-nitrophenyl phosphate <strong>and</strong><br />

protein substrates phosphocasein <strong>and</strong> apoptosis signal-regulating kinase 1 (ASK1, thought to<br />

be a physiological substrate of PP5). Inhibition of polyamine biosynthesis in COS-7 cells by<br />

alpha-difluoromethylornithine (DFMO) led to accelerated dephosphorylation of oxidative<br />

stress-activated ASK1. This effect of DFMO was suppressed by okadaic acid <strong>and</strong> by siRNAmediated<br />

PP5 depletion, indicating that the effect of polyamine levels on ASK1<br />

dephosphorylation was mediated by PP5. Polyamine depletion in COS-7 cells abrogated<br />

oxidative stress-induced activation of caspase-3 (which executes ASK1-induced apoptosis), as<br />

well as caspase-3 activation induced by ASK1 overexpression, but had no effect on basal<br />

caspase-3 activity. These results implicate polyamines, emerging intracellular <strong>signaling</strong><br />

molecules, as potential physiological regulators of PP5. Our findings also suggest a novel<br />

mechanism of the antiapoptotic action of a decrease in polyamine levels via de-inhibition of<br />

PP5 <strong>and</strong> accelerated dephosphorylation <strong>and</strong> deactivation of ASK1.<br />

- 271 -


V. Phosphatases as key cell <strong>signaling</strong> intermediates Poster V, 6<br />

Regulation of SHP-2 protein tyrosine phosphatase by Angiotensin II-derived reactive<br />

oxygen species in hypertensive <strong>and</strong> normotensive rats- Evidence of oxidation of protein<br />

tyrosine phosphatases in hypertension<br />

Fatiha Tabet1, Ernesto L. Schiffrin2, Rhian M. Touyz1.<br />

1Ottawa Health Research Institute, University of Ottawa, Health Sciences Building,<br />

304/451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada. E-mail: ftabet@uottawa.ca,<br />

E-mail: rtouyz@uottawa.ca<br />

2Experimental Hypertension Laboratory, Clinical Research Institute of Montreal, 110<br />

Pine Av. West, Montreal, Quebec H2W 1R7, Canada. E-mail:<br />

ernesto.schiffrin@ircm.qc.ca<br />

Reactive Oxygen Species (ROS) act as <strong>signaling</strong> molecules that regulate activation of MAP<br />

kinases. We recently demonstrated that angiotensin II (Ang II)-stimulated tyrosine kinases<br />

play a role in redox-sensitive activation of MAP kinases <strong>and</strong> that in vascular smooth muscle<br />

cells (VSMCs) from hypertensive rats (SHR) responses are enhanced. In the present study we<br />

tested the hypothesis that Ang II-induced ROS production augments tyrosine<br />

phosphorylation-dependent <strong>signaling</strong> in SHR as compared to normotensive rats WKY through<br />

differential activation of intracellular SHP-2 tyrosine phosphatase expression <strong>and</strong> activation,<br />

<strong>and</strong> through reversible inactivation of cellular PTPs. Mesenteric artery VSMCs of 16-week<br />

WKY <strong>and</strong> SHR rats were studied. Cells were stimulated with Ang II (10-7mol/l). The<br />

expression <strong>and</strong> activation (0-30 min) of SHP-2 was evaluated using immunoblotting analysis.<br />

The role of AT-1 <strong>and</strong> AT-2 receptors in Ang II-induced activation de SHP-2 was evaluated<br />

using AT-1 <strong>and</strong> AT-2 receptor inhibitors Valsartan <strong>and</strong> PD123319, respectively. The “In-Gel”<br />

PTP Assay was used to determine whether PTPs in response to Ang II (10-7mol/l, 0-60 min)<br />

or H2O2 (0.1-200µM, 10 min), are susceptible to oxidation. GST-FER fusion protein was<br />

used as a protein tyrosine kinase s<strong>our</strong>ce for phosphorylation of the [$-32P]-labeled poly (4:1)<br />

Glu-Tyr substrate. In the basal state, SHP-2 expression (p


Notes<br />

- 273 -


Notes<br />

Notes<br />

- 274 -


- 275 -


Session VI. Proteasome degradation pathways<br />

- 276 -


VI. Proteasome degradation pathways Poster VI, 1<br />

Dactylin, a F-box/WD ubiquitin ligase regulates osteoblast differentiation <strong>and</strong> limb<br />

development<br />

Steven Y Cheng1, 2 <strong>and</strong> Ying E. Zhang2<br />

1 Cellogenetics. Inc., Gaithersburg, MD 20898<br />

2 Laboratory of Cellular & Molecular Biology, National Cancer Institute, NIH,<br />

Bethesda, MD 20892<br />

Dactylin is a member of the F-box/WD-40 gene family, which recruit specific target proteins<br />

through their WD-40 protein-protein binding domains for ubiquitin mediated degradation.<br />

Disruption of the mouse dactylin gene results in the absence of central digits, underdeveloped<br />

or absent metacarpal/metatarsal bones <strong>and</strong> syndactyly. This phenotype is remarkably similar<br />

to split h<strong>and</strong>-split foot malformation in humans, a clinically heterogeneous condition with a<br />

variety of modes of transmission. We found BMP <strong>signaling</strong> downregulates dactylin<br />

expression through Smad pathways. Moreover, we showed that downregulation of Dactylin<br />

is required for BMP-2-mediated C2C12 myoblast differentiation into osteoblast since<br />

overexpression of Dactylin inhibited osteoblast differentiation. Thus, through its F-Box <strong>and</strong><br />

WD protein interaction domains, Dactylin regulates osteoblast differentiation by modulating<br />

BMP <strong>signaling</strong>. These results offer new insights into how BMP elicits biological effects, in<br />

particular into the mechanism of inhibition of myoblast differentiation <strong>and</strong> stimulation of<br />

osteoblast differentiation, <strong>and</strong> important role of Dactylin in controlling mesenchymal cell fate.<br />

- 277 -


VI. Proteasome degradation pathways Poster VI, 2<br />

p14ARF triggers G2 arrest through ERK-mediated Cdc25C phosphorylation,<br />

ubiquitination <strong>and</strong> proteasomal degradation.<br />

Beatrice Eymin, Christian Brambilla, Elisabeth Brambilla <strong>and</strong> Sylvie Gazzeri.<br />

INSERM U578, Institut Albert Bonniot, 38706 La Tronche Cedex, France. Université<br />

Joseph F<strong>our</strong>ier, Faculté de Medecine, Groupe de Recherche sur le Cancer du Poumon,<br />

La Tronche Cedex, France. E-mail: Beatrice.Eymin@ujf-grenoble.fr<br />

The Cdc25C phosphatase is a key regulator of mitotic entry which activity is highly regulated<br />

by phosphorylation. In response to DNA damage, phosphorylation of Cdc25C at serine 216<br />

induces cytosolic retention through 14-3-3 binding. We previously reported the ability of the<br />

p14ARF tumor suppressor to induce accumulation of inactive phospho-Cdc25C(Ser216)<br />

protein as well as decrease of Cdc25C steady state level <strong>and</strong> correlated these events with a<br />

p53-independent G2 arrest. The aim of this study was to investigate the cellular <strong>signaling</strong><br />

pathways involved in this process. By using specific pharmacological inhibitors, we<br />

demonstrate that activation of the MAP kinases ERK1/2 pathway is involved in the p53independent<br />

G2 checkpoint induced by p14ARF. Moreover, we identify Cdc25C as a new<br />

ERK1/2 target by providing evidence that activated P-ERK1/2 bind <strong>and</strong> phosphorylate<br />

Cdc25C on its ser216 residue following p14ARF expression. Importantly, we further show<br />

that phosphorylation at Ser216 by phospho-ERK1/2 promotes Cdc25C ubiquitination <strong>and</strong><br />

proteasomal degradation, suggesting that Cdc25C proteolysis is required for a sustained G2<br />

arrest in response to p14ARF. Taken together, these results demonstrate that the MAPK ERK<br />

<strong>signaling</strong> pathway plays a role in the p53-independent antiproliferative functions of p14ARF.<br />

Furthermore, they identify a new mechanism by which phosphorylation at serine 216<br />

contributes to Cdc25C inactivation.<br />

- 278 -


VI. Proteasome degradation pathways Poster VI, 3<br />

Ubiquitination as a priming process of PKC alpha <strong>and</strong> epsilon degradation in the<br />

alphalphaT3-1 gonadotrope cell line.<br />

Benoit Poulin, Hélène Maccario, Brice Junoy, Bénédicte Boyer, Alain Enjalbert <strong>and</strong><br />

Sophia V. Drouva.<br />

CNRS UMR 6544, Université de la Méditerranée, Faculté de Médecine, IFR Jean Roche<br />

Bd Pierre Dramard 13916 Marseille, France. E-mail: drouva.sv@jean-roche.univ-mrs.fr<br />

Protein kinase C (PKC) is a family of isoenzymes playing a key role in the regulation of<br />

gonadotrope cell functions. We have previously demonstrated that gonadotrope cell lines<br />

(alphaT3-1, LbT2) possess several PKC isoenzymes which are selectively sensitive to<br />

gonadotropin-releasing hormone (GnRH) <strong>and</strong> phorbol ester TPA. Indeed, specific PKC<br />

isoforms are differentially phosphorylated, activated <strong>and</strong> down regulated by GnRH (delta,<br />

epsilon, theta) or TPA (alpha, betaII, delta, epsilon, theta). Furthermore, we have shown that<br />

under desensitization conditions the degradation of PKC alpha, epsilon <strong>and</strong> delta isoenzymes<br />

may involve proteolytic processing implicating ubiquitin/proteasome system. Specific<br />

proteasome inhibitors prevented GnRH- <strong>and</strong> TPA-elicited kinase depletion <strong>and</strong> induced<br />

membrane accumulation of the respective isoenzymes in a phosphorylated form. In the<br />

present study we investigated in alphaT3-1 gonadotrope cells, the mechanisms underlying<br />

proteasome-dependent TPA-sensitive down-regulation of the PKC alpha <strong>and</strong> epsilon<br />

isoenzymes, <strong>and</strong> of the GnRH-activated PKC epsilon isoform. By a pull-down assay using<br />

beads conjugated with a GST-fusion protein containing ubiquitin-associated domains (Rad23)<br />

that bind polyubiquitinated proteins, we show that TPA induced rapid (15min) <strong>and</strong> sustained<br />

(up to 4h) PKC alpha <strong>and</strong> PKC epsilon polyubiquitination. Moreover, GnRH provoked a<br />

selective polyubiquitination of PKC epsilon, in a receptor-dependent manner, but was unable<br />

as expected to induce any polyubiquitination of PKC alpha The GnRH-evoked PKC epsilon<br />

polyubiquitination was important <strong>and</strong> rapid (as early as 10min) <strong>and</strong> progressively decreased<br />

over time (but remained detectable after a 4h treatment). The empty beads, used as control,<br />

did not retain any polyubiquitinated protein. Thus in alphaT3-1 gonadotrope cells, the event<br />

priming TPA-induced PKC alpha <strong>and</strong> PKC epsilon down-regulations as well as GnRHevoked<br />

PKC epsilon desensitization, is their respective polyubiquitination, it preceeds the<br />

isoenzymes degradation by the proteasome complex. Surprisingly proteasome inhibition<br />

counteracted the TPA- as well as GnRH- induced polyubiquitination of their target PKC<br />

isoenzymes ; this may reflect either high activity of deubiquitinating enzymes or direct<br />

interaction with ubiquitin conjugation at the proteasome level.<br />

- 279 -


Notes<br />

- 280 -


Session VII. Stem cell specific cell <strong>signaling</strong> mechanisms<br />

- 281 -


VII. Stem cell specific cell <strong>signaling</strong> mechanisms Poster VII, 1<br />

Murine marrow-derived mesenchymal stem cells:Isolation,Characterisation<br />

*Raza Haji Hosseini ,**Samad Nadri<br />

*Biology Dept. Faculty of science, Payame Noor University, Tehran, Iran 19569<br />

*E.mail:hosseini@pnu.ac.ir,<br />

**Biology Dept. Faculty of science, Payame Noor University, Tehran, Iran 19569<br />

**E.mail:nadrisamad@yahoo.co.uk<br />

Introduction: Many researchers have employed different methods to isolate <strong>and</strong> purify<br />

mesenchymal stem cells in mouse bone marrow but there is a lack of consensus on<br />

mesenchymal stem cells express surface markers. There is evidence for the affect cell density<br />

on surface markers in human mesenchymal stem cells. IN present study, Expression of<br />

CD34,CD44,Sca-1,c-Kit,Vcam-1 on MSCs purified by Low density <strong>and</strong> High density from<br />

NMRI <strong>and</strong> BALB/c was studied.<br />

Method <strong>and</strong> Material:6-8 week-old NMRI <strong>and</strong>BALB/c were killed by cervical dislocation <strong>and</strong><br />

bone marrow were flushed <strong>and</strong> cultured using low glucose DMEM medium by Low density<br />

<strong>and</strong> High density methods then MSCs were purified by continue passages.To examine the<br />

mesenchymal nature of isolated cells,The cells were differentiated into bone <strong>and</strong><br />

adipocyte.the cells were abeled by CD34,CD44,Sca-1,c-Kit,Vcam-1 antibody conjugated to<br />

PE or FITC <strong>and</strong> analyzed by flow cytometry method using FACS-analysis Machine.<br />

Result:Spindle –shaped purified cells were observed in Low density <strong>and</strong> High density cultures<br />

at the end of the f<strong>our</strong>th <strong>and</strong> seven passage,respectively.The cells from both NMRI<br />

<strong>and</strong>BALB/c were easily differentiated into osteogenic <strong>and</strong> adipogenic Lineages.Flow<br />

cytometry analysis indicated that The MSCs from either NMRI <strong>and</strong> BALB/c mice are CD44+<br />

,Sca-1+ CD34- <strong>and</strong> c-Kit- in Low density <strong>and</strong> High density culture.The mMSCs from tow<br />

strains differed in their expression of the cell-surface epitope Vcam-1.<br />

Conclusion:In present investigation,The presence of five surface markers was investigated on<br />

MSCs purified by Low density <strong>and</strong> High density culture from NMRI <strong>and</strong> BALB/c.The results<br />

presented here,indicated up to 90% cells is CD44+ , It seems That It could be useful markers<br />

in isolation of the MSCs.Comparisons of MSCs isolated from 2 strains demonstrated That<br />

some features of The cells were strain specific also They isolation more rapidly if plated at<br />

Low density culture. In this studies, In spite of previously research,Cell density is not affected<br />

on expression surface markers.<br />

Key Word:marker,mesenchymal stem cell,bone marrow<br />

- 282 -


Notes<br />

- 283 -


Session VIII. <strong>Inflammation</strong> specific <strong>signaling</strong><br />

- 284 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 1<br />

Analysis of tissue distribution of TNF-alpha, TNF-alpha-Receptors <strong>and</strong> the activating<br />

TNF-alpha-Converting Enzyme (TACE) suggest activation of the TNF-alpha system in<br />

the ageing intervertebral disc<br />

B.Bachmeier (1), A. Nerlich (2), Ch. Weiler (3), N. Boos (4)<br />

Dept. of Clinical Chemistry Clin. Biochemistry, Univ. Munich, Germany,<br />

bachmeier@med.uni-muenchen.de<br />

Dept. of Pathology, Academic Hospital Munich-Bogenhausen, Germany<br />

Institute of Pathology, Ludwig-Maximilians-University Munich, Germany<br />

Spinal Surgery Unit, Orthopaedic Clinic, Univ. Zürich, Switzerl<strong>and</strong><br />

The expression <strong>and</strong> up-regulation of the pro-inflammatory cytokine TNF-alpha recently has<br />

been suggested to play a major role in the degeneration of the intervertebral disc. In order to<br />

further elucidate the role of TNF-alpha in intervertebral discs <strong>and</strong> to find out, if the TNFalpha<br />

activation by the corresponding converting enzyme TACE <strong>and</strong> the two TNF-alphareceptors<br />

I <strong>and</strong> II are indeed potentially involved in the cytokine action, we<br />

immunohistochemically analyzed the expression <strong>and</strong> localization of all f<strong>our</strong> factors in human<br />

discs. In parallel, the quantitative levels of TNF-alpha-mRNA (by quantitative RT-PCR) were<br />

determined.<br />

Therefore, a series of 43 human lumbar intervertebral disc samples with complete crosssection<br />

of the motion segment were immunohistochemically stained for TNF-alpha, TNFR-I<br />

<strong>and</strong> –II <strong>and</strong> TACE (all antibodies: Santa Cruz Biotech., CA, USA) <strong>and</strong> morphometrically<br />

evaluated for the proportion of positively labelled cells in five anatomic sub-settings: nucleus<br />

pulposus, inner <strong>and</strong> outer anterior <strong>and</strong> posterior annulus fibrosus. In a further series of<br />

surgical material from 20 symptomatic patients the level of TNF-alpha-mRNA was<br />

determined by quantitative RT-PCR (Light Cycler, Roche, Penzberg) <strong>and</strong> correlated to the<br />

corresponding immunohistochemical staining pattern.<br />

Thereby, we detected a significant expression of TNF-alpha beginning in advanced<br />

adolescence (ca. 17 years) <strong>and</strong> being most extensive in the nucleus pulposus. Likewise, the<br />

TNF-alpha expression in adult nucleus pulposus ranges between 27 – 80% of all cells until<br />

senile age. In the inner <strong>and</strong> outer annulus fibrosus significantly less cells are labelled (0 –<br />

50%). In fetal <strong>and</strong> juvenile discs no TNF-alpha expression is detectable. The parallel analyses<br />

both TNF-alpha-receptors revealed a similar distribution pattern with respect to age <strong>and</strong><br />

localization. Additionally, the TNF-alpha converting enzyme TACE, which is essential for the<br />

shedding <strong>and</strong> activation of the cytokine, are up-regulated similarly. The number of TNFalpha-transcripts<br />

was elevated in most cases with immunohistochemical TNF-alpha<br />

expression confirming the synthesis of TNF-alpha protein in the discs.<br />

Our present study clearly supports the recent notion that TNF-alpha is expressed in discs of<br />

increasing age. Furthermore, we also can confirm the previous observation that TNF-alpha<br />

correlates with histomorphological signs of disc degeneration. Additionally, the present study<br />

provides unambiguous evidence that TNF-alpha is activated (by the converting enzyme<br />

TACE). Finally, the presence of both biologically active TNF-alpha receptors (TNFR-I <strong>and</strong> –<br />

II) in similar proportion to each other supports the idea that TNF-alpha can also exhibit its<br />

action within the disc.<br />

- 285 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 2<br />

A role for TNF-alpha in VLDL apoB but not lipid secretion modulation in primary rat<br />

hepatocytes under non-promoting triacylglycerol production conditions<br />

Nerea Bartolomé, Lorena Rodríguez, Cristina López, Virginia Martínez, Begoña Ochoa,<br />

María J. Martínez <strong>and</strong> Yol<strong>and</strong>a Chico<br />

Department of Physiology, Faculty of Medicine <strong>and</strong> Dentistry, University of the Basque<br />

Country, P. Box 669, Bilbao, Bizkaia, Spain. ofbbaesn@lg.ehu.es<br />

The cytokine TNF-alpha has been reported to play a pivotal role in the septic inflammatory<br />

response. It is rapidly liberated to bloodstream triggering the production of other cytokines<br />

<strong>and</strong> initiating the liver acute phase reaction. Hipertriglyceridemia is an early hallmark of<br />

sepsis associated to high levels of VLDL particles, due to both enhanced secretion by the liver<br />

<strong>and</strong> decreased peripheral clearance, but the time response of hepatocytes <strong>and</strong> the underlying<br />

mechanisms are not completely understood. The biogenesis of VLDL is a complex <strong>and</strong> timerequiring<br />

process that depends on lipid availability <strong>and</strong> the activity of microsomal triglyceride<br />

transfer protein (MTP) for correct apoB lipidation. The aim of this work was to asses in vitro<br />

whether TNF-alpha has a sustained role in VLDL production regulation <strong>and</strong> the putative<br />

implication of apoB <strong>and</strong> MTP transcription. Rat primary hepatocytes were cultured in<br />

conditions mimicking the fed situation, with 5, 20 or 100 ng/ml TNF-alpha for 8 or 16 h.<br />

VLDL were isolated from the medium <strong>and</strong> apoB-48, apoB-100 <strong>and</strong> lipids were quantified.<br />

Total RNA was extracted from cells <strong>and</strong> apoB <strong>and</strong> MTP mRNA levels measured by Northern<br />

blotting. At 8 h, there was an increase of 17%, 22% <strong>and</strong> 24% in the secretion of VLDL<br />

particles by 5, 20 or 100 ng/ml TNF-alpha-treated hepatocytes, respectively. However, basal<br />

secretion was recorded after 16 h. Cell apoB mRNA content increased by 1.88-, 1.54- <strong>and</strong><br />

1.44-fold in cells at 8 h of TNF-alpha treatment with 5, 20 or 100 ng/ml, <strong>and</strong> 1.43 <strong>and</strong> 1.38<br />

fold after 16 h of treatment with 20 or 100 ng/ml. The total amount of lipids secreted into<br />

VLDL, as well as the lipid composition of each particle <strong>and</strong> the MTP transcript levels did not<br />

change significantly with any of the TNF-alpha doses <strong>and</strong> periods of treatment tested. In<br />

conclusion, this is another finding supporting for a correlation between the hepatocyte level of<br />

apoB mRNA <strong>and</strong> the secretion of its protein without enhanced lipid secretion vinculated to a<br />

sepsis-promoted cytokine.<br />

Supported by grants G03/015, PE02UN04 <strong>and</strong> a personal grant to N.B.<br />

- 286 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 3<br />

Immune-complexes induce monocyte survival through defined intracellular pathways<br />

Giordano Bianchi, Fabrizio Montecucco, Maria Bertolotto, Luciano Ottonello <strong>and</strong><br />

Franco Dallegri.<br />

Clinic of Internal Medicine I, Department of Internal Medicine, University of Genoa<br />

Medical School, Genoa, Italy<br />

Regulation of inflammation is a crucial event since its alteration determines severe tissue<br />

damage. The fine balance among several microenvironment components establishes the<br />

resolution or perpetuation of inflammatory reactions. In a variety of diseases, immunecomplex<br />

(IC) deposition induces an inflammatory response with tissue injury which is<br />

mediated by inflammatory cell infiltration, proliferation, survival or apoptosis. In particular,<br />

the interaction between ICs <strong>and</strong> the Fc gamma receptor triggers the activation of monocytes<br />

<strong>and</strong> macrophages, favoring their tissue accumulation by inhibiting the apoptosis. To elucidate<br />

the intracellular pathways governing this process, on the basis of <strong>our</strong> previous findings<br />

regarding the dose-dependent inhibition of apoptosis in IC-activated monocytes, we have<br />

investigated the role of phosphatidylinositol 3-kinase (PI3K)/Akt pathway showing its<br />

involvement in survival of activated monocytes. As confirmation, molecular inhibitors of<br />

PI3K <strong>and</strong> Akt suppressed the anti-apoptotic effect of ICs. In parallel we have demonstrated<br />

the activation of the NF-kB system in triggered monocytes, directly by EMSA assay <strong>and</strong><br />

indirectly by showing the increased expression of X-IAP. Evenly NF-kB <strong>and</strong> IKK-b inhibitors<br />

induced the apoptosis of activated monocytes at a rate comparable to that of resting<br />

monocytes. In agreement with previous data showing the Fc triggered activation of MAPK<br />

pathway, we have found that ICs induce the activation of erk1/2 <strong>and</strong> p38 conferring apoptosis<br />

resistance to activated monocytes. Finally the activity of caspase 3, 8 <strong>and</strong> 9 resulted inhibited<br />

in IC-activated monocytes. Even though further experiments are needed to identify a possible<br />

link among these pathways, these results disclose a <strong>signaling</strong> route triggered by ICs which can<br />

be involved in the pato-physiology of inflammatory diseases.<br />

- 287 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 4<br />

Modulation of Host Responses to Bacterial Endotoxin<br />

Herbert Bosshart <strong>and</strong> Michael Heinzelmann<br />

Innate Immunity Research Laboratory, Department of Surgery, Zurich University<br />

Hospital, CH-8091 Zurich, Switzerl<strong>and</strong>, e-mail: h_bosshart@bluewin.ch<br />

The term sepsis describes a potentially lethal clinical condition that develops as a result of a<br />

dysregulated host response to bacterial infection. The commonest bacterial component<br />

implicated in initiating the septic syndrome is a cell wall molecule derived from Gramnegative<br />

bacteria, known as lipopolysaccharide (LPS) or endotoxin. Like all mammals,<br />

humans are equipped with an LPS-sensing machinery consisting, primarily, of LPS-binding<br />

protein (LBP), CD14, a glycosylphosphatidylinositol-anchored monocyte differentiation<br />

antigen, <strong>and</strong> Toll-like receptor 4 (TLR4), a signal transducing integral membrane protein.<br />

Modest stimulation of TLR4 facilitates the elimination of invading microorganisms. Potent<br />

TLR4 stimulation, however, produces severe reactions in the host, often leading to multiple<br />

organ failure <strong>and</strong> death. The search for pharmaceuticals that reduce mortality in septic<br />

patients has been a painstaking process. Thus far, only a few compounds have been found to<br />

significantly reduce mortality rates. Perhaps one of the more promising therapeutic strategies<br />

currently pursued is based on the identification of synthetic or naturally-occurring substances<br />

that neutralize LPS or inhibit LPS-mediated activation of host immune cells such as<br />

monocytes <strong>and</strong> macrophages. Here, we describe a number of divers molecular structures with<br />

a capacity to either enhance or blunt LPS-induced monocyte activation. The underlying<br />

molecular mechanisms are discussed.<br />

- 288 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 5<br />

CD3 activation alters T cell responses to LFA-1 engagement: regulation of T cell<br />

polarization <strong>and</strong> migration<br />

Eva M. Cernuda-Morollón1, Federica Marelli-Berg2 <strong>and</strong> Anne J. Ridley1.<br />

(1) Ludwig Institute for Cancer Research-UCL Branch, 91, Riding House Street, W1W<br />

7BS London, UK. eva@ludwig.ucl.ac.uk; anne@ludwig.ucl.ac.uk (2) Department of<br />

Immunology, Division of Medicine, Imperial College London, Du Canne Road, London<br />

W12 ONN, UK. f.marelli@imperial.ac.uk.<br />

Recognition of cognate antigen by the T cell receptor induces T cell proliferation. The<br />

recruitment of these activated T cells to <strong>and</strong> retention at sites of inflammation are crucial for<br />

the inflammatory response <strong>and</strong> therefore play an important role in pathological conditions<br />

such as cardiovascular disorders <strong>and</strong> chronic inflammation. T cell recruitment requires them<br />

to cross the endothelium, a process that involves multiple receptors including the integrin<br />

LFA-1 on T cells, which binds to ICAM-1 on endothelial cells. We have observed that T cell<br />

receptor activation, mimicked by cross-linking with anti-CD3 antibody, inhibits T cell<br />

transendothelial migration. CD3 activation also reduces T cell migration speed on ICAM-1.<br />

This effect is not due to changes in T cell adhesive properties <strong>and</strong> correlates with an<br />

impairment in T cell polarization, as revealed by staining for different markers, including<br />

CD44, ICAM-3 <strong>and</strong> lipid raft markers. Rho GTPases regulate actin <strong>and</strong> microtubule dynamics<br />

<strong>and</strong> therefore cell polarization <strong>and</strong> migration. We are investigating the activation status of the<br />

Rho GTPases RhoA, Rac1 <strong>and</strong> Cdc42. Pre-treatment with CD3 antibodies increases Rac1<br />

activity <strong>and</strong> modulates T cell responses to LFA-1 engagement. Furthermore, CD3 activation<br />

promotes stathmin/Op18 phosphorylation <strong>and</strong> increases the levels of acetylated alpha-tubulin,<br />

both of which indicate an increase in microtubule stability.<br />

We propose that CD3 activation affects actin <strong>and</strong> microtubule dynamics, thereby impairing<br />

normal T cell polarization <strong>and</strong> migration.<br />

- 289 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 6<br />

The stress-activated protein kinase pathway in dendritic cell activation.<br />

Benjamin M. Chain, Matt H<strong>and</strong>ley, Jane Rasaiyaah, Gabriel Pollara <strong>and</strong> David R Katz<br />

Dept. Immunology <strong>and</strong> Molecular Pathology, UCL, London, UK E.mail :<br />

b.chain@ucl.ac.uk<br />

The activation <strong>and</strong> subsequent maturation of dendritic cells via Toll-like receptors (TLRs) is a<br />

key step in the initiation of adaptive immunity. The canonical pathway requires activation <strong>and</strong><br />

nuclear translocation of NFkB transcription factor. However there is accumulating data that<br />

activation of members of the stress-activated kinase p38 family is also necessary. In contrast,<br />

the role of another family of stress activated kinases, the c-Jun N-terminal kinases (JNKs) , in<br />

TLR signalling remains obscure.<br />

Our laboratory has re-examined the function of the stress-activated protein kinase pathway in<br />

human in vitro cultured monocyte-derived dendritic cells. Activation of dendritic cells with<br />

high concentrations of lipopolysacharide (LPS, a TLR4 agonist) or poly I:C (TLR3 agonist)<br />

induced both p38 <strong>and</strong> JNK phosphorylation. Low concentrations of these agonists activated<br />

p38 but not JNK. In contrast zymosan (TLR2/dectin agonist) induced selective p38 but no<br />

JNK activation at any concentration. The activation of p38 by LPS correlated with the degree<br />

of dendritic cell maturation, while activation of JNK was associated with some cell death.<br />

Both kinases were strongly induced by reactive oxygen (H2O2) species, but this was not<br />

associated with any dendritic cell maturation, but rather with extensive dendritic cell<br />

apoptosis. A selective competitive inhibitor of p38 blocked dendritic cell maturation induced<br />

via TLR2, 3 or 4. In contrast a selective competitive inhibitor of JNK had no effect on<br />

dendritic cell maturation, but partially protected dendritic cells from reactive oxygen induced<br />

apoptosis.<br />

Unexpectedly, an inhibitor of mixed linage kinases (MLKs) , previously considered to be<br />

predominantly upstream activators of JNK activation, strongly inhibited dendritic cell<br />

maturation in response to LPS. Dendritic cells were found to express several members of the<br />

MLK family, <strong>and</strong> furthermore, both LPS <strong>and</strong> poly I:C induced MLK3 phosphorylation. In<br />

contrast, zymosan induced no MLK3 phosphorylation. Inhibition of MLK activity resulted in<br />

a block in maturation (as measured by cell surface phenotype) <strong>and</strong> cytokine secretion in<br />

response to LPS <strong>and</strong> poly I:C, but not in response to zymosan.<br />

In conclusion, we have documented selective activation of individual modules of the stress<br />

activated kinase family in dendritic cells depending both on TLR lig<strong>and</strong> <strong>and</strong> signal strength.<br />

This plasticity in molecular response may be crucial in allowing these key sentinels of the<br />

immune system to optimise the quantitative <strong>and</strong> qualitative parameters of the ensuing immune<br />

response.<br />

H<strong>and</strong>ley et al. Free Radic Biol Med. 2005 Jun 15;38(12):1637-52. Epub 2005 Mar 30. JNK<br />

activation limits dendritic cell maturation in response to reactive oxygen species by the<br />

induction of apoptosis.<br />

- 290 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 7<br />

Elastin peptides pro-inflammatory role in melanoma progression.<br />

Romain Debret#§, Richard Le Na<strong>our</strong>§, Jean Michel Sallenave‡, William Hornebeck#,<br />

Moncef Guenounou§, Philippe Bernard# <strong>and</strong> frank Antonicelli#<br />

# Department of Dermatology, CNRS UMR 6198, § Department of Immunology, EA<br />

3796, IFR 53 Biomolécules, University of Reims, Champagne-Ardenne, France. ‡ Rayne<br />

Laboratories, Medical School, The University of Edinburgh, Edinburgh, UK.<br />

E-mail: frank.antonicelli@univ-reims.fr<br />

Background: Melanoma is a serious skin cancer. If not treated, it can be fatal. Melanoma is<br />

not the most common cancer, but its incidence is increasing faster than any other. Although<br />

surgery can be curative for early-stage melanoma, complete surgical excision of advanced<br />

disease is often followed by recurrence. This is because surgery is a local treatment; it reduces<br />

tum<strong>our</strong> burden (<strong>and</strong> tum<strong>our</strong>-associated immune suppression), but it does not eliminate tum<strong>our</strong><br />

cells that have entered the circulation <strong>and</strong> spread to distant sites. Prior to metastasize to the<br />

regional nodes which are the single most important prognostic factor in solid neoplasms,<br />

melanoma cells ought to degrade the surrounding extra-cellular matrix releasing therefore<br />

protein fragments so-called matrikines because of their biological activities. Initial studies of<br />

the “architecture” of the tum<strong>our</strong> front indicated certain structural changes such as elastin<br />

fibres degradation in this front. We, then, focussed <strong>our</strong> work on the effects of these elastin<br />

fragments on the complex interaction between immune system (lymphocytes) <strong>and</strong> melanoma<br />

progression in the purpose to help to explain how certain tum<strong>our</strong>s are more aggressive than<br />

others.<br />

Results: We investigated the contribution of EFs on IL-1 beta expression, a cytokine known<br />

for playing a key role in melanoma cells activation. Our results first evidenced that high<br />

tumorigenic melanoma cells (M3Da cells) treated with EFs led to IL-1 beta mRNA <strong>and</strong><br />

protein up-regulation. The effects of EFs on M3Da cells were found to be mediated by<br />

receptor (S-gal) occupancy, as being suppressed by lactose <strong>and</strong> reproduced by cell stimulation<br />

with the VGVAPG peptide. Binding of EFs to their receptor induced a rapid activation of Erk<br />

& <strong>and</strong> p38 MAP Kinase pathways. However, activation of these pathways was not associated<br />

with IL-1 beta mRNA up-regulation by EFs. Concomitantly, we demonstrated that EFs<br />

stimulation induced NF-kB nuclear translocation (Western blot) <strong>and</strong> DNA binding on IL-1<br />

beta promoter region (EMSA <strong>and</strong> ChIP techniques) whereas inhibition of NF-kB with the<br />

specific chemical inhibitor SN-50 or by over-expression of IkB, the endogenous inhibitor of<br />

NF-kB pathway, totally abolished EFs-mediated IL-1 beta mRNA over-expression. In setting<br />

with IL-1 beta results, EFs also enhanced IL-8 <strong>and</strong> GRO-alfa expressions in melanoma cells.<br />

However, increased concentration of IL-1 beta in culture media inhibited this EFs-induced IL-<br />

8 expression. Interestingly, EFs also enhanced PHA-stimulated lymphocyte IL-1 beta<br />

expression, but addition of this media on melanoma cells failed to activate IL-8 expression<br />

compared to media from PHA-treated lymphocytes.<br />

Conclusion: These data further illustrate that elastic fibres are important s<strong>our</strong>ces of matrikines<br />

in pathologic conditions such as melanoma invasion by interfering in the relation<br />

cancer/inflammation. Depending on their concomitant localisation with large amount of IL-1<br />

beta, EFs could intervene in different stages of melanoma progression according to the<br />

presence or not of infiltrated inflammatory cells in melanoma microenvironment.<br />

- 291 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 8<br />

Specific contribution of the signalling pathways NF-kappaB <strong>and</strong> PI3K / mTOR on the<br />

gene expression profile of LPS stimulated murine macrophages<br />

Sofia Dos Santos1, Anne-Isabelle Delattre, Françoise de Longueville2 <strong>and</strong> Martine<br />

Raes1<br />

1Laboratory of Biochemistry <strong>and</strong> Cellular Biology, University of Namur (F.U.N.D.P.),<br />

Rue de Bruxelles, 61, 5000 Namur, Belgium. Email : martine.raes@fundp.ac.be,<br />

2Eppendorf Array Technologies, Rue du Séminaire, 20a, 5000 Namur, Belgium.<br />

<strong>Inflammation</strong> is the first response of the immune system to a variety of external or internal<br />

insults, such as infectious agents, physical injury, hypoxia, or pathological processes.<br />

In this study, we have simulated an inflammatory response by incubating RAW264.7 murine<br />

macrophages with LPS (lipopolysaccharide), a major component of the outer membrane of<br />

Gram-negative bacteria. The DNA microarray "DualChipTM Mouse <strong>Inflammation</strong>"<br />

(Eppendorf) was used to characterize the gene expression profiles of LPS-stimulated<br />

macrophages in the presence or not of BAY 11-7082 (a specific inhibitor of NF-kappaB) or<br />

LY294002 (a specific inhibitor of the PI3K / mTOR pathway).<br />

NF-kappaB is known to be a key transcription factor in the response to inflammation, but<br />

little is known about the specific contribution of the PI3K / mTOR pathway in LPS signalling.<br />

Results suggest a particularly important role for the transcription factor NF-kappaB, as<br />

expected, but to a lesser extent the PI3K pathway is also involved. Differences between BAY<br />

11-7082 <strong>and</strong> LY294002 were underlined. For example, genes encoding the PAF receptor,<br />

PAI-1, PLA2 (group V), IL13 receptor (alpha 2) <strong>and</strong> GTP cyclohydrolase 1, which were upregulated<br />

after LPS treatment, were mainly repressed by LY294002.<br />

NF-kappaB plays an important role in the macrophage response to LPS, but we also have<br />

shown that the PI3K pathway partially contributes to it. Further experiments with the specific<br />

inhibitor of mTOR (rapamycin) will provide more information on the specific contribution of<br />

the PI3K / mTOR pathway in the inflammatory response in LPS stimulated macrophages.<br />

S. Dos Santos is a Research Fellow of FNRS (Fonds National de la Recherche Scientifique,<br />

Brussels, Belgium).<br />

- 292 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 9<br />

Interleukin-31: Characterisation of signalling capacities <strong>and</strong> relevance for inflammatory<br />

skin diseases<br />

Alex<strong>and</strong>ra Dreuw1, Barbara Lippok1, Mark M. Neis2, Bettina Peters3, Hans F. Merk2,<br />

Andreas Bosio3, Jens M. Baron2, Peter C. Heinrich1 <strong>and</strong> Heike M. Hermanns1<br />

1Institut für Biochemie <strong>and</strong> , Uniklinik der RWTH Aachen, Aachen, Germany;<br />

2Hautklinik, Uniklinik der RWTH Aachen, Aachen, Germany<br />

3Miltenyi Biotec GmbH, MACSmolecular Business Unit, Köln, Germany<br />

Interleukin-31 is a recently identified cytokine which is produced by activated T lymphocytes,<br />

preferentially by Th2 cells <strong>and</strong> to a minor extent by Th1 cells. It signals via a receptor<br />

complex containing the specific IL-31 receptor <strong>and</strong> as the second signal transducing receptor<br />

the oncostatin M-receptor #. In the present study we characterised in depth the molecular<br />

mechanisms underlying IL-31R-mediated signal transduction. IL-31R is a strong activator of<br />

STAT3 <strong>and</strong> STAT5, while STAT1 is only marginally tyrosine phosphorylated. We identify<br />

tyrosine residues 652 <strong>and</strong> 721 in the cytoplasmic region of the longest isoform of IL-31R as<br />

the major STAT5 <strong>and</strong> STAT3 activating sites, respectively. Additionally, we demonstrate<br />

Jak1 binding to IL-31R <strong>and</strong> its activation in heteromeric complexes with the OSMR#, but<br />

also in a homomeric receptor complex. Interestingly, unlike OSMR# <strong>and</strong> gp130, IL-31R is<br />

insufficient to mediate ERK1/2 phosphorylation. We propose that this is due to a lack of<br />

recruitment of the tyrosine phosphatase SHP-2 or the adapter protein Shc to the cytoplasmic<br />

domain of IL-31R. Since transgenic mice overexpressing IL-31 develop a phenotype closely<br />

resembling atopic dermatitis in humans we evaluated the potential importance of this novel<br />

cytokine in the pathogenesis of T cell-mediated skin diseases. Confirming the results so far<br />

obtained only in transgenic mice we find statistically elevated mRNA levels of IL-31 in<br />

biopsies taken from patients with atopic dermatitis. However, no increased transcription of<br />

IL-31 can be detected in biopsies taken from psoriatic plaques, which strengthens the<br />

hypothesis, that IL-31 might be an important Th2-cytokine.<br />

This work was supported by the Deutsche Forschungsgemeinschaft (SFB 542; TP B6 <strong>and</strong><br />

C11)<br />

- 293 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 10<br />

Mitochondrial uncoupling protein UCP2 regulates the LPS-induced ROS <strong>signaling</strong> in<br />

innate imunity.<br />

Yalin Emre, Corinne Hurtaud, Tobias Nübel, François Criscuolo, Daniel Ricquier, <strong>and</strong><br />

Anne-Marie Cassard-Doulcier<br />

CNRS-UPR 9078, Faculté de Médecine Paris 5 Necker-Enfants Malades, 156 rue de<br />

Vaugirard, 75730 Paris Cedex 15, France. E-mail : emre@necker.fr,<br />

hurtaud@necker.fr, nuebel@necker.fr, criscuolo@wanadoo.fr, ricquier@necker.fr <strong>and</strong><br />

cassard-doulcier@necker.fr<br />

Macrophages constitute the first line of defense against pathogens in innate immunity. The<br />

recognition by Toll-like receptor-4 (TLR4) of lipopolysaccharides (LPS), a chief pathogenassociated<br />

molecular pattern of Gram negative bacteria, triggers innate activation of<br />

macrophages. Efficient activation is largely dependent upon the concomitant reactive oxygen<br />

species (ROS) <strong>signaling</strong>. Since mitochondria is the main site of ROS production in resting<br />

cells <strong>and</strong> the inner mitochondrial membrane protein UCP2 (uncoupling protein 2) is a<br />

negative regulator of mitochondrial ROS production, we investigated a possible involvement<br />

of UCP2 in macrophage innate activation <strong>and</strong> more precisely in the underlying ROS<br />

<strong>signaling</strong>. We showed that UCP2 was quickly downregulated in macrophages in response to<br />

LPS in order to increase the mitochondrial ROS production thus promoting MAPK<br />

phosphorylation <strong>and</strong> activation. Consistent with this, UCP2-deficient macrophages have<br />

increased nitric oxide production, cytokine release <strong>and</strong> their ablity to migrate. Based on <strong>our</strong><br />

data we conclude that the mitochondrial UCP2 plays an important role in pathogen-mediated<br />

innate activation of macrophages by negatively regulating the LPS-induced ROS <strong>signaling</strong>.<br />

- 294 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 11<br />

Specific integrin alpha <strong>and</strong> beta chain phosphorylations regulate LFA-1 activation<br />

through affinity-dependent <strong>and</strong> –independent mechanisms<br />

Susanna C. Fagerholm, Susanna M. Nurmi, Tiina J. Hilden, <strong>and</strong> Carl G. Gahmberg§<br />

Division of Biochemistry, Faculty of Biosciences, PB56 (Viikinkaari 5), 00014 University<br />

of Helsinki, Finl<strong>and</strong><br />

Integrins are heterodimeric adhesion receptors that are crucial for the functions of<br />

multicellular organisms. The activation of integrin-mediated adhesion is a complex process<br />

that involves both affinity regulation <strong>and</strong> cytoskeletal coupling, but the molecular<br />

mechanisms have remained incompletely understood. The f<strong>our</strong> members of the beta2integrins<br />

are expressed exclusively on leukocytes <strong>and</strong> mediate adhesion <strong>and</strong> <strong>signaling</strong> events<br />

that are essential for leukocyte function. In T cells, the main beta2-integrin is LFA-1<br />

(alphaLbeta2-integrin), which can become activated after cell stimulation through the T cell<br />

receptor, chemokine receptors or other signals. LFA-1 mediates crucial T cell functions like<br />

the contact between the T cell <strong>and</strong> the antigen presenting cell <strong>and</strong> T cell extravasation from<br />

the blood stream. Here, we report that phosphorylation of each cytoplasmic domain of the<br />

leukocyte integrin LFA-1 mediates different modes of integrin activation. Alpha-chain<br />

phosphorylation on Ser1140 is needed for conformational changes in the integrin after<br />

chemokine- or integrin lig<strong>and</strong>-induced activation, or activation induced by active Rap1<br />

(Rap1V12). In contrast, the beta-chain Thr758 phosphorylation mediates selective binding to<br />

14-3-3 proteins in response to inside-out activation through the T cell receptor, resulting in<br />

cytoskeletal rearrangements. Thus, site-specific phosphorylation of the integrin cytoplasmic<br />

domains is important for the dynamic regulation of these complex receptors in cells.<br />

- 295 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 12<br />

Molecular characterization of synovial fibroblasts from rheumatoid arthritis <strong>and</strong> their<br />

reponse to diverse anti-inflammatory drugs<br />

Valerie Gossye1, Karolien De Bosscher1, Dirk Elewaut2 <strong>and</strong> Guy Haegeman1<br />

1Laboratory for Eukaryotic Gene Expression <strong>and</strong> Signal Transduction (LEGEST),<br />

Department of Molecular Biology, Ghent University, Gent, Belgium. E-mail:<br />

valerie.gossye@ugent.be, karolien.debosscher@ugent.be, guy.haegeman@ugent.be;<br />

2Laboratory for Molecular Immunology <strong>and</strong> <strong>Inflammation</strong>, Department of<br />

Rheumatology, Ghent University Hospital, Gent, Belgium. E-mail:<br />

dirk.elewaut@ugent.be<br />

Objective. <strong>Inflammation</strong> is a central feature of many chronically developing inflammatory <strong>and</strong><br />

immune diseases, including rheumatoid arthritis (RA). Although the cause of RA is at present not<br />

completely understood, previous studies have shown that fibroblast-like synoviocytes (FLS) as part of<br />

a complex cellular network, are crucial for disease progression as well as for joint destruction.<br />

To date, the goal of treatment is to reduce joint inflammation <strong>and</strong> pain, maximize joint function, <strong>and</strong><br />

prevent joint destruction <strong>and</strong> deformity. However, current treatment regimens suffer major drawbacks.<br />

For instance, the usage of glucocorticoids (GCs) is overshadowed by severe side effects, such as<br />

diabetes, glaucoma <strong>and</strong> osteoporosis. Furthermore, occurrences of resistance to the therapeutic effects<br />

of corticosteroids, as well as disease reactivation after cessation of GC therapy, have been reported.<br />

Therefore, there is a particular need for the development of a new generation of drugs to treat these<br />

diseases.<br />

Hence, we undertook a detailed study of the molecular characteristics of primary FLS isolated<br />

from the inflamed synovium of RA patients <strong>and</strong> investigated whether anti-inflammatory drugs, such as<br />

GCs <strong>and</strong> Compound A (CpdA), a fully dissociated compound of plant origin for inflammatory gene<br />

repression (see De Bosscher et al. PNAS, 102, 15827-15832, 2005), exert their anti-inflammatory<br />

effects on these effector cells <strong>and</strong> examined their putative mode of action.<br />

Results <strong>and</strong> Conclusions<br />

1. Immunofluorescence studies demonstrate that, upon dexamethasone (DEX) or CpdA stimulation for<br />

1 h<strong>our</strong>, the glucocorticoid receptor (GR) shifts from the cytoplasm to the nucleus, indicating that GR<br />

translocation proceeds normally.<br />

2. RT-PCR results showed that in FLS93 <strong>and</strong> FLS118 TNF-induced Interleukin-6 mRNA expression<br />

levels were repressed by DEX more adequately than by CpdA, but vice versa in FLS120. Along the<br />

same line, CpdA was found to be more effective in downregulating RANTES in FLS93 <strong>and</strong> FLS120,<br />

whilst in FLS118 DEX appeared to be more effective. These data suggest that different patients show<br />

a different response/sensitivity towards different therapeutics. Furthermore, we can also conclude that<br />

in the same FLS different cytokines are differently modulated by either therapeutic agent.<br />

3. Finally, we observed in all investigated FLS a fast ‘homologous’ down-regulation of GR, already<br />

apparent after 7 h<strong>our</strong>s of DEX induction. In addition, Western blot analysis results showed that<br />

induction of FLS with TNF, in combination with DEX, results in downregulation of IkappaB-alpha<br />

protein expression, as opposed to the TNF-only condition. We thus confirmed previous results of <strong>our</strong><br />

group, demonstrating that in L929sA fibroblasts DEX stimulation does not lead to upregulation of<br />

IkappaB-alpha protein expression (De Bosscher et al., PNAS, 94, 13504-13509, 1997).<br />

Altogether, these findings highlight the complexity of molecular <strong>signaling</strong> cascades suitable for<br />

therapeutic intervention that underly the chronic inflammatory <strong>and</strong> destructive processes in RA.<br />

- 296 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 13<br />

The JAK1 SH2 domain plays a structural role in the upregulation of OncostatinM<br />

receptor surface expression<br />

Serge Haan1*, Simone Radtke1*, Angela Jörissen1*, Heike M. Hermanns1, S<strong>and</strong>ra<br />

Dieffenbach1, Tanja Smyczek1, Peter C. Heinrich1, Iris Behrmann2* <strong>and</strong> Claude<br />

Haan1,2*<br />

1 Institut für Biochemie, Uniklinik Aachen, Pauwelsstraße 30, 52074 Aachen, Germany<br />

2 Laboratoire de Biologie et Physiologie Intégrée, Fac. Sciences Techno. & Com.,<br />

Université du Luxemb<strong>our</strong>g, 162A avenue de la Faïencerie, L-1511 Luxemb<strong>our</strong>g<br />

*contributed equally<br />

The Janus kinase family of protein tyrosine kinases comprises f<strong>our</strong> mammalian members.<br />

Three, Jak1, Jak2 <strong>and</strong> Tyk2, are expressed in a wide variety of tissues, whereas Jak3<br />

expression is restricted to cells of the haematopoietic system. Jak1 is involved in signal<br />

transduction of the interferons IFN" <strong>and</strong> IFN$ <strong>and</strong> many other cytokines such as IL-6-type<br />

cytokines (IL-6, OSM, LIF, IL-11, CNTF, CT-1, CLC) signal via the receptor chains gp130,<br />

LIFR <strong>and</strong> OSMR. All these signal transducing receptors have been described to bind Jak1,<br />

Jak2 <strong>and</strong> Tyk2. Among these, Jak1 is essential for signal transduction. Interestingly the<br />

surface expression of the OncostatinM receptor (OSMR) has been described to be dependent<br />

on Jak binding. Recent data also suggest that Jak1 <strong>and</strong> gp130 form an indissociable complex<br />

which might best be compared to a receptor tyrosine kinase. It is still under debate which<br />

functional domains exist in the Jaks <strong>and</strong> the interplay of these domains in kinase activation is<br />

not well understood. Although the presence of an SH2 domain is discussed since the first<br />

decription of Jaks elaborate functional studies have not yet been published.<br />

In the present study we investigated the functionality of the Jak1 SH2 domain using an<br />

inactivating mutant of the Jak1 SH2 domain, Jak1-R466K. No effect on cytokine signal<br />

transduction could be observed. The R466K mutant also had no effect on receptor binding, on<br />

subcellular distribution as well as on Jak1-mediated upregulation of the OSMR. However, the<br />

SH2 domain seems to be structurally important to ensure cytokine receptor binding <strong>and</strong><br />

surface expression.<br />

(This work has been supported by the Deutsche Forschungsgemeinschaft (SFB542 TP B1 <strong>and</strong><br />

HA3433/1) <strong>and</strong> the Fonds der Chemischen Industrie (Frankfurt am Main, Germany))<br />

- 297 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 14<br />

Real-time analysis of Jak/STAT signal transduction in single cells<br />

Andreas Herrmann 1 , Michael Vogt 1 , Martin Mönnigmann 2 , Bernd Giese 1 , Michael<br />

Sommerauer 1 , Peter C. Heinrich 1 , Gerhard Müller-Newen 1<br />

1 Institut für Biochemie, Universitätsklinikum der RWTH Aachen, Pauwelsstraße 30,<br />

52057 Aachen, Germany. e-mail: mueller-newen@rwth-aachen.de<br />

2 Lehrstuhl für Prozesstechnik, RWTH Aachen, 52056 Aachen, Germany<br />

The Jak/STAT signal transduction pathway plays a central role in acute <strong>and</strong> chronic<br />

inflammation. The pro-inflammatory cytokine interleukin-6 (IL-6) signals through the<br />

cytokine receptor gp130. Upon activation, the receptor-associated tyrosine kinases of the<br />

Janus kinase (Jak) family phosphorylate STAT transcription factors. STAT3 (signal<br />

transducer <strong>and</strong> activator of transcription 3) is preferentially activated in response to IL-6.<br />

Activated STAT3 translocates into the nucleus where it induces target genes that exert<br />

multiple functions in inflammation <strong>and</strong> carcinogenesis.<br />

We have tagged the cytokine IL-6, its receptor gp130 <strong>and</strong> the transcription factor STAT3 with<br />

different fluorescent proteins (yellow fluorescent protein (YFP) or cyan fluorescent protein<br />

(CFP)). These fusion proteins enabled us to study lig<strong>and</strong>-binding <strong>and</strong> receptor dimerization<br />

(Giese et al. (2005) J. Cell. Sci. 118, 5129-40) as well as nuclear translocation of STAT3<br />

(Pranada et al. (2004) J. Biol. Chem. 279, 15114-23) in single cells by the use of confocal<br />

laser-scanning microscopy <strong>and</strong> advanced photo-bleaching techniques. We demonstrated that<br />

non-phosphorylated STAT3 constitutively shuttles between the cytoplasm <strong>and</strong> the nucleus.<br />

Persistent activation of STAT3 is observed in chronic inflammation <strong>and</strong> cancer. To analyze<br />

persistent activated STAT3 we cotransfected double labelled STAT3-CFP-YFP with v-Src<br />

resulting in constitutive tyrosine phosphorylation <strong>and</strong> nuclear accumulation of the fluorescent<br />

transcription factor. By bleaching selectively the YFP moiety of STAT3-CFP-YFP in one<br />

cellular compartment <strong>and</strong> by monitoring the distribution of the CFP <strong>and</strong> YFP fluorescence<br />

over time with high spatial resolution, we show that persistently activated STAT3 shuttles<br />

between cytoplasm <strong>and</strong> nucleus. Computational evaluation of the data by model-based<br />

parameter estimations revealed that activated STAT3 shuttles more rapidly than non-activated<br />

STAT3. We propose that inhibition of nucleocytoplasmic shuttling of persistently activated<br />

STAT proteins is a new target for the treatment of chronic inflammation <strong>and</strong> cancer.<br />

- 298 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 15<br />

Sulforaphane inhibits phorbol ester-induced COX-2 expression in human breast<br />

epithelial cells: I!B kinase # as a potential target<br />

Ha-Na Kim, Eun-Hee Kim, Hye-Kyung Na, Young-Joon Surh<br />

National Research Laboratory of Molecular Carcinogenesis <strong>and</strong> Chemoprevention,<br />

College of Pharmacy, Seoul National University, Seoul 151-742, South Korea<br />

Sulforaphane (SFN; 1-isothiocyanato-4-(methylsulfinyl)-butane), occurring naturally as a<br />

precursor glucosinolate in cruciferous vegetables, is a member of the isothiocyanate family of<br />

chemopreventive agents. Recent studies suggest that inflammation is causally linked to<br />

carcinogenesis. Cyclooxygenase-2 (COX-2), a rate-limiting enzyme in the biosynthesis of<br />

prostagl<strong>and</strong>ins, is inappropriately expressed in various cancers. In the present study, we<br />

investigated the effect of SFN on the 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced<br />

COX-2 expression <strong>and</strong> underlying molecular mechanisms in human breast epithelial (MCF-<br />

10A) cells. Treatment of MCF-10A cells with SFN (12.5 <strong>and</strong> 25 µM) significantly reduced<br />

TPA-induced COX-2 expression. The DNA binding of NF-!B <strong>and</strong> degradation of IkappaBalpha<br />

(I!B-") were suppressed by SFN in TPA-stimulated MCF-10A cells. SFN dosedependently<br />

inhibited TPA-induced I!B kinas # (IKK#) activity. In addition, SFN inhibited<br />

the phosphorylation of extracellular signal-regulated kinase (ERK) <strong>and</strong> the activity of ERK<br />

<strong>and</strong> p38 mitogen-activated protein kinase (MAPK) in TPA-treated MCF-10A cells. U0126<br />

<strong>and</strong> SB203580 that are specific inhibitors of ERK <strong>and</strong> p38 MAPK, respectively blunted TPAinduced<br />

COX-2 expression. In conclusion, SFN inhibited TPA-induced COX-2 expression<br />

<strong>and</strong> NF-!B activation in MCF-10A cells by targeting IKK# <strong>and</strong> MAPKs.<br />

- 299 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 16<br />

Redirecting adenovirus encoding dominant-negative inhibitor kappaB to inflamed<br />

endothelial cells inhibits endothelial gene expression<br />

J.M. Ku#do1, S.A. Asgeirsdottir1, A.R. Bellu2, M.G. Rots2, K.I. Ogawara3, T. Peneder1,<br />

C. Trautwein4, H. Haisma2, <strong>and</strong> G. Molema1<br />

University Medical Center Groningen, University of Groningen, The Netherl<strong>and</strong>s;<br />

1Department of Pathology <strong>and</strong> Laboratory Medicine, Medical Biology Section,<br />

Hanzeplein 1, 9713 GZ Groningen, j.m.kuldo@med.umcg.nl; 2Department of<br />

Therapeutic Gene Modulation; 3Department of Pharmaceutics, Faculty of<br />

Pharmaceutical Sciences, Okayama University, Japan; 4Medical School of Hanover,<br />

Germany<br />

In glomerulonephritis endothelium expresses inflammatory molecules responsible for harmful<br />

leukocyte infiltration. We propose that inhibition of expression of a broad spectrum of genes<br />

by endothelial cells will inhibit disease progression by strong suppression of leukocyte<br />

recruitment into the kidney. To accomplish this multiple gene inhibition, we used an<br />

adenovirus encoding a therapeutic transgene that interferes with nuclear factor kappaB<br />

<strong>signaling</strong>, a major pathway involved in inflammatory gene expression control. For retargeting<br />

to the activated endothelial cells, we introduced endothelial cell-specific anti-E-selectin or<br />

anti-VCAM-1 antibody-polyethylene glycol (PEG) modifications <strong>and</strong> studied effects of<br />

constructs in endothelial <strong>and</strong> messangial cells in vitro, as well as in vivo in mouse model of<br />

glomerulonephritis. In vitro retargeted adenoviruses selectively infected cytokine-activated<br />

endothelial cell <strong>and</strong> no transgene was detected in (activated or non-activated) messangial<br />

cells. E-selectin directed adenovirus was able to deliver more transgene to activated<br />

endothelium than VCAM-1 directed virus. In vivo, PEGylated, anti-E-selectin-retargeted<br />

adenovirus selectively homed to glomeruli in glomerulonephritis, resulting in downregulation<br />

of inflammatory genes at two days after start of disease. These studies demonstrate the<br />

potential of tropism-modified adenovirus to deliver therapeutic genes to endothelial cells in<br />

inflammation.<br />

- 300 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 17<br />

Novel Serine Protease-Induced Signaling in Human Peripheral Monocytes<br />

Yves Laumonnier, Tatiana Syrovets, Thomas Simmet<br />

Department of Pharmacology of Natural Products & Clinical Pharmacology, University<br />

of Ulm, D-89081 Ulm, Germany. E-mail: yves.laumonnier@uni-ulm.de<br />

Besides its important role in fibrinolysis, plasmin is a potent proinflammatory serine protease<br />

activating human peripheral monocytes. Plasmin induces lipid mediator release, expression of<br />

proinflammatory genes <strong>and</strong> chemotaxis. However, the exact nature of its receptor in<br />

monocytes remains elusive. In this context we have now investigated the role of annexin A2<br />

described as a surface protein involved into the binding of the inactive zymogen plasminogen.<br />

In contrast to neutrophils, primary monocytes respond to plasmin <strong>and</strong> express the annexin A2<br />

heterotetramer consisting of annexin A2 <strong>and</strong> S100A10 as shown by Western immunoblot,<br />

flow cytometry, immunofluorescence microscopy <strong>and</strong> co-immunoprecipitation. Using a<br />

biotin-labeled tri-functional plasmin cross-linker, we were able to biotinylate both annexin A2<br />

<strong>and</strong> S100A10 suggesting that plasmin is indeed binding to the subunits of the annexin A2<br />

heterotetramer. To assess the role of the annexin A2 heterotetramer as a receptor for plasmin<br />

<strong>signaling</strong>, plasmin-induced chemotaxis was analyzed after treatment with neutralizing<br />

antibodies. Antibodies directed against either annexin A2 or S100A10 impaired the plasmininduced<br />

<strong>signaling</strong> but not that induced by FMLP. Similarly, antisense oligodeoxynucleotides<br />

(ODN) directed against annexin A2 or S100A10, but not the respective sense controls,<br />

reduced the plasmin-induced chemotaxis, but not that to FMLP. Consistently, monocytes<br />

pretreated with antisense ODN showed a decreased release of the proinflammatory cytokine<br />

tumor necrosis factor (TNF)-alpha confirming that annexin A2 <strong>and</strong> S100A10 are essential for<br />

the plasmin-induced <strong>signaling</strong>. At the molecular level plasmin induced cleavage of the<br />

annexin A2 heterotetramer at position 27 as demonstrated by point mutation. Finally, we<br />

found that the plasmin-mediated cleavage triggers dissociation of the annexin A2<br />

heterotetramer.<br />

These data identify the annexin A2 heterotetramer as a <strong>signaling</strong> receptor for plasmin in<br />

human peripheral monocytes. The dissociation of the heterotetramer after the plasmininduced<br />

cleavage of annexin A2 is apparently the molecular initiator for the plasmin-induced<br />

proinflammatory <strong>signaling</strong> in human peripheral monocytes.<br />

- 301 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 18<br />

Molecular mechanisms underlying inflammatory responses to Helicobacter pylori<br />

infection<br />

Jeong-Sang Lee1, Ki-Baik Hahm2, Jeffrey A. Johnson3, <strong>and</strong> Young-Joon Surh1<br />

1National Research Laboratory of Molecular Carcinogenesis <strong>and</strong> Chemoprevention,<br />

College of Pharmacy, Seoul National University. Seoul 151-742, 2Genomic Research<br />

Center for Gastroenterology, School of Medicine, Ajou University, Suwon 442-749,<br />

South Korea, <strong>and</strong> 3School of Pharmacy, University of Wisconsin, Madison, WI, USA<br />

The Helicobacter pylori (H. pylori) were identified by Marshall <strong>and</strong> Warren in 1983. The H.<br />

pylori survive in the forbidding harsh acid environment of the stomach <strong>and</strong> duodenum by<br />

hiding in the mucus layer <strong>and</strong> neutralizing stomach acid in its local surrounding environment.<br />

Multiple lines of evidences suggest that H. pylori infection is one of the primary causes of<br />

gastritis <strong>and</strong> peptic ulcer, which are provoked by oxidative stress <strong>and</strong> inflammation. More<br />

than 50% of the world's population is infected by this bacterium. The H. pylori–induced<br />

inflammation has been implicated in the pathogenesis <strong>and</strong> progression of gastric cancer. In the<br />

present study, we investigated molecular mechanisms responsible for H. pylori-induced<br />

inflammation in human gastric cancer (AGS) cells. H. pylori induced inflammatory response<br />

accompanied up-regulation of cyclooxygenase-2 (COX-2) <strong>and</strong> inducible nitric oxide synthase<br />

(iNOS), <strong>and</strong> both the secretion <strong>and</strong> protein expression of interleukin-8 (IL-8) in AGS cells.<br />

The nuclear translocation <strong>and</strong> subsequent DNA binding of nuclear factor-kappaB (NF-!B), a<br />

eukaryotic transcription factor known to regulate aforementioned pro-inflammatory enzymes<br />

<strong>and</strong> mediators, were increased after H. pylori treatment. Similarly, the DNA binding of<br />

eukaryotic transcription factor activator protein-1 (AP-1) was induced by H. pylori treatment.<br />

In addition, c-Jun that is the major component of AP-1 was detected by super shift assay, <strong>and</strong><br />

the level of phospho-c-Jun was found to be increased. H. pylori infection accelerated the<br />

DNA binding of another transcription factor CREB, whose interacting partners were detected<br />

as CREB <strong>and</strong> CBP as determined by the super shift assay. In another experiment, H. pylori –<br />

infected AGS cells exhibited increased phosphorylation of upstream kinases, ERK <strong>and</strong> AKT.<br />

However, long term administration of H. pylori resulted in the activation of proliferating cell<br />

nuclear antigens (PCNA) <strong>and</strong> Terminal deoxynucleotidyl Transferase Biotin-dUTP Nick End<br />

Labeling (TUNEL) in C57BL6 mice, suggesting that H. pylori infection may create a balance<br />

between proliferation <strong>and</strong> apoptotic process in the stomach. Taken together, H. pylori<br />

treatment can cause the inflammatory response through up-regulation of proteins involved in<br />

pro-inflammatory signal transduction.<br />

- 302 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 19<br />

Lack of association of the cyclooxygenase-2 <strong>and</strong> inducible nitric oxide synthase gene<br />

polymorphism with risk of cervical cancer in Korean population.<br />

Taek Sang Lee a,b, Jae Weon Kima,b, Noh Hyun Parka,b, Soon Beom Kanga,b, Hyo<br />

Pyo Leea,b, Yong Sang Songa,b*<br />

aDepartment of Obstetrics <strong>and</strong> Gynecology, bCancer Research Institute, College of<br />

Medicine, Seoul National University, Seoul, 110-744, Korea<br />

E-mail: yssong@snu.ac.kr<br />

In recent years, plenty of studies have shown that nitric oxide (NO) <strong>and</strong> prostagl<strong>and</strong>in (PG) as<br />

the main inflammatory mediators are involved in various pathophysiological processes<br />

including inflammation <strong>and</strong> carcinogenesis. In this study, we have explored the impact of<br />

polymorphisms of two representative inflammatory mediators on the risk of cervical cancer.<br />

This study includes 176 cases of histologically confirmed invasive cervical cancer <strong>and</strong> 172<br />

healthy controls. Allele frequency of twelve SNPs of COX-2 <strong>and</strong> 5 of iNOS gene in 43<br />

normal populations were analyzed. 14 of 17 shows monomorphic or minimal minor allele<br />

frequency, <strong>and</strong> therefore we did not perform further analysis for them. Three SNPs ( 2 for<br />

COX-2, 1 for iNOS) were thus chosen for the study. Genotyping of three SNPs (SNP-rs5275<br />

in the untranslated region of exon 10 <strong>and</strong> rs5277 in the coding region of exon 3 of COX-2,<br />

<strong>and</strong> iNOS Ser608Leu allele C/T polymorphism within exon 16 of the iNOS reductase<br />

domain) was performed. No significant increase was found in any genotype of the COX-2 <strong>and</strong><br />

iNOS in cancer group. We analyzed the correlation between the genotypes <strong>and</strong> the<br />

clinicopathologic parameters of cervical cancer. No significant association of genotype was<br />

found with respect to each phenotype including clinical stage, lymph node (LN) status,<br />

histologic type, or parametrial invasion. In conclusion, <strong>our</strong> data shows the lack of significant<br />

evidence of association of the COX-2 <strong>and</strong> iNOS polymorphisms with cervical cancer in<br />

Korean population.<br />

- 303 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 20<br />

Helicobacter pylori induced IFN gamma production in CD8- NK cells<br />

Åsa Lindgren, Åsa Sjöling & Samuel Lundin<br />

Department of Medical Microbiology <strong>and</strong> Immunology, Göteborg University,<br />

Medicinaregatan 7A, 413 90 Göteborg, Sweden. E-mail: asa.lindgren@microbio.gu.se<br />

Helicobacter pylori is a gram-negative bacterium which causes a chronic infection of the<br />

gastric <strong>and</strong> duodenal mucosa <strong>and</strong> is associated with both development of ulcers <strong>and</strong> an<br />

increased risk of gastric adenocarcinomas.<br />

Natural killer (NK) cells are lymphocytes that play a critical role for the innate immunity<br />

against viral <strong>and</strong> bacterial infections as well as against tum<strong>our</strong> cells. NK cells are present in<br />

the mucosa of both healthy <strong>and</strong> H. pylori infected individuals <strong>and</strong> because of their ability to<br />

secrete IFN gamma (IFNg), the presence of NK cells in the gastric mucosa may indicate that<br />

this cell type is involved in the immune response to H. pylori infection.<br />

It has been shown that there are two different subpopulations of NK cells; CD8+ <strong>and</strong> CD8-<br />

<strong>and</strong> we found that CD8- dominates in the stomach mucosa. Our attention is thus directed<br />

towards elucidating how CD8- <strong>and</strong> CD8+ NK cells respond to H. pylori <strong>and</strong> if there are any<br />

differences between the two populations.<br />

Stimulations of NK cells with H. pylori have revealed that only the CD8- subpopulation<br />

seems to respond with IFNg production. Inhibition of MAPK p38 <strong>and</strong> of PI3K revealed<br />

opposite effects in CD8- NK cells, where inhibition of p38 gave rise to decreased <strong>and</strong><br />

inhibition of PI3K gave rise to increased IFNg secretion. We have also detected a degradation<br />

of the NFkB inhibitor IkBa upon stimulation of NK cells with H. pylori, which strengthens<br />

the hypothesis that the above mentioned signalling cascades are associated with the immune<br />

response to<br />

H. pylori. Upstream recognition of H. pylori <strong>and</strong> initiation of the response are also studied.<br />

We are currently studying Toll-like receptors (TLRs) that are bacterial recognition receptors<br />

that might be involved in the induction of IFNg production.<br />

In conclusion, we have shown that CD8- NK cells produce IFNg in response to H. pylori<br />

stimulation, in a p38-dependent manner. We believe that elucidation of the signalling<br />

pathways in NK cells activated by H. pylori will be important for the further study of the<br />

development of H. pylori-induced diseases.<br />

- 304 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 21<br />

Insulin primes human neutrophils for CCL3-induced migration: crucial role for JNK.<br />

Fabrizio Montecucco, Giordano Bianchi, Luciano Ottonello, Maria Bertolotto, Giorgio<br />

L. Viviani <strong>and</strong> Franco Dallegri.<br />

Clinic of Internal Medicine I, Department of Internal Medicine, University of Genoa<br />

Medical School, I-16132 Genoa, Italy. E-mail: fabriziomontecucco@tiscali.it<br />

Recent studies showing that neutrophil infiltration in unstable atherosclerotic lesions is<br />

associated with plaque rupture have suggested a pathogenetic role for neutrophils in<br />

atherosclerosis. Nevertheless, the mechanisms regulating the recruitment of neutrophils at<br />

sites of atherosclerotic lesions are still unknown. In this regard, it has been recently shown<br />

that insulin is involved in actin reorganization of neutrophil locomotory response. Thus, we<br />

studied the effects of > 50 microU/ml insulin, i.e. concentrations occurring in<br />

hyperinsulinemic conditions characterized by an increased atherosclerotic risk, on neutrophil<br />

migration to CCL3, a chemokine produced in atherosclerotic plaques <strong>and</strong> considered to be<br />

incapable of attracting human neutrophils. Neutrophils from healthy volunteers became<br />

capable of migrating to CCL3 after a short (15 min) exposure to 300 microU/ml insulin.<br />

Accordingly, although no [Ca2+]i mobilization was observed in response to CCL3 or insulin,<br />

the combination of insulin <strong>and</strong> CCL3 stimulation results in a well detectable [Ca2+]i spike.<br />

Furthermore, the exposure of neutrophils to serum from hyperinsulinemic <strong>and</strong> euglycaemic<br />

adult patients with metabolic syndrome (as defined by ATP III <strong>and</strong> WHO criteria), induced<br />

neutrophil responsiveness to CCL3. A strict correlation (p:


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 22<br />

Streptococcus pneumoniae-induced p38 MAPK <strong>and</strong> NF-kappaB dependent COX-2<br />

expression in human lung epithelium<br />

P. Dje N´Guessan, S. Hippenstiel, M. O. Etouem, J. Zahlten, W. Beermann, D. Lindner,<br />

B. Opitz, M. Witzenrath, S. Rosseau, N. Suttorp, B. Schmeck<br />

Department of Internal Medicine/Infectious Diseases, Charité – Universitätsmedizin<br />

Berlin, 13353 Berlin, E-mail: Stefan.hippenstiel@charite.de<br />

S. pneumoniae is a major cause of community-acquired pneumonia <strong>and</strong> death due to<br />

infectious diseases in industrialized countries. Cyclooxygenase (COX) derived prostagl<strong>and</strong>ins<br />

like prostagl<strong>and</strong>in E2 (PGE2) are considered as important regulators of lung function. Herein<br />

we tested the hypothesis that pneumococci induced COX-2 dependent PGE2 production in<br />

pulmonary epithelial cells. Pneumococci infected human pulmonary epithelial BEAS-2B cells<br />

released PGE2. Expression of COX-2 but not COX-1 was dose- <strong>and</strong> time-dependently<br />

increased in S. pneumoniae-infected BEAS-2B cells as well as in lungs of mice with<br />

pneumococcal pneumonia. S. pneumoniae induced degradation of IkappaBalpha <strong>and</strong> DNAbinding<br />

of NF-kappaB. Specific inhibition of the IKK kinase complex blocked pneumococciinduced<br />

PGE2 release <strong>and</strong> COX-2 expression. p38 MAP kinase <strong>and</strong> cJun-NH2-terminal<br />

kinase (JNK) were activated in pneumococci infected cells. PGE2 release <strong>and</strong> COX-2<br />

expression was reduced by p38 MAP kinase-inhibion but not by JNK-inhibition. Dominant<br />

negative mutants of p38 MAP kinase isoforms alpha, beta2, gamma, <strong>and</strong> delta blocked S.<br />

pneumoniae-induced NF-kaapB activation. In addition, recruitment of NF-kappaB subunit<br />

p65/RelA <strong>and</strong> RNA polymerase II to the cox2 promoter depended on p38 MAP kinase but not<br />

on JNK activity. In summary, p38 MAP kinase <strong>and</strong> NF-kappaB controlled COX-2 expression<br />

<strong>and</strong> subsequent PGE2 release by lung epithelial cells may contribute significantly to the host<br />

response in pneumococcal pneumonia.<br />

Supported by German Federal Ministry of Education <strong>and</strong> Research, Competence Network<br />

CAPNETZ.<br />

- 306 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 23<br />

Non-receptor tyrosine kinases play a role in zymosan (yeast) induced macrophage<br />

activation<br />

S<strong>and</strong>ra Olsson <strong>and</strong> Roger Sundler<br />

Macrophage Signaling, Exp. Med. Science, Lund University, BMC B12, 221 84 Lund,<br />

Sweden. E-mail: S<strong>and</strong>ra.Olsson@med.lu.se, Roger.Sundler@med.lu.se<br />

Macrophages play an essential role in defending us against microorganisms, by phagocytosis<br />

<strong>and</strong> by production of proinflammatory mediators, such as TNF-" <strong>and</strong> eicosanoids. The<br />

proinflammatory mediators also play a role in chronic inflammation. It is therefore of<br />

importance to study the <strong>signaling</strong> to regulation of their production.<br />

Fungi are typical microorganisms causing opportunistic infections <strong>and</strong> zymosan beads,<br />

carbohydrate-rich cell wall preparations from Saccharomyces cerevisea, are often used to<br />

study the interaction of yeast with the innate immune system. Members of the Src family<br />

kinases are known to play a critical role in many <strong>signaling</strong> pathways, with a putative role in<br />

inflammation. We therefore studied their role in zymosan-induced activation of macrophages<br />

using different Src family kinase inhibitors. Our results demonstrate that Src kinases play<br />

differential roles in both arachidonate release for eicosanoid formation <strong>and</strong> TNF-"<br />

production. Zymosan may signal through several receptors <strong>and</strong> the postulated receptors<br />

include mannose receptor, Toll-like receptor 2 (TLR2), #-glucan receptors (Dectins) <strong>and</strong><br />

SIGNR1. We show that the Dectin-1 receptor is involved in zymosan <strong>signaling</strong> to arachidonic<br />

acid release, while the receptor used for TNF-" formation may be different. Tec kinase, a<br />

downstream kinase of Src, also seems to be involved in zymosan-induced activation of<br />

macrophages, since it is phosphorylated upon zymosan stimulation <strong>and</strong> arachidonate release is<br />

reduced after inhibition of Tec. MAP kinases are able to contribute to the production of<br />

eicosanoids <strong>and</strong> TNF-" <strong>and</strong> we show here that inhibition of Src kinases, but not Btk,<br />

decreased the zymosan-induced phosphorylation of MAP kinases. These data indicate that<br />

non-receptor tyrosine kinases such as the Src family kinases <strong>and</strong> Btk play a role in zymosaninduced<br />

formation of inflammatory mediators in macrophages.<br />

- 307 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 24<br />

Microsomal prostagl<strong>and</strong>in E synthase-1 expression induced by TNFalpha does not<br />

involve PKC or p38 MAP kinase in gingival fibroblasts<br />

Tove Olsson, Tomomi Kawakami <strong>and</strong> Tülay Yucel-Lindberg<br />

Department of Pediatric Dentistry, Karolinska Institutet, Box 4064, SE-141 04<br />

Huddinge, Sweden. E-mail: tove.olsson@ki.se; tomomi.kawakami@ki.se;<br />

tulay.lindberg@ki.se<br />

Prostagl<strong>and</strong>in E2 (PGE2) plays a pivotal role in several inflammatory conditions including<br />

rheumatoid arthritis <strong>and</strong> periodontitis. The inducible microsomal prostagl<strong>and</strong>in E synthase-1<br />

(mPGES-1) is the recently identified terminal enzyme regulating the synthesis of PGE2. We<br />

have previously reported that the cytokine tumor necrosis factor alpha (TNFalpha) induces the<br />

expression of mPGES-1 in human gingival fibroblasts. In this study we further investigated<br />

the regulation of mPGES-1 with special regards to protein kinase C (PKC) <strong>and</strong> p38 MAP<br />

kinase (p38 MAPK) in relation to PGE2. The results showed that TNFalpha induced the<br />

expression of mPGES-1 in gingival fibroblasts accompanied by increased PGE2 production.<br />

Treatment of the cells with the PKC activator PMA (phorbol-12-myristate-13-acetate)<br />

increased the production of PGE2 as well as upregulated the stimulatory effect of TNFalpha,<br />

without increasing mPGES-1 expression. In addition, the PKC inhibitor Bisindolylmaleimide<br />

markedly decreased the TNFalpha-induced PGE2 production but not mPGES-1 expression.<br />

Furthermore, the p38 MAPK inhibitor SB203580 caused an abrogation of the TNFalphainduced<br />

PGE2 production whereas mPGES-1 expression remained unaffected. These findings<br />

indicate a lack of co-regulation of mPGES-1 <strong>and</strong> its product PGE2 regarding PKC <strong>and</strong> p38<br />

MAPK signal pathways in human gingival fibroblasts.<br />

- 308 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 25<br />

The effect of Epidermal Growth Factor (EGF) implantation in incisional wounds of<br />

rabbits on leucocyte chemotaxis<br />

Nesrin Özsoy 1, $ule Co%kun 2, Nursel Gül 1<br />

1 Department of Biology, Faculty of Science, Ankara University, 06100, T<strong>and</strong>o&an,<br />

Ankara, Turkey. E-mail: ozsoy@science.ankara.edu.tr<br />

2 Department of Biology, Faculty of Science <strong>and</strong> Art, Gazi University, 06500 Be%evler,<br />

Ankara, Turkey.<br />

Epidermal growth factor (EGF) is mitogenic for many cells of mesodermal <strong>and</strong> ectodermal<br />

origin. It is a polypeptide that increases ion change, glycolysis, RNA <strong>and</strong> DNA production<br />

<strong>and</strong> wound healing in effected cells. It especially promotes cell migration into the wound area.<br />

EGF plays a fundamental role in the process of wound repair by stimulating chemotaxis. In<br />

this study, the impact of EGF to the leucocyte chemotaxis at the wounded tissues has been<br />

studied. Mucous membranes of rabbits were wounded by incision. EGF formulation was<br />

submucosally localized <strong>and</strong> afterwards, the wound was closed with double suture. The<br />

significance of this study is that it is the first wound model applied to the mucosa of rabbits.<br />

Oral implantation of EGF is original in this wound model. In this research, New-Zeal<strong>and</strong> male<br />

rabbits were used weighting to 2.5 ± 0.4 kg. After submucosal incisions were made, the<br />

rabbits were divided into three groups. 1- Only incision group 2- polyethylene glycol (PEG)<br />

implanted group 3-EGF into PEG implanted wounds. On the 5th day of operation, the rabbits<br />

were killed by excess of sodium pentobarbitale anesthesia <strong>and</strong> the wounded tissue was<br />

immediately removed for Transmission Electron Microscope (TEM) preparation. The tissues<br />

were embedded into araldite <strong>and</strong> blocked. Semi-thin <strong>and</strong> thin sections were prepared from the<br />

blocks, <strong>and</strong> leucocyte chemotaxis was investigated. It was observed that, in the connective<br />

tissues of the “incision only” group, leucocyte cells were in minority. Leucocytes<br />

accumulated in the connective tissues of the group that received both incision <strong>and</strong> EGF<br />

application. That all leucocytes were observable attracted attention. It was found that EGF<br />

stimulated leucocyte chemotaxis during wound healing <strong>and</strong> attracted all leucocytes to the<br />

healing area.<br />

- 309 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 26<br />

Regulations of Notch2, 3, 4 <strong>and</strong> Dll1 expression by TNF" in Endothelial Cells are<br />

specifically dependant on PI3K, NFkB <strong>and</strong> JNK MAPK pathways.<br />

Thibaut Quillard, Stéphanie Coupel, Juliette Fitau <strong>and</strong> Béatrice Charreau<br />

INSERM, U643, Nantes, France ; ITERT, Institut de Transplantation Et de Recherche<br />

en Transplantation, CHU, Nantes, France ; Université de Nantes, UFR de médecine,<br />

Nantes, France. E-mail : thibaut.quillard@univ-nantes.fr<br />

Notch <strong>signaling</strong> pathway is a key player in cell communication through molecular cell/cell<br />

interactions. Vertebrates express multiple Notch receptors (Notch1 - 4) <strong>and</strong> lig<strong>and</strong>s including<br />

Delta-like (Dll) (1-4) <strong>and</strong> Jagged 1 & 2. Notch plays a role in adult in several contexts<br />

involving cell plasticity including vascular morphogenesis <strong>and</strong> remodeling. However,<br />

regulation of the Notch receptors <strong>and</strong> lig<strong>and</strong>s in vascular endothelial cells (EC) upon<br />

inflammation remains unknown. AIM : In the present study, we investigated the expression<br />

<strong>and</strong> regulation of Notch receptors <strong>and</strong> lig<strong>and</strong>s in EC upon activation with TNF" in vitro <strong>and</strong><br />

in vivo. METHODS & RESULTS : Expression of Notch receptors <strong>and</strong> lig<strong>and</strong>s was<br />

investigated in primary cultures of human EC, treated with TNF", by quantitative RT-PCR<br />

<strong>and</strong> Western Blotting. We show that TNF" down-regulated Notch1,3,4 <strong>and</strong> up-regulated<br />

Notch2 <strong>and</strong> the lig<strong>and</strong> Dll1. A concomitant decrease in mRNA level for effector genes Herp1<br />

<strong>and</strong> Hes1 was observed reflecting a reduced activity of Notch <strong>signaling</strong>. Similar regulations<br />

were obtained in HUVEC <strong>and</strong> vascular SMC. Moreover, comparison with IL1", IFN$ <strong>and</strong><br />

VEGF indicate a regulation pattern specific of inflammatory stimuli. In EC, TNF" activates<br />

several <strong>signaling</strong> pathways including the PI3Kinase (PI3K), NF!B <strong>and</strong> JNK MAPK<br />

pathways. The respective involvement of these <strong>signaling</strong> pathways in Notch regulations<br />

mediated by TNF" in EC was examined. Our findings suggest that TNF"-mediated<br />

regulation of Notch2 transcription was dependant of PI3K as changes for Notch3, 4 <strong>and</strong> Dll1<br />

mRNA levels involved NF!B <strong>and</strong> JNK MAPK. Notch regulations were further confirmed in<br />

vivo, in rats treated with TNF"..<br />

CONCLUSION : Our results show for the first time that inflammation triggers a dysregulated<br />

expression of Notch molecules in endothelium that could contribute to EC/immune cells<br />

interactions <strong>and</strong> promote vascular remodeling as observed in chronic inflammatory disorders.<br />

- 310 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 27<br />

Melphalan reduces the severity of experimental colitis in mice by blocking tumor<br />

necrosis factor-alpha <strong>signaling</strong> pathway<br />

Galina V. Shmarina, Alex<strong>and</strong>er L. Pukhalsky, Vladimir A. Alioshkin <strong>and</strong> Alex<br />

Sabelnikov<br />

Research Centre for Medical Genetics, Moscow 115478, Gabrichevsky Institute of<br />

Epidemilogy <strong>and</strong> Microbilogy, Moscow 125212, Russia, East Carolina University,<br />

Greenvill, NC 27858, U.S.A. E-mail: osugariver@medgen.ru<br />

In acute DSS-induced colitis nuclear factor (NF)-kappaB-dependent inflammatory cytokines<br />

including tumor necrosis factor (TNF)-alpha are known to be up-regulated. We examined the<br />

effects of alkylating drug melphalan on the sensitivity of mouse fibroblasts to TNF-alphainduced<br />

cytitoxicity in vitro <strong>and</strong> on intestinal inflammation <strong>and</strong> NF-kappa B activation in<br />

vivo. In vitro, low concentrations of melphalan protected fibroblastoid cells (L929) against<br />

TNF-alpha-mediated apoptosis. The cells were pre-incubated with 0.0001 mg/ml of<br />

melphalan for 1 h <strong>and</strong> then treated with recombinant TNF-alpha. A significant decrease in the<br />

number of dead cells in comparison to the control (TNF-alpha treated cells without preincubation<br />

with melphalan) was observed. In vivo, daily administration of melphalan<br />

markedly reduced the severity of murine experimental colitis as determined by clinical <strong>and</strong><br />

quantitative histological criteria. Male BALB/c mice were exposed to 5 % (w/v) dextran<br />

sulfate sodium (DSS) dissolved in drinking water for 5 days with or without daily melphalan<br />

injections (0.025 mg/kg body weight, i.p.). Time-matched controls consisted of mice received<br />

melphalan only, <strong>and</strong> those injected with phosphate buffered saline. At day 6 the mice were<br />

sacrificed <strong>and</strong> the general signs of colitis had been evaluated. DSS-mice exhibited a<br />

significant loss of the intestine length, pronounced elevation in blood white cells <strong>and</strong> marked<br />

increase in relative spleen weight. Such changes were not so evident in DSS-mice treated with<br />

melphalan. Histological analysis of colon biopsies showed that melphalan protected against<br />

both the neutrophil infiltration <strong>and</strong> mucosal destruction. Furthermore, colonic NF-kappaB<br />

DNA-binding activity, up-regulated in DSS-induced colitis, was suppressed by melphalan<br />

treatment. Taken together, these results suggest that alkylating drug melphalan applied in noncytotoxic<br />

concentrations may reduce the severity of experimental colitis in mice by blocking<br />

TNF-alpha <strong>signaling</strong> pathway.<br />

- 311 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 28<br />

Experimental sepsis: characteristics of activated macrophages <strong>and</strong> apoptotic cells in the<br />

rats spleen<br />

Helle E. Simovart1, Andres Arend1, Kersti Kokk1, Helle Tapfer1, Marina Aunapuu1,<br />

Elle Poldoja1, Gunnar Selstam2, Aade Liigant1<br />

1Department of Anatomy, University of Tartu, Biomedicum , 19, Ravila St. Tartu<br />

50411, Estonia. E-mail : helle-evi.simovart@ut.ee<br />

2Department of Molecular Biology, University of Umea, Sweden<br />

Sepsis, being characterized by massive translocation of bacteria into tissues, induces the<br />

suppression of the function of both leukocytes <strong>and</strong> macrophages.<br />

The aim of the study was to count activated macrophages (AM) <strong>and</strong> apoptotic (Ao) cells in<br />

the rat spleen during the period of experimental sepsis <strong>and</strong> to clarify the associations of these<br />

parameters with the leukocyte migration <strong>and</strong> the bacterial translocation into different organs.<br />

The Wistar rats were intraperitoneally inoculated with E. coli <strong>and</strong> were sacrificed after 2, 6,<br />

24, 48 <strong>and</strong> 120 h<strong>our</strong>s. Bacteria <strong>and</strong> leukocytes in tissues were specifically stained. AM were<br />

identified by immunohistological staining <strong>and</strong> Ao cells were detected with the «In Situ Cell<br />

Death Detection Kit».<br />

The high counts of E. coli were strongly associated with a low level of the total counts of<br />

leukocytes at 6h, accompanied by the high translocation of microbes <strong>and</strong> migration of<br />

leukocytes into tissues. In the spleen, the count of AM was highest at 24h after the inoculation<br />

with E. coli, at the same time the Ao cell count began to rise <strong>and</strong> achieved the highest level 24<br />

h later when the AM amount has diminished.<br />

In conclusion, <strong>our</strong> investigation indicates that the molecular peculiarities of macrophages <strong>and</strong><br />

their response to the inflammation process are tissue-specific. In the spleen the activation<br />

process was remarkable at the late stage of sepsis accompanied by a high count of apoptotic<br />

cells, the process which involved the haematopoietic cells, localized in the spleen as<br />

macrophages themselves.<br />

- 312 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 29<br />

Angiotensin II Type 1 Receptor as a target for the resolution of disease induced<br />

wounding<br />

Gary R. Smith<br />

Chemistry Department, The Open University, Walton Hall, Milton Keynes MK7 6AA,<br />

UK.<br />

E-mail : gary.smith@persescomms.com<br />

The critical role of inappropriate inflammation is becoming accepted in many diseases,<br />

including cardiovascular diseases, inflammatory <strong>and</strong> autoimmune disorders,<br />

neurodegenerative conditions, infection <strong>and</strong> cancer.<br />

A review of laboratory <strong>and</strong> clinical evidence demonstrates that cancer up-regulates the<br />

angiotensin II type 1 (AT1) receptor through oxidative, hypoxic <strong>and</strong> steer stress mechanisms,<br />

thereby triggering a wound response that remodels surrounding tissue <strong>and</strong> subdues the<br />

immune system.<br />

With regard to cancer being a systemic disease, an examination of supporting evidence for a<br />

systemic role of AT1 in relationship to the wound response is presented as a logical<br />

progression. The evidence suggests that regulation of the mutually antagonistic angiotensin II<br />

receptors (AT1 <strong>and</strong> AT2) is an essential process in the management of ‘chronic’ inflammation<br />

<strong>and</strong> wound recovery, <strong>and</strong> that an imbalance in the expression of these receptors will lead to<br />

progress in disease conditions.<br />

Manipulation of the angiotensin system with existing anti-hypertensive drugs has been found<br />

to have therapeutic effect in clinical studies in type 2 diabetes, arteriosclerosis, renal disease,<br />

sarcoidosis <strong>and</strong> hormone refractory prostate cancer. It is anticipated that in the foreseeable<br />

future Angiotensin Receptor Blockade will provide a new approach to the treatment of many<br />

of the diseases that afflict mankind.<br />

- 313 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 30<br />

A role for Src <strong>and</strong> receptor tyrosine kinases but not for Janus kinases in SOCS3 tyrosine<br />

phosphorylation<br />

Ulrike Sommer1, Christine Schmid1, Radoslaw M. Sobota1, Ute Lehmann1, James A.<br />

Johnston2, Fred Schaper1, Peter C. Heinrich1 <strong>and</strong> Serge Haan1<br />

1 Institut für Biochemie, Rheinisch-Westfälische Technische Hochschule Aachen,<br />

Pauwelsstrasse 30, D-52074 Aachen, Germany<br />

2 Dept. Immunology, Queen's University Belfast, 97 Lisburn Rd., Belfast BT9 7BL,<br />

Northern Irel<strong>and</strong><br />

The suppressors of cytokine signalling (SOCS) are negative feedback inhibitors of cytokine<br />

signal transduction. SOCS3 is a key negative regulator of IL-6 signal transduction.<br />

Furthermore, SOCS3 was shown to be phosphorylated upon treatment of cells with IL-2 <strong>and</strong><br />

this has been reported to regulate its function <strong>and</strong> half-life. We set out to investigate whether<br />

SOCS3 phosphorylation may play a role in IL-6 signalling. Tyrosine phosphorylated SOCS3<br />

was detected upon treatment of mouse embryonic fibroblasts (MEF) with IL 6. Interestingly,<br />

the observed SOCS3 phosphorylation does not require SOCS3 recruitment to phosphotyrosine<br />

pY759 of gp130 <strong>and</strong> the kinetics of SOCS3 phosphorylation do not match the activation<br />

kinetics of the Janus kinases. This suggests that other kinases may be involved in SOCS3<br />

phosphorylation. Using Src <strong>and</strong> Janus kinase inhibitors as well as Src kinase deficient MEF<br />

cells, we provide evidence that Src kinases, which we found to be constitutively active in<br />

these cells, are involved in the phosphorylation of IL-6 induced SOCS3. In addition, we found<br />

that receptor tyrosine kinases (RTKs) such as PDGFR or EGFR can very potently<br />

phosphorylate IL-6 induced SOCS3. Taken together, these results suggest that SOCS3<br />

phosphorylation is not a JAK-mediated phenomenom but is dependent on the activity of other<br />

kinases such as Src kinases or RTKs which can either be constitutively active or activated by<br />

an additional stimulus.<br />

This work has been supported by the Deutsche Forschungsgemeinschaft (HA3433/3) <strong>and</strong> the<br />

Fonds der Chemischen Industrie (Frankfurt am Main, Germany).<br />

- 314 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 31<br />

Increased Cyclooxygenase-2 Expression Associated with Inflammatory Cellular<br />

Infiltration in Elderly Patients with Vulvar Cancer<br />

Taek Sang Lee a,b, Jae Weon Kima,b, Jae Kyung Wonc, Noh Hyun Parka,b,<br />

In Ae Parkc, Soon Beom Kanga,b, Hyo Pyo Leea,b, Yong Sang Songa,b*<br />

aDepartment of Obstetrics <strong>and</strong> Gynecology, bCancer Research Institute, cDepartment<br />

of pathology, College of Medicine, Seoul National University, Seoul, 110-744, Korea<br />

E-mail: yssong@snu.ac.kr<br />

As the relationship between inflammation <strong>and</strong> carcinogenesis grows stronger, the role of<br />

cyclooxygenase-2 (COX-2), <strong>and</strong> epidermal growth factor (EGFR) has been highlighted in the<br />

pathogenesis <strong>and</strong> progression of human cancer. In view of the fact that vulvar cancer is<br />

characterized by pre-cancerous inflammatory changes in elderly patients, the expressions of<br />

COX-2 <strong>and</strong> EGFR are expected to show different pattern of distribution according to age <strong>and</strong><br />

other prognostic factors. To verify whether there was a relationship between their expressions<br />

<strong>and</strong> clinico-pathologic parameters in vulvar cancer, we investigated the inflammatory cellular<br />

infiltration <strong>and</strong> the expression of COX-2 <strong>and</strong> EGFR <strong>and</strong> by immunohistochemical analysis.<br />

Eleven of 19 samples (57.8%) were stained positive for COX-2, <strong>and</strong> 17(89.4%) for EGFR.<br />

The portion of inflammatory cellular infiltration in adjacent normal tissue was also higher in<br />

old age group <strong>and</strong> showed strong correlation with COX2 positivity (p=0.002). Furthermore,<br />

COX-2 expression was significantly more frequent in over 60 year old group than in under 50<br />

(p=0.009). COX-2 revealed high expression in the moderately <strong>and</strong> well differentiated cases,<br />

whereas, poorly differentiated carcinoma were negative (p=0.023). On the other h<strong>and</strong>, EGFR<br />

expression was not differently distributed according to stage, age, tumor grading, or presence<br />

of lymph node metastasis. Our study suggests that vulvar cancer in elderly patients may be<br />

associated with inflammation, <strong>and</strong> thus with increased COX-2 expression. In light of these<br />

data, a clinical trial designed to assess the addition of COX-2 targeted therapy to conventional<br />

treatment in vulvar cancer seemed worthwhile, avoiding serious surgical morbidity in the<br />

elderly patients.<br />

- 315 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 32<br />

Role of the host body cytokine TNF-alpha in the mechanism of bacterial growth<br />

activation.<br />

Alex<strong>and</strong>ra A. Tomova, Yulia M. Romanova, Alex<strong>and</strong>r L. Gintsburg<br />

Laboratory of Genetic Engineering of Pathogenic Microorganisms, The Gamaleya<br />

Research Institute of Epidemiology <strong>and</strong> Mycrobiology, Russian Academy of Medical<br />

sciences, 18 Gamaley street, Moscow 123098, Russia; e-mail: altomova@abv.bg<br />

Resting or nonculturable microbes are thought to represent about 60% of the biomass on<br />

Earth. How to stimulate such microbes to exit from dormant state may aid the culturing of<br />

such organisms. Why <strong>and</strong> how nonculturable forms of bacteria resuscitate <strong>and</strong> propagate in<br />

the host body with a lot of defense factors of immunity? The aim of this work was to identify<br />

the role of the host body cytokine TNF-" in the mechanism of bacterial growth activation in<br />

vitro <strong>and</strong> in vivo. During evolutional coexistence of micro- <strong>and</strong> macroorganisms pathogenic<br />

bacteria developed the adaptive mechanisms that allowed them to utilize even immune<br />

defense factors (cytokines) as growth activation factors. We studied the dynamics of the<br />

reproduction of Salmonella vegetative forms <strong>and</strong> the resuscitation of Salmonella<br />

nonculturable forms in infected animals. We demonstrated that the increase in the level of<br />

TNF-" in host body leads to the activation of the Salmonella cell’s growth. We have<br />

discovered that the incubation of Salmonella cells in vitro in a media complemented with<br />

TNF-" leads to a higher growth rate of bacterial cells. To realize the mechanism of<br />

pathogenic bacteria activation in host body we investigated the growth rates of vegetative <strong>and</strong><br />

resuscitation of Salmonella nonculturable forms after action of TNF-". Direct interaction of<br />

the bacteria with cytokines in vitro during incubation the cells with TNF-" must be occurring<br />

through the bacterial receptor <strong>and</strong> the increase in the bacterial growth rate in the presence of<br />

cytokines must be a result of cytokine induced bacterial cell gene <strong>and</strong> protein expression. By<br />

the method of molecular display we identified several genes that were significantly up<br />

regulated in the presence of TNF-". By the proteome analysis of S. typhimurium cultures<br />

incubated with/without addition of TNF-" we have shown the increase expression of some<br />

proteins. We identified the protein that might participate in the binding of Salmonella to<br />

cytokine in vitro. The investigation of mechanisms of reproduction <strong>and</strong> growth activation of<br />

culturable <strong>and</strong> nonculturable forms of pathogenic bacteria in host body is very important <strong>and</strong><br />

actual problem.<br />

- 316 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 33<br />

Receptor interacting protein RIP1, a crucial initiator of inflammation <strong>and</strong> necrotic cell<br />

death<br />

Authors: Tom V<strong>and</strong>en Berghe, Miki Kalai, Nele Festjens, Jill Demeulemeester, Kathleen<br />

D’Hondt <strong>and</strong> Peter V<strong>and</strong>enabeele<br />

Molecular Signaling <strong>and</strong> Cell Death Unit, Department of Molecular Biomedical<br />

Research, VIB, Ghent University, Technologiepark 927, 9052 Zwijnaarde, Belgium<br />

Receptor interacting protein (RIP1) is recruited to tumor necrosis factor-" receptor 1<br />

(TNFR1) complex upon stimulation <strong>and</strong> known to play a crucial role in the receptor-induced<br />

NF-!B activation. Interaction of RIP1 with the scaffold protein Hsp90 protects the adaptor<br />

kinase from proteasome-dependent degradation. We showed that pretreatment of L929<br />

fibrosarcoma cells with the Hsp90 inhibitor geldanamycin, induces a shift of TNF-induced<br />

necrosis to apoptosis, suggesting a crucial role of RIP1 also in necrotic <strong>signaling</strong>. Using an<br />

RNA interference approach, we demonstrate that RIP1 is crucial for the initiation of necrosis.<br />

Moreover, retroviral transduction of kinase-death mutants shows that RIP1-induced necrosis<br />

is dependent on its kinase activity. The importance of RIP1 has also been shown in FasL- <strong>and</strong><br />

dsRNA-induced caspase-independent cell death. Analogous to the formation of the<br />

apoptosome, one can propose the formation of a “necrosome-complex” in which the<br />

activation of RIP1 is a crucial event. We are currently trying to unravel the activation<br />

mechanism of RIP1 <strong>and</strong> how RIP1 recruitment finally triggers downstream execution<br />

mechamisms.<br />

- 317 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 34<br />

ABIN-3 is a novel LPS/TNF-inducible inhibitor of NF-kB activation<br />

Lynn Verstrepen, Andy Wullaert, Sofie Van Huffel&, Mira Haegman, Matthew<br />

S<strong>and</strong>ers, Karen Heyninck <strong>and</strong> Rudi Beyaert<br />

Unit of Molecular Signal Transduction in <strong>Inflammation</strong>, Department for Molecular<br />

Biomedical Research, Fl<strong>and</strong>ers Interuniversity Institute for Biotechnology (VIB), Ghent<br />

University, Technologiepark 927, B-9052 Ghent (Zwijnaarde), Belgium, E-mail:<br />

lynnv@dmbr.ugent.be<br />

& Current address: Membrane Biology Lab (HWJ), Institute of Molecular <strong>and</strong> Cell<br />

Biology, 61 Biopolis Drive, Proteos, Singapore 138673<br />

The transcription factor NF-kB is responsible for upregulation of different genes involved in<br />

inflammation, immune responses, differentiation, proliferation <strong>and</strong> apoptosis. Dysregulation<br />

of NF-kB activation plays an important role in a number of diseases like asthma, Crohn’s<br />

disease, <strong>and</strong> rheumatoid arthritis. Therefore, a tight regulation of the NF-kB <strong>signaling</strong><br />

pathway is necessary. Via in silico cloning, we identified a novel protein (termed ABIN-3)<br />

that shows partial sequence homology to ABIN-1 <strong>and</strong> ABIN-2, which were previously<br />

identified in <strong>our</strong> group as NF-kB modulatory proteins. Similar to the other ABIN’s, ABIN-3<br />

bound the ubiquitin-editing protein A20 <strong>and</strong> inhibited NF-kB activation induced by TNF, IL-<br />

1 <strong>and</strong> TPA upon overexpression. ABIN-3 was also found to inhibit the LPS/TLR4-initiated<br />

pathway to NF-kB. Unlike the other ABIN’s, ABIN-3 expression could only be detected<br />

upon stimulation with LPS <strong>and</strong> to a lesser extend with TNF. Furthermore, we found a NF-kB<br />

site in the promoter of ABIN-3. Luciferase-reporter <strong>and</strong> gelretardation assays showed that the<br />

ABIN-3 promoter was activated upon stimulation with TNF or LPS in a NF-kB dependent<br />

way. Taken togheter, these results implicate ABIN-3 as a novel negative feedback regulator<br />

of LPS-/TNF- induced NF-kB activation.<br />

- 318 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 35<br />

Stimulus Specificity of Gene Expression Programs Determined by Temporal Control of<br />

IKK activity<br />

Shannon L. Werner, Derren Barken, <strong>and</strong> Alex<strong>and</strong>er Hoffmann<br />

Signaling Systems Laboratory, Department of Chemistry <strong>and</strong> Biochemistry, University<br />

of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0375, USA<br />

Email: slwerner@ucsd.edu; dbarken@ucsd.edu; ahoffmann@ucsd.edu<br />

The transcription factor nuclear factor-kappa B (NF-kB) regulates genes having critical roles<br />

in cellular stress responses, adaptive <strong>and</strong> innate immunity, lymphoid organ development, cell<br />

growth, survival, <strong>and</strong> apoptosis. Misregulation of NF-kB plays a role in chronic<br />

inflammatory diseases <strong>and</strong> cancers. Many different stimuli induce the NF-kB, including<br />

inflammatory cytokines (TNF, IL-1), bacterial endotoxin (LPS), oxidative stress, UVirradiation,<br />

<strong>and</strong> chemotherapeutic drugs. This begs the question: how do different stimuli<br />

elicit unique gene expression programs by activating the same <strong>signaling</strong> pathway? One<br />

possibility is that the same <strong>signaling</strong> pathway can transmit temporally distinct signals that<br />

have different physiological effects.<br />

NF-kB-inducing stimuli utilize specific <strong>signaling</strong> pathways that converge at IKK (inhibitor of<br />

kappaB kinase). Do different stimuli result in unique IKK activity profiles? Do these, in<br />

turn, generate temporally distinct, stimulus-specific nuclear NF-kB activity <strong>and</strong> gene<br />

expression profiles? This study provides evidence that the temporal control of IKK is<br />

modulated by stimulus-specific signal processing mechanisms that play a role in determining<br />

downstream specificity in gene expression. While TNF-induced IKK activity is rapidly<br />

attenuated by negative feedback by the A20 protein, LPS-<strong>signaling</strong> <strong>and</strong> LPS-specific gene<br />

expression programs are dependent on a cytokine-mediated positive feedforward mechanism.<br />

- 319 -


VIII. <strong>Inflammation</strong> specific <strong>signaling</strong> Poster VIII, 36<br />

T cell aggregation induced through CD43: Intracellular signals <strong>and</strong> inhibition by the<br />

immunomodulatory drug leflunomide.<br />

Esther Layseca-Espinosa1,2, Gustavo Pedraza-Alva1, José Luis Montiel1, Roxana del<br />

Río1, Nora A. Fierro1, Roberto González-Amaro2, <strong>and</strong> Yvonne Rosenstein1.<br />

1Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca,<br />

Mor., <strong>and</strong> 2Department of Immunology, Facultad de Medicina, Universidad Autónoma<br />

de San Luis Potosí, San Luis Potosí, S.L.P., México<br />

The CD43 co-receptor molecule has been shown to participate in lymphocyte adhesion <strong>and</strong><br />

activation. Leukocyte homotypic aggregation results from a cascade of intracellular signals<br />

delivered to the cells following engagement of different cell surface molecules with their<br />

natural lig<strong>and</strong>s. This phenomenon requires an active metabolism, reorganization of the<br />

cytoskeleton <strong>and</strong> relocalization of cell surface molecules. The aim of this study was to<br />

identify some of the key members of the <strong>signaling</strong> cascade leading to T lymphocyte<br />

homotypic aggregation following CD43 engagement. CD43-mediated homotypic aggregation<br />

of T lymphocytes required the participation of Src kinases, PLC-$2, PKC, PI3-K as well as<br />

ERK1/2 <strong>and</strong> p38. Data shown here suggest that these <strong>signaling</strong> molecules play a central role<br />

in regulating actin cytoskeleton remodeling after CD43 ligation. We also evaluated the ability<br />

of immunomodulatory drugs such as leflunomide to block the CD43-mediated homotypic<br />

aggregation. Leflunomide blocked the recruitment of targets of the Src family kinases as well<br />

as actin polymerization, diminishing the ability of T lymphocytes to aggregate in response to<br />

CD43 specific signals, suggesting that this drug might control the migration <strong>and</strong> recruitment<br />

of lymphoid cells to inflamed tissues.<br />

- 320 -


Notes<br />

- 321 -


Notes<br />

Notes<br />

- 322 -


- 323 -


Session IX. Novel compounds targeting inflammatory<br />

<strong>signaling</strong> pathways<br />

- 324 -


Session IX. Novel compounds targeting inflammatory <strong>signaling</strong> pathways Poster IX, 1<br />

Oxidized phospholipids reduce the vascular leak <strong>and</strong> inflammation in the rat model of<br />

LPS-induced acute lung injury<br />

1Anna A. Birukova, 2Stephanie Nonas, 1Joe G.N. Garcia <strong>and</strong> 1Konstantin G. Birukov<br />

1Department of Medicine, University of Chicago, Chicago, Illinois 60637, U.S.A. Emails:<br />

abirukov@medicine.bsd.uchicago.edu, jgarcia@medicine.bsd.uchicago.edu,<br />

kbirukov@medicine.bsd.uchicago.edu, 2Department of Medicine, Johns Hopkins<br />

University, Baltimore, Maryl<strong>and</strong> 21224, U.S.A. E-Mail: snonas1@jhmi.edu<br />

Acute inflammation <strong>and</strong> vascular leak are cardinal features of acute lung injury (ALI) <strong>and</strong> the<br />

acute respiratory distress syndrome (ARDS). Non-specific tissue inflammation <strong>and</strong> injury in<br />

response to a variety of infectious <strong>and</strong> non-infectious insults leads to oxidative stress <strong>and</strong> the<br />

generation lipid oxidation products. We showed that, besides antagonistic effects on toll-like<br />

receptor 4 (TLR4) which triggers LPS-induced inflammatory cascade, oxidized 1-palmitoyl-<br />

2-arachidonoyl-sn-glycero-3-phosphorylcholine (OxPAPC) enhances lung endothelial cell<br />

(EC) barrier via activation of small GTPases Rac <strong>and</strong> Cdc42. Using EC cultures <strong>and</strong> rat<br />

models of lipopolyscharide (LPS)-induced lung injury, we tested the hypothesis that OxPAPC<br />

may affect acute lung inflammatory response to LPS <strong>and</strong> recover lung vascular barrier<br />

properties. LPS induced 200-fold increase in broncho-alveolar lavage (BAL) cell count<br />

(p


Session IX. Novel compounds targeting inflammatory <strong>signaling</strong> pathways Poster IX, 2<br />

NAC prevents cardiac maladaptative responses to hypertension <strong>and</strong> promotes TNFR-2<br />

<strong>signaling</strong> in cardiomyocytes<br />

Nicole Defer1, Gabriele C<strong>and</strong>iani1, Jin-Bo Su2, Marie B<strong>our</strong>raindeloup2, Luc Hittinger2,<br />

Françoise Pecker1 <strong>and</strong> Catherine Pavoine1<br />

1INSERM U581; Université Paris 12, Faculté de Médecine; Créteil, F-94000 France;<br />

2INSERM U660; Université Paris 12, Faculté de Médecine; Créteil, F-94000 France;<br />

The aim: Our previous in vivo studies showed that N-acetylcysteine (NAC) was a potential<br />

therapeutical tool for the management of hypertension–induced heart failure. However, early<br />

mechanisms of NAC action remained to be defined.<br />

Results <strong>and</strong> Methods: Rats were given a f<strong>our</strong> week treatment with the nitric oxide synthase<br />

inhibitor, NG-nitro-L-arginine methyl ester (L-NAME, 50 mg/kg/day in drinking water) <strong>and</strong><br />

high salt diet (HS, 8% NaCl). HS/L-NAME rats displayed hypertension-induced adaptative<br />

responses, characterized by thickening of ventricular walls <strong>and</strong> eccentric hypertrophy of<br />

cardiomyocytes, without left ventricular dysfunction. This was associated with changes in two<br />

early biochemical markers of adaptative cardiac responses, namely decrease in beta-1adrenergic<br />

(AR) stimulation of adenylyl cyclase (AC) <strong>and</strong> increase in the number of the type<br />

1 TNF receptors (TNFR-1) over TNFR-2. NAC treatment (125 mg/kg/day in drinking water)<br />

given during weeks 3 <strong>and</strong> 4 of HS/L-NAME administration, had no effect on hypertension,<br />

but triggered a proportional hypertrophy of cardiomyocytes <strong>and</strong> prevented alterations of both<br />

beta-1-AR/AC <strong>signaling</strong> <strong>and</strong> TNFR-1/TNFR-2 ratio. Imaging studies performed on<br />

electrically stimulated cardiomyocytes isolated from control rats treated for 2 weeks with<br />

NAC, showed that NAC blunted TNF- induced production of ROS <strong>and</strong> its associated negative<br />

effect on Ca transient amplitude. Neutralizing anti-TNFR-1 antibodies mimicked these effects<br />

of NAC in cardiomyocytes isolated from control untreated rats. Furthermore, NAC<br />

uncovered a strong enhancing effect of TNF on Ca transients, that was associated with<br />

phosphorylation of Erk, Akt <strong>and</strong> PKC zeta <strong>and</strong> activation of the cPLA2. These TNF effects<br />

were impaired by anti TNFR-2 antibodies.<br />

Conclusion: Our results highlighted NAC as a tool fav<strong>our</strong>ing TNFR-2 while blunting TNFR-1<br />

<strong>signaling</strong> pathways, <strong>and</strong> counteracting early maladaptative responses to hypertension.<br />

- 326 -


Session IX. Novel compounds targeting inflammatory <strong>signaling</strong> pathways Poster IX, 3<br />

Wogonin prevents immunosuppressive action but not anti-inflammatory effect induced<br />

by glucocorticoid<br />

Riyo Enomoto1, Chie Suzuki1, Chika Koshiba1, Takayuki Nishino1, Mikiko<br />

Nakayama1, Hiroyuki Hirano2, Toshio Yokoi2 <strong>and</strong> Eibai Lee1.<br />

1Department of Pharmacology <strong>and</strong> 2Department of Pharmaceutical Chemistry, Faculty<br />

of Pharmaceutical Sciences, Kobe Gakuin University, Ikawadani-cho, Nishi-ku, Kobe<br />

651-2180 Japan. E-mail : enomoto@pharm.kobegakuin.ac.jp<br />

Glucocorticoid such as dexamethasone has anti-inflammatory <strong>and</strong> immunosuppressive action<br />

as major pharmacological effects. The latter action caused by lymphocyte apoptosis is not<br />

only therapeutic effect but also adverse reaction. Wogonin, a plant flavone, inhibited<br />

dexamethasone-induced apoptotic changes such as DNA fragmentation, nuclear condensation,<br />

phosphatidylserine exposure <strong>and</strong> caspase activation in rat thymocytes. Since wogonin<br />

inhibited the dexamethasone-induced DNA fragmentation in the noncompetitive manner, a<br />

target of this flavone is unlikely to the glucocorticoid receptor. This flavone also protected<br />

apoptosis induced by other glucocorticoids. Since wogonin reduced one of major<br />

pharmacological effects of dexamethasone, we examined whether this flavone diminishes the<br />

anti-inflammatory action, another pharmacological effect. The anti-inflammatory action was<br />

measured by the carrageenan-induced edema model. Although dexamethasone significantly<br />

suppressed paw edema induced by carrageenan, wogonin had no effect on the antiinflammatory<br />

action of dexamethasone. These results suggest that wogonin may be an useful<br />

compound to reduce the immunosuppressive side effect of glucocorticoids.<br />

- 327 -


Session IX. Novel compounds targeting inflammatory <strong>signaling</strong> pathways Poster IX, 4<br />

The Cyclooxygenase-2 Selective Inhibitor Celecoxib Suppresses matrix<br />

Metalloproteinase <strong>and</strong> Inhibits in Vitro Invasion of the Human Oral Squamous<br />

Carcinoma cells<br />

Young E. Kwak, Nam K. Jeon, Jin Kim <strong>and</strong> Eun J. Lee<br />

Department of Oral Pathology, Oral Cancer Research Institute, BK21 project for<br />

Medical Science, Yonsei University College of Dentistry, Seoul, KOREA, 120-752<br />

E-mail: e16lee@yumc.yonsei.ac.kr<br />

Cyclooxygenease-2(COX-2) expression is a critical factor in inflammation, <strong>and</strong> plays an<br />

important role in defense against exogenous stimuli, while overexpression of COX-2 causes<br />

cells to exhibit changes in tumor phenotype. Recently, celecoxib was shown to reduce the risk<br />

of development of cancer <strong>and</strong> mortality from it. Tumor metastasis is the most important cause<br />

of cancer death. The present study attempted to determine the effects of celecoxib with regard<br />

to human oral squamous cell carcinoma(OSCC) cell growth <strong>and</strong> invasion/migration. The YD-<br />

10B cells represented a highly-invasive human oral squamous cell carcinoma(OSCC) cell line<br />

which was found to express the COX-2 protein. Celecoxib inhibited cell invasion/migration<br />

through the type 1 collagen matrix by ~ 60% within 24 h<strong>our</strong>s. Gelatin-based zymography<br />

assay reveal that, in the presence of 10uM celecoxib, both MMP-2 <strong>and</strong> MMP-9 enzyme<br />

activity decreased by ~25-30%. The current in vitro study indicated that the inhibition of<br />

invasion/migration in OSCC cell lines by the cyclooxgenase-2 specific inhibitor, celecoxib,<br />

results in anticancerous effects via a MMP-2 <strong>and</strong> MMP-9 suppression. In addition, YD-10B<br />

cells could be useful to study the pathological mechanism of OSCC. (This study was<br />

supported by the BK21 project for Medical Science, <strong>and</strong> by a grant of the national Cancer<br />

control R&D Program 2003(No. 0320230), Ministry of Health & Welfare, Republic of<br />

Korea.)<br />

- 328 -


Session IX. Novel compounds targeting inflammatory <strong>signaling</strong> pathways Poster IX, 5<br />

Inhibition of ICAM-1-mediated metastasis by thalidomide in human lung cancer<br />

Yi-Chu Lin <strong>and</strong> Ching-Chow Chen<br />

Department of Pharmacology, College of Medicine, National Taiwan University,<br />

Taipei 10018, Taiwan. E-Mail: f92443016@ntu.edu.tw<br />

Our previous study demonstrated that TNF-alpha-induced expression of Intercellular<br />

Adhesion Molecule-1 (ICAM-1) not only increased monocyte adherence to the lung epithelial<br />

cells, but also elicited tumor cell invasion. In this report, we examined the role of ICAM-1 in<br />

carcinogenesis <strong>and</strong> the effect of thalidomide. The in vitro cell invasion <strong>and</strong> the in vivo tumor<br />

metastasis induced by ICAM-1-overexpressing A549 cells were both inhibited by<br />

thalidomide. The tumors grown in nude mice induced by s.c. human lung cancer xenografts<br />

were detected to express ICAM-1 <strong>and</strong> attenuated by oral administration of thalidomide (200<br />

microg/kg/day). Moreover, highly expressed ICAM-1 was detected in the human specimens<br />

of lung cancer patients. The inhibitory effect of thalidomide on the TNF-alpha-induced<br />

protein <strong>and</strong> mRNA expressions of ICAM-1 was seen, <strong>and</strong> this inhibition attributed to the<br />

reduced activity <strong>and</strong> binding of NF-kappaB to the ICAM-1 promoter. However, IkappaBalpha<br />

degradation <strong>and</strong> translocation of NF-kappaB to the nucleus were not affected by<br />

thalidomide. These studies provide a framework for targeting ICAM-1 gene by thalidomide as<br />

a biologically based therapy for lung cancer.<br />

- 329 -


Session IX. Novel compounds targeting inflammatory <strong>signaling</strong> pathways Poster IX, 6<br />

Statins selectively upregulate TNFR2 expression in primary human endothelial cells<br />

Tobias Nübel1*, Steffen Schmitt2 <strong>and</strong> Gerhard Fritz1<br />

1 fritz@mail.uni-mainz.de, Department of Toxicology, University of Mainz, Obere<br />

Zahlbacher Str. 67, D-55131 Mainz, Germany <strong>and</strong> 2 facslab@uni-mainz.de, University<br />

of Mainz, Naturwissenschaftlich-Medizinisches Forschungszentrum, Obere Zahlbacher<br />

Str. 67, D-55131 Mainz, Germany<br />

* Current address: nuebel@necker.fr, CNRS-UPR 9078, Faculté de Médecine Necker-<br />

Enfants malades, 156 rue de Vaugirard, 75730 Paris Cedex 15, France<br />

3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors (statins) are<br />

clincally widely used as lipid-lowering drugs. Apart from their cholesterol lowering capacity,<br />

statins exert pleiotropic effects which are clearly independent from lipid-lowering. Besides<br />

influencing angiogenesis, thrombosis, apoptosis <strong>and</strong> metastasis-related mechanisms, an antiinflammatory<br />

effect is also ascribed to statins. The cytokine tumor necrosis factor alpha<br />

(TNFalpha) plays a key role in inflammatory processes. Upon binding of TNFalpha to its<br />

receptors TNFR1 <strong>and</strong> TNFR2 the activity of the transcription factor NF-kB gets stimulated<br />

leading to transcriptional activation of genes involved in the regulation of inflammatory<br />

processes.<br />

Here we addressed the question of whether modulation of TNFR expression might be a<br />

molecular mechanism of statins contributing to their anti-inflammatory potency. To this end,<br />

we investigated the effect of the HMG-CoA reductase inhibitor lovastatin on the cell surface<br />

expression of TNFRs in primary human endothelial cells (HUVEC). We showed by ELISA,<br />

FACS <strong>and</strong> immunocytochemical analyses that lovastatin selectively upregulates TNFR2<br />

expression in HUVEC without affecting the cell surface expression of TNFR1. This effect of<br />

lovastatin appears to be independent of statin mediated inhibition of cell cycle progression<br />

from G1 to S-phase since cells both in G1 <strong>and</strong> G2 phase showed elevated levels of TNFR2<br />

expression after lovastatin treatment. Analyzing TNFalpha provoked induction of the cell<br />

adhesion molecule E-selectin as an important endothelial inflammatory stress response, we<br />

observed a dose dependent promoting effect of lovastatin on the TNFalpha stimulated<br />

increase in the frequency of E-selectin positive endothelial cells.<br />

This finding indicates that lovastatin sensitizes HUVEC towards TNFalpha induced <strong>signaling</strong><br />

mechanisms by upregulation of TNFR2 expression. Based on <strong>our</strong> data we therefore suggest<br />

that statins have impact on endothelial responses to inflammatory stress by modulating the<br />

expression of cytokine receptors.<br />

- 330 -


Session IX. Novel compounds targeting inflammatory <strong>signaling</strong> pathways Poster IX, 7<br />

DiC14-amidine cationic lipid induces the production of IL-12 by murine bone-marrow<br />

derived dendritic cells: importance of TLR-4 <strong>and</strong> JNK<br />

Tetsuya Tanaka1), Michel V<strong>and</strong>enbr<strong>and</strong>en2), Jean-Marie Ruysschaert2) <strong>and</strong> Alain<br />

Jacquet1).<br />

1)Service de Génétique Appliquée, Institut de Biologie et de Médecine Moléculaires,<br />

Université Libre de Bruxelles, B-6041 Gosselies, Belgium <strong>and</strong> 2)Laboratoire "Structure<br />

et Fonction des Membranes Biologiques," Université Libre de Bruxelles, Campus<br />

Plaine, B-1050 Brussels, Belgium E-mail : tetsuyat2003@hotmail.com<br />

DiC14-amidine cationic lipid has been shown to act as a Th1-adjuvant. A recombinant form<br />

of the major mite allergen Der p 1 (ProDer p 1) complexed with diC 14-amidine prevented the<br />

development of typical Th2-biased allergic response in mice. The present study investigates<br />

the effect of diC14-amidine on murine bone-marrow-derived dendritic cells (BMDC) <strong>and</strong><br />

particularly at the level of the pro-Th1 cytokine IL-12 production.<br />

BMDC exhibited IL-12 but not IL-10 secretion when stimulated with diC14-amidine,<br />

complexed or not with ProDer p 1. The recombinant allergen did not induce cytokine<br />

production. Using BMDCs from TLR4- <strong>and</strong> TLR2-deficient mice, we clearly demonstrated<br />

that diC14-amidine stimulation of dendritic cells involved TLR4 but not TLR2. Although all<br />

three MAP kinases, ERK1/2, p38 <strong>and</strong> JNK were activated, diC14-amidine induced IL-12<br />

release exclusively via the JNK <strong>signaling</strong> pathway. Finally, IL-12 release induced by the<br />

cationic lipid correlated with degradation of IkB, a critical step in NF-kB activation. In<br />

conclusion, the activation of dendritic cells by the complex ProDer p 1-amidine leading to IL-<br />

12 production could explain the development of Th1-biased immune response by this vaccine.<br />

- 331 -


Notes<br />

- 332 -


Session X : Cell death in cancer<br />

- 333 -


Session X : Cell death in cancer Poster X, 1<br />

Rosiglitazone antagonizes IGF-I effects <strong>and</strong> induces growth arrest <strong>and</strong> apoptosis in<br />

anaplastic cancer cells<br />

Aurora Aiello, Giuseppe P<strong>and</strong>ini*, Francesco Frasca*, Marco Genua, Antonella<br />

Murabito, Riccardo Vigneri*, Antonino Belfiore<br />

Dip. di Medicina Clinica e Sperimentale, University of Catanzaro, Policlinico Mater<br />

Domini, via T. Campanella 115. 88100 Catanzaro, Italy <strong>and</strong> *Dip. di Medicina Interna e<br />

Medicine Specialistiche, University of Catania, Catania, Italy. E-mail: belfiore@unicz.it<br />

Anaplastic thyroid cancer (ATC) is an extremely aggressive tumor characterized by resistance<br />

to chemotherapy <strong>and</strong> a very poor prognosis. PPAR-$ agonists have recently emerged as<br />

potential antineoplastic drugs. In order to establish whether ATC could be a target of PPARgamma<br />

agonists we examined PPAR-gamma protein expression in a panel of six ATC cell<br />

lines <strong>and</strong> then studied the biologic effects of a PPAR-gamma agonist, rosiglitazone, currently<br />

used as antidiabetic drug. PPAR-gamma protein was present <strong>and</strong> functional in all ATC cell<br />

lines. Rosiglitazone induced complex biological effects in ATC cells, including inhibition of<br />

anchorage dependent <strong>and</strong> independent growth <strong>and</strong> migration, <strong>and</strong> increased apoptosis rate.<br />

Moreover, rosiglitazone antagonized the mitogenic <strong>and</strong> anti-apoptotic effect of IGF-I. These<br />

effects of rosiglitazone were associated with up-regulation of the lipid phosphatase PTEN <strong>and</strong><br />

inhibition of the PI3K/AKT <strong>signaling</strong> pathway. Moreover, rosiglitazone induced changes in<br />

cell cycle regulators, such as an increase of cdk inhibitors p21cip1 <strong>and</strong> p27kip1, a decrease of<br />

cyclin D1 <strong>and</strong> inactivation of Rb protein <strong>and</strong> changes in apoptosis regulators, such as a<br />

decrease of Bcl-XL expression <strong>and</strong> caspase 3/7 activation.<br />

In conclusions, these data suggest that rosiglitazone induces multiple beneficial effects in<br />

ATC cells, among which up-regulation of PTEN <strong>and</strong> inhibition of IGF-I effects, <strong>and</strong> may<br />

have, therefore, a role in the multimodal therapy currently used to slow down ATC growth<br />

<strong>and</strong> dissemination.<br />

This work was partially supported by grants from AIRC <strong>and</strong> MIUR (Cofin 2003) to A.B.<br />

- 334 -


Session X : Cell death in cancer Poster X, 2<br />

Induction of apoptosis <strong>and</strong> activation of p53-dependent <strong>and</strong> independent pathways by a<br />

new mithramycin analogue<br />

Veronica Albertini1, Sara Vignati1, Sara Napoli1, Jurgen Rohr2, Giuseppina M.<br />

Carbone1 <strong>and</strong> Carlo V. Catapano1<br />

1Laboratory of Experimental Oncology, IOSI, Bellinzona, CH <strong>and</strong> 2Department of<br />

Pharmaceutical Sciences, University of Kentucky, Lexington, KY<br />

Aureolic acid antibiotics, like chromomycin <strong>and</strong> mithramycin (MTM), bind selectively to<br />

GC-rich DNA <strong>and</strong> block preferentially activity of GC-rich DNA binding transcription factors,<br />

like Sp1. Combinatorial biosynthesis approaches applied to the MTM producer S. Argillaceus<br />

have recently yielded new analogues that exhibit greater transcriptional inhibitory activity <strong>and</strong><br />

might have an improved therapeutic index compared to MTM. A new compound, MTM SDK,<br />

down-regulated transcription of multiple genes implicated in cancer pathogenesis, inhibited<br />

growth of a variety of human cancer cell lines <strong>and</strong> was a potent inducer of apoptosis. SDK<br />

was equally active against cancer cell lines with wild type or mutated p53. Interestingly, the<br />

pro-apoptotic activity of SDK in cancer cells was not associated with significant toxicity in<br />

normal fibroblasts. Although the activity of SDK was apparently p53-independent, we<br />

investigated whether the p53 pathway could modulate cell responses to SDK <strong>and</strong> MTM in<br />

p53wt cells. Both compounds increased p53 protein level by a post-transcriptional<br />

mechanism. Activation of p53 by MTM resulted in up-regulation of p21, while the level of<br />

p21 did not increase in SDK-treated cells due to inhibition of p21 transcription. Increased<br />

levels of p21 attenuated the apoptotic response to MTM, while its absence in SDK-treated<br />

cells was associated with increased apoptosis. The protective role of p21 could also explain<br />

the limited cytotoxicity of SDK in normal fibroblasts, in which SDK did not block p21<br />

induction. Thus, SDK is a potent antiproliferative <strong>and</strong> pro-apoptotic agent due to its ability to<br />

interfere with critical growth <strong>and</strong> survival genes. SDK activity is also modulated by p53dependent<br />

pathways in p53wt cells. The ability of SDK to simultaneously activate p53 <strong>and</strong><br />

block p53-dependent activation of p21 favors apoptosis in cancer cells <strong>and</strong> contributes to its<br />

greater activity compared to MTM. On the other h<strong>and</strong>, in normal fibroblasts p53-dependent<br />

up-regulation of p21 can have a protective role <strong>and</strong> contribute to the reduce toxicity of SDK.<br />

- 335 -


Session X : Cell death in cancer Poster X, 3<br />

Comaprative Proteomics of Ovarian Epithelial Tumors<br />

!<br />

Hee Jung An, Nam Hoon Cho, Park, Choi YP, Kang S, Ding O, Kim DS<br />

Dept of Pathology, Bundang CHA Medical Hosiptal<br />

Dept of Pathology, Yonsei Univresity College of Medicine<br />

Brain Korea 21 Project for Medical Science, Yonsei University!!<br />

!<br />

We analyzed twelve ovarian epithelial tumors using 2D PAGE-based comparative proteomics<br />

to construct intra- <strong>and</strong> inter-tumoral distance map trees, <strong>and</strong> to discover surrogate biomarkers<br />

indicative of an ovarian tumor. The analysis was performed after laser microdissection of 12<br />

fresh-frozen tissue samples, including 4 serous, 5 mucinous <strong>and</strong> 3 endometrioid tumors, with<br />

correlation with their histopathological characteristics. Ovarian epithelial tumors <strong>and</strong> normal<br />

tissues showed an apparent separation on the distance map tree. Mucinous carcinomas were<br />

closest to the normal group, whereas serous carcinomas were located furthest from the normal<br />

group. All mucinous tumors with aggressive histology were separated from the LMP group.<br />

The benign-looking cysts adjacent to the IEC showed an expression pattern identical to that of<br />

the IEC area. The extent of change on the lineages leading to the mucinous <strong>and</strong> serous<br />

carcinoma was 1.98-fold different. The overall gene expression profiles of serous or<br />

endometrioid carcinomas appeared to be less affected by grade or stage than by histologic<br />

type. The surrogate biomarkers screened in ovarian tumors <strong>and</strong> found to be significantly upregulated<br />

in comparison to normal tissues were as follows: NM23, annexin-1, protein<br />

phosphatase-1, ferritin light chain, proteasome alpha-6, <strong>and</strong> NAGK (N-acetyl glucosamine<br />

kinase). In conclusion, ovarian mucinous tumors are distinct from other ovarian epithelial<br />

tumors. LMP mucinous tumors showing histologically aggressive features belong to<br />

mucinous carcinoma on the proteomic basis.<br />

- 336 -


Session X : Cell death in cancer Poster X, 4<br />

Endosulfan inhibits cell growth <strong>and</strong> apoptosis through activation of ERK1/2 <strong>and</strong> AKT<br />

<strong>signaling</strong> pathways<br />

S. Antherieu 1, N. Ledirac 1, P. Lenorm<strong>and</strong> 2 <strong>and</strong> R. Rahmani 1<br />

1 INRA UMR-1112, Equipe de Toxicologie Cellulaire et Moléculaire, et Génomique,<br />

Sophia-Antipolis, France, e-mail: antherieu@antibes.inra.fr<br />

2 CNRS UMR-6543, Centre Antoine Lacassagne, Nice, France.<br />

Endosulfan is an organochlorine insecticide reported to be a potential carcinogen in humans.<br />

This insecticide was previously described to alterate MAP kinases <strong>signaling</strong> pathways <strong>and</strong><br />

suspected to affect cell growth <strong>and</strong> differentiation in keratinocytes. In the present study, we<br />

assessed the mitogenic <strong>and</strong> apoptogenic potentials of endosulfan in HaCaT keratinocytes.<br />

We first confirmed its mutagenicity by using the AMES assay, which demonstrated that<br />

unchanged endosulfan rather than metabolites, was at the origin of mutagenic effects<br />

(negative results in the presence of S9 hepatic fractions).<br />

Moreover, we showed that endosulfan led to a persistent ERK1/2 phosphorylation with an<br />

accumulation of the phosphorylated form both in cytosol <strong>and</strong> nucleus. This sustained<br />

activation is responsible for a decreased cell proliferation, since untreated cells reached<br />

confluency after 3 days, whereas confluency is reached after 4 days in endosulfan-treated<br />

cells. These data were also confirmed by BrdU incorporation which was strongly decreased<br />

after insecticide treatment.<br />

In addition, reactive oxygen species (ROS) has been shown to be involved in endosulfaninduced<br />

ERK1/2 activation; we demonstrated that blockade of this oxidative stress by NAC<br />

(N-acetylcystein) strongly prevented inhibition of proliferation by endosulfan. These results<br />

clearly showed that endosulfan-induced ROS were involved in cell growth decrease.<br />

Finally, endosulfan apoptogenic potential was investigated <strong>and</strong> a reduction of caspases 3/7<br />

activation <strong>and</strong> PARP cleavage was evidenced. This inhibition of apoptosis is probably<br />

correlated to endosulfan-induced AKT pathway, since a 3-fold induction of phospho-AKT<br />

was observed after 1 h of treatment.<br />

On the whole, this study demonstrates that endosulfan is a mutagenic agent, responsible of a<br />

ROS-induced proliferation decrease <strong>and</strong> apoptosis inhibition. These processes in turn prevent<br />

elimination of altered cells <strong>and</strong> therefore may contribute to carcinogenesis.<br />

- 337 -


Session X : Cell death in cancer Poster X, 5<br />

Apoptosis associated with reduced Bcl-2 expression in colorectal cancer tumors<br />

Márcia M. Aranha1, Pedro M. Borralho1, Paula Ravasco2, Isabel B. Moreira da Silva1,<br />

Luís Correia3, Afonso Fern<strong>and</strong>es4, Maria E. Camilo2, Cecília M.P. Rodrigues1<br />

1Centro de Patogénese Molecular, Faculty of Pharmacy, University of Lisbon, 1600-083<br />

Lisbon, Portugal; 2Unidade de Metabolimo e Nutrição, IMM; Centro de<br />

3Gastrenterologia e de 4Anatomia Patológica, Santa Maria Hospital, Lisbon, Portugal;<br />

E-mail: maranha@ff.ul.pt<br />

Compelling evidence implicates loss of cell-cycle control <strong>and</strong> changes in apoptotic pathways<br />

in the development <strong>and</strong> progression of most human cancers. In this study, several apoptotic<br />

factors were examined in normal <strong>and</strong> tumor tissues from patients diagnosed for colorectal<br />

cancer. Apoptosis as assessed by the transferase mediated dUTP-digoxigenin nick-end<br />

labeling (TUNEL) assay was significantly higher in tumor tissue compared with normal<br />

mucosa (p < 0.01). TUNEL-positive cells were increased from adenoma to poorly<br />

differentiated adenocarcinoma, but decreased in association with higher degree of<br />

adenocarcinoma differentiation. Similar results were observed for NF-!B/RelA expression<br />

detected by immunohistochemistry (p < 0.01). Further, Bcl-2 expression was consistently<br />

higher in normal mucosa than in tumor tissue (p < 0.01). In both adenoma <strong>and</strong><br />

adenocarcinoma tissues, Bcl-2 immunoreactivity was almost undetectable, which inversely<br />

correlated with the percentage of apoptotic cells (r = - 0.54; p < 0.01). In addition, expression<br />

analysis of phosphorylated p53 <strong>and</strong> Bax by western blot revealed almost no variation of<br />

protein expression in tumor tissue compared to normal mucosa. These results demonstrate<br />

increased apoptosis in pre-malignant <strong>and</strong> poorly differentiated malignant tissues. Apoptosis<br />

may proceed through a p53- <strong>and</strong> Bax-independent fashion but is correlated with suppression<br />

of Bcl-2 expression. The histological progression of colorectal cancer from adenoma to<br />

adenocarcinoma involves also NF-!B activation. Whether these changes are associated with<br />

a better prognosis deserves further investigation. (Supported by Sociedade Portuguesa de<br />

Gastrenterologia, Portugal)<br />

- 338 -


Session X : Cell death in cancer Poster X, 6<br />

Expression of Bcl-xL, Bax <strong>and</strong> P53 in primary tumors <strong>and</strong> lymph node metastases in<br />

oral squamous cell carcinoma.<br />

Marek Baltaziak1, Ewa Duraj1, Mariusz Koda1, Mariola Sulkowska1, Marcin<br />

Musiatowicz2, Luiza Kanczuga-Koda1, Andrzej Wincewicz1, Boguslaw Musiatowicz1,<br />

Stanislaw Sulkowski1.<br />

Departments of Pathology1, Pediatric Otorhinolaryngology2; Medical University of<br />

Bialystok, Waszyngtona 13, 15-269 Bialystok, Pol<strong>and</strong>. E-mail: sulek@zeus.amb.edu.pl<br />

Disturbances in expression of apoptosis associated proteins, take part in development <strong>and</strong><br />

progression of many human malignancies. The aim of the present study was assessment of<br />

correlations among proteins involved in apoptosis: Bcl-xL, Bax <strong>and</strong> P53 as well as<br />

relationships of these proteins with selected clinicopathological features in oral squamous cell<br />

carcinoma. Consequently, we examined by immunohistochemistry, using the avidin-biotinperoxidase<br />

method, Bcl-xL, Bax <strong>and</strong> P53 expression in 56 samples of primary oral squamous<br />

cell carcinoma <strong>and</strong> in 22 matched pairs of primary <strong>and</strong> metastatic tumors. The evaluation of<br />

immunostaining of Bcl-xL, Bax <strong>and</strong> P53 was analyzed in 10 different tumor fields <strong>and</strong> the<br />

mean percentage of tumor cells with positive staining was evaluated. The significance of the<br />

associations were determined using Spearman correlation analysis <strong>and</strong> the Chi-square test.<br />

We found positive, Bcl-xL, Bax <strong>and</strong> P53 immunostaining in 44.6%, 28.6% <strong>and</strong> 58.9% of<br />

studied primary tumors <strong>and</strong> in 63.6%, 45.5% <strong>and</strong> 72.7% of lymph node metastases,<br />

respectively. Analysis of associations among studied proteins revealed positive correlation<br />

between Bcl-xL <strong>and</strong> Bax in primary tumors (p


Session X : Cell death in cancer Poster X, 7<br />

Salicylic acid <strong>and</strong> oxidative stress susceptibility within colon epithelial cell cultures<br />

Charles S. Bestwicka, Lesley Milnea, Mary P. Moyerc, Garry Duthiea <strong>and</strong> Susan J.<br />

Duthieb<br />

Molecular Nutrition Groupa <strong>and</strong> Nutrition <strong>and</strong> Epigenetics Groupb, The Rowett<br />

Research Institute, Aberdeen AB21 9SB, Scotl<strong>and</strong>, UK. INCELL Corporationc, LLC<br />

12000 Network Blvd., Ste B-200 San Antonio, TX 78249 210-877-0100.<br />

Email:C.Bestwick@rowett.ac.uk<br />

Salicylic acid (SA) is a widely occurring dietary phyto-phenol that may contribute to the<br />

cancer preventative effects of a fruit <strong>and</strong> vegetable rich diet. In planta, SA is a critical<br />

component of pathogen resistance responses, affecting the generation of reactive oxygen<br />

species (ROS) <strong>and</strong> programmed cell death development. The colonic epithelium may be<br />

unusually resistant to stress-induced apoptosis, thereby potentially increasing the propensity<br />

for tum<strong>our</strong> development <strong>and</strong> we hypothesise that SA exposure may enhance the response to<br />

oxidative DNA damage, ultimately facilitating more effective entry into apoptosis. Here, we<br />

describe preliminary observations on the effect of non-cytotoxic SA exposure on cellular<br />

ROS, antioxidant activity <strong>and</strong> resistance to oxidative insult within both non-transformed<br />

(NCM460) <strong>and</strong> malignantly transformed (HT-29) colon epithelial cells. <strong>Expo</strong>sure to SA for<br />

24h decreased intracellular ROS levels within both cell types. By 72h, no significant<br />

difference was observed between control <strong>and</strong> SA treated NCM460 cells but ROS levels in<br />

HT-29 were slightly elevated in response to >100micromolar SA. No change to catalase<br />

activity was detected but glutathione peroxidase activity had declined by 72h in both SA<br />

treated cell types. DNA str<strong>and</strong> breaks were significantly increased in NCM460 cells but not<br />

within HT-29 cells variously exposed to SA. Following a 72h SA exposure, cultures were<br />

challenged with varying levels of hydrogen peroxide. SA significantly enhanced DNA<br />

damage within NCM460 but not HT-29cells subject to mild oxidative stress. In conclusion,<br />

SA modifies antioxidant <strong>and</strong> ROS levels within both transformed <strong>and</strong> non-transformed colon<br />

epithelial cells, though there is no intuitive correlation between these changes <strong>and</strong> effects on<br />

DNA stability. Nevertheless, SA does marginally enhance DNA susceptibility to oxidative<br />

damage within the non-transformed colon cell line.<br />

- 340 -


Session X : Cell death in cancer Poster X, 8<br />

Bisnaphthalimidopropylspermidine induces DNA instability <strong>and</strong> apoptosis within colon<br />

carcinoma derived cell lines<br />

Charles S. Bestwick a, Lynda Ralton,c Lesley Milne,a Susan J. Duthie,b <strong>and</strong> Paul Kong<br />

Thoo Linc<br />

Molecular Nutrition Groupa <strong>and</strong> Nutrition <strong>and</strong> Epigenetics Groupb, The Rowett<br />

Research Institute, Greenburn Road, Aberdeen AB29 9SB, Scotl<strong>and</strong>, UK. The Robert<br />

Gordon Universityc, School of Life Sciences, St. Andrew Street, Aberdeen AB25 1HG,<br />

Scotl<strong>and</strong>, UK. Email: C.Bestwick@rowett.ac.uk<br />

DNA intercalators are potentially potent apoptosis inducers. Bisnaphthalimido compounds<br />

bisintercalate to DNA via the major groove. However, solubility is poor <strong>and</strong> we have<br />

modified the linker chain connecting the two naphthalimido ring moieties to increase<br />

solubility. One such modification has been the incorporation of polyamines within the linker.<br />

Polyamines are ubiquitous polycations critical for cell growth <strong>and</strong> also exhibit apoptotic<br />

regulatory activity. Thus, in addition to facilitating cellular uptake, incorporating polyamines<br />

within the linker may lead to an altered or extended spectrum of activity. Here, we describe<br />

the effect of a spermidine analogue, bisnaphthalimidopropylspermidine (BNIPSpd), on DNA<br />

stability <strong>and</strong> survival of Caco-2 <strong>and</strong> HT-29 colon carcinoma derived cells. After 48h<br />

exposure, an IC50 of 0.15 <strong>and</strong> 1.64 micromolar was determined for Caco-2 <strong>and</strong> HT-29<br />

respectively. Analogue uptake was rapid <strong>and</strong> essentially homogenous within the cell<br />

populations by 4h. Uptake was associated with a dose dependent increase in DNA str<strong>and</strong><br />

breaks (0.1 –100micromolar) <strong>and</strong> decreased intracellular polyamines (0.01-100micromolar).<br />

Following treatment with 0.5micromolar BNIPSpd or greater, cells expressed enhanced active<br />

caspase-3, increased nuclear instability, internucleosomal DNA fragmentation <strong>and</strong> apoptoticmorphological<br />

features. Pre-treatment with non-overtly cytotoxic or direct DNA damaging<br />

concentrations of BNIPSpd retarded DNA repair capacity, as well as inducing a depletion of<br />

cellular polyamines. The potential to exploit such analogues as chemotherapeutic agents <strong>and</strong><br />

as probes to assess the role of polyamines in cell survival regulation is discussed<br />

- 341 -


Session X : Cell death in cancer Poster X, 9<br />

Par-4-mediated Recruitment of Amida to the Actin Cytoskeleton Leads to the Induction<br />

of Apoptosis<br />

Meike Boosen1, Susanne Vetterkind1, Ansgar Koplin1, Susanne Illenberger2 <strong>and</strong> Ute<br />

Preuss1*<br />

1Institute of Genetics, University of Bonn, Roemerstr. 164 D-53117 Bonn, Germany,<br />

2Cell Biology, Zoological Institute, Technical University of Braunschweig, D-38092<br />

Braunschweig, Germany<br />

email:meike.boosen@uni-bonn.de<br />

Par-4 (prostate apoptosis response-4) sensitizes cells to apoptotic stimuli, but the exact<br />

mechanisms are still poorly understood. Using Par-4 as bait in a yeast two-hybrid screen, we<br />

identified Amida as a novel interaction partner, a ubiquitously expressed protein which has<br />

been suggested to be involved in apoptotic processes. Complex formation of Par-4 <strong>and</strong> Amida<br />

occurs in vitro <strong>and</strong> in vivo <strong>and</strong> is mediated via the C-termini of both proteins, involving the<br />

leucine zipper of Par-4. Amida resides mainly in the nucleus, but displays nucleo-cytoplasmic<br />

shuttling in heterokaryons. Upon co-expression with Par-4 in REF52.2 cells, Amida<br />

translocates to the cytoplasm <strong>and</strong> is recruited to actin filaments by Par-4, resulting in<br />

enhanced induction of apoptosis. The synergistic effect of Amida/Par-4 complexes on the<br />

induction of apoptosis is abrogated when either Amida/Par-4 complex formation or<br />

association of these complexes with the actin cytoskeleton are impaired, indicating that the<br />

Par-4-mediated relocation of Amida to the actin cytoskeleton is crucial for the pro-apoptotic<br />

function of Par-4/Amida complexes in REF52.2 cells. The latter results in enhanced<br />

phosphorylation of the regulatory light chain of myosin II (MLC) as has previously been<br />

shown for Par-4-mediated recruitment of DAP-like kinase (Dlk), suggesting that the<br />

recruitment of nuclear proteins involved in the regulation of apoptotic processes to the actin<br />

filament system by Par-4 represents a potent mechanism how Par-4 can trigger apoptosis.<br />

- 342 -


Session X : Cell death in cancer Poster X, 10<br />

2-Methoxyestradiol induces autophagy in Ewing sarcoma-derived cell lines:<br />

an anti-apoptotic mechanism<br />

Amélie Borges1, Chantal Bauvy1, Sylvie Souquére2, Gérard Pierron2, Patrice<br />

Codogno1, Mojgan Djavaheri-Mergny1<br />

1INSERM U504, Institut André Lwoff, Villejuif<br />

mergny@vjf.inserm.fr<br />

2 UPR-1983, Institut André Lwoff, Villejuif,<br />

2-Methoxyestradiol (2-Me) is a naturel estrogen metabobolite which acts as antiproliferative<br />

agent in various tumor cell lines. We have previously shown that 2-Me induced apoptosis in<br />

Ewing sarcoma cells through mitochondrial death pathway. In the present study, we report<br />

that 2-Me-induced macroautophagy in Ewing sarcoma-derived cells independently of their<br />

p53 status. Macroautophagy has been characterized by ultrastructural analysis, accumulation<br />

of LC3-II protein <strong>and</strong> proteolysis assay. In addition, we found that the inhibition of autophagy<br />

by either using 3-methyladenine or by silencing of autophagy genes enhances 2-Me-induced<br />

apoptosis. These results suggest that 2-Me-induced autophagy is an anti-apoptotic<br />

mechanism.<br />

- 343 -


Session X : Cell death in cancer Poster X, 11<br />

5-Fluor<strong>our</strong>acil activates death receptor <strong>and</strong> mitochondrial pathways of apoptosis in<br />

colon cancer cells in a p53 proficiency-dependent manner<br />

Pedro M. Borralho,1 Isabel B. Moreira da Silva,1 Márcia M. Aranha,1 Cristina<br />

Albuquerque,2 Carlos Nobre Leitão,2 Cecília M.P. Rodrigues1<br />

1Centro de Patogénese Molecular, Faculty of Pharmacy, University of Lisbon, 1600-083<br />

Lisbon, Portugal; 2Centro de Patobiologia Molecular, IPO, Lisbon, Portugal; E-mail:<br />

borralho@ff.ul.pt<br />

5-Fluor<strong>our</strong>acil (5-FU), the most important drug used for colorectal cancer (CRC)<br />

chemotherapy, is a potent antitumor agent that affects pyrimidine synthesis by inhibiting<br />

thymidylate synthase. Treatment of cancer cells with 5-FU is thought to induce apoptosis.<br />

However, resistance to 5-FU is still a major limitation to its clinical use. In these studies, we<br />

used HCT116 <strong>and</strong> SW480 human colorectal cancer cell lines, with wild-type <strong>and</strong> mutant-type<br />

p53 expression, respectively. Supplementation with 8 µM 5-FU was more cytotoxic in<br />

HCT116 than in SW480 cells (p < 0.001). Nuclear fragmentation <strong>and</strong> caspase-3, -8 <strong>and</strong> -9<br />

activities were markedly increased in HCT116 cells after 5-FU exposure (p < 0.01). In<br />

addition, wild-type p53 expression in 5-FU-treated HCT116 cells was almost 25-fold<br />

increased compared with control cells (p < 0.001), whereas mutant p53 expression was not<br />

altered in SW480 cells. Pro-apoptotic Bax <strong>and</strong> anti-apoptotic Bcl-2 remained unchanged<br />

during 5-FU treatment. Interestingly, 5-FU resulted in a 4-fold increased expression of the<br />

death receptor Fas in HCT116 cells (p < 0.05). siRNA-mediated Fas gene expression<br />

knockdown significantly reduced nuclear fragmentation as well as caspase-3, -8 <strong>and</strong> -9<br />

activities. Finally, western blot analysis of mitochondrial extracts showed a 6-fold increase in<br />

Bax, together with a 3-fold decrease in cytochrome c in HCT116 cells exposed to 5-FU (p <<br />

0.05). In conclusion, these results suggest that 5-FU exerts its cytotoxic effects through a<br />

p53/Fas-dependent apoptotic pathway, which is independent from modulation of Bax <strong>and</strong><br />

Bcl-2 protein expression. In addition, p53 might also play a transcriptional independent role,<br />

tipping the Bax/Bcl-2 balance, allowing Bax to translocate to the mitochondria <strong>and</strong><br />

consequently activating the mitochondrial pathway of apoptosis. Thus, 5-FU activates <strong>and</strong><br />

modulates classical pathways of apoptosis in a p53 proficiency-dependent manner.<br />

(Supported by Fundação Calouste Gulbenkian, Portugal)<br />

- 344 -


Session X : Cell death in cancer Poster X, 12<br />

Antiproliferative effects of retinoic acid <strong>and</strong> histone deacetylase inhibitor Bml-210 on<br />

human cervical cancer HeLa cells<br />

Veronika V. Borutinskaite1, 2, Ruta Navakauskiene 1 <strong>and</strong> Karl-Eric Magnusson2<br />

1Department of Developmental Biology, Institute of Biochemistry, LT-08662 Vilnius,<br />

Lithuania. E-mail: verbo@imk.liu.se ruta.navakauskiene@bchi.lt<br />

2Division of Medical Microbiology, Department of Molecular <strong>and</strong> Clinical Medicine,<br />

Linköping University, SE-581 85 Linköping, Sweden. E-mail: karma@imk.liu.se<br />

Human papillomaviruses (HPVs) have been associated with a number of neoplastic lesions,<br />

most notably cervical cancer which is one of the major forms of cancer world wide. Human<br />

cervical carcinoma HeLa cells contain integrated human papillomavirus type 18 (HVP18).<br />

Retinoic acid is a key regulator of epithelial cell differentiation <strong>and</strong> a growth inhibitor in vitro<br />

of HeLa cervical carcinoma cells. Cellular responses to RA are mediated by nuclear retinoic<br />

acid receptors (RARs) <strong>and</strong> retinoid X receptors (RXRs). On the other h<strong>and</strong>, histone<br />

deacetylase inhibitors have been shown to be chemopreventive agents for the treatment of<br />

cancer cells.<br />

In this study, we have studied the antiproliferative effect of retinoic acid (RA) <strong>and</strong> histone<br />

deacetylase inhibitor Bml-210 on HeLa cells, <strong>and</strong> particularly the effects on the protein<br />

expression that may be involved in the cell cycle control <strong>and</strong> apoptosis. Our data suggest that<br />

a combination of RA <strong>and</strong> Bml-210 leads to cell growth inhibition with following apoptosis in<br />

a treatment time- dependent manner. We confirm that Bml-210 alone or in combination with<br />

RA cause a marked increase in the level of p21, which coincides with the increased level of<br />

transcription factor Sp1. The changes in p53 level are only under RA influence. We also<br />

discovered that histone deacetylase inhibitor Bml-210 causes increased levels of antiapoptotic<br />

protein Bcl-2 <strong>and</strong> phosphorylated p38 MAPK; these link in cell cycle arrest with<br />

response to extracellular stimuli. Our results suggest that RA <strong>and</strong> Bml-210 are involved in<br />

different signalling pathways that regulate cell cycle arrest <strong>and</strong> lead to apoptosis of HeLa<br />

cells.<br />

- 345 -


Session X : Cell death in cancer Poster X, 13<br />

Targeting of the IFNgammaR2 dileucine-based (LI276-277) internalization motif<br />

reinstates T cell sensitivity to the IFNgamma/STAT1-dependent apoptosis in vitro <strong>and</strong><br />

in vivo<br />

Daniela Boselli1,2, Josiane Ragimbeau4, Paola Bernabei1, Roberto Chiarle1,3, S<strong>and</strong>ra<br />

Pellegrini4, Mirella Giovarelli1,2, <strong>and</strong> Francesco Novelli1,2<br />

1Center for Experimental Research <strong>and</strong> Medical Studies (CeRMS), San Giovanni<br />

Battista Hospital, Via Santena 5, 10126 Turin, Italy; 2Department of Medicine <strong>and</strong><br />

Experimental Oncology, 3Department of Pathology, University of Turin, 10126 Turin,<br />

Italy; 4Unit of Cytokine Signaling, Institut Pasteur, 75724 Paris, France<br />

E-mail: daniela.boselli@unito.it<br />

Activation of the IFNgamma (IFNg)/STAT1 pathway switches on many pro-apoptotic <strong>and</strong><br />

anti-proliferative genes. The downregulation of the IFNg receptor 2 chain (IFNgR2) has been<br />

described in T lymphocytes <strong>and</strong> is thought to prevent activation of the apoptotic pathway. We<br />

have previously shown that in human T lymphocytes IFNgR2 is internalized in a clathrindependent<br />

manner <strong>and</strong> that this internalization is lig<strong>and</strong>-independent. Here we show that, in<br />

Jurkat T cells, an IFNgR2 mutant in which the LI267-277 motif had been substituted with two<br />

alanines (LI276-277AA) accumulated at the cell surface. The defective internalization of the<br />

LI267-277AA mutant determined a strong <strong>and</strong> sustained IFNg-dependent response in terms of<br />

STAT1 activation, IRF-1 <strong>and</strong> MHC class I induction, Fas <strong>and</strong> FasL cell surface up-regulation<br />

<strong>and</strong> apoptosis. All these functions were weakly or not induced by IFNg in Jurkat cells<br />

expressing wild type IFNgR2. The growth into SCID mice of ST4 malignant T cells<br />

expressing the LI276-277AA mutant receptor, but not that of cells expressing the wild type<br />

receptor, was inhibited by IFNg. These data indicate that the LI276-277 motif critically<br />

controls lig<strong>and</strong>-independent internalization of IFNgR2 in T cells. This internalization motif<br />

may provide a valuable therapeutic target for the reinstatement of IFNg/STAT1 apoptotic<br />

<strong>signaling</strong> in autoreactive or neoplastic T cells.<br />

- 346 -


Session X : Cell death in cancer Poster X, 14<br />

Endogenous FGF1 inhibits p53-dependent apoptosis<br />

Sylvina Bouleau, Vincent Rincheval, Bernard Mignotte, Jean-Luc Vayssière, Flore<br />

Renaud.<br />

Laboratoire de Génétique et Biologie Cellulaire, CNRS FRE2445, Ecole Pratique des<br />

Hautes Etudes, Université de Versailles, 45 Av des Etats Unis, 78035 Versailles, France.<br />

E-mail : bouleau@genetique.uvsq.fr<br />

The oncosuppressor p53 is an important regulator of apoptosis <strong>and</strong> is associated with few<br />

diseases (like cancers, neurodegenerative diseases…). In contrast to p53, survival factors<br />

inhibit the cell death process. Their action is usually mediated via paracrine <strong>and</strong>/or autocrine<br />

pathways. However, some survival factors, as FGF1 <strong>and</strong> FGF2, do not contain secretion<br />

peptide signal but a nuclear localization signal (NLS) <strong>and</strong> may act via an intracrine pathway.<br />

We have initiated the study of the role of FGF1 in p53-dependent apoptosis in a rat embryo<br />

fibroblast cellular model. We have shown that : (1) p53 represses fgf1 gene expression; (2)<br />

FGF1 overexpression inhibits pro-apoptotic <strong>and</strong> anti-proliferative p53 activities via an<br />

intracrine pathway; (3) FGF1 increases the level of MDM2, which induced p53 degradation;<br />

(4) FGF1 inhibits the p53-dependent trans-activation of the bax gene, which code a major<br />

pro-apoptotic protein (Bouleau S. et al. , 2005, Oncogene, 24(53) : 7839-7849).<br />

Then, we persue this study in a neuronal cellular model: the PC12 cells (issued from a rat<br />

pheocromocytoma). These cells can differentiate in sympathetic like neurons in presence of<br />

neurotrophic factors like NGF or FGF1. First, we have characterized the apoptosis process<br />

induced by etoposide, to ascertain the implication of p53 in this process. Second, we have<br />

shown that the addition of recombinant FGF1 in the culture medium protects the cells from<br />

p53-dependent cell death, suggesting a protective activity of exogenous FGF1. Actually, we<br />

initiate the study of the activity of endogenous FGF1. In particular, we currently analyze the<br />

activity of intracellular <strong>and</strong> nuclear FGF1 on p53-dependent apoptosis.<br />

Bouleau S, Grimal H, Rincheval V, Godefroy N, Mignotte B, Vayssière JL, Renaud F. “FGF1<br />

inhibits p53-dependent apoptosis <strong>and</strong> cell cycle arrest via an intracrine pathway”. 2005,<br />

Oncogene, 24(53) : 7839-7849.<br />

- 347 -


Session X : Cell death in cancer Poster X, 15<br />

hGTSE-1 protein can modulate the cellular response to stress by regulating the stability<br />

of the cyclin-dependent kinase inhibitor p21CIP1/WAF1<br />

Bublik DR, Scolz M, Monte M <strong>and</strong> Schneider C.<br />

Human GTSE-1 (G2 <strong>and</strong> S phase-expressed-1) is a cell cycle-regulated protein with increased<br />

expression during S <strong>and</strong> G2 phases of the cell cycle. We have previously demonstrated that<br />

hGTSE-1 can negatively regulate p53 transactivation function, protein levels <strong>and</strong> p53dependent<br />

apoptosis during the S/G2 cell cycle window. More recently we have observed<br />

that, although hGTSE-1 represses p53 activity, it’s able to stabilize the cyclin-dependent<br />

kinase inhibitor p21CIP1/WAF1. p21CIP1/WAF1 protein was shown to be strongly<br />

regulated by hGTSE-1: p21CIP1/WAF1 levels were increased in cells with Ponasterone Ainducible<br />

expression of hGTSE-1, <strong>and</strong> concomitantly downregulated in hGTSE-1-knocked<br />

down cells, in a proteasome-dependent manner. As a functional consequence,<br />

p21CIP1/WAF1 stabilization observed in hGTSE-1-induced cells, conferred resistance to<br />

taxol-induced apoptosis; conversely, loss of p21CIP1/WAF1 after hGTSE-1 knock-down by<br />

small interference RNA sensitized cells to taxol-induced apoptosis.<br />

Besides, we have also found out that hGTSE-1-dependent stabilization of p21CIP1/WAF1<br />

requires a functional Heat Shock Protein 90 (Hsp90) given that the specific Hsp90 inhibitor<br />

Geldanamycin completely prevented hGTSE-1’s effects on p21CIP1/WAF1. We propose the<br />

recently discovered Hsp90-binding <strong>and</strong> p21CIP1/WAF1-interactor TPR protein WISp39, as a<br />

mediator that acts downstream of hGTSE-1 in regulating p21CIP1/WAF1 stability; indeed in<br />

the absence of WISp39 hGTSE-1 failed to induce p21CIP1/WAF1 accumulation. Moreover,<br />

a direct interaction between hGTSE-1, p21CIP1/WAF1 <strong>and</strong> WISp39 was confirmed in vitro<br />

<strong>and</strong> in vivo suggesting that hGTSE-1 might be associated to the multi-protein Hsp90<br />

chaperone complex.<br />

These findings may support an important role of hGTSE-1 in maintaining <strong>and</strong> tightly<br />

regulating p21CIP1/WAF1 levels thereby modulating its functions, <strong>and</strong> indicate that hGTSE-<br />

1 could fine tune the cellular response to stress through regulation of p21CIP1/WAF1<br />

turnover.<br />

- 348 -


Session X : Cell death in cancer Poster X, 16<br />

The effects of selenium diet on expression of selenoproteins in the respiratory tract of<br />

the rat<br />

Katarzyna Bukalis, , Dorothea Alber, Dietrich Behne, Antonios Kyriakopoulos<br />

Hahn-Meitner-Institut, Department “Molecular Trace Element Research in the Life<br />

Sciences”, Glienicker Str. 100, D-14109 Berlin, Germany, e-mail: k.bukalis@hmi.de)<br />

The organs of the respiratory tract lung <strong>and</strong> trachea, are constantly exposed to many gases <strong>and</strong><br />

particular matter present in the atmosphere or toxins such as asbestos or tobacco smoke.<br />

Many of these pollutants are powerful oxidants. Several lung <strong>and</strong> trachea diseases have been<br />

associated with oxidative stress <strong>and</strong> linked to oxidant insults. In addition, long-term exposure<br />

to environmental toxicants can induce mutation, or changes in the DNA of the exposed cells,<br />

resulting in altered gene functions <strong>and</strong> in the further step lead to altered patterns of protein<br />

production that might allow for growth of precancerous cells. The tissues of the respiratory<br />

tract therefore require a specific defence system against oxidants <strong>and</strong> free radicals.<br />

The trace element selenium plays an important role in this defence system. In the form of<br />

selenocysteine it is an essential component of glutathione peroxidase, an enzyme involved in<br />

the detoxification of peroxides. Selenium has been also reported to have diverse anticancer<br />

actions <strong>and</strong> to inhibit cancer growth in animals. In the patients with many cancers including<br />

those of respiratory tract the low blood levels of selenium have been observed. This finding<br />

indicate that low selenium levels can associated with increased cancer incidence in humans.<br />

There is evident relationship between selenium deficiency, oxidative stress <strong>and</strong> lung <strong>and</strong><br />

trachea diseases. Therefore it is necessary to investigate the impact of the selenium status in<br />

the diet on the incidence of the lung <strong>and</strong> trachea diseases. It is of grate interest to analyse the<br />

selenium containing proteins to underst<strong>and</strong> the role of the selenium <strong>and</strong> its compounds in the<br />

protective mechanisms. In addition, there may be further selenoproteins which may likewise<br />

be of significance in the antioxidant defence system of the respiratory tract.<br />

- 349 -


Session X : Cell death in cancer Poster X, 17<br />

Pirfenidone suppresses TGF-beta <strong>and</strong> is cytotoxic in malignant glioma cells<br />

Isabel Burghardt, Steffen A. Aulwurm, Brigitte Frank, Michael Weller <strong>and</strong> Wolfgang<br />

Wick<br />

Laboratory of Molecular Neuro-Oncology, Department of General Neurology, Hertie<br />

Institute for Clinical Brain Research, University of Tuebingen, Medical School, Otfried-<br />

Mueller-Str. 27, 72076 Tuebingen, Germany<br />

Presenting author: Tel. +49 7071 2981969, FAX: +49 7071 295260,<br />

E-mail: isabel.burghardt@gmx.net<br />

Among the essential characteristics of human malignant gliomas are infiltrative growth,<br />

angiogenesis <strong>and</strong> suppression of antitumor immune surveillance. Transforming growth factorbeta<br />

(TGF-beta) is intimately involved in the regulation of these processes.<br />

The novel antifibrotic drug 5-methyl-1-phenyl-2-(1H)-pyridone (Pirfenidone) significantly<br />

reduces TGF-beta2 mRNA <strong>and</strong> protein levels in f<strong>our</strong> human glioma cell lines (LN18, LNT-<br />

229, LN-308 <strong>and</strong> T98G) in vitro. In good accordance, Pirfenidone between 1 <strong>and</strong> 5 mM also<br />

shows a dose dependent decrease in the growth inhibition of TGF-beta-sensitive CCL-64 cells<br />

mediated by TGF-beta-containing glioma cell supernatant. TGF-beta2, the major isoform in<br />

malignant gliomas, is synthesized as a pre-pro-TGF-beta polypeptide at 55 kDa that is<br />

processed to a 12,5 kDa polypeptide by cytoplasmic <strong>and</strong> secreted furin-like proteases. 5 mM<br />

Pirfenidone decreases activity of recombinant furin by 40% in vitro.<br />

In addition, Pirfenidone at 1, 2.5 <strong>and</strong> 5 mM leads to reduced cell density in LN18, LNT-229,<br />

LN-308 <strong>and</strong> T98G by 10, 20 <strong>and</strong> 40% after 48 h<strong>our</strong>s of treatment.<br />

The interaction with cell survival pathways is further demonstrated by reduction of the levels<br />

of the endogenous phosphorylation of protein kinase B(PKB)/Akt <strong>and</strong> ribosomal protein S6<br />

(RPS6) at a concentration of 5 mM, thus possibly influencing the phosphatidyl-inositol-3<br />

kinase (PI3K) cascade downstream of the epidermal growth factor receptor (EGFR).<br />

Investigation of effects on the invasive phenotype of glioma reveals that Pirfenidone has no<br />

effect on protein levels of “pro-migratory” matrix metalloproteinases matrix<br />

metalloproteinase (MMP)-2 <strong>and</strong> MMP-9, as well as on tissue inhibitor of metalloproteinase<br />

(TIMP)-2.<br />

- 350 -


Session X : Cell death in cancer Poster X, 18<br />

Dosage Effects of Ginkgolide B on Ethanol-Induced Cell Death in Human Hepatoma G2<br />

Cells<br />

Wen-Hsiung Chana <strong>and</strong> Yan-Der Hsuuwb<br />

a. Department of Bioscience Technology, Chung Yuan Christian University, Chung Li,<br />

Taiwan 32023. E-mail: whchan@cycu.edu.tw<br />

b. Department of Animal Science, National Pingtung University of Science <strong>and</strong><br />

Technology, Neipu, Pingtung, Taiwan 91201. E-mail: hsuuw@yahoo.com.tw<br />

Ginkgolide B is a major active component of Ginkgo biloba extracts, which have been shown<br />

to confer anti-cancer effects by inducing apoptosis or inhibiting oxidative stress generation.<br />

Ethanol induces a wide range of cellular toxicities, many of which have been linked to free<br />

radical generation. To further elucidate the cellular effects of ginkgolide B, we examined the<br />

dose-response effect of ginkgolide B on ethanol-induced toxicity in human Hep G2 cells.<br />

TUNEL <strong>and</strong> MTT assays revealed that ethanol (50-400 mM) induced apoptotic cell death in<br />

human Hep G2 cells, <strong>and</strong> that this effect was inhibited by low (5-25 µM) doses of ginkgolide<br />

B, but enhanced by high (50-100 µM) doses of ginkgolide B. Additional experiments revealed<br />

that ethanol treatment directly increased intracellular oxidative stress; this effect was<br />

enhanced by high doses of ginkgolide B but remained unchanged following treatment with<br />

low concentrations of ginkgolide B. The dose-response effects of ginkgolide B on reactive<br />

oxygen species (ROS) generation was directly correlated with cell apoptotic biochemical<br />

changes including c-Jun N-terminal kinase (JNK) activation, caspase-3 activation, <strong>and</strong> DNA<br />

fragmentation. These results indicate that treatment dosage may determine the effect of<br />

ginkgolide B on ethanol-induced ROS generation <strong>and</strong> cell apoptosis, <strong>and</strong> support the notion<br />

that an appropriate dosage of ginkgolide B may aid in decreasing the toxic effects of ethanol.<br />

- 351 -


Session X : Cell death in cancer Poster X, 19<br />

Downregulation of Lifeguard leads to sensitization against TRAIL induced apoptosis in<br />

tumor cell lines<br />

Claudia YU Choi, Kerstin Reimers, Susanne Kall, Peter M. Vogt<br />

Department of Plastic, H<strong>and</strong> <strong>and</strong> Reconstructive Surgery, Medical School Hannover,<br />

Podbielskistr. 380, 30659 Hannover, Germany, E-mail: Choi.Claudia@MH-<br />

Hannover.de<br />

A dysbalance between apoptosis <strong>and</strong> proliferation plays an essential role in tumor<br />

development. The expression of anti-apoptotic proteins promotes tumorigenesis <strong>and</strong><br />

resistance to chemotherapy. TRAIL is the most promising c<strong>and</strong>idate for tumor selective death<br />

receptor-activation. It activates the death receptors TRAIL-1 <strong>and</strong> TRAIL 2 inducing apoptosis<br />

preferentially in tumor cells but not in normal tissue. Specific means to sensitize tumor cells<br />

for TRAIL are desirable. Lifeguard (LFG) is a protein with inhibitory function on Fasmediated<br />

apoptosis. We hypothesized that LFG inhibits TRAIL-induced apoptosis. We<br />

determined high LFG-expression-levels in A549 cells <strong>and</strong> comparing tumor cell lines of<br />

different origins we found that TRAIL-sensitivity correlated with high LFG-expressionlevels.<br />

To selectively interfere with expression of LFG, an antisense construct with effective<br />

down-regulation was introduced to A549 cells.After treatment, the sensitivity to TRAIL was<br />

enhanced with up-regulation of the apoptotic activity. In conclusion, LFG is expressed in<br />

many TRAIL-resistant tumor cell lines <strong>and</strong> the inhibition of LFG-expression was sufficient to<br />

sensitize A549 cells from TRAIL-mediated apoptosis. Our findings indicate that LFG is a<br />

protein with pathogenic function in tumorigenesis with new therapeutic options in cancer<br />

therapy targeting LFG.<br />

- 352 -


Session X : Cell death in cancer Poster X, 20<br />

Relationship between apoptotic <strong>and</strong> autophagic programmed cell death: role of Bcl-2<br />

<strong>and</strong> Beclin 1 proteins<br />

Iwona A. Ciechomska, Christoph Goemans, Aviva Tolkovsky<br />

Department of Biochemistry, University of Cambridge, Tennis C<strong>our</strong>t Road, Cambridge,<br />

CB2 1QW, UK<br />

Email: iac23@mole.bio.cam.ac.uk; cgg20@mole.bio.cam.ac.uk;<br />

amt@mole.bio.cam.ac.uk<br />

Programmed cell death (PCD) mechanisms can be divided into several categories including<br />

type I (apoptosis) <strong>and</strong> type II (autophagic cell death). The mutual relationship between<br />

apoptotic <strong>and</strong> autophagic death is currently debated because Beclin 1, an important regulator<br />

of autophagy, is also a Bcl-2 interacting protein. Recent studies revealed that the interaction<br />

between Bcl-2 <strong>and</strong> Beclin 1 regulates starvation-induced autophagy.<br />

In this study we investigated how the interactions between Bcl-2 <strong>and</strong> Beclin-1 influence the<br />

apoptotic machinery. In order to investigate this effect first we have studied how each of these<br />

proteins individually, Bcl-2 or Beclin 1, influence the level of apoptosis in HeLa cells induced<br />

by a variety of apoptotic stimuli. The Bcl-2 protein is localized in the different cellular<br />

membranes, therefore wild-type <strong>and</strong> Bcl-2 variants that are exclusively targeted to the<br />

endoplasmic reticulum (ER) membrane or the mitochondrial outer membrane (MOM) were<br />

used. We have confirmed that Bcl-2 decreases the level of apoptosis induced by staurosporine<br />

independently on the localization, although we have found some toxic effect of wild-type Bcl-<br />

2 after transient overexpression in untreated cells. Overexpression of either wild-type or<br />

Beclin 1 mutants that cannot bind to Bcl-2 resulted in increased apoptosis. Furthermore, Bcl-2<br />

protein binds to Beclin 1 <strong>and</strong> the intracellular localization of Beclin 1 was changed according<br />

to Bcl-2 localisation. Next we examined how the interaction between Beclin 1 <strong>and</strong> Bcl-2<br />

influence the level of apoptosis. Beclin 1 mutants that are defective in Bcl-2 binding induced<br />

less apoptosis than wild-type Beclin 1 after overexpression of ER- or MOM-targeted Bcl-2.<br />

Thus, <strong>our</strong> data suggest that Beclin 1 not only functions as a proautophagy protein, but also as<br />

a proapoptotic protein via disruption of the anti-apoptotic function of Bcl-2.<br />

- 353 -


Session X : Cell death in cancer Poster X, 21<br />

Human soluble TRAIL/Apo2L induces apoptosis in chemotherapy refractory nodal<br />

diffuse large B-cell lymphomas.<br />

Saskia AGM Cillessen1, Chris JLM Meijer1, Gert J Ossenkoppele2, Kitty CM<br />

Castricum1, August H Westra2, Petra Niesten1, Jettie JF Muris1, Hoite F Nijdam3,<br />

Klaas G van der Hem4, Marcel Flens5, Erik Hooijberg1, Joost J Oudejans1<br />

Department of Clinical Pathology1 <strong>and</strong> Hematology2, VU University Medical Center,<br />

Amsterdam. Department of Otolaryngology, Westfries Gasthuis, Hoorn3, Department<br />

of Internal Medicine4 <strong>and</strong> Clinical Pathology5, Zaans Medical Center de Heel,<br />

Za<strong>and</strong>am. The Netherl<strong>and</strong>s. Email: SAGM.Cillessen@vumc.nl<br />

Part of Diffuse Large B-Cell Lymphomas (DLBCL) is resistant to chemotherapy. Human<br />

soluble (hs) TRAIL/Apo2L might be an alternative form of therapy for these lymphomas. In<br />

isolated lymphoma cells of DLBCL <strong>and</strong> B-cell lines we investigated whether<br />

hsTRAIL/Apo2L induces apoptosis in lymphomas that are resistant to chemotherapy induced<br />

cell death.<br />

Twelve out of twenty-two DLBCL samples including seven chemotherapy refractory<br />

lymphomas were sensitive for hsTRAIL/Apo2L. hsTRAIL/Apo2L induced apoptosis was<br />

preferentially observed in DLBCL samples <strong>and</strong> B-cell lines that were relatively or completely<br />

resistant to Etoposide induced apoptosis. Furthermore, hsTRAIL/Apo2L induced apoptosis in<br />

DLBCL <strong>and</strong> B-cell lines showing high expression levels of inhibitors of the caspase 9<br />

mediated pathway Bcl-2 <strong>and</strong>/or XIAP. In hsTRAIL/Apo2L sensitive cells expression of the<br />

TRAIL receptor R1 <strong>and</strong>/or R2 <strong>and</strong> absence of R3 <strong>and</strong> R4 was observed.<br />

We conclude that hsTRAIL/Apo2L induces apoptosis in part of chemotherapy refractory<br />

nodal DLBCL <strong>and</strong> that disruption of the caspase 9 mediated pathway <strong>and</strong> expression of Bcl-2<br />

<strong>and</strong> XIAP does not confer resistance to hsTRAIL/Apo2L induced apoptosis in DLBCL <strong>and</strong><br />

B-cell lines. Thus, based on <strong>our</strong> results hsTRAIL/Apo2L appears to be a valuable alternative<br />

treatment for patients with chemotherapy refractory DLBCL.<br />

- 354 -


Session X : Cell death in cancer Poster X, 22<br />

In the absence of Insulin-like Growth Factor (IGF)-1 <strong>signaling</strong>, Interferon-gamma (IFNg)<br />

inhibits the growth of human malignant T cells<br />

Laura Conti1,2, Angela Longo1, Gabriella Regis1,2, Mirella Giovarelli1,2, Roberto<br />

Chiarle1 <strong>and</strong> Francesco Novelli1,2.<br />

1Center for Experimental Research <strong>and</strong> Medical Studies (CERMS), San Giovanni<br />

Battista Hospital-Molinette, via Santena 5, 10126 Turin, Italy, <strong>and</strong> 2Department of<br />

Medicine <strong>and</strong> Experimental Oncology, Section of Pathology, University of Turin, C.so<br />

Raffaello 30, 10125 Turin, Italy. E-mail: laura.conti@unito.it.<br />

IGF-1 is a growth factor that promotes the survival <strong>and</strong> proliferation of many tumors <strong>and</strong> cell<br />

types, <strong>and</strong> several approaches to target IGF-1 <strong>signaling</strong> have resulted in the induction of<br />

apoptosis in a broad range of tumor cells. However, we have found that human malignant T<br />

cells, which are insensitive to the antiproliferative <strong>and</strong> apoptotic effects of IFN-g, still grow in<br />

the absence of IGF-1 <strong>signaling</strong>. T cell refractoriness to the IFN-g/Signal Transducer <strong>and</strong><br />

Activator of Transcription (STAT) 1 pathway mainly rests on internalization of the IFN-g<br />

Receptor 2 (IFN-gR2) <strong>signaling</strong> chain, which is critically induced by IGF-1. Here we show<br />

that retrovirus-mediated gene transfer of a dominant negative IGF-1 Receptor (IGF-1R DN)<br />

inhibits IFN-gR2 internalization <strong>and</strong> induces its cell surface accumulation. This results in a<br />

strong <strong>and</strong> sustained IFN-g-induced STAT1 activation <strong>and</strong> consequently in the high<br />

expression of pro-apoptotic molecules that render malignant T cells susceptible to the<br />

antiproliferative <strong>and</strong> apoptotic effects of IFN-g, both in vitro <strong>and</strong> in vivo. These data indicate<br />

that inhibition of IGF-1 <strong>signaling</strong> associated with IFN-g administration could be a useful<br />

approach to inhibit the growth of neoplastic T cells resistant to each treatment on its own.<br />

- 355 -


Session X : Cell death in cancer Poster X, 23<br />

Mechanisms of Bcl-2 up-regulation in normal <strong>and</strong> tumor monocytes.<br />

Silvia Cristofanon#*, Silvia Nuccitelli*, Maria D’Alessio*, Marc Diederich# <strong>and</strong> Lina<br />

Ghibelli*.<br />

#Laboratoire de Biologie Moléculaire et Cellulaire du Cancer (LBMCC), Fondation<br />

Recherche sur le Cancer et les Maladies du Sang, Hôpital Kirchberg, 9, rue Edward<br />

Steichen, L-2540 Luxemb<strong>our</strong>g<br />

*Dipartimento di biologia, Università di Roma “Tor Vergata”, Via della Ricerca<br />

Scientifica, 00133 Rome, Italy<br />

Glutathione (GSH) depletion (i.e., with BSO) stimulates Bcl-2 up-regulation in a set of tumor<br />

cells (D’Alessio et al, 2004). These cells are able to survive to BSO <strong>and</strong> to resist to BSOinduced<br />

chemo-sensitisation: indeed, a) inhibition of Bcl-2 trans-activation sensitises U937<br />

cells to apoptosis; <strong>and</strong> b), GSH depletion up-regulates Bcl-2 in BSO resistant but not in BSOsensitive<br />

cells. Here we show that freshly isolated peripheral blood monocytes from of<br />

healthy donors up-regulate Bcl-2 as a response to the strong ROS production due to the<br />

separation protocol. BSO treatment further up-regulate Bcl-2, with a mechanism involving $glutamyl-transpeptidase<br />

(GGT) <strong>and</strong> the transcription factor NF!B. On the one side, acivicin, a<br />

specific inhibitor of GGT, though speeding up BSO-induced GSH depletion, completely<br />

abrogates Bcl-2 up-regulation. On the other side, the involvement of NF!B in the BSOdependent<br />

Bcl-2 up-regulation was previously suggested in <strong>our</strong> system by the ability of its<br />

inhibitor CAPE of impairing Bcl-2 over-expression <strong>and</strong> sensitizing U937 to BSO. The<br />

mechanism involved seems to imply a non-canonical activation of a p50-p50 NF!B<br />

homodimer (as opposed to the canonical p50-p65 heterodimer), recently demonstrated to be<br />

responsible of the NF!B mediated BCL-2 up-regulation (Kurl<strong>and</strong> et al, 2001).<br />

- 356 -


Session X : Cell death in cancer Poster X, 24<br />

Apoptotic GSH extrusion as a switch between extrinsic <strong>and</strong> intrinsic apoptotic pathway:<br />

possible involvement of caspase 2<br />

Milena De Nicola*, Claudia Cerella*, Maria D’Alessio*, Antonio Bergamaschiª, Andrea<br />

Magriniª, Lina Ghibelli*<br />

* Dipartimento di Biologia; ª Cattedra Medicina del Lavoro, Universita' di Roma Tor<br />

Vergata, Via Ricerca Scientifica 1, 00133 Roma, Italia. E.mail:<br />

milena.de.nicola@uniroma2.it<br />

A current paradigm of apoptotic <strong>signaling</strong> is that different types of apoptogenic stimuli,<br />

physiologic vs damaging, trigger different apoptotic pathways, extrinsic or intrinsic,<br />

respectively. Many recent reports describe cross-talks between these pathways, depending on<br />

cells <strong>and</strong>/or inducers. Here we describe that damaging agents such as puromycin or etoposide<br />

are able to elicit both apoptotic pathways in a mutually exclusive fashion on human tumor<br />

monocytic U937 cells, <strong>and</strong> that the switch between the two pathways depends on the<br />

extrusion vs non-extrusion of glutathione. In the first instance, the redox disequilibrium<br />

following GSH loss activates Bax which in turn causes cytochrome c release <strong>and</strong> caspase 9<br />

activation, i.e., the intrinsic pathway. In the second instance, apoptosis occurs without redox<br />

imbalance <strong>and</strong> requires caspase 8, in a fashion that is undistinguishable from a canonical<br />

extrinsic pathway triggered by Fas stimulation. Thus, glutathione extrusion is a trigger of the<br />

intrinsic pathway; as such, it is upstream to caspase 9 or 3 activation, but requires caspase 2.<br />

We propose that caspase 2 activates the intrinsic pathway by promoting GSH extrusion; it<br />

may additionally promote caspase 8 activation, possibly explaining why puromycin or<br />

etoposide are able to elicit also the extrinsic pathway.<br />

- 357 -


Session X : Cell death in cancer Poster X, 25<br />

The secret life of caspase-14<br />

G. Denecker, E. Hoste, T. Hochepied, B. Gilbert, P. Ovaere, K. D’Herde1, C.<br />

V<strong>and</strong>enBroucke, S. Lippens, L. Schoonjans2, C. Libert, P. V<strong>and</strong>enabeele <strong>and</strong> W.<br />

Declercq<br />

Molecular Signalling <strong>and</strong> Cell Death Unit, Department for Molecular Biomedical<br />

Research, Technologiepark 927, B-9052 Ghent-Zwijnaarde (http://www.dmbr.ugent.be),<br />

VIB, Ghent University. E-mail: wim.declercq@dmbr.Ugent.be<br />

1 Department of Anatomy, Embryology, Histology, Medical Physics, Ugent. 2<br />

Department of Transgene Technology, KUL, Leuven, Belgium.<br />

Caspase-14 is expressed in the differentiating keratinocytes of the epidermis <strong>and</strong> the<br />

hairfollicles. In addition, caspase-14 could also be detected in the Hassall's body's of the<br />

thymus. In the skin caspase-14 activation occurs in the uppermost layers of the epidermis <strong>and</strong><br />

correlates with epidermal cornification. To unravel the possible role of caspase-14 during skin<br />

differentiation, we have generated caspase-14-deficient mice. Caspase-14 knock-out mice<br />

were born normally with the expected Mendelian ratio. The absence of caspase-14 protein<br />

was confirmed both by western blot <strong>and</strong> immunohistochemistry. Histological sections of the<br />

back skin <strong>and</strong> thymus of newborn wild-type <strong>and</strong> caspase-14-deficient mice revealed no<br />

macroscopical differences. Immunohistochemical analysis of other early <strong>and</strong> late<br />

differentiation markers demonstrated a normal expression pattern of K1, K10, K14, loricrin,<br />

involucrin <strong>and</strong> filaggrin. However, the skin of caspase-14-deficient mice had a more shiny<br />

appearance <strong>and</strong> showed a lichiniform phenotype. Electron microscopic analysis indicated the<br />

presence of high numbers composite keratohyalin granules in the caspase-14-deficient<br />

epidermis. These granules are normally used as profilaggrin stores. In accordance, caspase-14<br />

mice have an altered profilaggrin-processing pattern. These data argue for a role of caspase-<br />

14 in the final steps of keratinocyte differentiation. Further analysis of different physiological<br />

parameters is ongoing in order to unravel the secret life of caspase-14.<br />

- 358 -


Session X : Cell death in cancer Poster X, 26<br />

Involvement of PI-3 Kinase <strong>signaling</strong> in polyunsaturated fatty acid-induced promotion<br />

of apoptosis in CaCo-2 cells.<br />

Joe-Lin du Toit, Louise Louw <strong>and</strong> Anna-Mart Engelbrecht<br />

Department of Physiological Sciences, University of Stellenbosch, Stellenbosch, 7600,<br />

South Africa. E-mail: jl@sun.ac.za<br />

The phosphoinositide 3-kinase (PI3-kinase) <strong>signaling</strong> pathway plays a pivotal role in the<br />

regulation of cell growth. A downstream component of the PI3-kinase pathway, PKB/Akt,<br />

phosphorylates <strong>and</strong> regulates the function of various substrates involved in metabolism,<br />

proliferation <strong>and</strong> apoptosis. Evidence has shown that PKB/Akt <strong>signaling</strong> also contributes to<br />

tum<strong>our</strong>igenesis <strong>and</strong> cancer progression <strong>and</strong> that PI3-kinase <strong>signaling</strong> is up regulated in colon<br />

cancer cells. This study aims to elucidate the mechanisms by which various fatty acids<br />

influence the PI3-kinase pathway in order to promote apoptosis of colon adenocarcinoma<br />

cells <strong>and</strong> slow down cancer progression.<br />

NCM 460 <strong>and</strong> CaCo-2 cells were cultured <strong>and</strong> treated with arachidonic acid (AA), oleic acid<br />

(OA), palmitic acid (PMA) <strong>and</strong> docosahexanoic acid (DHA) respectively, <strong>and</strong> incubated for<br />

48 h<strong>our</strong>s. Samples were analysed by Western blotting with antibodies directed against total<br />

PBK/Akt, phospho-PKB/Akt (Ser473 <strong>and</strong> Thr308), PI3-kinase <strong>and</strong> various substrates of<br />

PKB/Akt as well as their phosphorylated counterparts. Wortmannin (100 nM) was used to<br />

inhibit PI3-kinase. Cell viability was assessed with MTT assays.<br />

DHA was most effective to significantly increase NCM 460 cell viability whilst decreasing<br />

CaCo2 cell viability, followed by AA. With fatty acid treatment, PI3-kinase <strong>signaling</strong> was up<br />

regulated in NCM460 cells <strong>and</strong> down regulated in CaCo-2 cells, <strong>and</strong> for both cell lines this<br />

was most profound in cells treated with DHA. Phosphorylation of the substrates of PKB/Akt<br />

involved in apoptosis (Bad, FKHR) as well as other substrates such as GSK-3# <strong>and</strong> mTOR<br />

were altered in CaCo-2 cells treated with DHA. We propose a potential role for DHA as a<br />

therapeutic agent in the treatment of colon cancer.<br />

- 359 -


Session X : Cell death in cancer Poster X, 27<br />

Cationic surfactants induced apoptosis in normal <strong>and</strong> cancer cells.<br />

Riyo Enomoto 1 , Chie Suzuki 1 , Masataka Ohno 1 , Toshinori Ohasi 1 , Ryoko Futagami 1 ,<br />

Keiko Ishikawa 1 , Mika Komae 1 , Takayuki Nishino 1 , Yasuo Konishi 2 <strong>and</strong> Eibai Lee 1 .<br />

1 Department of Pharmacology, Faculty of Pharmaceutical Sciences, Kobe Gakuin<br />

University, Ikawadani-cho, Nishi-ku, Kobe 651-2180 Japan, 2 Biotechnology Research<br />

Institution, National Research Council Canada, 6100 Royalmount Avenue, Montreal,<br />

Quebec, H4P 2R2, Canada. E-mail : suzuki@pharm.kobegakuin.ac.jp<br />

Cationic surfactants such as benzethonium chloride which possess quaternary ammonium salt<br />

are used as a preservative <strong>and</strong> an antimicrobial agent. Recently, these compounds in eye drops<br />

have been reported to induce apoptosis in conjunctive cells. Here, we examined whether<br />

various types of surfactants induces apoptosis or not in normal <strong>and</strong> cancer cells.<br />

Benzethonium chloride induced apoptosis at the concentration lower than its critical micelle<br />

concentration (CMC) in rat thymocytes. Other quaternary ammonium surfactants such as<br />

cetyltrimethylammonium bromide similarly increased biochemical <strong>and</strong> morphological<br />

features of apoptosis whereas both anionic <strong>and</strong> amphoteric surfactants had no significant<br />

effect on these apoptotic changes. These results suggest that the cationic charge of quaternary<br />

ammonium surfactants rather than the surface action of these surfactants is involved with<br />

onset of the apoptotic process. Benzethonium chloride increased the release of cytochrome c<br />

<strong>and</strong> followed the activation of caspase-9 <strong>and</strong> -3. The treatment of benzethonium chloride also<br />

led to apoptotic cell death in Jurkat cells. These results indicate that quaternary ammonium<br />

surfactants induce apoptosis in normal <strong>and</strong> cancer cells.<br />

- 360 -


Session X : Cell death in cancer Poster X, 28<br />

Butylated hydroxyanisole is more than a reactive oxygen species scavenger<br />

Nele Festjens1, Michael Kalai1,2, Joél Smet3, Ann Meeus1, Rudy Van Coster3, Xavier<br />

Saelens1,4 <strong>and</strong> Peter V<strong>and</strong>enabeele1<br />

1Molecular Signalling <strong>and</strong> Cell Death Unit, Department for Molecular Biomedical<br />

Research, Ghent University <strong>and</strong> Flemish Interuniversity Institute for Biotechnology<br />

(VIB), Technologiepark 927, 9052 Ghent, Belgium; E-mail:<br />

Nele.Festjens@dmbr.Ugent.be<br />

2Laboratory of Cellular Microbiology, Unit of Molecular Microbiology, Institute<br />

Pasteur of Brussels, Engel<strong>and</strong>straat 642, 1180 Brussels, Belgium<br />

3Department of Pediatrics, Division of Pediatric Neurology <strong>and</strong> Metabolism, Ghent<br />

University Hospital, De Pintelaan 185, 9000 Ghent, Belgium<br />

4Molecular Virology Unit, Department for Molecular Biomedical Research, Ghent<br />

University <strong>and</strong> VIB, Technologiepark 927, 9052 Ghent, Belgium<br />

L929 fibrosarcoma cells treated with TNF or dsRNA die by necrosis, a process in which<br />

mitochondria-produced reactive oxygen species (ROS) are crucial. We studied the effects of<br />

two related chemical antioxidants, butylated hydroxyanisole (BHA) <strong>and</strong> butylated<br />

hydroxytoluene (BHT). Although they reduced ROS levels similarly, BHA was clearly more<br />

anti-necrotic than BHT, showing that inhibition of necrosis does not exclusively depend on<br />

scavenging ROS. While BHA delays TNF-induced necrosis, it apparently does not block<br />

dsRNA-induced cytotoxicity. However, cell death was shifted from necrosis to apoptosis.<br />

BHT did not modulate cell death type indicating that inhibition of necrotic cell death does not<br />

exclusively depend on scavenging ROS <strong>and</strong> implying that BHA has additional features<br />

lacking in BHT. We then compared the effects of both antioxidants on the activities of<br />

complexes of the mitochondrial electron transport chain (ETC) with those of specific ETC<br />

inhibitors. BHA is more effective than BHT, but less than rotenone, in the inhibition of<br />

NADH-dehydrogenase activity. However, rotenone is less efficient as anti-necrotic agent,<br />

suggesting that other features of BHA contribute to its protective effect. We finally<br />

demonstrate that phospholipase A2 <strong>and</strong> lipoxygenases play a role in signalling to or execution<br />

of necrotic cell death. We conclude that the strong anti-necrotic effect of BHA reflects not<br />

only its ROS scavenging property, but also its ability to inhibit complex I <strong>and</strong> lipoxygenases.<br />

- 361 -


Session X : Cell death in cancer Poster X, 29<br />

Combination of doxorubicin <strong>and</strong> sulforaphane for reversing doxorubicin-resistant<br />

phenotype in mice fibroblasts with p53Ser220 mutation<br />

Carmela Fimognari, Giorgio Cantelli-Forti, Patrizia Hrelia.<br />

Department of Pharmacology, University of Bologna, Bologna, Italy. E-mail:<br />

carmela.fimognari@unibo.it<br />

Traditional cytotoxic chemotherapeutic approaches cannot cure most advanced solid<br />

malignancies. The major factor that limits the effectiveness of chemotherapy in patients with<br />

advanced cancer is the acquisition of resistance. Chemoresistance is a multifactorial process,<br />

which includes alterations in drug accumulation, increased activity of gluthatione Stransferases,<br />

loss of function <strong>and</strong> mutations of p53, etc. One strategy for reversing drug<br />

resistance is the concomitant use of chemopreventive agents (i.e. non-cytotoxic agents able to<br />

block the progression to invasive cancer) that are by themselves non-toxic but that cause a<br />

better response rates than either reagent alone. Sulforaphane is one of the most promising<br />

chemopreventive agent. Sulforaphane inhibits cell-cycle progression <strong>and</strong> induces apoptosis in<br />

different tumor cell lines. The pro-apoptotic potential of sulforaphane could be effective<br />

either alone or in combination with other therapeutic strategies in reversing chemoresistance.<br />

We firstly investigated the effects of sulforaphane on mouse fibroblasts bearing a different<br />

p53 status (wild-type, knock-out, mutated) for underst<strong>and</strong>ing whether its activity is prevented<br />

by a mutated p53 status. p53-knock-out fibroblasts from newborn mice transfected with the<br />

p53Ser220 mutation, observed in different human cancers, were used as a model of mutated<br />

p53 status. Moreover, since p53Ser220 mutation fibroblasts showed a doxorubicin-resistant<br />

phenotype, we secondly treated the cells with a combination of doxorubicin plus<br />

sulforaphane. To clarify the optimal schedule of combination, we studied the effects of<br />

simultaneous <strong>and</strong> sequential exposure to doxorubicin <strong>and</strong> sulforaphane. Taken together, <strong>our</strong><br />

results suggest that a mutated p53 status did not prevent the induction of apoptosis by<br />

sulforaphane <strong>and</strong> that sulforaphane was able to reverse the resistance to doxorubicin when<br />

administered simultaneously or after doxorubicin. The association sulforaphane-doxorubicin<br />

may therefore allow doxorubicin to be administered at lower doses, thereby reducing its<br />

potential toxicity.<br />

- 362 -


Session X : Cell death in cancer Poster X, 30<br />

Alterations in the mRNA expression of the novel, apoptosis-related gene, BCL2L12,<br />

after treatment of human leukemic cell line HL60 with the antineoplasitic agent<br />

etoposide.<br />

Kostas V. Floros1, Hellinida Thomadaki1, Dimitra Florou1, Maroulio Talieri2 , <strong>and</strong><br />

Andreas Scorilas1<br />

1 Department of Biochemistry <strong>and</strong> Molecular Biology, Faculty of Biology, University of<br />

Athens, GR-15701 Athens, Greece. E-mail: ascorilas@biol.uoa.gr<br />

2‘G. Papanicolaou’ Research Center for Oncology, Saint Savas Hospital, GR-11522<br />

Athens, Greece.<br />

Apoptotic cell death is a high-regulated process, which plays a crucial role in many biological<br />

events. Etoposide is an antineoplastic drug which targets the DNA unwinding enzyme,<br />

topoisomerase II. It is observed that in specific concentrations it rapidly causes death of<br />

myeloid leukemia cells by apoptosis. HL-60, a human promyelocytic cell line, is very<br />

sensitive to a number of apoptosis-inducing agents. We investigated the possible alterations in<br />

the expression of the novel apoptosis-related gene, BCL2L12, which was cloned by <strong>our</strong><br />

group, after treatment of the HL-60 cell line with etoposide. Apoptosis was assessed by DNA<br />

laddering <strong>and</strong> cell toxicity was investigated by the MTT method. Total RNA was extracted<br />

<strong>and</strong> cDNA was prepared by reverse-transcription. BCL2L12 <strong>and</strong> some other apoptosis-related<br />

genes were amplified by PCR, using specific primers. #-Actin was used as a control gene.<br />

Analysis of RT-PCR product demonstrated a gradual downregulation of BCL2L12 <strong>and</strong> more<br />

than 5 fold decrease 4h after drug treatment. Our results suggest that mRNA expression<br />

analysis of BCL2L12 in human promyelocytic leukemia cells HL-60 after treatment with<br />

etoposide, may serve, after further studies, as new molecular factor predicting chemotherapy<br />

outcome in human leukemia in the future.<br />

- 363 -


Session X : Cell death in cancer Poster X, 31<br />

Apoptotic cell death after Foscan(-induced photosensitization in respect of Foscan<br />

subcellular localization<br />

Aurélie François, Sophie Marchal, François Guillemin, Lina Bolotine<br />

Centre Alexis Vautrin, CRAN UMR 7039 CNRS, INPL UHP Nancy I, V<strong>and</strong>oeuvre-Lès-<br />

Nancy<br />

Subcellular localisation of photosensitizers is a main factor involved in cell death in response<br />

to the photodynamic therapy (PDT), since the primary damage sites are closely related to the<br />

distribution of sensitizers. In this study, we have studied the influence of the time of<br />

incubation on the subcellular localisation of Foscan( <strong>and</strong> on the apoptotic cell death after<br />

PDT.<br />

At short incubation times (0.5 h to 3 h), Foscan( displays diffuse pattern with preferential<br />

localisation in the endoplasmic reticulum (ER) <strong>and</strong> the Golgi apparatus. From 12h incubation<br />

Foscan( is progressively excluded from the Golgi, <strong>and</strong> is tightly confined to ER by 24 h<strong>our</strong>s.<br />

Considering ER localization we further evaluated the response to ER stress induced by<br />

Foscan(-PDT through the expression of the ER lumen protein GRP 78 (Bip). For the same<br />

levels of photoinduced cell death the GRP 78 expression, assessed by Western Blot, was<br />

much more pronounced for 24h Foscan( incubation. Apoptotic cell death after Foscan(-<br />

PDT was assessed by the activation of caspases 12, 6 <strong>and</strong> 7 along with the PARP cleavage.<br />

Similar to GRP 78 expression, a greater activation of caspase-dependent apoptotic pathway<br />

was observed for 24h incubation compared to 3h.<br />

In conclusion, for the given level of photocytotoxicity with Foscan( after 3h or 24h<br />

incubation, the better ER stress was observed at prolonged incubation time, consistent with<br />

ER localization of Foscan(. An enhanced apoptosis through caspase-12 activation with the<br />

subsequent involvement of post-mitochondria caspases at 24h of incubation indicates that<br />

apoptotic cell death is fav<strong>our</strong>ed from ER rather than Golgi localization.<br />

- 364 -


Session X : Cell death in cancer Poster X, 32<br />

Alternative splicing of TRAIL <strong>and</strong> DR5: first clues for their role in human renal cell<br />

carcinoma<br />

N Göbel, A Krieg, U Ramp, N Wethkamp, T Kempf, HE Gabbert, C Mahotka<br />

Institute of Pathology, Heinrich Heine-University, Moorenstr.5, 40225 Düsseldorf,<br />

Germany, nadine@wcv-mail.de<br />

Objective: Tumor necrosis factor (TNF) related apoptosis-inducing lig<strong>and</strong> (TRAIL/APO2L)<br />

induces programmed cell death in a variety of neoplastic cell types, but only in a few nonneoplastic<br />

cells. In this study, we report on the tumor progression dependent expression of<br />

two novel alternative splice variants of TRAIL – identified by <strong>our</strong> group – in neoplastic <strong>and</strong><br />

non-neoplastic human cells lacking either exon 3 (TRAIL-beta) or exons 2 <strong>and</strong> 3 (TRAILgamma).<br />

Moreover, we analyzed the expression of the TRAIL receptor DR5 (TRICK2) <strong>and</strong><br />

its alternative splice variants TRICK2a <strong>and</strong> TRICK2b during tumor progression of renal cell<br />

carcinoma (RCC). Methods: Determination of tum<strong>our</strong> grading <strong>and</strong> staging by<br />

histopathological examination, total RNA-isolation, RT-PCR, statistical analysis by the<br />

Mann-Whitney <strong>and</strong> Wilcoxon test. Results: 1. TRAIL-alpha, -beta <strong>and</strong> -gamma are<br />

significantly (p=0.029, 0.024, 0.008) higher expressed in non-neoplastic than in neoplastic<br />

cells. 2. The expression ratio of TRAIL-beta <strong>and</strong> –gamma shows a significant (p=0.002)<br />

difference in normal vs. tumor cells. 3. All TRAIL variants were not regulated during tumor<br />

progression from pT1/2 to advanced pT3 stage. 4. The expression levels of the DR5 receptor<br />

variants TRICK2a <strong>and</strong> TRICK2b were not affected in normal vs. tumor cells. Interestingly,<br />

the expression levels of TRICK2a significantly (p= 0.012) increased in tumor stage pT1/2 vs.<br />

non-neoplastic cells. 5. The expression of TRICK2b in pT1/2 stages significantly (p=0.037)<br />

decreased in advanced tumor stages (pT3). Conclusions: Our data suggest that all three<br />

TRAIL variants - mainly TRAIL-gamma - <strong>and</strong> TRICK2b are significantly decreased in tumor<br />

cells, indicating their potential role in RCCs. Alternative splicing of the immature TRAIL-<br />

RNA as well as the TRICK2-RNA might be involved in fine tuning of TRAIL-induced<br />

apoptosis <strong>and</strong> underlines the complexity of the TRAIL-system in renal cell carcinoma.<br />

- 365 -


Session X : Cell death in cancer Poster X, 33<br />

Membrane fluidity changes are associated with benzo[a]pyrene-induced apoptosis in<br />

F258 cells: protection by exogenous cholesterol<br />

1Morgane Gorria, 1Xavier Tekpli, 2Odile Sergent, 1Laurence Huc, 3François Gaboriau,<br />

1Mary Rissel, 1Marie-Thérèse Dimanche-Boitrel, 1Dominique Lagadic-Gossmann<br />

1Inserm U620, Université de Rennes 1; 2UPRES EA 3891, Université de Rennes 1;<br />

3Inserm U522, Hôpital Ponchaillou, 35000 Rennes, FRANCE. E-mail:<br />

morgane.gorria@univ-rennes1.fr<br />

Polycyclic aromatic hydrocarbons (PAHs) are important environmental pollutants, to which<br />

humans are largely exposed. Besides their well-known carcinogenic effects, PAHs also<br />

induce apoptosis in different cell types such as hepatic cells (Huc et al., Faseb J, 2004;<br />

Solhaug et al., Carcinogenesis, 2004). Although activation of p53 plays a primordial role in<br />

this apoptosis, permissive pathways might also occur; indeed, we have recently evidenced a<br />

parallel activation of Na+/H+ exchanger (NHE1) with consequences on apoptosis occurrence.<br />

Such an activation might result from changes in membrane characteristics, especially as<br />

PAHs are highly lipophilic molecules <strong>and</strong> have been shown to interact with biological<br />

membranes (Jimenez et al., Environ Toxicol Chem, 2002). This study was therefore to<br />

investigate the effects of benzo[a]pyrene (B[a]P) on the membrane fluidity of F258 cells <strong>and</strong><br />

to test the role of these changes in the related apoptosis.<br />

Using the fluorescence polarization technique <strong>and</strong> the DPH probe, we first demonstrated that<br />

B[a]P (50 nM) induced an increase in bulk membrane fluidity following 48 h of exposure.<br />

This result was confirmed using EPR technique <strong>and</strong> the 12-DSA spin label. Using cariporide<br />

(30 µM) to inhibit NHE1 activation observed at 48 h (Huc et al., Faseb J, 2004), we further<br />

showed that this change in fluidity was partly related to this transporter. Exogenous<br />

application of cholesterol (30 mg/mL), a well known membrane stabilizer, was then used to<br />

test the involvement of membrane fluidization in B[a]P-induced apoptosis. Our data<br />

demonstrated that a co-treatment with this compound, besides inhibiting any membrane<br />

fluidization, significantly reduced apoptosis, as evidenced by a decrease of cell population<br />

exhibiting nuclear fragmentation <strong>and</strong> of caspase 3/7 activity. We further showed that the<br />

protective effect of cholesterol was mainly through inhibition of B[a]P-induced iron uptake,<br />

leading to subsequent reduction of oxidative stress, <strong>and</strong> hence apoptosis.<br />

In conclusion, this work suggest a role for membrane fluidity in early events of B[a]P-induced<br />

apoptosis <strong>and</strong> an implication of NHE1 activation in B[a]P-induced membrane fluidization.<br />

- 366 -


Session X : Cell death in cancer Poster X, 34<br />

INVOLVEMENT OF TRACE ELEMENT CONTAINING-PROTEINS IN THE<br />

APOPTOSIS AND REDOX PROCESSES IN THE PROSTATE AND HUMAN<br />

PROSTATE CELLS<br />

I. Grbavac, C. Wolf, N. Wenda, D. Alber, M. Kühbacher, D. Behne, A. Kyriakopoulos<br />

Hahn-Meitner-Institut Department for molecular trace element research in the life<br />

science Glienicker Str. 100, 14109 Berlin, Germany<br />

It is well known that trace elements play a significant role in many functions of the human<br />

body. Most of them are bound to proteins <strong>and</strong> have an important function like selenium <strong>and</strong><br />

zinc in the prostate. Trace elements like copper <strong>and</strong> zinc are known for their role in the<br />

apoptosis of different cells. In order to obtain more information about several trace elements<br />

in this organ, analytical methods such as instrumental neutron activation analysis (INAA) <strong>and</strong><br />

ICP-MS, which are appropriate procedures to determine trace elements, were used. To find<br />

out, whether some of these elements are bound to proteins of the prostate, the gl<strong>and</strong> was<br />

homogenized <strong>and</strong> the proteins in the homogenate were separated by electrophoretic<br />

techniques. Several antibodies against specific prostate proteins were tested. By means of<br />

these antibodies several trace element containing-proteins such as selenoproteins <strong>and</strong> Cu- Znproteins<br />

could be identified which are involved in the apoptosis <strong>and</strong> redox processes. By<br />

combining of SEC <strong>and</strong> ICP-MS some trace elements, which are bound to proteins were<br />

detected in the prostate cytosol <strong>and</strong> human prostate cell lines. The spectra of the SEC-ICP-MS<br />

let assume that trace elements like copper, zinc, manganese <strong>and</strong> others have a strong affinity<br />

to several proteins in the prostate. The finding of some new metals- <strong>and</strong> metalloids containing<br />

proteins in the micro-organelles of the prostate, let assume the existence of metalloproteins<br />

<strong>and</strong> metalloidproteins in this organ which might be play a role in the apoptosis <strong>and</strong> redox<br />

system. In this paper some characteristics are present with regard to the newly found trace<br />

element -containing proteins <strong>and</strong> their relationship to the redox processes.<br />

- 367 -


Session X : Cell death in cancer Poster X, 35<br />

A Bench-to-Bed-Side Approach for the Identification <strong>and</strong> Validation of Novel<br />

Compounds to Induce Apoptosis of Cancer Cells<br />

Maria Hägg1, Takayuki Ueno1, Maria Berndtsson1, Aleks<strong>and</strong>ra M. Havelka1, Frank<br />

Neumann2, Heiko van der Kuip3, Thomas E. Murdter3, Maria C. Shoshan1, Masakazu<br />

Toi4 <strong>and</strong> Stig Linder1<br />

1Cancer Center Karolinska, Department of Oncology <strong>and</strong> Pathology, R8:03, Karolinska<br />

Institute <strong>and</strong> Hospital, S-171 76 Stockholm, Sweden. 2BIOAXXESS Technology, Moor<br />

C<strong>our</strong>t Farm, Upper Pendock WR13 6JW, UK, fneumann@bioaxxess.com, 3Dr<br />

Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstr. 112, D-<br />

70376 Stuttgart, Germany, 4Department of Surgery <strong>and</strong> Breast Oncology, Tokyo<br />

Metropolitan Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, 113-8677,<br />

Tokyo, Japan<br />

Cell based screening offers the advantage that active, cell permeable, agents such as those that<br />

induce tumor apoptosis can directly be identified. Two-dimensional tissue culture conditions<br />

are, however, very different from those of the human tumor environment <strong>and</strong> it is difficult to<br />

decide which compounds should be developed for further pre-clinical <strong>and</strong> clinical studies.<br />

We here describe an approach based on the assessment of a caspase-cleaved protein fragment<br />

of cytokeratin-18 released from apoptotic cells that can be used to identify apoptosis-inducing<br />

drugs, assess their potency to induce apoptosis in organ cultures of human tumors, <strong>and</strong> finally<br />

monitor carcinoma cell apoptosis in vivo by using plasma samples from drug-treated mice<br />

bearing human tumor xenografts. Finally, this assay can be used to address nearly in real time<br />

the drug-induced tumor cell apoptosis by using patient blood samples.<br />

This method should be useful to facilitate the drug development process by bridging the gap<br />

between screening of drugs to clinical phase I studies.<br />

[1] Linder S, Havelka AM, Ueno T, Shoshan MC. Determining tumor apoptosis <strong>and</strong> necrosis<br />

in patient serum using cytokeratin 18 as a biomarker.<br />

Cancer Lett. (2004) 214: 1-9<br />

- 368 -


Session X : Cell death in cancer Poster X, 36<br />

Antigens <strong>and</strong> cytokine gene in antitumoral vaccines: the importance of the temporal<br />

delivery sequence in the antitumor signals<br />

Maria Jose Herrero, Rafael Botella, Francisco Dasi, Rosa Algas, Maria Sanchez,<br />

Salvador F. Aliño.<br />

Laboratory of Gene Therapy, Department of Pharmacology, Faculty of Medicine,<br />

University of Valencia. Avda Blasco Ibáñez, 15. 46010 Valencia-España. E-mail:<br />

mariajoseherrero@ono.com<br />

Different studies against cancer in the last years, including clinical trials, have shown that a<br />

correct activation of the immune system can lead to tumor rejection but also, on the other<br />

h<strong>and</strong>, an incorrect signalling results in no positive effects or even anergy. How the different<br />

signals must be given to the immune system in order to get one or the other response is not<br />

clear yet. Knowing that the genetic cell vaccines with cytokine genes as gm-csf (granulocyte<br />

<strong>and</strong> macrophage colony stimulating factor) are the best to mediate efficient antitumoral<br />

responses, we have worked assuming that both signals, GM-CSF <strong>and</strong> tumor antigens, are<br />

crucial. In order to study which is the ideal temporal sequence for their administration we<br />

have worked in a murine model of antimelanoma vaccine, with B16 cells <strong>and</strong> C57/BL6 mice,<br />

using tumor membrane protein antigens (TMP) or whole tumor cells in combination with gmcsf<br />

transfer before or after the antigen delivery. B16 cells were transfected using PEI<br />

(polyethyleneimine) polyplexes (0.02 mg/ml). Tisular transfection in TMP experiments was<br />

performed by sc. injection of 0.01 or<br />

0.05 mg m-gmcsf in 0.5 ml saline solution. Our results show that: 1) when gmcsf tisular<br />

transfection is performed before TMP delivery, a good dose-dependent correlation in tumor<br />

volume decrease is observed, but with a limit effect; in contrast, when higher doses of antigen<br />

were administered before DNA transfection, more antitumor efficacy was obtained. 2) A<br />

similar behavi<strong>our</strong>, but with stronger positive results, was observed employing whole tumor<br />

cells as antigens. Thus, an inefficient or efficient (total survival of treated mice) antitumoral<br />

response was observed when DNA was administered before or after the cell vaccine,<br />

respectively. We conclude that optimal antitumoral response can be obtained when a high<br />

antigen signal is given before (or simultaneously) to GMCSF production, while the inversion<br />

of the signals could result in the non desired inhibition or anergy of the immune response.<br />

- 369 -


Session X : Cell death in cancer Poster X, 37<br />

Epigallocatechin Gallate Dose-Dependently Induces Apoptosis or Necrosis in Human<br />

MCF-7 Cells<br />

Yan-Der Hsuuw1 <strong>and</strong> Wen-Hsiung Chan2<br />

1. Department of Animal Science, National Pingtung University of Science <strong>and</strong><br />

Technology, Neipu, Pingtung, Taiwan 91201. E-mail: hsuuw@yahoo.com.tw<br />

2. Department of Bioscience Technology <strong>and</strong> Center for Nanotechnology, Chung Yuan<br />

Christian University, Chung Li, Taiwan 32023. E-mail: whchan@cycu.edu.tw<br />

The catechins, a family of polyphenols found in tea, can evoke various responses, including<br />

cell death. However, the precise molecular mechanisms of these effects are unknown. Here,<br />

we demonstrate that treatment of human MCF-7 cells with 50 µM (-)-Epigallocatechin-3gallate<br />

(EGCG), a catechin that is highly abundant in green tea, can induce apoptotic changes,<br />

including mitochondrial membrane potential changes <strong>and</strong> activation of c-Jun N-terminal<br />

kinase (JNK), caspase-9 <strong>and</strong> caspase-3. In contrast, higher concentrations of EGCG (100-400<br />

µM) do not induce apoptosis, but rather trigger necrotic cell death in MCF-7 cells.<br />

Investigations of the possible mechanisms underlying these differences revealed that<br />

treatment with lower concentrations of EGCG (10-50 µM) directly increased intracellular<br />

oxidative stress, while higher concentrations (100-400 µM) did not. Immunoblotting revealed<br />

that treatment of MCF-7 cells with 10-50 µM EGCG caused increases in Bax protein levels<br />

<strong>and</strong> decreases in Bcl-2 protein levels, shifting the Bax/Bcl-2 ratio to favor apoptosis, while<br />

treatment with 100-400 µM EGCG had no such effect. Moreover, we observed a dosedependent<br />

decrease in intracellular ATP levels in cells treated with high-dose EGCG.<br />

Blockade of reactive oxygen species (ROS) generation <strong>and</strong> ATP synthesis using antioxidants<br />

<strong>and</strong> ATP synthesis inhibitors revealed that ROS <strong>and</strong> ATP play important roles to switch cell<br />

death types with apoptosis or necrosis. Collectively, these results indicate for the first time<br />

that EGCG treatment has a dose-dependent effect on ROS generation <strong>and</strong> intracellular ATP<br />

levels in MCF-7 cells, leading to either apoptosis or necrosis, <strong>and</strong> that the apoptotic cascade<br />

involves JNK activation, Bax expression, mitochondrial membrane potential changes, <strong>and</strong><br />

activation of caspase-9 <strong>and</strong> caspase-3.<br />

- 370 -


Session X : Cell death in cancer Poster X, 38<br />

Cooperation of Amphiregulin <strong>and</strong> IGF1 Inhibits Bax- <strong>and</strong> Bad-mediated Apoptosis via a<br />

PKC-dependent pathway in non-small cell lung cancer cells<br />

Am<strong>and</strong>ine HURBIN, Carole NIANG, Jean-Luc COLL <strong>and</strong> Marie C. FAVROT<br />

Lung Cancer Research Group, INSERM U578, Institut Albert Bonniot, 38706 La<br />

Tronche Cedex, France. E-mail : Am<strong>and</strong>ine.Hurbin@ujf-grenoble.fr<br />

Amphiregulin (AR) <strong>and</strong> insulin-like growth factor-1 (IGF1) are growth factors known to<br />

promote non-small cell lung cancer (NSCLC) survival. We have previously published that 1)<br />

AR <strong>and</strong> IGF1, secreted by H358 NSCLC cells, cooperate to protect those cells <strong>and</strong> H322<br />

NSCLC cells from serum-starved apoptosis; 2) H358 cells resist to Bax-induced apoptosis<br />

through an inhibition of Bax conformational change. We show here that the anti-apoptotic<br />

activity of AR/IGF1 combination is specifically abolished by the PKC inhibitors calphostin C<br />

<strong>and</strong> staurosporine, but not by the MAPK <strong>and</strong> PI3K inhibitors PD98059 <strong>and</strong> wortmannin,<br />

suggesting the involvement of a PKC-dependent, MAPK- <strong>and</strong> PI3K-independent survival<br />

pathway. The PKC% inhibitor rottlerin restores apoptosis induced by serum deprivation. In<br />

addition, phosphorylation of PKC <strong>and</strong> PKC / , but not of PKCalpha/#II, increases in<br />

serum-starved H358 cells <strong>and</strong> in H322 cells treated with AR/IGF1 combination <strong>and</strong> is<br />

blocked by calphostin C. Combination of AR <strong>and</strong> IGF1 increases p90Rsk <strong>and</strong> Bad<br />

phosphorylation as well as it inhibits the conformational change of Bax by a PKC-dependent<br />

mechanism. Finally, PKC , PKC or p90Rsk siRNAs block the anti-apoptotic activity of<br />

AR/IGF1 combination but have no effect on partial apoptosis inhibition observed with each<br />

factor used alone. Constitutively active PKC expression inhibits serum deprivation-induced<br />

apoptosis, whereas a catalytically inactive form of p90Rsk restores it. Thus, AR <strong>and</strong> IGF1<br />

cooperate to prevent apoptosis by activating a specific PKC-p90Rsk-dependent pathway,<br />

which leads to Bad <strong>and</strong> Bax inactivation. This signalling pathway is different to that used by<br />

single factor.<br />

- 371 -


Session X : Cell death in cancer Poster X, 39<br />

Non-autonomous p53/p21 mediated <strong>signaling</strong> in tumor – stroma interactions<br />

Hippokratis Kiaris, George Trimis & Ioulia Chatzistamou<br />

Department of Biochemistry, University of Athens Medical School, M. Asias 75, 115 27<br />

Athens, Greece. E-mail: hkiaris@med.uoa.gr<br />

During carcinogenesis stromal fibroblasts, in parallel with the neoplastic cells, undergo<br />

certain changes <strong>and</strong> progressively enter a cancer-associated state. Despite however recent<br />

progress in underst<strong>and</strong>ing the role of fibroblasts in tumor biology, how this process is being<br />

regulated remains obscure. The finding that tumor fibroblasts frequently harbor p53 mutations<br />

in breast <strong>and</strong> other cancers prompted us to address the role of fibroblasts’ p53 in<br />

tumorigenesis. Tumor transplantation experiments showed that that indeed, p53 ablation in<br />

the hosts induced reduced the latency for the development of MCF7 human breast tumors.<br />

Co-innoculation of MCF7 cells with fibroblasts differing in the status of p53 showed that this<br />

finding is at least in part due to the stromal fibroblasts. Subsequently we asked if these effects<br />

of fibroblasts’ p53 are mediated by a p21waf1/Cip1 (p21)/dependent mechanism. Therefore,<br />

analogous tumor co-inoculation experiments have been performed in SCID mice with MCF7<br />

cells <strong>and</strong> fibroblasts isolated from wild type <strong>and</strong> p21 deficient animals. Our results show that<br />

p21 deletion in stromal fibroblasts accelerates tumorigenesis by non-autonomous<br />

mechanisms. This non-autonomous stimulation of MCF7 growth by p21 ablation in<br />

fibroblasts was confirmed in vitro in co-culture experiments <strong>and</strong> also showed that at least in<br />

part it might be due to the increased expression of transforming growth factor-b <strong>and</strong> stromal<br />

cell derived factor-1. Apparently, these findings were biologically relevant because in human<br />

benign fibroadenomas p21 expression is clustered proximal to the mammary epithelial cells<br />

while in invasive breast cancers stromal fibroblasts express p21 r<strong>and</strong>omly, in a mosaic<br />

pattern. Collectively <strong>our</strong> results imply that stromal p53/p21 play important rolesin<br />

tumorigenesis <strong>and</strong> that modulation of endogenous p21 expression in stromal fibroblasts of<br />

primary tumors might be implicated, in the regulation of neoplastic growth.<br />

- 372 -


Session X : Cell death in cancer Poster X, 40<br />

ER stress induces Jpk, which inhibits cell cycle progression in F9 teratocarcinoma cell<br />

Hye Sun Kim, Kyoung-Ah Kong, Hyunjoo Chung, Sungdo Park, <strong>and</strong> Myoung Hee Kim<br />

Department of Anatomy, Embryology Lab., Brain Korea 21 Project for Medical<br />

Science, Yonsei University College of Medicine, Seoul 120-752, Korea.<br />

E-mail: goldfish79@hanmail.net; mhkim1@yumc.yonsei.ac.kr<br />

A Jopock (Jpk), a trans-acting factor associating with the position-specific regulatory element<br />

of murine Hoxa-7, has shown to induce cell death in both prokaryotic <strong>and</strong> eukaryotic cells<br />

when introduced <strong>and</strong> overexpressed. Since Jpk protein harbors a transmembrane domain<br />

(TM) <strong>and</strong> a putative ER-retension signal at the N-terminus, a subcellular localization of the<br />

protein was analyzed after fusing it into the green fluorescent protein (GFP). Both N-term<br />

(Jpk-EGFP) <strong>and</strong> C-term fused-Jpk (EGFP-Jpk) showed to be localized in the endoplasmic<br />

reticulum (ER) when analyzed under the fluorescence microscopy after staining the cells with<br />

ER- <strong>and</strong> MitoTracker. Through deletion analysis TM turned out to be important for ER<br />

localization of Jpk. When flow cytometric analysis was performed, both cells expressing Jpk-<br />

EGFP <strong>and</strong> EGFP-Jpk led cell cycle arrest <strong>and</strong> subsequent apoptotic cell death. In order to see<br />

whether Jpk is expressed during ER-stress mediated apoptosis, F9 cells were treated with<br />

DTT, an ER-stress inducer. In the presence of 4 mM of DTT, about 50% of cells died<br />

strongly expressing (7 fold) Jpk as well as Grp78, a molecular chaperone <strong>and</strong> Chop-10, a well<br />

known protein arresting cells at the G0/G1 phase <strong>and</strong> apoptosis. In summery, excess ER stress<br />

inducing apoptosis upregulated the expression of Jpk, which seemed to inhibit the cell cycle<br />

progression. These results altogether suggest that Jpk could be a useful cell death triggering<br />

molecule applicable for cancer therapy.<br />

- 373 -


Session X : Cell death in cancer Poster X, 41<br />

Buddlejasaponin IV <strong>and</strong> Pleurospermum kamtschaticum extract induce apoptosis by<br />

reducing alpha(2)beta(1) integrin-mediated adhesion in HT-29 human colon cancer cells<br />

Jin-Eun Kim1,2,3, Won-Yoon Chung1,2, Hee-Juhn Park4, Hyun-Ju Jung4 <strong>and</strong> Kwang-<br />

Kyun Park1,2,3<br />

1Department of Oral Biology, 2Oral Science Research Center, College of Dentistry,<br />

3Brain Korea 21 Project for Medical Science, Yonsei University, Seoul 120-752,<br />

4Department of Botanical Res<strong>our</strong>ces, Sangji University, Wonju 220-702, E-mail:<br />

judyjean@hanmail.net<br />

Buddlejasaponin " (BS-") is one of the active components of<br />

Pleurospermum kamtschaticum (Umbelliferae). The aerial part of P. kamtschaticum is used<br />

traditionally to treat cold, atherosclerosis, arthritis <strong>and</strong> to overcome fatigue in Korea.<br />

However, the antitumor effect of the aerial part of P. kamtschaticum extract (Pl-K) <strong>and</strong> BS-"<br />

has not been studied. Integrins are cell surface adhesion receptors that regulate cell survival<br />

<strong>and</strong> cell growth in response to cues derived from the extracellular matrix. Alpha(2)beta(1)<br />

integrin downregulation induces apoptosis in various colon cancer cell lines. We investigated<br />

whether apoptosis induced by Pl-K <strong>and</strong> BS-" was associated with alterations in integrin<br />

<strong>signaling</strong>. Pl-K <strong>and</strong> BS-" induced apoptosis in HT-29 cells through mitochondrial dependent<br />

pathway. Additionally, Pl-K <strong>and</strong> BS-" decreased the cell attachment to type# <strong>and</strong> "<br />

collagen, resulting from the downregulation of alpha(2)beta(1) integrin. Furthermore, Pl-K<br />

<strong>and</strong> BS-" inhibited expression <strong>and</strong> phosphorylation of FAK, Akt, <strong>and</strong> ERK. These results<br />

suggest that Pl-K <strong>and</strong> BS-" induce apoptosis <strong>and</strong> inhibit survival by reducing alpha(2)beta(1)<br />

integrin-mediated adhesion in HT-29 cells.<br />

- 374 -


Session X : Cell death in cancer Poster X, 42<br />

Caspase activation <strong>and</strong> extracelluar signal regulated kinase inhibition were involved in<br />

luteolin induced apoptosis in Lewis lung carcinoma cells<br />

Jin-Hyung Kim, Eun-Ok Lee, Hyo-Jung Lee, Sung-Hoon Kim<br />

Department of Oncology, Graduate School of East-West Medical Science, KyungHee<br />

University, Yongin 449-701, Republic of Korea. E-mail:sungkim7@khu.ac.kr<br />

Luteolin, a naturally occurring flavonoid, was reported to inhibit angiogenesis, DNA<br />

topoisomerase I <strong>and</strong> induce apoptosis in different cancer cells. However, the apoptotic<br />

mechanism <strong>and</strong> in vivo efficacy of luteolin still remains unclear. Thus, in the present study,<br />

we examined the underlying molecular mechanism of luteolin <strong>and</strong> its effect on in vivo tumor<br />

growth of Lewis lung carcinoma (LLC) cells. Luteolin exhibited anti-proliferative activity<br />

against LLC cells with IC50 of 12 µ). Luteolin effectively increased Annexin V positive<br />

cells as well as sub G1 DNA peaks by flow cytometric analysis. Western blotting has revealed<br />

that luteolin effectively activates the expression of caspase 9 <strong>and</strong> 3, cleaves poly(ADP-ribose)<br />

polymerase (PARP) <strong>and</strong> increases the ratio of Bax/Bcl-2. Furthermore, mitochondrial<br />

membrane potential was reduced in a concentration dependent manner by luteolin. However,<br />

it did not affect the expression of protein kinase c (PKC) "/#, PKC & <strong>and</strong> pan PKC, while it<br />

down-regulated the expression of extracellular signal-regulated kinase (ERK). In addition,<br />

luteolin significantly inhibited the growth of LLC cells implanted on the flank of mice to 40<br />

% <strong>and</strong> 60% of untreated control group at 2 mg/kg <strong>and</strong> 10 mg/kg, respectively. Similarly,<br />

luteolin significantly reduced the expression of proliferating cell nuclear antigen (PCNA) as<br />

well as increased the expression of Terminal deoxynucleotidyl transferase biotin-dUTP nick<br />

end labeling (TUNEL) in tumor section of LLC bearing mice by immunohistochemistry.<br />

Taken together, these results suggest that luteolin exerts anti-tumor activity by caspase<br />

activation <strong>and</strong> ERK inhibition.<br />

- 375 -


Session X : Cell death in cancer Poster X, 43<br />

The anti-cancer activity of decursin <strong>and</strong> decursinol isolated from Angelica gigas Nakai<br />

Mi-Jeong Kim1,2, Won-Yoon Chung1, Jin-Eun Kim1,2, Seung Hwa Son1, Sang Kook<br />

Lee3, Kwang-Kyun Park1,2<br />

1Department of Oral Biology, Oral Science Research Institute, Oral Cancer Research<br />

Institute, Yonsei university College of Dentistry, Seoul 120-752, Republic of Korea,<br />

2Brain Korea 21 Project for Medical Science, Yonsei University, Seoul Republic of<br />

Korea 120-752, Republic of Korea, 3College of Pharmacy , Ewha Womans University,<br />

Seoul Republic of Korea 120-750 E-mail: kimmi780507@hanmail.net<br />

Angiogenesis, the process of vascular growth by sprouting of preexisting vessels, plays a<br />

crucial role in tumor growth <strong>and</strong> metastasis. Vascular endothelial cell proliferation <strong>and</strong><br />

migration are critical steps in angiogenesis <strong>and</strong> are regulated by various growth factors such<br />

as vascular endothelial growth factor (VEGF). Angelica gigas Nakai root has been<br />

traditionally used in Korean folk medicine as a tonic <strong>and</strong> for treating anemia <strong>and</strong> other<br />

common disease. In the present study, we observed that decursin <strong>and</strong> decursinol isolated from<br />

A. gigas inhibited VEGF-induced proliferation <strong>and</strong> migration of human umbilical vein<br />

endothelial cells (HUVECs). Also, these compounds showed antiangiogenic activity in a<br />

mouse Matrigel plug assay <strong>and</strong> CAM assay. In addition, decursin <strong>and</strong> decursinol inhibited the<br />

formation of tumor nodules in spontaneous lung metastasis induced with murine colon cancer<br />

CT-26 cells in BALB/c mice. Our results suggest that decursin <strong>and</strong> decursinol possess an<br />

antimetastatic potential by suppressing VEGF-induced angiogenesis. Furthermore, to explain<br />

the mechanism for how decursin <strong>and</strong> decursinol block VEGF-induced angiogenesis, we<br />

investigated their effect on PI3k/Akt/eNOS <strong>signaling</strong> in HUVECs.<br />

- 376 -


Session X : Cell death in cancer Poster X, 44<br />

ER stress induces Jpk, which inhibits cell cycle progression in F9 teratocarcinoma cell<br />

Hye Sun Kim, Kyoung-Ah Kong, Hyunjoo Chung, Sungdo Park, <strong>and</strong> Myoung Hee Kim<br />

Department of Anatomy, Embryology Lab., Brain Korea 21 Project for Medical<br />

Science, Yonsei University College of Medicine, Seoul 120-752, Korea.<br />

E-mail: goldfish79@hanmail.net; mhkim1@yumc.yonsei.ac.kr<br />

A Jopock (Jpk), a trans-acting factor associating with the position-specific regulatory element<br />

of murine Hoxa-7, has shown to induce cell death in both prokaryotic <strong>and</strong> eukaryotic cells<br />

when introduced <strong>and</strong> overexpressed. Since Jpk protein harbors a transmembrane domain<br />

(TM) <strong>and</strong> a putative ER-retension signal at the N-terminus, a subcellular localization of the<br />

protein was analyzed after fusing it into the green fluorescent protein (GFP). Both N-term<br />

(Jpk-EGFP) <strong>and</strong> C-term fused-Jpk (EGFP-Jpk) showed to be localized in the endoplasmic<br />

reticulum (ER) when analyzed under the fluorescence microscopy after staining the cells with<br />

ER- <strong>and</strong> MitoTracker. Through deletion analysis TM turned out to be important for ER<br />

localization of Jpk. When flow cytometric analysis was performed, both cells expressing Jpk-<br />

EGFP <strong>and</strong> EGFP-Jpk led cell cycle arrest <strong>and</strong> subsequent apoptotic cell death. In order to see<br />

whether Jpk is expressed during ER-stress mediated apoptosis, F9 cells were treated with<br />

DTT, an ER-stress inducer. In the presence of 4 mM of DTT, about 50% of cells died<br />

strongly expressing (7 fold) Jpk as well as Grp78, a molecular chaperone <strong>and</strong> Chop-10, a well<br />

known protein arresting cells at the G0/G1 phase <strong>and</strong> apoptosis. In summery, excess ER stress<br />

inducing apoptosis upregulated the expression of Jpk, which seemed to inhibit the cell cycle<br />

progression. These results altogether suggest that Jpk could be a useful cell death triggering<br />

molecule applicable for cancer therapy.<br />

- 377 -


Session X : Cell death in cancer Poster X, 45<br />

Akt involvement in paclitaxel chemoresistance of human ovarian cancer cells<br />

Su-Hyeong Kim 1, Yong-Sung Juhnn 1,2, Yong-Sang Song1,3<br />

1Cancer Research Institute, 2Department of Biochemistry <strong>and</strong> Molecular Biology,<br />

3Department of Obstetrics <strong>and</strong> Gynecology, Seoul National University, College of<br />

Medicine Seoul, 110-799, Korea, E-mail:yssong@snu.ac.kr<br />

Paclitaxel (taxol) is extensively used clinically for chemotherapy of various cancers including<br />

ovarian cancer. Although paclitaxel induces apoptosis in cancer cells, its exact mechanism of<br />

action still remains to be determined. Akt mediates survival signals that might protect cancer<br />

cells from apoptosis <strong>and</strong> thus is a potentially important therapeutic target. Here, we<br />

demonstrated that inhibition of Akt increases the efficacy of the paclitaxel-induced apoptosis<br />

using SKOV3 <strong>and</strong> PA-1 human ovarian cancer cells.<br />

The sensitivity to paclitaxel of SK0V3 <strong>and</strong> PA-1 cells was examined using the MTT assay. At<br />

a concentration of 30 µM, PA-1 cells were more sensitive to paclitaxel than SKOV3 cells.<br />

Apoptosis was accompanied by release of cytochrome c into the cytoplasm <strong>and</strong> cleavage of<br />

poly (ADP-ribose) polymerase (PARP). To further elucidate the mechanism of sensitization<br />

by paclitaxel, we explored the difference in the Akt phosphorylation between paclitaxelresistant<br />

SKOV3 cells <strong>and</strong> paclitaxel–sensitive PA-1 cells. The higher level of phosphorylated<br />

Akt was shown in SKOV3 cells than in PA-1 cells in response to paclitaxel.<br />

Inhibition of Akt by specific phosphatidyinostiol-3-kinase (PI3K)-Akt (Wortmannin, <strong>and</strong><br />

LY294002) increased the efficacy of the paclitaxel-induced apoptosis in both cells.<br />

These results suggest that combination therapy of paciltaxel with the Akt inhibitor may<br />

increase the therapeutic efficacy of paclitaxel.<br />

- 378 -


Session X : Cell death in cancer Poster X, 46<br />

Cytoplasmic fraction of Lactococcus latic ssp. Lactis induces apoptosis <strong>and</strong> G0/G1 cell<br />

cycle arrest in SNU1 human stomach cancer cells<br />

Seo Young Kim, Ki Won Lee, Min A Jeong, Ji Yeon Kim, <strong>and</strong> Hyong Joo Lee*<br />

Department of Food Biotechnology, School of Agricultural Biotechnology, <strong>and</strong> Center<br />

for<br />

Agricultural Biomaterials, Seoul National University, Seoul 151-742, Republic of Korea<br />

Lactic acid bacteria are known to have antitumor activity, but the underlying mechanisms<br />

remain unclear. The present study investigated antiproliferative activity of cytoplasmic<br />

fraction of Lactococcus latic ssp. (L.lac CF) on SNU-1 human stomach cancer cell line (SNU-<br />

1 cells) <strong>and</strong> underlying molecular mechanisms. The proliferation of SNU-1 cells was<br />

inhibited by the treatment of L.lac CF in a time- <strong>and</strong> dose-dependent. In addition, L.lac CF<br />

showed a significantly more relevant G0/G1 cell cycle arrest, which associated increase of<br />

p21 <strong>and</strong> reduction of cyclin D1 <strong>and</strong> phosphorylation level of Rb. Furthermore, L.lac CF<br />

induced apoptosis of SNU-1 cells, but not SNU-C2A human colon cancer cells, as evidenced<br />

by nuclei condensation, increase of sub-G1 peak, <strong>and</strong> DNA fragmentation. In particular, p53<br />

<strong>and</strong> bcl-2 appears to play a critical role in the apoptosis of SNU-1 cells by L.lac CF. We<br />

further investigated if arginine deiminase (ADI) is involved in the antiproliferative activity of<br />

L.lac CF, since arginine is known as essential amino acid for growing cells. Surprisingly, only<br />

L.lac CF, which showed significantly strong antiproliferative activity <strong>and</strong> ADI activity among<br />

tested six LAB. The concentration of L.lac CF showed linear relationship with both ADI<br />

activity <strong>and</strong> antiproliferative activity. Furthermore, addition of L-arginine recover the<br />

antiproliferative activity of L.lac CF. We purified ADI from L.lac CF by subsequent<br />

fractionations, <strong>and</strong> the IC50 value of final ADI fraction was 2 µg/ml. The ADI fraction also<br />

exerted apoptosis as evidenced by nuclei condensation in cells. These results, taken together,<br />

indicate that the antiproliferative activity of L.lac CF on SNU-1 is attributable to induction of<br />

G0/G1 cell cycle arrest <strong>and</strong> apoptosis, particularly the latter one by ADI.<br />

- 379 -


Session X : Cell death in cancer Poster X, 47<br />

Involvement of AMPK <strong>signaling</strong> cascade in capsaicin-induced apoptosis of<br />

HT-29 colon cancer cells<br />

Young Min Kim 1 Jin-Taek Hwang 2 , Dong Wook Kwak 1 , Yun Kyung Lee 1 <strong>and</strong><br />

Ock Jin Park 3<br />

1 Department of Biological Sciences, Hannam University, Daejeon 306-791, Korea<br />

2 Department of Biochemistry <strong>and</strong> Molecular Biology, Medical Research Center for<br />

Bioreaction to Reactive Oxygen Species, Kyung Hee University College of Medicine,<br />

Seoul 130-791, Korea<br />

3 Department of Food <strong>and</strong> Nutrition, Hannam University, 133 Ojeong-dong Daedeok-gu,<br />

Daejeon 306-791, Korea, E-mail: ojpark@hannam.ac.kr<br />

Capsaicin has been used in food additives <strong>and</strong> drugs. In animal studies there are conflicting<br />

reports of the effects of capsaicin on carcinogenesis. Capsaicin itself was mutagenic <strong>and</strong><br />

promoted tumor formation, whereas it showed anticarcingenic <strong>and</strong> antimutagenic properties.<br />

Capsaicin induces apoptosis in certain types of cancers, but the molecular mechanism of<br />

apoptosis caused by capsaicin has not been elucidated. In this study, we have investigated the<br />

effects of capsaicin on apoptosis in relation to AMPK (AMP-activated protein kinase)<br />

activation in colon cancer cell. Capsaicin-induced apoptosis was revealed by presence of<br />

nucleobodies in the capsaicin-treated HT-29 colon cancer cells. Concomitantly, the activation<br />

of AMPK <strong>and</strong> the increased expression of the inactive form of acetyl-CoA carboxylase<br />

(ACC) were detected in capsaicin-treated colon cancer cells. We showed that both capsaicin<br />

<strong>and</strong> AICAR, an AMPK activator possess the AMPK activating capacity as well as apopotosisinducing<br />

properties. Also, compared to genistein or EGCG, capsaicin exhibited the similar<br />

AMPK activating capacity. We suggest that AMPK is an important component of apoptosis<br />

induced by capsaicin in colon cancer cells further implying AMPK as a possible target of<br />

cancer control.<br />

- 380 -


Session X : Cell death in cancer Poster X, 48<br />

Ceramide–modulation of antigen-triggered Ca2+-signals <strong>and</strong> cell fate: a diversity in the<br />

responses by different lymphoid <strong>and</strong> myeloid cells.<br />

Endre Kiss, Gabriella Sármay <strong>and</strong> János Matkó<br />

Eötvös Lor<strong>and</strong> University, Institute of Biology, Department of Immunology, Budapest,<br />

Hungary<br />

Release of ceramide (Cer) from plasma membrane sphingomyelin upon cell death, stress <strong>and</strong><br />

inflammation stimuli is a signal mediating the mitochondrial cell death pathway in various<br />

cell types. We have shown recently that Cer-accumulation differentially affects T-cell fate<br />

(apoptosis vs. survival) depending on its strength <strong>and</strong> duration [Detre et al, Cellular Signalling<br />

2006. 18:294-306]. Moderate, non-apoptotic Cer stimuli suppressed the early <strong>and</strong> late events<br />

of both antigen-specific or polyclonal T-cell activation signalling. K+ channels (Kv1.3, KCa)<br />

as regulators <strong>and</strong> Ca2+ channels (CRAC, VDCC) as executors of the calcium influx were<br />

proposed so far as potential targets of Cer-mediated inhibition. The molecular background of<br />

the ceramides’ modulatory effect, however, still remained unresolved. Here we analyzed how<br />

general is this modulation among further antigen-dependent cells of the immune system, with<br />

attention to their maturation or differentiation stage, as well. The antigen-dependent calcium<br />

signals of murine (IP12-7) <strong>and</strong> human (Jurkat) T cells, as well as of mast cells (RBL-2H3)<br />

were remarkably inhibited by short (10 min) C2-Cer stimuli, in a concentration dependent<br />

manner. Similar, but much less inhibition was observed in murine B splenocytes <strong>and</strong><br />

immature or mature B cell lines, 38C13 <strong>and</strong> A20, respectively. In contrast, in two human<br />

Burkitt’s lymphoma B cell lines (BL41 <strong>and</strong> ST486) or the X16C murine B cell line of<br />

marginal zone origin the magnitude of the antigen-BCR mediated calcium response was<br />

uneffected or enhanced by Cer compared to the untreated cells. Interestingly, the sensitivity of<br />

the above cells to Cer-mediated apoptosis (long duration C2-Cer), monitored through<br />

mitochodrial depolarization <strong>and</strong> DNA-fragmentation, showed a similar diversity. The<br />

Burkitt’s lymphoma cells were highly resistant, in contrast to the high <strong>and</strong> Cer dosedependent<br />

apoptotic rate of T cells. The murine immature <strong>and</strong> mature B cells did not display<br />

any apoptotic marker either, but became PI-positive (necrotic), depending on the ceramide<br />

dose. Our data together suggest that the antigen-dependent immune cells have differential<br />

sensitivity to ceramide-generating death or stress signals. Underst<strong>and</strong>ing the mechanisms<br />

underlying these differences needs further investigations, particularly on Cer-induced<br />

reorganization of the plasma membrane <strong>and</strong> identification of further molecular targets for<br />

ceramide action involved in modulation of antigen-induced activation signals.<br />

- 381 -


Session X : Cell death in cancer Poster X, 49<br />

Sensitivity of human glioma cells to pro-apoptotic cannabinoid treatment is due to<br />

specific expression of cannabinoid receptors<br />

Liliana Konarska 1, Aleks<strong>and</strong>ra Ellert-Miklaszewska 1, Bozena Kaminska 2, Wieslawa<br />

A. Grajkowska 3, Konrad Gabrusiewicz 2, Malgorzata Danilkiewicz 2<br />

1Dept. of Biochemistry <strong>and</strong> Clinical Chemistry, Medical University, Banacha 1 str., 02-<br />

091 Warsaw, Pol<strong>and</strong>, Email: konarska@nencki.gov.pl 2Lab. of Transcription<br />

Regulation, Nencki Institute, Pasteura 3 str., 02-093 Warsaw, Pol<strong>and</strong>, 3Dept. of<br />

Pathology, Children’s Memorial Health Institute, Dzieci Polskich 20 str., 04-730<br />

Warsaw, Pol<strong>and</strong><br />

Cannabinoids, originally derived from Cannabis sativa, have been recently extensively<br />

studied as potential antitumoral agents. In the current study we examined whether synthetic<br />

cannabinoids with different receptor specificity are able to induce apoptosis in human glioma<br />

cells <strong>and</strong> whether selective CB2 receptor activation is sufficient to effectively kill tumor cells.<br />

Human glioma cell lines derived from highly malignant brain tumors, including T98G,<br />

U373MG, U87MG <strong>and</strong> LN229 cells as well as 3 primary human glioma cell lines T3, T10<br />

<strong>and</strong> T1Y1 were exposed to WIN55,212-2 (non-selective CB1/CB2 agonist) <strong>and</strong> JWH133<br />

(CB2-selective agonist). The expression of CB1 <strong>and</strong> CB2 cannabinoid receptors was<br />

investigated by RT-PCR <strong>and</strong> immunocytochemistry. WIN-55,212-2 decreased cell viability in<br />

all examined cell lines <strong>and</strong> induced severe changes in cell morphology. Susceptibility of the<br />

cells to JWH133 treatment correlated with the CB2 cannabinoid receptor expression. Both<br />

cannabinoids, once effective, triggered a decrease of mitochondrial membrane potential,<br />

cleavage of caspase-9 <strong>and</strong> effector caspases. Using immunohistochemistry we evaluated the<br />

CB2 receptor expression in paraffin sections from various histopathological types of human<br />

gliomas. Most of the analyzed tumors expressed significant levels of CB2 receptor. The<br />

extent of CB2 expression in the tum<strong>our</strong> specimens was related to tum<strong>our</strong> malignancy, which<br />

was in line with <strong>our</strong> RT-PCR <strong>and</strong> immunocytochemistry studies. We conclude that<br />

cannabinoids are efficient inhibitors of human glioma cells growth, once the cells express<br />

specific type of cannabinoid receptor. Due to the presence of the CB1 receptors on normal<br />

cells, the use of a selective CB2 receptor agonist, such as JWH133, seems to be a better<br />

choice for in vivo studies <strong>and</strong> in terms of potential therapeutic approaches. Supported by the<br />

grant No. 06/2002 from the Polish Pharmacy <strong>and</strong> Medicine Development Foundation by<br />

Polpharma S.A.<br />

- 382 -


Session X : Cell death in cancer Poster X, 50<br />

Role of MAPK kinase signalling pathways in photoinduced apoptosis in<br />

cancer cells<br />

Jarmila Kralova1, Michal Dvorak1, <strong>and</strong> Vladimir Kral2<br />

1 Institute of Molecular Genetics, Academy of Sciences of the Czech Republic,<br />

Flemingovo nám 2, 166 37 Prague 6, E-mail: kralova@img.cas.cz, mdvorak@img.cas.cz;<br />

2 Department of Analytical Chemistry, Institute of Chemical Technology, Technická 5,<br />

166 28 Prague 6, Czech Republic. E-mail: Vladimir.Kral@vscht.cz<br />

Oxidative stress, such as photodynamic therapy (PDT), can act as an apoptosis inducer. In the<br />

present study we investigated apoptotic pathways activated by novel photosensitizer<br />

tetrakis(p-oligoethylenglycol) pentafluorophenyl porphyrin-mediated PDT in human<br />

promyelocytic leukemia HL-60 <strong>and</strong> mouse mammary carcinoma 4T1. We show that p38 <strong>and</strong><br />

JNK MAP kinases are markedly activated during this process. Blocking the p38 MAPK<br />

activation by specific inhibitor SB203580 or PD169316 resulted in the suppression of<br />

apoptosis contrary to JNK blocking by specific inhibitor SP600125 that rather led to its<br />

enhancement. The onset of apoptotic events + cytochrome c release, caspase activation <strong>and</strong><br />

dramatic changes in the expression of some Bcl-2 family members (down-regulation of Bcl-2<br />

<strong>and</strong> Bak, cleavage of Bad <strong>and</strong> Mcl-1, activation of pro-apoptotic Bid protein) appeared<br />

concurrently within 30 min following PDT treatment in a time-response manner. Apoptosis<br />

could also be partly inhibited by overexpression of Bcl-2. Pre-incubation of cells with caspase<br />

family inhibitor, fluoromethylketone (Z-VAD-FMK), significantly reduced apoptosis, while<br />

individual caspase-specific inhibitors had only a partial effect. These observations indicate<br />

that multiple signalling pathways, which play diverse roles in the apoptotic process, are<br />

activated during PDT in a cell type-specific manner. The p38 MAPK signalling pathway<br />

seems to be directly involved in the activation, while the Akt – JNK1 pathway in the cellular<br />

resistance against PDT-induced apoptosis. Multi-functional signalling network enables<br />

additional augmentation of the apoptotic process e.g. through activation of pro-apoptotic Bcl-<br />

2 family protein Bid <strong>and</strong> caspase 8.<br />

- 383 -


Session X : Cell death in cancer Poster X, 51<br />

Analysis of interleukin-24-induced signalling in melanoma cells<br />

Stephanie Kreis*, Georg A. Munz**, Laure Dumoutier***, Claude Haan*, Waraporn<br />

Komyod**, Jean-Christophe Renauld***, Peter C. Heinrich** <strong>and</strong> Iris Behrmann*<br />

*Laboratoire de Biologie et Physiologie Integrée, Université du Luxemb<strong>our</strong>g, 162a,<br />

avenue de la Faïencerie, L-1511 Luxemburg, ** Dept. of Biochemistry, RWTH Aachen<br />

Medical School, D-52074 Aachen, Germany, ***Ludwig Institute for Cancer Research,<br />

University Catholique de Louvain, B-1200 Brussels, Belgium<br />

Interleukin (IL)-24, which was initially named “melanoma differentiation-associated gene 7”<br />

(mda-7), is an IL-10-type cytokine that has been discovered by means of differentiation<br />

therapy <strong>and</strong> subtraction hybridization in melanoma cells. Via two receptor complexes (IL-<br />

20R1/IL-20R2 <strong>and</strong> IL-22R/IL-20R2), IL-24 can activate tyrosine kinases of the Janus family<br />

(Jaks) <strong>and</strong> STAT transcription factors in target cells. In addition to this „classical“ signalling<br />

pathway, IL-24 has the unique property to induce apoptosis in many different cancer cells, but<br />

not in normal cells, when applied via an adenoviral vector or as GST fusion protein.<br />

Moreover, it promotes anti-oncogenic byst<strong>and</strong>er activity, it inhibits angiogenesis, synergises<br />

with radiation, <strong>and</strong> modulates immune responses. However, the mechanisms of cancer cell<br />

selectivity as well as the receptor-independent signalling events leading to IL-24-induced<br />

apoptosis remain unclear.<br />

A panel of 20 melanoma cell lines as well as normal human keratinocytes <strong>and</strong> primary human<br />

melanocytes were analysed with respect to their Jak-STAT signalling capacity in response to<br />

IL-24 stimulation. Although all cells are RT-PCR-positive for expression of specific cytokine<br />

receptor subunits, only keratinocytes responded to IL-24 stimulation by phosphorylation of<br />

STAT3. Neither the primary melanocytes nor the 20 tested melanoma lines reacted to IL-24<br />

stimulation. Of note, we did not observe STAT3 phosphorylation in<br />

unstimulated melanoma cells.<br />

To further investigate the cancer apoptosis-inducing function of IL-24, we have generated <strong>and</strong><br />

expressed a GST-IL-24 fusion protein <strong>and</strong> variants thereof, based on differential splicing <strong>and</strong><br />

on a structure/function comparison of IL-10 like cytokines. Furthermore, inducible stable<br />

melanoma cell lines expressing IL-24 are being generated <strong>and</strong> first resulting data will be<br />

discussed.<br />

- 384 -


Session X : Cell death in cancer Poster X, 52<br />

Caspase 3 activation, Bcl-2 contents <strong>and</strong> soluble Fas-lig<strong>and</strong> are appear to be<br />

independent of the inflammatory marker profile in patients with sepsis <strong>and</strong> septic shock<br />

Fabian Kriebel1, Silke Wittemann2, Hsin-Yun Hsiu2, Thomas Joos2, Manfred Weiss3,<br />

E. Marion Schneider1<br />

Manfred.weiss.ulm@online.de<br />

1Sektion Experimentelle Anaesthesiologie, Universitaetsklinikum Ulm, Ulm, Germany;<br />

2NMI Natural <strong>and</strong> Medical Sciences Institute at the University of Tuebingen, Reutlingen<br />

, Germany; 3Abteilung Klinische Anaesthesiologie, Universitaetsklinikum Ulm, Ulm,<br />

Germany.<br />

Objective: In order to extend treatment <strong>and</strong> diagnostics in intensive care patients suffering<br />

from systemic inflammatory response syndrome (SIRS), sepsis <strong>and</strong>/or septic shock, we asked<br />

whether apoptosis plays a role in hyperinflammation.<br />

Patients: Twenty intensive care unit (ICU) patients were analyzed daily: 2 had systemic<br />

inflammatory response syndrome (SIRS), 5 suffered from sepsis (2 died), <strong>and</strong> 13 had septic<br />

shock (5 died).<br />

Methods: EDTA-blood was lysed on ice with Cell Lysis Buffer (BD-Germany; 10mM Tris-<br />

Hcl, 10mM NaH2PO4/NaHPO4, 130mM NaCl, 1% Triton X-100, 10mM PPi, protease<br />

inhibitor cocktail of 800 µg/ml benzamidine- HCL, 500 µg/ml o-phenanthroline, 500 µg/ml<br />

aprotinin, 500 µg/ml leupeptin, 500 µg/ml pepstatin A, 50nM PMSF). The lysate was<br />

centrifuged at 14,000rpm for 10min <strong>and</strong> stored at –20˚C. Active caspase-3 was measured with<br />

the R&D Quantikine Active Caspase-3 Immunoassay (R&D Systems, Hamburg, Germany).<br />

Cytokines <strong>and</strong> active metalloproteinases (MMPs) were quantified by the Beadlyte(<br />

Multiplex Cytokine Detection System (Upstate USA, Lake Placid, NY, USA).<br />

Metalloproteinases (MMPs) were quantified using Luminex assisted Beadlyte Assays (Qiagen<br />

LiquiChip, Hilden, Germany). Soluble FAS-Lig<strong>and</strong> (sCD178) was determined by ELISA<br />

(MBL, purchased via Beckman-Coulter, Krefeld, Germany).<br />

Results: Laboratory data obtained by plasma <strong>and</strong> cell lysate analysis demonstrate that active<br />

caspase-3 was identified in defined samples of whole blood lysates, (but never in EDTAplasma)<br />

covering (e.g. 5/7, 8/18, 6/11) consecutive days during the patients’ stay on the ICU.<br />

Caspase-3 antigen contents were not found to be related to any of the inflammatory markers,<br />

including the inflammatory cytokines (IL-1, IL-6, IL-8, TNF-a, etc.) the metalloproteinases<br />

(MMPs 1, 3, 7, 9, 13) <strong>and</strong> C-reactive protein (CRP). However, when comparing the kinetics<br />

of active caspase 3 <strong>and</strong> Bcl-2 protein quantified in whole blood lysates with plasma<br />

concentrations of soluble Fas-lig<strong>and</strong> (sCD178-L), all 3 parameters were found to be elevated<br />

either simultaneously or in close time window.<br />

Conclusions: We conclude that the activation of apoptosis can be determined in whole blood<br />

by active caspase-3 <strong>and</strong> by Bcl-2. Interestingly, the upregulation of caspase 3 coincides with<br />

high Bcl-2 contents <strong>and</strong> a consecutive peak of plasma soluble FAS-lig<strong>and</strong> in a patient with a<br />

successful reconstitution after septic shock. We therefore conclude that pro- <strong>and</strong> antiapoptotic<br />

effects during sepsis may not be related to inflammatory markers may indicate<br />

modelling of leukocyte subpopulations <strong>and</strong> may play an important role in immune<br />

reconstitution.<br />

- 385 -


Session X : Cell death in cancer Poster X, 53<br />

Cyclosporine A induced cytotoxicity, cell cycle arrest <strong>and</strong> mitochondrial apoptosis in a<br />

human monocytic leukaemia cell line<br />

Molay Kumar Roy*1, Makiko Takenaka1, Masuko Kobori1, Kazuhiko Nakahara2,<br />

Seiichiro Isobe1, Tojiro Tsushida1.<br />

1National Food Research Institute, 2-1-9, Kannondai, Tsukuba, Ibaraki, 305-0051,<br />

Japan.<br />

2Japan International Research Center for Agricultural Sciences (JIRCAS), 1-1 Owashi,<br />

Tsukuba, Ibaraki 305-8686, Japan.<br />

The immunosuppressive drug cyclosporine A (CsA) has been used in organ transplantation<br />

<strong>and</strong> in the treatment of autoimmune disorders. However, the drug causes adverse effects in<br />

kidney, liver <strong>and</strong> nervous system characterized by cellular loss in the affected area. Apoptosis<br />

was found to play a role in CsA induced cytotoxicity. Because mitochondrial membrane<br />

permeabilization is a common criterion in the most setting of apoptosis in vertebrate cells, the<br />

aim of this study was to evaluate whether CsA induces mitochondrial function loss in the<br />

pathway leading to cytotoxicity in a cell line. In this study we found that CsA caused a<br />

concentration <strong>and</strong> time dependent cell viability loss in U937 cell line. CsA treatment of cells<br />

at 10 µM dose resulted in G0/G1 arrest with concurrent decrease of cells in S <strong>and</strong> G2/M<br />

phases. In mechanistic studies related to its viability lost we found that treatments of cells<br />

with 10 µM CsA for 24 h resulted in DNA fragmentation <strong>and</strong> in the increase of annexin V<br />

positive cells. CsA also increased the activity of a cysteine protease caspase-3. In other<br />

studies, we observed that CsA treatment decreased mitochondrial membrane potential <strong>and</strong><br />

increased cytochrome c release into cytosol. Furthermore, CsA treatment increased the<br />

number of cells in sub-G0/G1 peak, an indicative of reduced DNA, however this effect was<br />

not observed, when cells were pre-treated with a broad caspase inhibitor. In the study, we<br />

also found that a higher dose of CsA induces LDH release when the cells were incubated for a<br />

longer period. The overall result suggests that the mode of cell death is dose <strong>and</strong> time<br />

dependent. Short-term incubation with lower doses of CsA arrests cell growth, this overlaps<br />

with the occurrence of apoptosis <strong>and</strong> then necrosis after longer treatment periods with higher<br />

doses.<br />

- 386 -


Session X : Cell death in cancer Poster X, 54<br />

Synergistic apoptosis induction of breast cancer cells by tamoxifen <strong>and</strong> TRAIL<br />

Lagadec C, Chopin V, Anderiaenssens E, Hondermarck H, Le B<strong>our</strong>his X<br />

ERI-8 INSERM "Signalisation des facteurs de croissance dans le cancer du sein.<br />

Proteomique fonctionnelle" UPRES-EA 1033, IFR-118 Université des Sciences et<br />

Technologies de Lille, France<br />

Although tamoxifen is widely used in the treatment of breast cancer expressing ostrogen<br />

receptors (ER); it has been recently reported that tamoxifen can induce apoptosis of ERnegative<br />

breast cancer cells. On the other h<strong>and</strong>, the TNF-related apoptosis-inducing lig<strong>and</strong><br />

(TRAIL), also called Apo2L, has been the subject of recent research as a potential new<br />

anticancer agent. We investigated the effect of tamoxifen alone or in combination with<br />

TRAIL in breast cancer cells. We showed that TRAIL-induced apoptosis was potentiated by<br />

tamoxifen in both ER-positive <strong>and</strong> ER-negative breast cancer cells. From a mechanistic<br />

st<strong>and</strong>point, combination treatment with tamoxifen <strong>and</strong> TRAIL resulted in increased cleavage<br />

of procasepases-8, 9 <strong>and</strong> Bid. Tamoxifen <strong>and</strong> TRAIL cooperated also to increase the<br />

expression of FADD, procaspase-8, Bax <strong>and</strong> Bid <strong>and</strong> to decrease the expression of FLIP <strong>and</strong><br />

Bcl-2. These modifications could constitute multiple amplification loops to result from the<br />

final synergistic induction of apoptosis. Importantly, the combination treatment with<br />

tamoxifen <strong>and</strong> TRAIL did not affect the viability of cultured normal breast epithelial cells.<br />

Moreover, tamoxifen <strong>and</strong> TRAIL were well tolerated in mice <strong>and</strong> the combination of<br />

tamoxifen <strong>and</strong> TRAIL dramatically reduced tumor burden in in vivo MDA-MB-231 tumor<br />

xenograf model. These data suggest that combination of tamoxifen <strong>and</strong> TRAIL may be useful<br />

in the treatment of ER negative breast cancers.<br />

- 387 -


Session X : Cell death in cancer Poster X, 55<br />

Forced expression of the novel heat shock protein H11 triggers cancer cell apoptosis<br />

through TAK1 activation providing a molecular target for cancer therapy<br />

Jennifer Laing, Baiquan Li, Cynthia Smith, <strong>and</strong> Laure Aurelian.<br />

Pharmacology <strong>and</strong> Experimental Therapeutics, University of Maryl<strong>and</strong> School of<br />

Medicine, Baltimore, MD, 21201, U.S.A. E-mail: laurelia@umaryl<strong>and</strong>.edu<br />

H11 is a small heat shock protein (Hsp) recently cloned in <strong>our</strong> laboratory. It retains the alphacrystallin<br />

motif, but differs from canonical family members in that its expression is inhibited<br />

in some cancer cells (notably melanoma, prostate <strong>and</strong> breast) relative to matched normal<br />

tissues. Inhibition is by aberrant DNA methylation, <strong>and</strong> can be forced by treatment with<br />

demethylating agents. The current studies were designed to examine the role of H11 overload<br />

in tumor cell fate determination. Melanoma was used as a model, because it is a prevalent<br />

cancer, which is resistant to most types of chemotherapy. To control specificity of gene<br />

upregulation, human melanoma cells were stably transfected with a retrovirus that expresses<br />

H11 under the control of a tetracycline inducible promoter. They were examined for H11<br />

expression <strong>and</strong> apoptosis at 1-3 days after doxycycline (Dox) treatment. H11 was rapidly (1<br />

day) expressed <strong>and</strong> expression was associated with activation (cleavage) of caspases-9 <strong>and</strong> -3<br />

<strong>and</strong> p38MAPK, as well as a significant increase in the % TUNEL+ cells.<br />

Immunopercipitation/immunoblotting assays indicated that H11, but not its apoptosis<br />

dominant negative mutant W51C, binds TAK1 resulting in the activation of its kinase activity<br />

(determined by immunocomplex PK assays with MKK6 as the phosphorylation substrate).<br />

Activated TAK1, in turn, activates p38MAPK, which is responsible for caspase-3 cleavage, as<br />

evidenced by inhibition with the TAK1 dominant negative mutant K63Wor the<br />

pharmacologic inhibitor SB203580. In addition, the H11-TAK1 complex binds <strong>and</strong><br />

phosphorylates #-catenin, thereby inhibiting its transcriptional activity <strong>and</strong> resulting in the<br />

inhibition of MITF <strong>and</strong> CDK2, both of which are required for melanoma cell proliferation.<br />

Preliminary data using animal xenograft models indicate that H11 overload inhibits tumor cell<br />

growth <strong>and</strong> triggers apoptosis, also in vivo. The data indicate that modulation of <strong>signaling</strong><br />

pathways by H11 overload is a promising novel paradigm for cancer therapy.<br />

- 388 -


Session X : Cell death in cancer Poster X, 56<br />

Regulation of autophagy by sphingosine kinase 1 <strong>and</strong> its role in cell survival during<br />

nutrient starvation*<br />

Grégory Lavieu†, Francesca Scarlatti§, Giusy Sala§, Thierry Levade‡, Riccardo<br />

Ghidoni§, Joëlle Botti† <strong>and</strong> Patrice Codogno†<br />

†INSERM U504, Institut André Lwoff, 16 avenue Paul-Vaillant-Couturier, 94807<br />

Villejuif Cedex France, §Laboratory of Biochemistry <strong>and</strong> Molecular Biology, San Paolo<br />

Medical School, via A. di Rudinì 8, 20142 Milan Italy, ‡INSERM U466, Institut Louis<br />

Bugnard, Centre Hospitalier Universitaire de Rangueil, BP 84225, 31432 Toulouse<br />

Cedex 4 France..<br />

The sphingolipid ceramide induces macroautophagy (here called autophagy) <strong>and</strong> cell death<br />

with autophagic features in cancer cells (1, 2). Here we show that overexpression of<br />

sphingosine kinase 1 (SK1), an enzyme responible for the production of sphingosine 1phosphate<br />

(S1P), stimulates autophagy by increasing the formation of LC3 positive<br />

autophagosomes <strong>and</strong> the rate of proteolysis sentitive to the autophagy inhibitor 3methyladenine.<br />

Furthermore, autophagy was blocked in the presence of dimethylsphingosine,<br />

an inhibitor of SK activity <strong>and</strong> in cells expressing a catalytically inactive form of SK1. In<br />

contrast to ceramide-induced autophagy, SK1(S1P)-induced autophagy was characterized : 1by<br />

the inhibition of mTOR <strong>signaling</strong> independently of the Akt/PKB <strong>signaling</strong> arm, 2-the lack<br />

of robust accumulation of the autophagy protein Beclin 1. In addition, nutrient starvation<br />

induced both the stimulation of autophagy <strong>and</strong> SK activity. Silencing the expression of the<br />

autophagy protein Atg7 or that of SK1 by siRNA abolished starvation-induced autophagy <strong>and</strong><br />

exacerbated cell death with apoptotic hallmarks. In conclusion, these results show that<br />

SK1(S1P)-induced autophagy protects cells from death with apoptotic features during nutrient<br />

deprivation.<br />

- 389 -


Session X : Cell death in cancer Poster X, 57<br />

Wogonin, a plant flavone, potentiates etoposide-induced apoptosis in cancer cells<br />

Eibai Lee 1 , Riyo Enomoto 1 , Chie Suzuki 1 , Masataka Ohno 1 , Toshinori Ohashi 1 , Azusa<br />

Miyauchi 1 , Eriko Tanimoto 1 , Kaori Maeda 1 , Hiroyuki Hirano 2 , Toshio Yokoi 2 <strong>and</strong><br />

Chiyoko Sugahara 1<br />

1 Department of Pharmacology <strong>and</strong> 2 Department of Pharmaceutical Chemistry, Faculty<br />

of Pharmaceutical Sciences, Kobe Gakuin University, Ikawadani-cho, Nishi-ku, Kobe<br />

651-2180 Japan. E-mail : elee@pharm.kobegakuin.ac.jp<br />

Etoposide, a podophylotoxin anticancer agent, induces apoptotic cell death in normal <strong>and</strong><br />

cancer cells. Etoposide-induced apoptosis plays a role in not only anticancer effect but also<br />

adverse reaction such as myelosuppression. Since we have found that wogonin, a flavone<br />

found in Scutellaria baicalensis, suppresses thymocyte apoptosis induced by various<br />

compounds including etoposide, we examined the effect of this flavone on etoposide-induced<br />

apoptosis in cancer cells. Although wogonin itself hardly affected biochemical <strong>and</strong><br />

morphological features of apoptosis in leukemia cells such as Jurkat <strong>and</strong> HL-60 cells, this<br />

flavone significantly potentiated etoposide-induced apoptosis in these cells. Similarly,<br />

wogonin accelerated etoposide-induced cell death in lung cancer cells. Since wogonin had no<br />

effect on the action of other anticancer agents such as 5-FU <strong>and</strong> cisplatin, this flavone seems<br />

to accelerate only apoptotic cell death induced by etoposide in cancer cells. These results<br />

suggest that the modification of etoposide-induced apoptosis by wogonin may be available to<br />

reduce the adverse reaction of this agent.<br />

- 390 -


Session X : Cell death in cancer Poster X, 58<br />

Jpk, a novel cell death inducer, regulates the expression of Hoxa7 in F9 teratocarcinoma<br />

cells, but not during apoptosis<br />

Eun Young Lee <strong>and</strong> Myoung Hee Kim<br />

Department of Anatomy, Embryology Lab., Brain Korea 21 Project for Medical<br />

Science, Yonsei University College of Medicine, Sodaemoongu Shinchondong 134, Seoul,<br />

120-752, Korea. E-mail: nannaya1210@paran.com ; mhkim1@yumc.yonsei.ac.kr<br />

Increased expression of both Meis1 <strong>and</strong> Hox genes, such as Hoxa7 <strong>and</strong> –a9 has been<br />

characterized in several acute myeloid leukemia. Although the overexpression of Meis1 alone<br />

was shown to induce massive apoptosis, coexpression of Meis1 <strong>and</strong> Hox was reported to<br />

suppress the apoptosis, suggesting that Hox protein might participate in the apoptotic process.<br />

Jpk was isolated as a putative regulatory factor associating with the upstream regulatory<br />

sequence of murine Hoxa7. Since overexpression of Jpk caused apoptosis in bacteria as well<br />

as in eukaryotic cells, we tried to analyze the relationship between Jpk <strong>and</strong> Hoxa7 during<br />

apoptosis after confirming the regulatory effect of Jpk on the expression of Hoxa7 in F9<br />

teratocarcinoma cells. For that purpose, an effector (pEGFP-Jpk) <strong>and</strong> reporter (pGL2-NM307)<br />

plasmid containing a luciferase gene under the 307 bp (NM307) of Hoxa7 upstream<br />

regulatory sequence were constructed. In the presence of Jpk (effector), luciferase activity<br />

was increased <strong>and</strong> this enhancement was decreased by siRNA against Jpk, suggesting that Jpk<br />

is a regulatory factor of Hoxa7. In order to see whether Jpk still regulate the expression of<br />

Hoxa7 during apoptosis, F9 cells were transiently transfected with pcDNA-Jpk, <strong>and</strong> analyzed<br />

the expression of Jpk, Hoxa7, <strong>and</strong> CHOP-10 using RT-PCR. Hoxa7 was not upregulated in<br />

the presence of Jpk whereas CHOP-10 was upregulated, indicating that the regulatory<br />

mechanism of Jpk on the expression of Hoxa7 might be different depending on the cell status;<br />

i. e., apoptotic or proliferative condition.<br />

- 391 -


Session X : Cell death in cancer Poster X, 59<br />

Isoliquiritigenin inhibits osteolytic bone metastasis through reduction of COX-<br />

2/RANKL expression<br />

Sun Kyoung Lee 1,2,3, Kwang Kyun Park 1,2,3, Jung Han Yoon Park 4, Soon Sung Lim<br />

4, Won Yoon Chung 1,2<br />

1Department of Oral Biology, 2Oral Science Research Center, College of Dentistry,<br />

3Brain Korea 21 Project for Medical Science Yonsei University, 120-752, Seoul,<br />

Republic of Korea, 4Division of Life Sciences <strong>and</strong> Silver Biotechnology Research Center,<br />

Hallym University, Chunchon, Republic of Korea, E-mail : l-pluto@hanmail.net<br />

Breast cancer metastasis to the bone occurs frequently, causing numerous complications<br />

including fracture <strong>and</strong> hypercalcemia. The bone destruction is mediated by the osteoclasts<br />

rather than directly by tumor cells. The interaction between tumor cells, tumor-derived<br />

humoral factors <strong>and</strong> bone cells, called to ‘vicious cycle’, is crucial for the initiation <strong>and</strong><br />

promotion of skeletal malignancies. We examined the effect of isoliquiritigenin(ISL) on<br />

vicious cycle of osteolytic bone metastasis by human breast cancer cells. ISL reduced the cell<br />

growth <strong>and</strong> the mRNA expression of IL-1beta, -6 <strong>and</strong> PTHrP in MDA-MB-231 cells. ISL<br />

also inhibited the expression of COX-2 <strong>and</strong> receptor activator of nuclear factor-kappa B<br />

lig<strong>and</strong>(RANKL), but induced the expression of osteoprotegerin(OPG) in human osteoblast<br />

cells, hFOB1.19 treated with conditioned medium of MDA-MB-231 cells. Consequently, the<br />

ratio of RANKL/OPG was decreased by ISL. Moreover, ISL inhibited RANKL-induced<br />

osteoclastogenesis in mouse bone marrow monocyte(BMM) <strong>and</strong> the formation of resorption<br />

pit. Taken together, <strong>our</strong> results suggest that ISL may block osteolytic bone metastasis by<br />

inhibiting RANKL expression through COX-2 expression.<br />

- 392 -


Session X : Cell death in cancer Poster X, 60<br />

Involvement of MAPKs <strong>and</strong> NF-kB in diosgenin-induced megakaryocytic differentiation<br />

<strong>and</strong> subsequent apoptosis in HEL cells<br />

David Y. Léger, Bertr<strong>and</strong> Liagre, Jeanne Cook-Moreau <strong>and</strong> Jean-L. Beneytout<br />

Laboratoire de Biochimie, UPRES EA 1085, Faculté de Pharmacie, Limoges, France. Email:<br />

bertr<strong>and</strong>.liagre@unilim.fr<br />

Recent reports demonstrated that NF-kB activation participated in megakaryocytic<br />

differentiation. A growing number of studies demonstrated the key role of the MAPK<br />

pathway during megakaryocytic differentiation. After differentiation, the fate of mature<br />

megakaryocytes is to produce platelets. Platelet shedding results from cytoplasmic<br />

fragmentation, which leads to the formation of denuded megakaryocytes constituted of the<br />

megakaryocyte nucleus, its envelope, <strong>and</strong> a ring of cytoplasm. These senescent<br />

megakaryocytes have been identified as apoptotic cells. Furthermore, platelet formation has<br />

recently been shown to require precise caspase activation. These data emphasized the<br />

involvement of apoptotic-related phenomena in thrombopoiesis.<br />

The treatment of HEL cells with 10 µM diosgenin caused an initial activation of ERK1/2<br />

within 5 min (5-fold over untreated cells), which was sustained up to 12h. Furthermore,<br />

diosgenin induced a rapid <strong>and</strong> sustained de-activation of p38. With diosgenin stimulation, we<br />

found an early slight activation of NF-!B at 24h. Then, diosgenin progressivly inhibited NF-<br />

!B translocation after 48-96h of treatment leading to a complete inhibition at 192h of<br />

treatment. During diosgenin treatment, two distinct bursts of caspase-3 activation were<br />

observed. At 48h stimulation, we observed a rapid <strong>and</strong> strong increase in active caspase-3.<br />

Then, active caspase-3 levels rapidly decreased at 96h but remained higher than controls.<br />

Afterwards, caspase-3 activity increased again <strong>and</strong> once more reached high levels by the end<br />

of the treatment. In addition, the intensity of the PARP cleaved 85 Kd fragment followed the<br />

kinetics of caspase-3 activation. Diosgenin treatment also led to the fragmentation of<br />

differentiated cells. Cellular fragmentation started at 96h post-stimulation <strong>and</strong> at the end of<br />

the treatment (at 192h), most of the cells were fragmenting or already fragmented.<br />

In conclusion, diosgenin induced the megakaryocytic differentiation of HEL cells through a<br />

combined activation of the ERK <strong>signaling</strong> pathway <strong>and</strong> inhibition of the p38 MAPK pathway.<br />

Afterwards, differentiated cells showed a marked inhibition of NF-kB nuclear translocation<br />

<strong>and</strong> an activation of caspase-3 together with PARP cleavage.<br />

- 393 -


Session X : Cell death in cancer Poster X, 61<br />

HGF/SF regulates expression of apoptotic genes in human mammary epithelial cells<br />

Catherine LEROY, Julien DEHEUNINCK, Sylvie REVENEAU, Bénédicte FOVEAU,<br />

Zongling JI, David TULASNE, Céline Villenet*, Sabine Quief*, <strong>and</strong> Jean-Pierre<br />

Kerckaert* <strong>and</strong> Véronique FAFEUR.<br />

CNRS UMR 8117, Institut de Biologie de Lille, Institut Pasteur de Lille, B.P.447, 59021<br />

Lille, FRANCE. E-mail : veronique.fafeur@ibl.fr<br />

* Plate-forme Biopuces, Université de Lille 2, Faculté de Médecine, Place de Verdun,<br />

59045, Lille, FRANCE<br />

Hepatocyte growth factor/scatter factor (HGF/SF) induces scattering, morphogenesis <strong>and</strong><br />

survival of epithelial cells through the activation of the MET tyrosine kinase receptor.<br />

HGF/SF <strong>and</strong> MET are involved in normal <strong>and</strong> tumoral development of many tissues <strong>and</strong><br />

organs, including the mammary gl<strong>and</strong>. At present, little is known of the target genes of<br />

HGF/SF implicated in mediating its biological responses.<br />

We searched for HGF/SF target genes using the human epithelial mammary cell line (MCF-<br />

10A), which is sensitive to its scattering <strong>and</strong> survival effect. We used a DNA microarray<br />

spotted with 60-mer oligonucleotide sets from Sigma-Genosys that include 1920 different<br />

genes. MCF-10A cells were treated or not with HGF/SF for 2 h. Total RNA was then<br />

extracted <strong>and</strong> purified, followed by reverse transcription to cDNA, with concomitant<br />

incorporation of fluorescent dCTP (Cy3 or Cy5). The probes were mixed <strong>and</strong> hybridized onto<br />

the microarray, <strong>and</strong> the fluorescent signals were detected using a GMS confocal scanner<br />

(Affymetrix). The ratios of Cy5/Cy3 were analyzed using Jaguar Software (Screensaver). The<br />

expression of 38 genes was found to be modified by HGF/SF. Among them, only three genes<br />

were clearly classified in apoptosis, with A20 being up-regulated by HGF/SF <strong>and</strong> DAXX <strong>and</strong><br />

SMAC being down-regulated by HGF/SF. Changes in expression of A20, DAXX <strong>and</strong> SMAC<br />

were confirmed by real time quantitative PCR using a Light Cycler (Roche). According to<br />

published data, A20 is anti-apoptotic, SMAC is pro-apoptotic, while a pro- or anti- apoptotic<br />

role of DAXX is still controversial. The fact that HGF/SF up-regulates an anti-apoptotic gene<br />

(A20) <strong>and</strong> down-regulates a pro-apoptotic gene (SMAC) is in agreement with its survival<br />

effect in MCF10A cells. This study identified novel target genes of HGF/SF that can<br />

contribute to its anti-apoptotic effect.<br />

- 394 -


Session X : Cell death in cancer Poster X, 62<br />

The nucleoside analogue cidofovir suppresses lung metastasis <strong>and</strong> induces apoptosis in<br />

FGF2-overexpressing endothelial cells.<br />

S<strong>and</strong>ra Liekens*, Sofie Gijsbers*, Erik De Clercq*, Erik Verbeken§, <strong>and</strong> Sigrid Hatse*<br />

*Rega Institute for Medical Research, Minderbroedersstraat 10, B-3000 Leuven,<br />

Belgium <strong>and</strong> §Division of Histopathology, K.U.Leuven. E-mail:<br />

S<strong>and</strong>ra.liekens@rega.kuleuven.be<br />

Cidofovir [(S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine, (S)-HPMPC] is an<br />

antiviral drug that is clinically used for the treatment of cytomegalovirus retinitis in AIDS<br />

patients (1). Cidofovir also possesses potent activity against human papillomavirus-induced<br />

tumors in animal models <strong>and</strong> patients (1). We have recently shown that cidofovir inhibits the<br />

development of vascular tumors induced by basic fibroblast growth factor (FGF2)overexpressing<br />

endothelial (3F2T) cells in mice (2). Here, we demonstrate that the cytotoxic<br />

activity of cidofovir in 3F2T cells may result from the specific induction of apoptosis. Cell<br />

cycle analysis revealed that cidofovir induces accumulation of cells in the S phase <strong>and</strong>, upon<br />

prolonged treatment, a significant increase in sub-G1 cells, exhibiting a sub-diploid DNA<br />

content. Moreover, annexin V binding, an early event in apoptosis induction, was increased in<br />

cidofovir-treated 3F2T cells. Cidofovir also caused nuclear fragmentation <strong>and</strong> the activation<br />

of caspase-3-like proteases in 3F2T cells, as evidenced by the cleavage of poly(ADPribose)polymerase.<br />

In addition, cidofovir treatment resulted in a pronounced up-regulation of<br />

p53 protein. Protein kinase B/Akt is recognized as a principal mediator of survival signals that<br />

protect cells from undergoing apoptosis. Cidofovir did not suppress the phosphorylation of<br />

Akt nor its downstream regulator Bad, indicating that the Akt pathway is not affected by<br />

cidofovir treatment of 3F2T cells. However, cidofovir inhibited the expression of FGF2 <strong>and</strong><br />

FGF2 <strong>signaling</strong> through Erk42/44, as shown by Western blot analysis. In vivo, cidofovir<br />

markedly suppressed the development of experimental lung metastasis, induced by<br />

inoculation of 3F2T cells in the tail vein of SCID mice. Our results indicate that cidofovir<br />

may inhibit the growth of 3F2T cells via inhibition of FGF2 expression <strong>and</strong> <strong>signaling</strong>, <strong>and</strong> via<br />

the induction of apoptosis through the caspase-3 pathway.<br />

1. De Clercq <strong>and</strong> Holy (2005) Nat Rev Drug Discov. 4: 928-940.<br />

2. Liekens et al. (2001) Cancer Res. 61: 5057-5064.<br />

- 395 -


Session X : Cell death in cancer Poster X, 63<br />

DMNQ S64 induces apoptosis via caspase activation <strong>and</strong> cyclooxygenase-2 inhibition in<br />

human non-small cell A549 lung cancer cells<br />

Eu-Soo Lim, Yun-Hee Rhee, Eun-Ok Lee, Eun-Young Lee, Sung-Hoon Kim<br />

Department of Oncology, Graduate School of East-West Medical Science, KyungHee<br />

University, Yongin 449-701, Republic of Korea. E-mail:sungkim7@khu.ac.kr<br />

Shikonin has been shown to induce apoptosis <strong>and</strong> inhibit angiogenesis in vivo <strong>and</strong> in vitro.<br />

However, it still has drawbacks of solubility <strong>and</strong> toxicity. Thus, in the present study, the<br />

underlying apoptotic mechanism of 6-ppim (1-propoxyiminoalkyl)-DMNQ-S64 (DMNQ<br />

S64), a shikonin derivative, was examined. DMNQ S64 exerted cytotoxicity against human<br />

A549 lung carcinoma cells with IC50 of 30 µM. Apoptotic bodies were observed in DMNQ<br />

S64 treated A549 cells. DMNQ S64 also increased sub G1 DNA peaks in a concentration<br />

dependent manner by flow cytometric analysis. Western blotting has revealed that DMNQ<br />

S64 effectively activates the expression of caspase 8, 9 <strong>and</strong> 3, cleaves poly(ADP-ribose)<br />

polymerase <strong>and</strong> increases the ratio of Bax/Bcl-2. Furthermore, mitochondrial membrane<br />

potential was reduced in a concentration dependent manner by DMNQ S64. It significantly<br />

inhibited the level of prostagl<strong>and</strong>in E2 by enzyme linked immunosorbent assay <strong>and</strong><br />

downregulated the expression of cyclooxygenase-2 (COX-2) in a concentration dependent<br />

manner by Western blotting. Taken together, DMNQ S64 may exhibit cytotoxicity against<br />

A549 cells via caspase activation <strong>and</strong> COX-2 inhibition.<br />

- 396 -


Session X : Cell death in cancer Poster X, 64<br />

The effect on cell death in drug <strong>and</strong> CD95-mediated apoptosis by the apoptosis enhancer<br />

Daxx in renal cell carcinoma<br />

C Mahotka, N Wethkamp, P Reinecke, <strong>and</strong> HE Gabbert<br />

Institute of Pathology, Heinrich Heine-University, Duesseldorf, Germany; e-mail:<br />

mahotka@med.uni-duesseldorf.de<br />

Introduction: Deregulation of apoptosis is involved in the development of cancer. Expression<br />

of Daxx is implicated in apoptosis but whether its function is pro- or anti-apoptotic is still<br />

discussed controversial. Daxx is involved in the regulation of p53 dependent transcription <strong>and</strong><br />

associates with the promyelocytic leukemia protein (PML) in speckled subnuclear structures<br />

so called PML oncogenic domains (PODs). Overexpression-based experiments in renal cell<br />

carcinoma (RCC) were used to explore the impact of Daxx on the CD95-dependent apoptosis<br />

<strong>and</strong> cell death induced by different anticancer agents such as Etoposide, Topotecan, Taxol <strong>and</strong><br />

Doxorubicine. Results: 1. Constitutive overexpression of Daxx in the CD95-lig<strong>and</strong> sensitive<br />

human RCC cell line clearCa-6 (clear-Ca-6/GFP-Daxx) has no influence on cell growth. 2.<br />

Treatment of clearCa-6/GFP-Daxx with the CD95-receptor agonistic antibody CH11 leads to<br />

no further enhancement of the CD95-dependent apoptosis compared to control cell line clear-<br />

Ca-6/GFP as analyzed by MTT Assay <strong>and</strong> Caspase-8 <strong>and</strong> PARP Western Blot, respectively.<br />

3. Daxx overexpression in the CD95-lig<strong>and</strong> resistent RCC cell line clearCa-2 (clearCa-2/GFP-<br />

Daxx) also causes no sensitization to the CD95-dependent apoptosis. 4. Although Daxx is<br />

known to trigger CD95-mediated apoptosis via the activation of JNK by a Caspase-8<br />

independent mechanism, even under Caspase-8 inhibition no Daxx-mediated modulation of<br />

the CD95-mediated apoptosis was detectable <strong>and</strong> moreover, no differences in CD95-mediated<br />

JNK aktivation were obvious as shown by in vitro kinase assays. 5. Treatment of both GFP-<br />

Daxx transduced cell lines with various anticancer drugs points to a protective role of Daxx<br />

during Taxol-dependent apoptosis of clearCa-2/GFP-Daxx, which is probably mediated by an<br />

enhanced activation of the p38/JNK pathway as shown by in vitro kinase assays.<br />

Conclusions: Our study suggests that the CD95-mediated apoptosis in renal cell carcinoma is<br />

not effected by the overexpression of the apoptosis enhancer Daxx. In contrast, the data<br />

indicate a rather anti-apoptotic than pro-apoptotic role for Daxx in human RCC as shown by<br />

the Daxx overexpression-mediated protective effect during Taxol-dependent apoptosis in the<br />

RCC cell line clearCa-2.<br />

- 397 -


Session X : Cell death in cancer Poster X, 65<br />

Increased expression of high mobility group box 1 (HMGB1) protein is associated with<br />

an elevated level of iNOS in lymphocytic thyroiditis <strong>and</strong> papillary thyroid cancer.<br />

Stefania Mardente, Gianluca Maiani, Emanuela Mari, Aless<strong>and</strong>ra Zicari, Massimo<br />

Realacci, Stefania Natalizi, Fabrizio Consorti*, Carlo Della Rocca <strong>and</strong> Alfredo<br />

Antonaci*.<br />

Department of Experimental Medicine <strong>and</strong> Pathology <strong>and</strong> * Department of Surgical<br />

Sciences <strong>and</strong> Applied Technologies, University “La Sapienza”, Viale Regina Elena 324,<br />

Rome Italy: email: stefania.mardente@uniroma1.it<br />

Some recent works have shown that autoimmune thyroiditis, although considered a benign<br />

condition often harb<strong>our</strong>s a genetic rearrangement that is highly specific for papillary<br />

carcinoma. Submicroscopic foci of papillary carcinoma exist in autoimmune thyroiditis<br />

although its behavi<strong>our</strong> is still benign. We have recently demonstrated that peripheral<br />

lymphocytes (type Tc1 <strong>and</strong> Tc2) infiltrate thyroids in autoimmune thyroiditis <strong>and</strong> in most<br />

papillary carcinomas. Direct cytotoxicity, secretion of cytokines, presence of intrathyroidal B<br />

secreting autoantibodies, would altoghether work as a defect in suppressing the immune<br />

process. The local inflammation would lead to secretion of NO that could exacerbate the<br />

autoimmune response <strong>and</strong> hypothyroidism. It has been demonstrated that high concentrations<br />

of NO are genotoxic in the sense of promoting mutations that could lead to cancer. Local<br />

hypoxia <strong>and</strong> high concentrations of NO may induce apoptosis of thyreocytes in chronic<br />

inflammation <strong>and</strong> cancer but it may also lead to necrotic death. HMGB1 is a non histone<br />

chromosomal protein implicated in a variety of processes including transcription,<br />

differentiation <strong>and</strong> development. An increased expression of this protein has been reported for<br />

several tumor types. Its overexpression inhibits apoptosis. According to <strong>our</strong> western blot<br />

analysis of primary cultures of thyreocytes from patients with thyroiditis <strong>and</strong> papillary cancer<br />

<strong>and</strong> immunohistochemistry of iNOS, we identified a molecular pathway triggered by HMGB1<br />

that could inhibit apoptosis of thyreocytes in a microenvironment rich of NO <strong>and</strong> other<br />

proinflammatory cytokines.<br />

- 398 -


Session X : Cell death in cancer Poster X, 66<br />

Arsenic trioxide induces a p53-independent cell death in Ewing sarcoma cells.<br />

Julie Mathieu, Michel Lanotte <strong>and</strong> Françoise Besançon<br />

INSERM Unité 685, Hopital St-Louis, 1 avenue Claude Vellefaux, 75010 Paris, France.<br />

E-mail : julie.mathieu@stlouis.inserm.fr; francoise.besancon@stlouis.inserm.fr<br />

Ewing sarcoma (ES), a highly malignant pediatric tumor, is consistently associated with<br />

translocations that fuse the EWS gene with a member of the ETS family gene, most<br />

commonly FLI-1. Despite significant advances with multi-agent chemotherapy, surgery <strong>and</strong><br />

radiotherapy, about 40% of ES patients still die from the disease. It is therefore necessary to<br />

explore novel agents for possible treatment of this tumor. Here we investigated the sensitivity<br />

of ES cells to arsenic trioxide (As2O3), a compound known to induce differentiation <strong>and</strong><br />

apoptosis of other types of malignant cells. We report that low doses of As2O3 (1-4<br />

micromolar) uniformly inhibited growth of six ES-derived cell lines irrespective of their p53<br />

status. As2O3 resulted in caspase activation, loss of mitochondrial potential <strong>and</strong> an apoptotic<br />

phenotype which was inhibited by the broad-spectrum caspase inhibitor ZVADfmk. These<br />

effects correlated with prolonged JNK activation, which is a signal for apoptosis in ES cells.<br />

As2O3 also decreased basal <strong>and</strong> cytokine-induced NF-kappa B activity, which was previously<br />

shown to protect ES cells from apoptosis induced by various stimuli. Together, <strong>our</strong> results<br />

demonstrate that clinically tolerable concentrations of As2O3 trigger p53-independent<br />

apoptosis of ES cells. They further suggest that inclusion of this compound in future trials of<br />

novel treatment strategies for Ewing sarcoma may be beneficial.<br />

- 399 -


Session X : Cell death in cancer Poster X, 67<br />

Functional Analysis of the Anti-Apoptotic Livin Protein<br />

Christina Mensger, Irena Crnkovic-Mertens, Claire Cullmann, Karin Butz <strong>and</strong> Felix<br />

Hoppe-Seyler<br />

Infection <strong>and</strong> Cancer, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 242,<br />

D-69120 Heidelberg, Germany. E-mail: c.mensger@dkfz.de<br />

Cancer cells are typically characterized by increased resistance to apoptosis, which enables<br />

their survival under abnormal growth stimulation. Moreover, deficiency in apoptosis is<br />

considered to be a major cause for therapeutic resistance of tum<strong>our</strong>s in the clinic, since many<br />

chemo- <strong>and</strong> radiotherapeutic agents act through the induction of apoptosis. The functional<br />

inhibition of specific anti-apoptotic factors should therefore provide a rational basis for the<br />

development of novel therapeutic strategies.<br />

Livin is a relatively new member of the "Inhibitors of Apoptosis Protein" (IAP) family.<br />

Initially associated with malignant melanoma, it recently has been found that Livin is also<br />

expressed in many additional cancers, such as lung cancer. Notably, livin gene expression is<br />

typically detectable in the tumor cells but not, or to substantially lower levels, in the<br />

corresponding normal tissue.<br />

We found that the targeted inhibition of Livin by RNA interference (RNAi) led to a proapoptotic<br />

sensitization towards different pro-apoptotic stimuli, which was specific for Livinexpressing<br />

tumor cells. Moreover, long-term inhibition of Livin expression in clonogenic<br />

survival assays led to the elimination of Livin-expressing tumor cells, even in the absence of<br />

additional pro-apoptotic stimuli. Isoform-specific RNAi showed that the biological<br />

significance of the two known Livin isoforms, Livin " <strong>and</strong> Livin #, can strongly differ,<br />

possibly in a cell-dependent manner. Ongoing work concentrates on the elucidation of the<br />

critical intracellular pathways <strong>and</strong> natural interaction partners which mediate the antiapoptotic<br />

activity of Livin.<br />

Based on its cancer-associated expression pattern, Livin may serve as a novel diagnostic <strong>and</strong><br />

prognostic tumor marker. In addition, the targeted inhibition of Livin could represent a new<br />

antitumor strategy.<br />

- 400 -


Session X : Cell death in cancer Poster X, 68<br />

EXTRACELLULAR SURVIVIN UPREGULATES ADHESION MOLECULES ON<br />

THE SURFACE OF LEUKOCYTES CHANGING THEIR REACTIVITY PATTERN<br />

Simona Mera, Mattias Magnusson, Andrej Tarkowski, <strong>and</strong> Maria Bokarewa<br />

Department of Rheumatology <strong>and</strong> <strong>Inflammation</strong> Research, Sahlgrenska University<br />

Hospital, Göteborg, Sweden<br />

Background. Rheumatoid arthritis (RA) is an autoimmune disease with joints as a principal<br />

target of inflammation. We have recently shown that the extracellular presence of antiapoptotic<br />

protein survivin was associated with unfav<strong>our</strong>able, i.e. destructive c<strong>our</strong>se of RA.<br />

Here we addressed the effects of extracellular survivin on peripheral leukocytes.<br />

Methods. Human peripheral blood leukocytes were cultured with <strong>and</strong> without recombinant<br />

survivin <strong>and</strong> assessed for the surface binding of survivin <strong>and</strong> the expression of adhesion<br />

molecules. The intracellular expression of survivin in non-activated human leukocytes was<br />

determined. Finally, the expression of adhesion molecules on leukocytes as a function of<br />

circulating survivin was analysed in blood of 24 patients with RA <strong>and</strong> compared to healthy<br />

individuals.<br />

Results. We found that the anti-apoptotic protein survivin expresses immuno-modulatory<br />

properties when released extracellularly. It binds to a 47% of neutrophils inducing the<br />

activation of #2-integrins <strong>and</strong> their lig<strong>and</strong>, ICAM-1. Survivin also binds to lymphocytes<br />

inducing the expression of L-selectin. Survivin-induced expression of #2-integrins could be<br />

abolished by the inhibitors of NFkB (parthenolide), PI3-kinase (LY294002), but not by MAPkinase<br />

inhibitors (PD98059, SB203580). Clinical relevance of <strong>our</strong> findings is proved by the in<br />

vivo association of extracellular survivin with an increased expression of CD11c on blood<br />

monocytes <strong>and</strong> neutrophils in RA patients.<br />

Conclusion. The results of <strong>our</strong> study demonstrate that extracellular survivin affects the<br />

function of leukocytes having possible impact on inflammatory responses during arthritis.<br />

- 401 -


Session X : Cell death in cancer Poster X, 69<br />

A new method to assess drug sensitivity on breast tumor acute slices preparation<br />

Pedro Mestres1, Andrea Morguet1, Werner Schmidt2, Axel Kob3, Ralf Ehret3<br />

Department of Anatomy <strong>and</strong> Cell Biology1, Clinic for Gynecology2, University of<br />

Saarl<strong>and</strong>, D-66421 Homburg, Bionas Inc. 3, D-18119 Rostock. Germany<br />

The method described is based on: 1) preparation of tissue slices <strong>and</strong> 2) use of silicon chips<br />

equipped with electrochemical sensors (multisensor array). The slices (200-300 µm thick) are<br />

prepared after surgery <strong>and</strong> incubated in a medium for recovery after slicing. The advantage,<br />

compared to other preparations, is that the original three-dimensional structure is retained.<br />

Multisensor arrays measure: 1) pericellular acidification (anaerobic metabolism) <strong>and</strong> 2)<br />

oxygen consumption (respiration). The innovative aspect is that such measurements can be<br />

made online, as opposed to the large battery of endpoint tests on cell vitality <strong>and</strong> proliferation.<br />

Electron microscopy of slices serves to determine cell density, structure <strong>and</strong> induction of<br />

apoptosis/necrosis.<br />

Over 200 breast tumors were used. Inhibition of metabolic activities was performed with<br />

sodium fluoride, which dose-dependently reduces glycolysis, <strong>and</strong> potassium cyanide which<br />

inhibits respiration. In other experiments cytostatics were applied, providing results obtained<br />

with Taxol, an anti-cytoskeleton agent. In addition, the application of beta-adrenergic receptor<br />

agonists, creates a situation in which metabolic patterns associated with lig<strong>and</strong> receptor<br />

interaction can be observed all the time. In conclusion, the methodology presented here, is<br />

able to provide information on drug sensitivity of a tumor, of assistance in designing<br />

individualized therapy <strong>and</strong> for drug-screening. (supported by BMBF to PM)<br />

- 402 -


Session X : Cell death in cancer Poster X, 70<br />

Cytotoxicity of TRAIL/anticancer agent combinations in human primary cells<br />

Olivier Meurette1, Anne Fontaine1, Amélie Rebillard1, Thierry Lamy2, Dominique<br />

Lagadic-Gossmann1 <strong>and</strong> Marie-Thérèse Dimanche-Boitrel1<br />

1 INSERM Unit 620, University of Rennes 1, Faculty of Pharmacy, 2 Av du Pr. Leon<br />

Bernard 35043 Rennes cedex France; 2 Department of Hematology, Pontchaillou<br />

Hospital, Rennes, France.<br />

TRAIL (TNF-"-Related Apoptosis Inducing Lig<strong>and</strong>) is a potential anticancer agent that<br />

induces apoptosis by binding to its death receptors TRAIL-R1 <strong>and</strong>/or -R2 in cancer cells but<br />

not in most normal cells. TRAIL also possesses two decoy receptors (TRAIL-R3/-R4). Some<br />

cancer cells are resistant to TRAIL-induced apoptosis. Combining TRAIL with a wide range<br />

of anticancer agents can restore cancer cell sensitivity to cell death, <strong>and</strong> combination of<br />

TRAIL with anticancer drugs is now tested in clinical trials. At present, the cytotoxic effect of<br />

such combinations in normal human primary cells is not well known.<br />

We studied here the sensitivity of several human primary cells: hepatocytes, lymphocytes <strong>and</strong><br />

neutrophils to combination of TRAIL with cisplatin or 5-fluor<strong>our</strong>acil in relation with TRAIL<br />

receptors membrane expression. We show that human primary hepatocytes express<br />

exclusively TRAIL-R4 decoy receptor on the cell membrane. In contrast to Fas lig<strong>and</strong>,<br />

TRAIL is not cytotoxic towards human primary hepatocytes. However, these normal cells<br />

become only sensitive to TRAIL/cisplatin but not to TRAIL/5-fluor<strong>our</strong>acil. Cisplatin or 5fluor<strong>our</strong>acil<br />

treatment has no effect on TRAIL receptors membrane expression in human<br />

primary hepatocytes. Freshly isolated lymphocytes express solely TRAIL-R4 at the cell<br />

membrane <strong>and</strong> are resistant to TRAIL. However, as human hepatocytes, lymphocytes are only<br />

sensitive to TRAIL/cisplatin <strong>and</strong> resistant to TRAIL/5-fluor<strong>our</strong>acil. Interestingly, these both<br />

combinations are cytotoxic towards PHA+IL-2 activated lymphocytes which express a little<br />

amount of TRAIL-R1 <strong>and</strong> TRAIL-R2 on the cell membrane. Finally, freshly isolated<br />

neutrophils express exclusively TRAIL-R3 on the plasma membrane <strong>and</strong> are resistant to<br />

TRAIL <strong>and</strong> to TRAIL/anticancer agent combinations.<br />

Altogether, these data demonstrate that human primary cells express exclusively TRAIL<br />

decoy receptors (TRAIL-R3 or TRAIL-R4) <strong>and</strong> are resistant to TRAIL. However, the<br />

cytotoxicity of certain TRAIL/anticancer drugs combinations towards human primary cells<br />

should be taken into account for the development of TRAIL-based anticancer therapy.<br />

- 403 -


Session X : Cell death in cancer Poster X, 71<br />

TRAIL induces a RIP (receptor interacting protein) dependent necrosis-like cell death<br />

at acidic extracellular pH (pH=6.5).<br />

Olivier Meurette1, Laurence Huc1, Amélie Rebillard1, Gwenaelle Le Moigne1, Olivier<br />

Micheau2, Delphine Merino2, Dominique Lagadic-Gossmann1 <strong>and</strong> Marie-Thérèse<br />

Dimanche-Boitrel1.<br />

1 INSERM Unit 620, University of Rennes 1, Faculty of Pharmacy 2 Av, du Pr. Leon<br />

Bernard 35043 Rennes cedex, France. 2 INSERM U517, Faculty of Medicine, University<br />

of Burgundy, 7 boulevard Jeanne d’Arc, 21033 Dijon Cedex, France.<br />

TRAIL (TNF-alpha-Related Apoptosis Inducing Lig<strong>and</strong>) is a potential anticancer agent that<br />

induces apoptosis in cancer cells but not in most normal cells. TRAIL is known to induce<br />

apoptosis via the extrinsic death pathway. In certain conditions a death signal amplification<br />

via the intrinsic death pathway is necessary. TRAIL has also been shown to induce necrosis<br />

independently of caspase activation. At present, necrosis-like programmed cell death<br />

signalisation is poorly understood. The Death Domain containing serine/threonine kinase RIP<br />

(Receptor Interacting Protein) has been involved both in necrosis-like cell death <strong>and</strong> in NFkB<br />

activation after death receptor ligation.<br />

We have recently shown that under acidic extracellular pH (6.5), TRAIL induced a cell death<br />

sharing some apoptotic <strong>and</strong> necrotic characteristics in human HT29 colon carcinoma <strong>and</strong><br />

HepG2 hepatocarcinoma cell lines. We demonstrate here, that in spite of a necrosis-like cell<br />

death phenotype, caspases are activated <strong>and</strong> are necessary in this cell death process. By using<br />

a geldanamycin pretreatment or a small interference RNA approach, we show that RIP is<br />

necessary for TRAIL-induced necrosis-like cell death in HT29 cells at acidic extracellular pH<br />

(6.5). Furthermore, the expression of RIP protein is increased in HT29 cells after TRAIL<br />

treatment at both physiological <strong>and</strong> acidic extracellular pH. Whereas RIP is cleaved after<br />

TRAIL treatment at physiological extracellular pH (7.4), it is not cleaved at acidic<br />

extracellular pH (6.5) despite high level of caspase activation. Finally, at acidic extracellular<br />

pH (6.5), TRAIL strongly activates NF-kB in HT29 cells, whose activation is dispensable for<br />

necrosis-like cell death induction.<br />

In conclusion, these data demonstrate that under acidic extracellular pH conditions TRAIL<br />

induces in HT29 cells a necrosis-like cell death which depends on caspase activation <strong>and</strong> on<br />

RIP expression.<br />

- 404 -


Session X : Cell death in cancer Poster X, 72<br />

Oxidative stress response in telomerized human fibroblasts from a centenarian<br />

Chiara Mondello1, Maria Grazia Bottone1,2, Sakon Noriki3, Cristiana Soldani2, Carlo<br />

Pellicciari2, A. Ivana Scovassi1<br />

1 Istituto di Genetica Molecolare del CNR, Via Abbiategrasso 207, 27100 Pavia, Italy, Email:<br />

mondello@igm.cnr.it, scovassi@igm.cnr.it; 2 Dipartimento di Biologia Animale,<br />

Piazza Botta 10, 27100, Pavia, Italy. E-mail: bottone@unipv.it, soldani@unipv.it,<br />

pelli@unipv.it; 3 Department of Oncological Pathology, Faculty of Medicine, Matsuoka,<br />

Yoshida-Gun, 9l0-ll93, Fukui, Japan. E-mail: noriki@fmsrsa.fukui-med.ac.jp<br />

It has been reported that cells with ectopic expression of telomerase (so-called telomerized)<br />

are more resistant to apoptotic cell death than their normal counterpart. However,<br />

controversial results have been obtained as to the response of telomerized cells to the<br />

oxidative stress. In the present research, we investigated the effects of the treatment with<br />

either tert-butylhydroperoxide (tBOOH) or 2-deoxy-D-ribose (D-ribose) on human fibroblasts<br />

from a centenarian individual (cen3) <strong>and</strong> on their counterpart with reactivated telomerase<br />

(cen3tel). Our results suggest that the effects of these drugs are modulated by telomerase<br />

reactivation. In fact, after drug treatment the cell number of both cell lines was lower than in<br />

controls, indicating that both agents impaired cell proliferation; remarkably, proliferation was<br />

always inhibited at a larger extent in cen3tel than in cen3 cells. Different parameters of<br />

apoptosis were also studied in situ (i.e., chromatin condensation, phosphatidylserine<br />

externalization, <strong>and</strong> DNA fragmentation) <strong>and</strong> showed that the percentage of apoptotic cells<br />

was lower in telomerized cell cultures, although apoptosis could not completely account for<br />

the observed cell loss in treated cultures. The evidence of active phagocytosis of apoptotic<br />

cells occurring in <strong>our</strong> fibroblast cultures provides an explanation for these contradictory<br />

results.<br />

- 405 -


Session X : Cell death in cancer Poster X, 73<br />

Met acts on Mdm2 through mTOR to signal cell survival in vivo<br />

Anice Moumen1, Almudena Porras2, Arm<strong>and</strong> Tasmadjian1, Rosanna Dono1 <strong>and</strong> Flavio<br />

Maina1<br />

1IBDML, Campus de Luminy-Case 907, 13288 Marseille Cedex 09, France - 2Dpto.<br />

Bioquimica y Biologia Molecular II, Facultad de Farmacia, Universidad Complutense,<br />

Ciudad Universitaria, 28040 Madrid, Spain.<br />

E-mail: maina@ibdm.univ-mrs.fr<br />

Coordination of cell death <strong>and</strong> survival is crucial during embryogenesis <strong>and</strong> adulthood <strong>and</strong><br />

alterations to this balance can result in degeneration or cancer. Growth factor receptors such<br />

as Met can activate phosphatidyl-inositol-3’ kinase (PI3K), a major intracellular mediator of<br />

growth <strong>and</strong> survival. PI3K can antagonise p53-triggered cell death but the underlying<br />

mechanisms are not fully understood. Using genetic <strong>and</strong> pharmacological approaches, we<br />

demonstrated that PI3K acts through mTOR to regulate p53 activity both in vitro <strong>and</strong> in vivo.<br />

mTOR inhibits p53 by promoting translation of Mdm2, the negative regulator of p53.<br />

Increased Mdm2 protein levels require the mTOR effector p70s6k/S6 ribosomal protein.<br />

Unexpectedly, although it is required for Mdm2 nuclear translocation, Akt is dispensable for<br />

Met-triggered mTOR activation <strong>and</strong> Mdm2 up-regulation. Inhibition of mTOR is sufficient to<br />

block cell survival induced by HGF/Met in vitro, to down-regulate Mdm2 protein levels <strong>and</strong><br />

to induce p53-dependent apoptosis in vivo. Our studies identify a novel mechanism for cell<br />

survival, involving translational regulation of Mdm2 by mTOR, thus reinforcing mTOR as a<br />

potential drug target in cancer.<br />

- 406 -


Session X : Cell death in cancer Poster X, 74<br />

HGF supports embryonic hepatocyte survival by suppressing apoptotic pathways<br />

involving p53 <strong>and</strong> TNFalpha/FasL<br />

Anice Moumen1, Aless<strong>and</strong>ro Ieraci1 <strong>and</strong> Flavio Maina1<br />

1IBDML, Campus de Luminy-Case 907, 13288 Marseille Cedex 09, France - 2Dpto.<br />

Bioquimica y Biologia Molecular II, Facultad de Farmacia, Universidad Complutense,<br />

Ciudad Universitaria, 28040 Madrid, Spain.<br />

E-mail: moumen@ibdm.univ-mrs.fr<br />

Survival of embryonic hepatocytes is controlled by a number of anti- <strong>and</strong> pro-apoptotic<br />

signals. To date, the only lig<strong>and</strong>/receptor couple known to induce hepatocyte survival in vivo<br />

is HGF <strong>and</strong> its cognate receptor tyrosine kinase Met. However, their downstream effectors<br />

involved in this process remain to be elucidated. We previously showed that met specificityswitch<br />

mutants are not able to mediate hepatocyte survival, thus resulting in massive<br />

apoptosis in developing livers as observed in met loss-of-function mutants. Taking advantage<br />

of the incomplete signalling of these mutant receptors, we asked which combination of<br />

signalling effectors is sufficient for proper hepatocyte survival. We found that Met makes use<br />

of distinct networks involving Flip <strong>and</strong> Mdm2 to block the pro-apoptotic signals TNF/FasL<br />

<strong>and</strong> p53, respectively. In contrast, modulators of cell survival such as CREB, Gab1 <strong>and</strong> Jun,<br />

are not sufficient. Thus, <strong>our</strong> results clearly show that during liver development, Met controls<br />

hepatocyte survival by modulating the cross-talk of pro- <strong>and</strong> anti-apoptotic signals.<br />

Furthermore, <strong>our</strong> use of defined mouse signalling mutants resolves some of the complexity<br />

underlying the mechanisms that regulate the survival of hepatocytes <strong>and</strong> their susceptibility to<br />

apoptotic signals.<br />

- 407 -


Session X : Cell death in cancer Poster X, 75<br />

The essential role of the mitochondria-dependent death <strong>signaling</strong> cascade in<br />

chemotherapy-induced potentiation of Apo2L/TRAIL cytotoxicity in cultured thoracic<br />

cancer cells: Amplified caspase 8 is indispensable for combination-mediated massive<br />

cell death.<br />

Dao M. Nguyen, Wen-Shuz Yeow, Rishindra M. Reddy, M. Firdos Ziauddin, Wilson<br />

Tsai, David S. Schrump.<br />

Section of Thoracic Oncology, Surgery Branch, Center for Cancer Research, National<br />

Cancer Institute, National Institutes of Health, Bethesda, MD, USA. Email address:<br />

dao_nguyen@nih.gov<br />

Chemotherapeutic drugs, regardless of their mechanism of action, sensitize cancer cells to<br />

Apo2L/TRAIL cytotoxicity in vitro <strong>and</strong> in vivo animal model. The molecular basis of this<br />

effect is diverse, ranging from modulation of the DISC activity to phenotypic alteration of the<br />

levels of anti-/pro-apoptotic proteins. We hypothesized that chemotherapeutic drugs, by<br />

altering the pro-apoptotic threshold of mitochondria, synergize with Apo2L/TRAIL to<br />

promote cytotoxicity via recruitment of the type II caspase activation cascade. Cisplatin,<br />

Paclitaxel, histone deacetylase inhibitors Trichostatin A or Valproic acid all sensitize multiple<br />

cultured thoracic cancer cells to Apo2L/TRAIL. Combining individual chemotherapeutics (at<br />

sublethal concentrations) with Apo2L/TRAIL results in 2- to >20-fold enhancement of<br />

cellular sensitivity to this lig<strong>and</strong> <strong>and</strong> massive synergistic induction of apoptosis (


Session X : Cell death in cancer Poster X, 76<br />

Modulation of p53 transcriptional activity by PRIMA-1 <strong>and</strong> Pifithrin-alpha : effects on<br />

staurosporine-induced apoptosis of wild-type <strong>and</strong> mutated p53 epithelial cells.<br />

Magali Nicolier, Jean-François Charlot, Jean-Luc Prétet <strong>and</strong> Christiane Mougin.<br />

IFR133, EA 3181, Carcinogenèse épithéliale, UFR Médecine et Pharmacie, 19 rue<br />

Ambroise Paré, 25000 Besançon, France. Email : magali.nicolier@univ-fcomte.fr<br />

The p53, a specific DNA-binding transcription factor, triggers multiple biological<br />

responses including cell cycle arrest <strong>and</strong> apoptosis. Inactivation of p53 functions by mutations<br />

or by viral oncoproteins such as E6 from high risk papillomavirus (HPV) is a common event<br />

in many human cancers. New chemotherapeutic approaches consist in the restoration of<br />

apoptosis through refolding p53 mutants by PRIMA-1 (p53 reactivation <strong>and</strong> induction of<br />

massive apoptosis). As a result, PRIMA-1 restores the transcriptional activity of mutated p53<br />

(p53mt). Anti-cancer therapies also damage surrounding normal tissues which highly express<br />

wild-type p53 (p53wt). That is why Pifithrin-alpha (p-fifty three inhibitor), a chemical<br />

inhibitor of p53wt, has been proposed as an adjunct to protect normal cells from otherwise<br />

lethal doses of chemo/radiotherapy.<br />

Recently, we argued for a major role of p53 in staurosporine (ST)-induced apoptosis of<br />

immortalized epithelial cells, according to p53 status (wt or mt) [Charlot et al., Apoptosis<br />

2004]. Here, we investigated the effects of PRIMA-1 <strong>and</strong> Pifithrin-alpha on ST-induced<br />

apoptosis of 5 different immortalized cell lines with variable p53 status (p53wt HeLa HPV<br />

18+ <strong>and</strong> CaSki HPV 16+ cells ; p53mt C33-A cells; p53-/- SaOs-2 cells) [Charlot, Nicolier et<br />

al., Apoptosis, in press]. As expected, PRIMA-1 has no effect on p53wt or p53-/- cells. On the<br />

other h<strong>and</strong>, it increases p21 <strong>and</strong> Bax expression, mitochondrial membrane potential<br />

dissipation <strong>and</strong> DNA fragmentation of p53mt cells. By contrast, Pifithrin-alpha does not<br />

modify ST-induced apoptosis of p53mt <strong>and</strong> p53-/- cells but readily decreases the apoptosis of<br />

cells harb<strong>our</strong>ing a transcriptionnally active p53.<br />

These data strength the evidence that PRIMA-1 could increase ST efficiency in tumors with<br />

mutant p53. Moreover, Pifithrin-alpha could be used in combination with ST <strong>and</strong> PRIMA-1 to<br />

prevent side effects of anti-tumor therapies on healthy tissues.<br />

- 409 -


Session X : Cell death in cancer Poster X, 77<br />

Reciprocal neutralization of the antiapoptotic effects of magnetic fields <strong>and</strong> melatonin<br />

due to interference between the two respective effectors, NO synthase <strong>and</strong> lipoxygenase<br />

Silvia Nuccitelli, Flavia Radogna, Claudia Cerella, Milena De Nicola, Maria D’Alessio,<br />

Andrea Magrini*, Antonio Bergamaschi*, Lina Ghibelli<br />

Dipartimento di Biologia;*Cattedra di Medicina del Lavoro, Università di Roma Tor<br />

Vergata, Via Ricerca Scientifica 1, 00133 Rome Italy. E-mail: silvia.nuccitelli@libero.it<br />

Chemical/physical agents able to prevent apoptosis are receiving much attention for their<br />

potential health hazard as tumor promoters <strong>and</strong> for their potential therapeutic role against<br />

tissue degenerations. Magnetic fields (MFs), which have been shown to increase the<br />

development of some tumors, reduce damage-induced apoptosis by a mechanism involving<br />

Ca2+ entry into cells [Fanelli et al., FASEB J. 13, 95-102, 1999]. The neuro-hormone<br />

melatonin, which is giving great expectations as therapeutic agent, also reduce damageinduced<br />

apoptosis, again by a mechanism involving Ca2+ entry. The pathways responsible for<br />

the two anti-apoptotic effects are complex <strong>and</strong> different (see accompanying presentations),<br />

involving NO synthase (NOS) as one effector of MFs protection; <strong>and</strong> lipoxygenase (LOX) as<br />

one of the effectors of melatonin protection. We wanted to know if the anti-apoptotic effects<br />

of MFs <strong>and</strong> melatonin were synergic. Thus, we induced apoptosis in the presence of both<br />

agents. Surprisingly we found that MFs <strong>and</strong> melatonin are not only not synergic, but abrogate<br />

their respective anti-apoptotic effects. In order to verify whether this was due to an<br />

interference between the effectors of the two different anti-apoptotic pathways, we inhibited<br />

one or the other effectors (NOS with L-NAME <strong>and</strong> LOX with AA861) <strong>and</strong> checked whether<br />

this would restore the protection mechanisms of melatonin or MFs, respectively. We found<br />

that in U937 induced to apoptosis in the presence of both MFs <strong>and</strong> melatonin, L-NAME<br />

abrogates MFs action <strong>and</strong> restores the anti-apoptotic effect of melatonin; vice versa, AA861<br />

abrogates melatonin action <strong>and</strong> restores the anti-apoptotic effects of MFs. These results<br />

indicate that the signal transduction pathways involving NOS <strong>and</strong> LOX interfere <strong>and</strong><br />

neutralise each other. The reciprocal interference implies that no direct inhibition is exerted<br />

between the two enzymes, but rather the products of each interfere with the downstream<br />

effectors of the other. No reports of effects of LOX or its products on NOS are available to<br />

the best of <strong>our</strong> knowledge whereas. it is well known that NO inactivates lipoxygenase<br />

[BBRC 219, 128-133, 1996]. These results recommend to explore the potential use of<br />

melatonin as a mean to contrast the harmful effects of unwanted MFs exposure.<br />

- 410 -


Session X : Cell death in cancer Poster X, 78<br />

Let’s flip the FLIP idea<br />

Beata Pajak, Arkadiusz Orzechowski<br />

Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw<br />

Agricultural University, Now<strong>our</strong>synowska 159, 02-776 Warsaw, Pol<strong>and</strong>, e-mail:<br />

bepaj@wp.pl<br />

The resistance of cancer cells to elimination by death lig<strong>and</strong>s (TNF-alpha, TRAIL, FasL) in<br />

the extrinsic apoptotic pathway allows a variety of tumors to grow <strong>and</strong> develop. The<br />

elucidation of the antiapoptotic mechanisms responsible for this phenomenon is the major<br />

challenge of the contemporary medicine. The last decade revealed several strategies of cancer<br />

cells (including colon adenocarcinomas) of how to avoid cell death <strong>and</strong> to maintain cell<br />

survival. Colorectal cancer cells inhibit death lig<strong>and</strong>-induced apoptosis by the expression of<br />

several antiapoptotic proteins, such as FLIP (FLICE-inhibitory protein), ,cIAPs (cellular<br />

inhibitors of apoptosis), <strong>and</strong> others which inhibit either signal transduction or execution of<br />

apoptosis. In colorectal cancer COLO 205 cell line the immunoprecipitation of FADD (Fasassociated<br />

death domain-containing protein) showed its assembly with FLIP. Usually, the<br />

presence of FLIP in the TNF-alpha signalosome inhibits the caspase 8 (FLICE - FADD-like<br />

interleukin-1#-converting enzyme) autoactivation, <strong>and</strong> blocks TNF-alpha-induced apoptosis.<br />

Several studies demonstrated that the reduction of FLIP level by the protein synthesis<br />

inhibitors, such as cycloheximide or bis-indolylmaleimide could sensitize cells to apoptotic<br />

stimuli. Similarly, actinomycin D, the inhibitor of DNA transcription exerts cell deathpromoting<br />

properties. In COLO 205 cell line the Western-blot <strong>and</strong> immunocytochemistry<br />

analyses demonstrated, that the use of cycloheximide decreases the FLIP protein level in a<br />

time-dependent manner, what correlates positively with the progression of cell death. In<br />

contrast, the use of siRNA-FLIP did not lead to cell death upon TNF-alpha treatment. It is<br />

concluded that protein(s) other than FLIP inhibited TNF-alpha-dependent cell death<br />

irrespective to the interaction of FLIP with the TNF-RI signalosome. Thus, unknown<br />

antiapoptotic protein(s) blocked apoptosis downstream to the TNF-R1 signalosome. This<br />

undefined protein(s) is essential for “immune escape” of COLO 205 cells. It is also assumed,<br />

that susceptibility of colorectal cancer cells to TNF-alpha-induced cell death upon<br />

concomitant cycloheximide treatment could be a consequence of oxidative stress <strong>and</strong> changes<br />

in the intracellular milieu. The latter is currently under detailed scrutiny.<br />

- 411 -


Session X : Cell death in cancer Poster X, 79<br />

Mitochondria role in the control of the activity of p53 <strong>and</strong> Rb<br />

Ioana Pârvu-Ferecatu, Nelly Godefroy, Vincent Rincheval, Nathalie Leleu, Bernard<br />

Mignotte <strong>and</strong> Jean-Luc Vayssière<br />

Laboratoire de Génétique et Biologie Cellulaire (FRE CNRS 2445), Université de<br />

Versailles Saint-Quentin-en-Yvelines et Laboratoire de Génétique Moléculaire et<br />

Physiologique,<br />

Ecole Pratique des Hautes Etudes, 45 avenue des Etats-Unis 78035 Versailles Cedex<br />

We previously showed in rat embryo fibroblasts that the p53 protein can activate two distinct<br />

two apoptotic pathways (1, 2 <strong>and</strong> 3). The first one involves p53’s targets gene transactivation,<br />

whose products trigger a mitochondrial apoptosis that is sensitive to Bcl-2 inhibition. The<br />

second pathway is mediated by the p53’s transcriptional repression activity, initiating a<br />

caspase-independent <strong>and</strong> Bcl-2-insensitive massive cell death. We also reported that the Rb<br />

tum<strong>our</strong> suppressor protein contributes to the outcome of p53 activation. Indeed, we observed<br />

that the full length form of Rb, named p110Rb, supports the transrepressional activity of p53,<br />

whereas its cleaved p76Rb form, generated by caspase-9, allows cell survival (3).<br />

Here, we describe a new role for mitochondria in regulating the activity of both the p53 <strong>and</strong><br />

Rb proteins. Indeed, in most of the cell types we tested, the inactive p53 protein displays a<br />

mitochondrial location under normal conditions (i.e. proliferating cells), suggesting that the<br />

docking of p53 to mitochondria contributes to its inactivation. After stress, posttranslational<br />

modifications (e.g.: phosphorylation) of p53 promotes its translocation towards the nucleus,<br />

followed by either apoptosis or growth arrest. Equally, we showed for the first time a<br />

mitochondrial location of the Rb protein which would be correlated to its ability to antagonize<br />

p53-induced apoptosis. Thereby, these data support a model in which mitochondria play a<br />

regulatory role of the activity of both p53 <strong>and</strong> Rb by trapping these proteins, to prevent at<br />

least their nuclear activity.<br />

- 412 -


Session X : Cell death in cancer Poster X, 80<br />

Over-expression of microRNA-21 modulates phosphoinositide-3 kinase <strong>and</strong> PTEN<br />

<strong>signaling</strong> <strong>and</strong> promotes tumor cell survival.<br />

Tushar Patel, Fanyin Meng, Roger Henson, Hania Wehbe, Molly Lang<br />

Scott <strong>and</strong> White Clinic, Texas A&M University System Health Science Center College of<br />

Medicine, 2401 South 31st Street, Temple, TX 76508, U.S.A. E-mail:<br />

tpatel@swmail.sw.org<br />

MicroRNAs (miRNAs) are endogenous RNA molecules that can regulate gene expression.<br />

Altered expression of miRNA such as miR-21 has been reported for several different cancers.<br />

However the role of miR-21 in tumor progression <strong>and</strong> the mechanisms involved remain<br />

unknown. Our aims were to assess the effect of miR-21 on critical survival <strong>signaling</strong><br />

pathways. Methods: A miRNA microarray was used to profile the miRNA expression in<br />

malignant (KMCH, Mz-ChA-1 <strong>and</strong> TFK) <strong>and</strong> non-malignant (H69) cholangiocyte cell lines.<br />

Expression of miR-21 was confirmed by Northern blot <strong>and</strong> real-time PCR. miR-21<br />

expression was altered using mir-21 specific antisense oligonulceotides or mir-21 precursor<br />

miRNAs. Expression of PTEN <strong>and</strong> p85 was assessed by Western blots. For in vivo studies,<br />

homogenates were obtained from xenografts of Mz-ChA-1 cells in nude mice under basal<br />

conditions or following treatment with gemcitabine. Results: miR-21 expression was<br />

increased in all malignant cell lines compared to non-malignant H69 cholangiocytes (2.74 ±<br />

0.11 fold in Mz-ChA-1 cells, 4.35 ± 0.14 fold in TFK cells, <strong>and</strong> 1.84 ± 0.03 fold in KMCH-1<br />

cells; all p


Session X : Cell death in cancer Poster X, 81<br />

Induction of osteoclasts apoptosis by using a decoy oligodeoxynucleotide in vivo<br />

mimicking a region of distal promoter C of ER alpha gene<br />

Letizia Penolazzi, Margherita Zennaro, Ercolina Bianchini, Eros Magri, Elisabetta<br />

Lambertini, Elisa Tavanti, Roberto Gambari <strong>and</strong> Roberta Piva.<br />

Department of Biochemistry <strong>and</strong> Molecular Biology, University of Ferrara, Italy. Email:<br />

piv@unife.it<br />

The function of osteoclasts (OCs), the multinucleated cells responsible for the resorption of<br />

extracellular bone matrix, are regulated by different kinds of hormones, cytokines,<br />

inflammatory <strong>and</strong> transcription factors. Of particular interest is the role of estrogen receptor<br />

alpha (ER) protein that mediates the action of estrogen. In particular, it suppresses OCs<br />

formation <strong>and</strong> activity <strong>and</strong> inhibits OCs-mediated bone resorption in part by stimulating OCs<br />

to undergo apoptosis through a receptor-mediated genomic action. In order to study<br />

therapeutic strategies to control bone formation, we investigated the possibility to inhibit<br />

osteoclastogenesis through a specific decoy strategy. We obtained an increase of ER<br />

expression by interfering with the activity of a negative transcription factor <strong>and</strong> by removing<br />

it with a decoy oligonucleotide (RA4-3’) mimicking a region of distal promoter C of ER gene.<br />

We demonstrated that this decoy was able to induce apoptosis in human primary OCs, but not<br />

in osteoblasts, in an estrogen dependent manner, increasing also Caspase 3 <strong>and</strong> Fas receptor<br />

levels. These findings may be of relevance for a possible therapeutical approach for tumors,<br />

bone metastasis <strong>and</strong> osteopenic diseases. At this purpose,the clinical orthodontic offers a good<br />

opportunity to study in vivo the efficacy of <strong>our</strong> approach. In fact, the increased number <strong>and</strong><br />

activity of OCs is involved in the regulation of alveolar bone resorption during orthodontic<br />

tooth movement, <strong>and</strong> also in the origin of dental problems in diseases of OCs activation<br />

affecting the maxillo-m<strong>and</strong>ibular bone. We designed in vivo experiments aimed at regulating<br />

alveolar bone resorption. Six Wistar male rats were subjected to orthodontic forces, in<br />

combination or not with RA4-3’ decoy treatment by using a split-mouth design. Examination<br />

of paraffin sections of the excised molars showed that orthodontic forces caused a percentage<br />

of apoptotic OCs that appears to be highly potentiated by RA4-3’, but not by scramble ODN.<br />

These data confirm the results obtained in vitro with human OCs from peripheral blood,<br />

suggesting an important correlation between ER <strong>and</strong> the possibility to modulate OCs<br />

functions.<br />

- 414 -


Session X : Cell death in cancer Poster X, 82<br />

Bcl-2 expession in oral squamous cell carcinoma<br />

Branka Popovi'1, Zvezdana Tepav(evi', Vladimir Juri)i'2 <strong>and</strong> Jelena. Mila)in1<br />

1Institute of Biology <strong>and</strong> Human Genetics, School of Stomatology, Belgrade, Serbia <strong>and</strong><br />

Montenegro, E-mail: jelena_milasin@yahoo.com<br />

2Institute of Pathophysiology, School of Medicine, Kragujevac, Serbia <strong>and</strong> Montenegro<br />

One of the mechanisms involved in the pathogenesis of oral squamous cell carcinoma<br />

(OSCC) is deregulation of the programmed cell death-apoptosis. Apoptosis is a genetically<br />

determined process controlled by bcl-2 gene family, with different genes promoting or<br />

inhibiting cell death depending on the ratio of proteins with opposite function in apoptotic<br />

athway. Bcl-2 gene belongs to antiapoptotic factors which overexpression may lead to<br />

tum<strong>our</strong> progression.<br />

The aim of this study was to estimate the bcl-2 immunoreactivity in OSSC <strong>and</strong> to assess its<br />

clinicopathological implication. A series of the 26 OSCC formalin fixed, paraffin embedded<br />

samples was analyzed for bcl-2 expression. The immunohistochemical staining was scored<br />

according to the percentage of positive stained tum<strong>our</strong> cells. Using TNM classification there<br />

were 7 tum<strong>our</strong>s in stage II, 14 in stage III <strong>and</strong> 5 in stage IV. The positive staining was present<br />

in all samples, with mean values <strong>and</strong> st<strong>and</strong>ard deviations 8.3±2.5, 7.5±1.1 <strong>and</strong> 8.4±5.8 within<br />

stages II, III <strong>and</strong> IV, respectively. All tum<strong>our</strong>s independently of the stage did not show<br />

statistically significant difference in positive cytoplasmic staining against bcl-2, but its lower<br />

expression was significantly associated with higher overall survival. Our results suggest that<br />

bcl-2 expression could be a valuable biological marker in predicting disease behavi<strong>our</strong>.<br />

- 415 -


Session X : Cell death in cancer Poster X, 83<br />

Histone deacetylase inhibitor LAQ824 downregulates beta-catenin, inhibits cell<br />

proliferation <strong>and</strong> induces apoptosis in colon carcinoma cell lines.<br />

Ignacio Portero-Robles, Martine Pape, Sina Droll, Dieter Holzer, Ulrich Böcker, Martin<br />

Ruthard, Oliver G. Ottman <strong>and</strong> Noriko Koyama.<br />

Universitäts-Klinikum Frankfurt. ZIM II Hämatologie. Theodor-Stern-Kai 7 60590<br />

Frankfurt am Main. Germany. E-Mail: robles@em.uni-frankfurt.de<br />

Histone deacetylase (HDAC) inhibitors represent a class of anticancer agents that act by<br />

promoting acetylation of histones, activating genes implicated in proliferation <strong>and</strong> apoptosis.<br />

Mutations in the Wnt <strong>signaling</strong> pathway result in a continuous translocation of beta-catenin to<br />

the nucleus acting as cofactor of TCF/LEF. Targets genes of these complex are constitutively<br />

expressed inhibiting apoptosis <strong>and</strong> promoting tumor cells proliferation.<br />

The aim of this study is to analyze the effect of LAQ824 (Novartis, Basel), a HDAC inhibitor,<br />

in colon carcinoma cell lines <strong>and</strong> the contribution of the Wnt-<strong>signaling</strong> pathway to HDAC<br />

inhibitor-induced apoptosis <strong>and</strong> inhibition of cellular proliferation.<br />

After treatment with 10 to 200nM of LAQ824, we show acetylation of histones -3 <strong>and</strong> -4 <strong>and</strong><br />

induction of p21Waf/Cip1. Under the same condition cell proliferation is inhibited, annexin-V<br />

expressing cells are increased, caspase -3, -9, -8, -2 as well as PARP cleavage is observed.<br />

Similar assays with HCT116 p53(-/-) cells reveal no significant differences with the parental<br />

HCT116.<br />

Treatment of HCT116 cells with 100nM LAQ824 leads to decrease the protein level of betacatenin<br />

<strong>and</strong> to downregulate consequently its target genes, c-jun, c-myc <strong>and</strong> cyclin D1.<br />

Similar effects are also observed when cells are treated with 0,5µM Trichostatin A <strong>and</strong> 5mM<br />

Valproic acid. HDAC2, an indirect target of beta-catenin which is overexpressed in colon<br />

cancer tissues, is downregulated by VPA but not by LAQ <strong>and</strong> TSA.<br />

Our results demonstrate that LAQ824 inhibit cell proliferation <strong>and</strong> induces apoptosis in a<br />

caspase-dependent <strong>and</strong> p53-independent manner <strong>and</strong> indicate that Wnt <strong>signaling</strong> could be<br />

involved in the anti-tumor effects of LAQ824.<br />

- 416 -


Session X : Cell death in cancer Poster X, 84<br />

APOPTOTIC PROPERTIES OF HYPERFORIN ON HUMAN B-CLL LEUKEMIC<br />

CELLS<br />

Claire Quiney, Christian Billard, Pezhman Mirshahi, Anne-Marie Faussat, Jean-<br />

Dominique F<strong>our</strong>neron *, <strong>and</strong> Jean-Pierre Kolb.<br />

INSERM U736, Centre Biomédical des Cordeliers, Paris <strong>and</strong> * UMR6171, Faculté des<br />

Sciences St Jérôme, Marseille<br />

The natural compound hyperforin (HF), purified from Saint John’s wort, was tested on<br />

leukemic cells from B cell lymphocytic leukemia (B-CLL) patients <strong>and</strong> on the ESKOL cell<br />

line derived from a patient with hairy cell leukemia. HF was found to inhibit the proliferation<br />

of ESKOL cells (IC50 around 1 µg/ml) <strong>and</strong> to elicit their apoptosis. Similarly, HF triggered a<br />

time- <strong>and</strong> dose-dependent induction of apoptosis in B-CLL leukemic cells that was detected<br />

by the increase in the percentage of annexinV-labelled cells <strong>and</strong> the augmentation of<br />

internucleosomal DNA fragmentation. This was accompanied by the activation of caspase 3,<br />

the disruption of the mitochondrial transmembrane potential &*m <strong>and</strong> the inhibition of nitric<br />

oxide (NO) production. HF-induced apoptosis was reverted in the presence of the general<br />

caspases inhibitor Z-VAD-fmk.<br />

Incubation of B-CLL cells with HF also resulted in a marked suppression of their capacity to<br />

release matrix metalloprotease 9 (MMP-9), an essential component in the degradation of the<br />

extra cellular matrix. In addition, HF prevented the formation of tubules by human bone<br />

marrow endothelial cells (HBMEC) cultured on matrigel, emphasizing its potential interest as<br />

an anti neo-vasculogenesis drug.<br />

Moreover, HF was found to impair the activity of several ABC pumps that are involved in the<br />

multidrug resistance of the leukemic cells against chemotherapeutic reagents.<br />

- 417 -


Session X : Cell death in cancer Poster X, 85<br />

Multiple, independent melatonin-engaged signal transduction pathways are required for<br />

melatonin anti-apoptotic effect.<br />

Flavia Radogna, Laura Paternoster, Silvia Nuccitelli, Maria D’Alessio, Claudia Cerella,<br />

Milena De Nicola, Antonio Bergamaschi*, Lina Ghibelli<br />

Dipartimento di Biologia; *Cattedra Medicina del Lavoro, Università di Roma Tor<br />

Vergata, Via Ricerca Scientifica 1, 00133 Rome Italy. E-mail: flavia.radogna@libero.it<br />

Melatonin, in addition to its main role as regulator of circadian rhythms, has recently been<br />

shown to modulate immune functions by controlling the behavi<strong>our</strong> of white blood cells<br />

(WBC), which are indeed able to synthesize melatonin <strong>and</strong> possess the specific high affinity<br />

(1nM) plasma membrane receptors (MT1 <strong>and</strong> MT2). Great interest is receiving the ability of<br />

melatonin to contrast apoptosis, a well accepted fact whose mechanisms however are still<br />

quite controversial. In this study, we analyze the mechanisms involved in the anti-apoptotic<br />

effect of melatonin in normal <strong>and</strong> tumor WBC. We have shown that this effect is due to two<br />

different, cooperating mechanisms, involving two primary targets melatonin directly interacts<br />

with, i.e., MT1/MT2 receptors; <strong>and</strong> calmodulin, a known melatonin low affinity (63uM)<br />

target. Receptor engagement <strong>and</strong> calmodulin binding give rise to two parallel signal<br />

transduction pathways, consisting of a canonical signal transduction on the one side, <strong>and</strong><br />

phospholipase A2 (a known calmodulin interactor) <strong>and</strong> consequent 5-lipoxygenase (LOX)<br />

activation <strong>and</strong> 5-HETE production, on the other. The two pathways converge in the inhibition<br />

of Bax dimerisation <strong>and</strong> activation, key event of the intrinsic apoptotic pathway. The antiapoptotic<br />

effect is completely abrogated if one or the other pathway is inhibited. The<br />

necessity of the low affinity calmodulin binding explains the requirement of high melatonin<br />

doses (>100uM), thus answering to the many perplexities as to whether melatonin antiapoptotic<br />

effect was indeed due to MT1/MT2 receptor stimulation. The involvement of 5-<br />

LOX in the anti-apoptotic effect of melatonin is particularly intriguing since 5-LOX has been<br />

reported to be involved in tumor progression by inhibiting apoptosis in some tumor types. In<br />

addition, the recruitment of a key enzyme of the inflammatory response may shed new lights<br />

on the role melatonin plays in the regulation of the immune response. Moreover, LOX<br />

activation implies a burst of free radicals that immediately (


Session X : Cell death in cancer Poster X, 86<br />

Effect of roscovitine on wt p53 protein in human MCF-7 breast cancer cells: uncoupling<br />

of p53 nuclear accumulation <strong>and</strong> its activation by site-specific phosphorylation<br />

Carmen Ranftler, Marieta Gueorguieva <strong>and</strong> Józefa W*sierska-G+dek<br />

Cell Cycle Regulation Group, Division: Institute of Cancer Research, Department of<br />

Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria.<br />

We have recently observed activation of wt p53 protein in human MCF-7 breast cancer cells<br />

upon treatment with roscovitine (ROSC), a potent cyclin-dependent inhibitor. ROSC is<br />

known to inhibit the activating phosphorylation of the carboxy-terminal domain of RNA<br />

polymerase II, thereby blocking the global transcription. It has been previously suggested that<br />

ROSC-induced upregulation of p53 protein is attributable to the inhibition of transcription of<br />

Mdm2, a negative p53 regulator. However, the analysis of the expression of p53-responsive<br />

genes revealed that Mdm2 was upregulated in ROSC-treated MCF-7 cells. The kinetics of the<br />

Mdm2 increase was slightly delayed as compared with that for p21waf1. <strong>Expo</strong>sure of human<br />

MCF-7 cells to different proteosome inhibitors resulted in the time-dependent increase of p53<br />

levels. However, unlike ROSC, they failed to modify p53 protein at Ser46 <strong>and</strong> to induce<br />

p53AIP1 protein. Moreover, whereas ROSC arrested MCF-7 cells in the G2 phase of the cell<br />

cycle, proteosome inhibitors blocked cells in the S-phase, presumably due to the prevention of<br />

cyclin degradation. Our results indicate that prevention of p53 degradation by proteasome<br />

inhibitors does not mimic the action of ROSC.<br />

- 419 -


Session X : Cell death in cancer Poster X, 87<br />

Role of plasma membrane fluidity in cisplatin-induced apoptosis in HT29 human colon<br />

cancer cells<br />

Amélie Rebillard1, Odile Sergent2, Olivier Meurette1, Gwénaëlle Le Moigne1, Morgane<br />

Gorria1, Laurent Vernhet1, Dominique Lagadic-Gossmann1 & Marie-Thérèse<br />

Dimanche-Boitrel1*<br />

1INSERM UMR 620 & 2Laboratoire de Biologie Cellulaire et Végétale, Faculté de<br />

Pharmacie, Université Rennes 1, 2 av du Prof Léon Bernard, 35043 Rennes cedex,<br />

France.<br />

Most current anticancer therapies induce tumor cell death through the induction of apoptosis.<br />

However, the biochemical lesions leading to cell death are not always understood. The plasma<br />

membrane has been considered as the most important target, other than DNA, for many<br />

anticancer drugs. From a structural viewpoint, the plasma membrane is an heterogeneous<br />

structure composed of distinct microdomains termed lipid rafts. These lipid rafts are enriched<br />

in cholesterol <strong>and</strong> sphingolipids (sphingomyelin <strong>and</strong> glycosphingolipids) thus defining more<br />

ordered area in the plasma membrane. These lipid microdomains actively participate in<br />

several <strong>signaling</strong> transduction pathways, particularly in the Fas death receptor pathway, <strong>and</strong><br />

also in drug-induced apoptosis. We have recently shown that an early <strong>and</strong> transient increase in<br />

plasma membrane fluidity, determined by a spin-labeling method using EPR (Electron<br />

Paramagnetic Resonance), has been associated to agregation <strong>and</strong> relocalisation of Fas receptor<br />

into plasma membrane lipid rafts during cisplatin-induced apoptosis in HT29 human colon<br />

cancer cells (Lac<strong>our</strong> et al., Cancer Res. 2004, 64 (10): 3593-3598).<br />

In order to study the role of plasma membrane fluidity, the effects of membrane stabilizing<br />

agents (ursodeoxycholic acid (UDCA), cholesterol (Chol) <strong>and</strong> monosyaloganglioside 1<br />

(GM1)) have been investigated in cisplatin-induced apoptosis in HT29 cells.<br />

UDCA, Chol <strong>and</strong> GM1 inhibit the early increase in plasma membrane fluidity following<br />

cisplatin treatment, <strong>and</strong> about 30 % the apoptosis induced by cisplatin, without modifying the<br />

entry of cisplatin into HT29 cells (measured by atomic absorption spectrometry). Moreover,<br />

we show that a pretreatment with stabilizing agents inhibits Fas agregation as well as lipid<br />

rafts agregation evidenced by a FITC-cholera toxin labeling on the cell surface of HT29 cells<br />

treated with cisplatin. Altogether, these data suggest that the transient increase in plasma<br />

membrane fluidity contribute to cisplatin-induced early plasma membrane events <strong>and</strong><br />

subsequently to cisplatin-induced apoptosis.<br />

- 420 -


Session X : Cell death in cancer Poster X, 88<br />

In T lymphocytes, ablation of STAT3 expression reinstates STAT1-dependent apoptosis<br />

by both IFNg <strong>and</strong> IL-6<br />

Gabriella Regis1,2, Laura Icardi1,2, Laura Conti1,2, Roberto Chiarle1, Paola<br />

Bernabei1, Valeria Poli1 <strong>and</strong> Francesco Novelli1,2<br />

1Center for Experimental Research <strong>and</strong> Medical Studies (CERMS), San Giovanni<br />

Battista Hospital, Via Santena 5, 10126, Turin, Italy <strong>and</strong> 2Department of Medicine <strong>and</strong><br />

Experimental Oncology, University of Turin, Corso Raffaello 30, 10125, Turin, Italy.<br />

The Interferon gamma (IFNg)/Signal Transducer <strong>and</strong> Activator of Transcription (STAT) 1<br />

pathway is mainly involved in the control of T lymphocytes homeostasis. However, these<br />

cells downregulate IFNgR2 <strong>signaling</strong> chain by both lig<strong>and</strong>-dependent <strong>and</strong> lig<strong>and</strong>-independent<br />

mechanisms <strong>and</strong> become refractory to the IFNg-mediated control of their expansion. Here, we<br />

investigate the role of STAT3, a transcriptional factor activated by many cytokines including<br />

IFNg, in the negative regulation of the IFNg/STAT1 <strong>signaling</strong> pathway. The expression of<br />

STAT3 in two human malignant T cell lines, PF383 <strong>and</strong> ST4, was almost completely<br />

inhibited by lentivirus that deliver small interfering (si) RNA against STAT3. The survival of<br />

many neoplastic cells is dependent on STAT3 expression, whereas the viability of malignant<br />

T cell lines we used was not influenced by STAT3 expression silencing. In T cells expressing<br />

normal STAT3 amounts, either IFNg or IL-6 induced a weak activation of STAT1. By<br />

contrast, in the absence of STAT3 both IFNg <strong>and</strong> IL-6 dependent STAT1 activation was<br />

enhanced. This increased activation of STAT1 by IL-6 confirms previous data that showed<br />

that this cytokine activates STAT1 <strong>and</strong> mediates an IFNg-like response in cells lacking<br />

STAT3. As STAT1 regulates the expression of many genes involved in the control of cellular<br />

proliferation <strong>and</strong>/or apoptosis we checked the proliferative responses to IFNg or IL-6 in T cell<br />

lines where STAT3 is silenced or not, but we didn’t observe any alteration. On the contrary,<br />

both IFNg <strong>and</strong> IL-6 switched on the apoptotic program in T cells devoid of STAT3<br />

expression, whereas they were ineffective in cells expressing normal amounts of STAT3. The<br />

apoptotic pathways are currently under investigation in <strong>our</strong> Laboratory. However, these data<br />

clearly show that the combined inhibition of STAT3 synthesis <strong>and</strong> the treatment with IFNg or<br />

IL-6 reinstate STAT1-dependent apoptosis in human T lymphocytes.<br />

- 421 -


Session X : Cell death in cancer Poster X, 89<br />

Overexpression of Lifeguard (LFG) suppresses TRAIL-induced apoptosis <strong>and</strong> activates<br />

NF-kB<br />

Kerstin Reimers, Claudia YU Choi, Susanne Kall, Peter M Vogt<br />

Department of Plastic, H<strong>and</strong> <strong>and</strong> Reconstructive Surgery, Medical School Hannover,<br />

Podbielskistr. 380, 30659 Hannover, Germany, E-mail: Reimers.Kerstin@MH-<br />

Hannover.de<br />

TRAIL is a promising cytokine for cancer therapies, which preferentially induces apoptosis in<br />

cancer cells by binding to death receptors TRAIL-R1/DR4 <strong>and</strong> TRAIL-R2/DR5. Other tumor<br />

cells, however, are resistant against TRAIL <strong>and</strong> require aditional treatment with radio- or<br />

chemotherapy. Lifeguard (LFG) is an anti-apoptotic protein related to the Bax-inhibitory<br />

protein 1 (BI-1). Cells expressing LFG are resistant against TRAIL- <strong>and</strong> Fas-induced cell<br />

death. We were able to show that this protective effect can be transmitted to non-LFGexpressing<br />

Fas-sensitive cell lines using transient transfection of LFG. In order to characterize<br />

the functional mechanism of this antiapoptotic protein we determined the DNA-binding<br />

activity of the NF-kB p65 subunit in cells overexpressing LFG. The cells showed an increased<br />

nuclear translocation of the p65 subunit. The resulting activation of NF-kB depends on IkBa<br />

degradation. An increased NF-kB activity is correlated with a sensitization of the treated cells<br />

against induction of apoptosis by TRAIL. Many cancer cells show constitutive activation of<br />

NF-kB leading to insensitiviy against therapeutically induced apoptosis. High expression of<br />

LFG might contribute to cell death prevention by activation of NF-kB.<br />

- 422 -


Session X : Cell death in cancer Poster X, 90<br />

THE INFLUENCE OF ECHINACEA PURPUREA L. MOENCH TO THE TOXICITY<br />

OF CADMIUM<br />

Nijole Savickiene3, Alina Smalinskiene1, Stanislovas Ryselis1, Oleg Abdrakhmanov1,<br />

Rima Kregzdyte1, Vaiva Lesauskaite2, Ilona Sadauskiene1, Leonid Ivanov1, , Virgilijus<br />

Zitkevi(ius3, Arunas Savickas4<br />

1Institute for Biomedical Research, Kaunas University of Medicine, 2Institute of<br />

Cardiology, Kaunas University of Medicine, 3Department of Pharmaceutical Chemistry<br />

<strong>and</strong> Pharmacognosy, Kaunas University of Medicine, 4Department of Drug Technology<br />

<strong>and</strong> Pharmaceutical Management, Kaunas University of Medicine, Kaunas, Lithuania<br />

E-mail : savickienenijole@takas.lt<br />

Cadmium has a diversity of toxic effects including nephrotoxicity, carcinogenicity,<br />

teratogenicity, endocrine toxicity. The aim of this study was to investigate the accumulation<br />

of various levels of Cd2+ in tissues, <strong>and</strong> the effects of Echinacea purpurea L. Moench liquid<br />

extract on Cd2+ induced changes in mice.<br />

Materials <strong>and</strong> methods. Experiments were carried out on the white laboratory mice (n=47).<br />

Chronic experiments were performed by giving to drink solution of different Cadmium <strong>and</strong><br />

EP concentrations: Cd2+ 25 mg/l, Cd2+ 250 mg/l, EP – 3.5 ml/l, daily for 8 weeks. Control<br />

group mice was giving to drink equal amount of deionisated water. Cd concentration was<br />

determined by atomic absorption spectrophotometer Perkin-Elmer/Zeeman 3030. The number<br />

of mitotic liver cells was counted in 10 r<strong>and</strong>omly selected reference areas (0.04 mm2).<br />

Apoptosis of liver cells was histochemically detected by the TUNEL assay using AP (Roche)<br />

in situ cell death detection kit. Preparation of extract from herb of Echinacea purpurea (L.)<br />

Moench was made according British herbal pharmacopoeia.<br />

Results. Cd2+ concentration in all explored organs <strong>and</strong> blood of mice, which were giving to<br />

drink CdCl2 was statistically significantly higher than that of control group mice. After 8<br />

weeks of experiment with mice giving to drink CdCl2 250 mg/l <strong>and</strong> EP extract solutions we<br />

estimated, that number of apoptotic liver cells statistically significantly (p


Session X : Cell death in cancer Poster X, 91<br />

Functional analysis of the tumorsuppressor Ras Association Family Domain 1<br />

(RASSF1A) reveals an important role of the SARAH-domain<br />

Claudia Seidel, Antje Klagge, Reinhard Dammann<br />

AWG Tumor Genetics of the Medical Faculty, Martin-Luther-University Halle-<br />

Wittenberg, 06097 Halle/Saale, Germany<br />

INTRODUCTION: RASSF1A is a tumor suppressor gene, which is often epigenetically<br />

inactivated in several cancer types. In lung cancer, a rate of between 30 to 45% of<br />

hypermethylation of the RASSF1A promoter has been found. In several cancer types a<br />

correlation between hypermethylation of RASSF1A <strong>and</strong> an impaired prognosis for cancer<br />

patients was reported. The promoter of the RASSF1C isoform is never hypermethylated. The<br />

two isoforms RASSF1A <strong>and</strong> RASSF1C only differ in the first domain, which is a<br />

diacylglycerol binding site <strong>and</strong> lacks in RASSF1C. Moreover RASSF1A encodes a Ras<br />

association domain <strong>and</strong> a SARAH domain, which is a protein-protein interaction domain.<br />

METHODS: In order to investigate the function of RASSF1A, we created mutations <strong>and</strong><br />

deletions of these different domains <strong>and</strong> performed functional experiments in the lung cancer<br />

cell line A549. Proliferation analyses as well as soft agar experiments were performed to<br />

investigate the influence of the aberrant RASSF1A forms. The ability of the different forms of<br />

RASSF1A to induce apoptosis was investigated. To analyse the localisation of RASSF1A <strong>and</strong><br />

its altered forms the cells were transient transfected using Yellow Fluorescent Protein (YFP)<br />

constructs. Additionally, the interaction of RASSF1A with the proapoptotic kinase MST1 was<br />

determined in precipitation experiments with protein extract from human cells.<br />

RESULTS: The reduced proliferation of A549 cells that express RASSF1A could be<br />

confirmed. Analysis of cells stable transfected with RASSF1A with a deletion of the SARAH<br />

domain (RASSF1ADelSARAH) showed a even lower proliferation rate in comparison to<br />

RASSF1A. A higher rate of apoptosis in cells with RASSF1ADelSARAH could be observed<br />

using transient transfected cells. RASSF1A is when overexpressed as a YFP-fusion<br />

colocalised to the cytoskeleton <strong>and</strong> during mitosis to the spindles <strong>and</strong> spindle poles. Using<br />

RASSF1ADelSARAH the mitoses show strong spindles no spindle poles <strong>and</strong> the<br />

chromosomes are not divided equally. In precipitation experiments MST1 interacts with<br />

RASSF1A but not with RASSF1ADelSARAH.<br />

CONCLUTIONS: The SARAH domain is necessary for the function of the tumor suppressor<br />

RASSF1A. It is responsible for the interaction with MST1 <strong>and</strong> plays an important role in<br />

regulation of mitotic processes as well as in induction of apoptosis.<br />

This study was supported by BMBF grant 01ZZ0104.<br />

- 424 -


Session X : Cell death in cancer Poster X, 92<br />

ASSESSMENT OF EFFECT OF ECHINACEA PURPUREA ON APOPTOTIC AND<br />

MITOTIC ACTIVITY OF LIVER CELLS DURING INTOXICATION BY CADMIUM<br />

Alina Smalinskiene1, Stanislovas Ryselis1, Oleg Abdrakhmanov1, Rima Kregzdyte1,<br />

Vaiva Lesauskaite2, Ilona Sadauskiene1, Leonid Ivanov1, Nijole Savickiene3, Virgilijus<br />

Zitkevi(ius3, Arunas Savickas4<br />

1Institute for Biomedical Research, Kaunas University of Medicine, 2Institute of<br />

Cardiology, Kaunas University of Medicine, 3Department of Pharmaceutical Chemistry<br />

<strong>and</strong> Pharmacognosy, Kaunas University of Medicine, 4Department of Drug Technology<br />

<strong>and</strong> Pharmaceutical Management, Kaunas University of Medicine, Kaunas, Lithuania<br />

E-mail : smalina@vector.kmu.lt<br />

Cadmium (Cd) is a very toxic <strong>and</strong> carcinogenic element. It disturbs the activity of<br />

biochemical systems of cells. Echinacea purpurea (L.) Moench (EP) can modify its influence.<br />

The aim of the study was to evaluate the effect of EP to accumulation of Cd in blood, liver,<br />

<strong>and</strong> kidney, mitotic <strong>and</strong> apoptotic activity of liver cells after the chronic intoxication by Cd2+.<br />

Materials <strong>and</strong> methods. Experiments were carried out on the white laboratory mice. Mice<br />

(n=57) were periodically i.p. injected for 6 weeks with CdCl2 (0.05 LD50 Cd2+) <strong>and</strong> EP<br />

extract solutions of two different concentrations (0.05 LD50 <strong>and</strong> 0.1 LD50) <strong>and</strong> their<br />

combinations. Control mice were injected with 0.9% saline. Cd concentration in blood <strong>and</strong><br />

tissue specimens was determined by atomic absorption spectrophotometer Perkin-<br />

Elmer/Zeeman 3030. The number of mitotic liver cells was counted in 10 r<strong>and</strong>omly selected<br />

reference areas (0.04 mm2). Apoptosis of liver cells was histochemically detected by the<br />

TUNEL assay using AP (Roche) in situ cell death detection kit. Preparation of extract from<br />

herb of Echinacea purpurea (L.) Moench was made according British herbal pharmacopoeia.<br />

Results. Cd2+ concentration in blood of mice in group EP(0.05)+Cd was 6.31-fold higher,<br />

therefore in group EP(0.1)+Cd was 8.25-fold higher comparing to Cd group, in liver that was<br />

1.78 fold <strong>and</strong> 2.11-fold, respectively, in kidney 2.1-fold <strong>and</strong> 1.92-fold, respectively. Higher<br />

concentration (0.1LD50) of EP extract <strong>and</strong> Cd+EP 0.1LD50 arise apoptotic activity of liver<br />

cells to compare with control group. The mitotic activity of liver cells induced by Cd2+ after<br />

injection of EP extract was the same as in control group.<br />

Conclusion. Long-term injections of extract of EP (0.1 LD50) combined with CdCl2 (0.05<br />

LD50) leads to the significant increase of cadmium concentration in blood <strong>and</strong> all<br />

investigated organs of experimental mice. EP decreases the mitotic activity of liver cells<br />

induced by cadmium <strong>and</strong> increases apoptotic activity of the liver cells.<br />

- 425 -


Session X : Cell death in cancer Poster X, 93<br />

Antiangiogenic effect of Isoliquiritigenin through down-regulation of vascular<br />

endothelial growth factor (VEGF) in breast cancer cells<br />

Seung Hwa Son1,2,3, Kwang Kyun Park1,2,3, Mi Jeong Kim1,2,3, Jung Han Yoon<br />

Park4, Soon Sung Lim4, Won Yoon Chung1,2,3<br />

1Department of Oral Biology, 2Oral Science Research Center, 3Oral Cancer Research<br />

Center, College of Dentistry, Yonsei University, Seoul 120-752, 4Division of Life Sciences<br />

<strong>and</strong> Silver Biotechnology Research Center, Hallym University, Chunchon, Republic of<br />

Korea, E-mail : hwaa7@msn.com<br />

Vascular endothelial growth factor (VEGF) is an intensively studied molecule that has<br />

significant potential, both in stimulating angiogenesis <strong>and</strong> as a target for antiangiogenic<br />

approaches. VEGF achieves its multiple functions by activating two receptor tyrosine kinases,<br />

Flt-1 <strong>and</strong> KDR, both of which are selectively expressed on primary vascular endothelium.<br />

The interaction of VEGF <strong>and</strong> its receptors regulates tumor angiogenesis. In the current study,<br />

we found that isoliquiritigenin (ISL) reduced VEGF <strong>and</strong> Flt-1 expression in two human breast<br />

cancer cells, MCF-7 <strong>and</strong> MDA-MB-231. ISL also inhibited dose- <strong>and</strong> time- dependently the<br />

proliferation of breast cancer cells in the presence <strong>and</strong> absence of VEGF. ISL completely<br />

reduced VEGF-stimulated expression of two receptor tyrosine kinases in human umbilical<br />

vein endothelial cells (HUVECs). Moreover ISL suppressed VEGF-induced cell proliferation,<br />

migration <strong>and</strong> tube formation in HUVECs. Antiangiogenic activity of ISL was confirmed in<br />

mouse Matrigel plug assay. In conclusion, these results suggest that ISL can be a c<strong>and</strong>idate<br />

agent for the prevention <strong>and</strong> treatment of breast cancer by blocking angiogenesis.<br />

- 426 -


Session X : Cell death in cancer Poster X, 94<br />

Apoptotic effect of celecoxib dependent upon p53 status in human ovarian carcinoma<br />

cells<br />

Yoo Cheol Song1, Su-Hyeong Kim1, Yong Sang Song1,2<br />

1Cancer Research Institute, 2Department of Obstetrics <strong>and</strong> Gynecology, Seoul National<br />

University, College of Medicine Seoul, Korea, E-mail: yssong@snu.ac.kr<br />

Celecoxib, a selective cyclooxygenase-2 inhibitor, induces the apoptosis in various cancers in<br />

COX-2 dependent <strong>and</strong>/or independent manners. The p53 protein is mutated in 50% of all<br />

human tumors <strong>and</strong> plays a key role in apoptosis, cell cycle, <strong>and</strong> the expression of several<br />

proteins. In ovarian cancer, the rate of p53 mutation has been shown to be very high <strong>and</strong><br />

associated with poor prognosis. To explore the importance of functional status of p53 in<br />

apoptosis by celecoxib in ovarian carcinoma cells, the cellular response to celecoxib was<br />

determined in SK-OV3 ovarian carcinoma cells with null type p53 <strong>and</strong> PA-1 with wild type<br />

p53. Our results showed that celecoxib inhibited cell growth more in PA-1 than in SK-OV3.<br />

The underlying antiproliferative mechanism may differ between these two cell types<br />

dependent upon the functional status of p53, which plays integral roles in regulating cell cycle<br />

<strong>and</strong> survival. Higher subG1 was shown in PA-1 than in SK-OV3 in response to celecoxib.<br />

Caspase -8, -9, <strong>and</strong> -3 were activated more in PA-1 cells than in SK-OV3. These results<br />

suggest that death receptor <strong>and</strong> mitochondria-mediated apoptotic pathways may be involved<br />

in celecoxib induced apoptosis irrespective of the functional status of p53. Upregulation of<br />

p21 were shown only in PA-1 cells. In summary, <strong>our</strong> study demonstrated that the celecoxib<br />

effectively inhibited cell growth <strong>and</strong> induced apoptosis in human ovarian carcinoma cells.<br />

However, apoptotic effect by celecoxib seemed to be different dependent upon the functional<br />

status of p53.<br />

- 427 -


Session X : Cell death in cancer Poster X, 95<br />

Effect of paclitaxel on intracellular localization of c-Myc <strong>and</strong> P-c-Myc in prostate<br />

carcinoma cell lines<br />

Rosanna Supino1, Giuditta Cuccuru1, Enrica Favini1, Franco Zunino1, A. Ivana<br />

Scovassi2<br />

1Istituto Nazionale per lo Studio e la Cura dei Tumori, Via Venezian 1, 20133 Milano,<br />

Italy;, E-mail: rosanna.supino@istitutotumori.mi.it; 2Istituto di Genetica Molecolare del<br />

CNR, Via Abbiategrasso 207, 27100 Pavia, Italy, E-mail: scovassi@igm.cnr.it<br />

The proto-oncogene c-myc is involved in multiple cell pathways with opposite effects on cell<br />

outcome of death or of proliferation. It has been proposed that these different roles depend on<br />

its sequestration in cellular compartments <strong>and</strong>/or its phosphorylation. We speculated that<br />

subcellular localization of c-Myc protein <strong>and</strong> of its phosphorylated form (P-c-Myc) could<br />

have a role in the different response to PTX in two prostate carcinoma cell lines, PC3 <strong>and</strong><br />

DU45 cells, which undergo multinucleation or c-myc-dependent apoptosis, respectively. cmyc<br />

is amplified only in PC3, but a similar extent of c-Myc phosphorylation was observed in<br />

both cell lines after PTX treatment. By immunofluorescence we found that PTX-induced<br />

upregulation of c-myc in DU145 cells, not occurring in PC3 cells, cannot be ascribed to<br />

different protein localization, <strong>and</strong> that similar c-Myc <strong>and</strong> P-c-Myc nuclear translocation<br />

occurs in both cell lines after drug treatment. Thus, subcellular localization of c-Myc <strong>and</strong> P-c-<br />

Myc is not crucial in determining the mode of cell death in these prostate carcinoma cell lines.<br />

- 428 -


Session X : Cell death in cancer Poster X, 96<br />

Breast cancer cells response to the antineoplastic agents cisplatin, carboplatin <strong>and</strong><br />

doxorubicin, at the mRNA expression levels of distinct apoptosis-related genes,<br />

including the new member, BCL2L12.<br />

Hellinida Thomadaki <strong>and</strong> Andreas Scorilas<br />

Department of Biochemistry <strong>and</strong> Molecular Biology, Faculty of Biology, University of<br />

Athens, Panepistimioupolis, Zografou, 15 701, Athens, Greece, Email:<br />

elthoma@biol.uoa.gr, ascorilas@biol.uoa.gr<br />

The drugs cisplatin, carboplatin <strong>and</strong> doxorubicin exhibit anticancer activity, the mechanism of<br />

which is not yet completely clarified, although these agents are known to modulate the<br />

expression of several genes including apoptosis-related genes, such as members of the BCL2<br />

family. Recently, BCL2L12, a new member of the BCL2 family, was cloned by <strong>our</strong> group. In<br />

order to define unequivocally the significance of the expression patterns of such genes as a<br />

response to anticancer drug cytotoxic activity, we studied the possible alterations in the<br />

mRNA expression levels of various apoptosis-related genes (BCL2, BAX, BCL2L12,<br />

CASPASE-9, FAS), after cell treatment with distinct anticancer drugs (cisplatin, carboplatin<br />

<strong>and</strong> doxorubicin), in the breast cancer cell line, MCF-7. The kinetics of cell toxicity were<br />

evaluated by the MTT method, whereas the expression levels of distinct apoptosis-related<br />

genes were analysed by RT-PCR, using gene specific primers. The percentage of non-viable<br />

cells was up regulated with increasing concentrations <strong>and</strong> cell exposure time to the different<br />

anticancer drugs. Distinct modulations of apoptosis-related genes, at the mRNA level, were<br />

also observed. Further work is required in order to ascertain whether the mRNA expression<br />

levels of distinct apoptosis-related genes may serve as a new prognostic factor predicting<br />

chemotherapy outcome in breast cancer. Acknowledgements: The present research was<br />

supported by an "EPEAEK II" grant, under the act “PYTHAGORAS I – SUPPORT OF<br />

UNIVERSITY RESEARCH GROUPS”, with co-funding of 75% from the European Social<br />

Fund <strong>and</strong> 25% from National Funds.<br />

- 429 -


Session X : Cell death in cancer Poster X, 97<br />

Effects of gamma <strong>and</strong> proton irradiation on human HTB140 melanoma cell growth,<br />

induction of apoptosis <strong>and</strong> cell cycle redistribution<br />

Danijela V. Todorovic1, Ivan M. Petrovic1, Lela B. Koricanac1, Lucia M. Valastro2,<br />

Luigi Raffaele2, Giacomo Cuttone2, Aleks<strong>and</strong>ra M. Ristic-Fira1<br />

1 Vin(a Institute of Nuclear Sciences, Belgrade, Serbia <strong>and</strong> Montenegro<br />

2 Istituto Nazionale di Fisica Nucleare, LNS, Catania, Italy<br />

E-mail: aristic@vin.bg.ac.yu<br />

Human HTB140 melanoma cells were irradiated with either gamma rays or protons, over the<br />

dose range of 8 to 24 Gy. Cell growth was investigated using microtetrasolium (MTT) <strong>and</strong><br />

sulforhodamine B (SRB) assays 7 days after irradiation, time needed for cell survival<br />

evaluation. With the increase of dose significant growth inhibition for both irradiation<br />

qualities was detected. The same dose level of protons induced stronger inactivation of<br />

HTB140 cells than single irradiation with gamma rays. Cell proliferation, detected by 5bromo-2<br />

-deoxyuridine (BrdU) incorporation assay 7 days after proton <strong>and</strong> gamma<br />

irradiation, has shown highly significant (p


Session X : Cell death in cancer Poster X, 98<br />

Inhibition of cell-free apoptosis in Xenopus oocyte extracts by Src kinase<br />

Alex<strong>and</strong>er A. Tokmakov1, Mikako Shirouzu1, Shigeyuki Yokoyama1,2,3<br />

1Genomic Sciences Center, RIKEN Yokohama Institute, Tsurumi, Yokohama, 230-0045<br />

Japan, 2RIKEN Harima Institute at Spring-8, 3University of Tokyo, Japan. E-mail:<br />

tokmak@gsc.riken.go.jp<br />

Oncoproteins can induce both cell proliferation <strong>and</strong> apoptosis, depending on cellular context.<br />

v-Src oncoprotein has been shown to activate several kinases, which can inhibit apoptosis,<br />

including ERK <strong>and</strong> PI3-K. To further characterize the involvement of Src kinase in apoptosis,<br />

we investigated the effect of Src overexpresion in the cell-free extracts prepared from oocytes<br />

<strong>and</strong> eggs of the African clawed frog, Xenopus laevis. These extracts can recapitulate a range<br />

of apoptotic events, including release of cytochrom C, caspase activation, <strong>and</strong> nuclear<br />

fragmentation (Newmeyer et al., 1994). Notably, interphase-arrested <strong>and</strong> metaphase-arrested<br />

extracts have different susceptibility to undergo apoptosis. The MAPK pathway active in<br />

metaphase-arrested extracts was shown to protect them from apoptosis (Tashker et al., 2002).<br />

In the present study we measured the caspase 3/7 activation <strong>and</strong> cytochrom C release in<br />

apoptotic cell-free Xenopus oocyte <strong>and</strong> egg extracts, overexpressing constitutively active (v-<br />

Src) <strong>and</strong> kinase negative (Src KN) mutants of Src kinase. We found that interphase-arrested<br />

oocyte extracts were highly apoptotic. They released cytochrom C <strong>and</strong> displayed elevated<br />

levels of caspase activity after several h<strong>our</strong>s’-long incubation at room temperature. Caspase<br />

activation in these extracts could be prevented by overexpression of either v-Src or Src KN<br />

protein kinase mutants. No or little effect of Src overexpression on cytochrom C release was<br />

detected. On the other h<strong>and</strong>, metaphase arrested egg extracts were resistant to apoptosis <strong>and</strong><br />

they could not be regulated by Src kinase. Our data suggest that the inhibition of apoptosis in<br />

Xenopus oocyte extracts by Src kinase is not mediated by activation of the MAPK pathway.<br />

- 431 -


Session X : Cell death in cancer Poster X, 99<br />

Hypoxia-induced cell death in human malignant glioma cells: the role of metabolic<br />

pathways<br />

Felix Tritschler, Michael W. Ronellenfitsch, Michael Weller <strong>and</strong> Joachim P. Steinbach<br />

Laboratory of Molecular Neuro-Oncology, Department of General Neurology, Hertie<br />

Institute for Clinical Brain Research, University of Tuebingen, Medical School, Otfried-<br />

Mueller-Str. 27, 72076 Tuebingen, Germany<br />

Presenting author: Tel. +49 7071 2981969, FAX: +49 7071 295260,<br />

E-mail: f.tritschler@gmx.de<br />

Adaptive responses to hypoxia are important determinants of tumor cell survival under the<br />

adverse conditions of the tumor microenvironment. We have previously established a<br />

paradigm of hypoxia-induced cell death of human malignant glioma cells. In this model,<br />

epidermal growth factor receptor (EGFR) inhibition <strong>and</strong> cycloheximide protect glioma cells<br />

from hypoxia by reducing energy consumption <strong>and</strong> maintaining ATP levels <strong>and</strong><br />

mitochondrial integrity. Here, we have examined the impact of glucose <strong>and</strong> amino acid<br />

restriction <strong>and</strong> withdrawal on cellular energy homeostasis <strong>and</strong> cell death.<br />

Restriction of glucose to 2 mM or less was necessary for cell death. Amino acid withdrawal<br />

had a smaller but definite independent influence. Notably, EGFR inhibition <strong>and</strong><br />

cycloheximide were only protective when glucose was present, suggesting that they exert<br />

their protective effect exclusively via reducing glucose consumption. Then, <strong>signaling</strong><br />

cascades downstream of EGFR were examined. Hypoxia suppressed the phosphorylation of<br />

protein kinase B (PKB)/Akt <strong>and</strong> the mammalian target of rapamycin (mTOR) effectors<br />

ribosomal protein S6 (RPS6) <strong>and</strong> eukaryotic initiation factor 4E-binding protein 1. Further,<br />

hypoxia induced phosphorylation of AMP-activated protein kinase (AMPK), a crucial<br />

metabolic regulator downstream of the tumor suppressor kinase LKB-1. The AMPK target<br />

acetyl-CoA carboxylase was also phosphorylated. Activation of AMPK by the synthetic<br />

compound 5-aminoimidazole-4-carboxamide-1-beta-4-ribofuranoside (AICAr), mimicking a<br />

starvation signal, protected glioma cells from hypoxia.<br />

These data delineate how glioma cells generate adaptive metabolic responses <strong>and</strong> suggest<br />

targets for therapeutic interventions.<br />

- 432 -


Session X : Cell death in cancer Poster X, 100<br />

Potential cytoprotective effects of new synthesised sulphur containing compounds<br />

Tzvetomira A. Tzanova1, 3, Ognyan Petrov2, Laurent Brault3, Denyse Bagrel3,<br />

Margarita H. Karaivanova1<br />

1. Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str, 1000 Sofia,<br />

Bulgaria; e-mail: tzvete_tzanova@abv.bg<br />

2. Faculty of Chemistry, University of Sofia"St. Kliment Ohriski", 1 James<br />

B<strong>our</strong>chier Blvd, Sofia 1164, Bulgaria; e-mail: OPetrov@chem.uni-sofia.bg<br />

3. Laboratoire d'Ingénierie Moléculaire et Biochimie Pharmacologique, UFR<br />

SciFA, Université Paul Verlaine - Metz, Campus Bridoux, rue du Général Delestraint,<br />

57070 Metz, France; e-mail: bagrel@univ-metz.fr<br />

Cancer pathogenic mechanisms are associated with DNA damage, oxidative stress, <strong>and</strong><br />

chronic inflammation. Therapies goal to maximizing tum<strong>our</strong> death while minimizing damage<br />

to normal tissues. The delivery of high doses of chemotherapy will increase survival though it<br />

is often limited by the development of severe adverse effects. Numerous studies have<br />

demonstrated that sulphur-containing nucleophiles can antagonize the dose–limiting effects of<br />

alkylating agents. The aim of this study is to assess the chemoprotective potential of seven<br />

new 1-(4-Bromo-2-mercaptophenyl)-3, 4-dimethyl-1, 3-dihydroimidazol-2-one derivatives.<br />

The chemoprotective effect of these synthesized sulphur-containing compounds was<br />

evaluated on HL-60, HL-60/Dox, SKW-3, K-562, EJ <strong>and</strong> MCF-7 malignant cell lines (MTT<br />

assay). F<strong>our</strong> of the tested molecules present an interesting effect since they did not display<br />

cytotoxic effects on cells at concentrations up to 100 µM when used as pre-treatment. They<br />

limit the cell mortality induced by cisplatin on MCF-7 breast carcinoma cell line. As expected<br />

cisplatin treatments resulted in accumulation of cells in S phase. Two compounds, containing<br />

mercaptoacetyl group, did not influence cisplatin effect on the cell cycle (DNA<br />

quantification) indicating that these molecules did not seem to interact directly with cisplatin.<br />

According to these results the oxidative stress was evaluated. A pre-treatment of the cells with<br />

these compounds followed by administration of cisplatin did not affect the level of GSH, the<br />

major intracellular antioxidant (HPLC method). As reactive oxygen species are important<br />

mediators of antineoplastic drugs, we actually focus on the evaluation of antiradical <strong>and</strong><br />

antioxidant activities of the most interesting compounds as well as on their implication in<br />

apoptosis induction.<br />

- 433 -


Session X : Cell death in cancer Poster X, 101<br />

Glycolysis inhibition in cancer cells by redox-cycling compounds<br />

Julien Verrax, Henrik S. Taper <strong>and</strong> Pedro Buc Calderon<br />

Pharmacokinetic, Metabolism, Nutrition <strong>and</strong> Toxicology Unit, School of Pharmacy,<br />

Université Catholique de Louvain, e-mail:julien.verrax@pmnt.ucl.ac.be<br />

Among all the different features of cancer cells, two of them are of particular interest: their<br />

nearly universal glycolytic phenotype <strong>and</strong> their sensitivity towards an oxidative stress, both<br />

resulting presumably from the combination between high anabolic needs <strong>and</strong> environmental<br />

growth conditions lacking O2. Therefore, we took advantage of these features to develop an<br />

experimental approach by exposing cancer cells to an oxidant insult induced by the<br />

association of menadione <strong>and</strong> ascorbate. This association develop a synergistic antitumor<br />

activity in vivo as well as in vitro, based on the potentiation of the menadione redox cycling<br />

by ascorbate. This leads to the intracellular generation of reactive oxygen species among them<br />

H2O2 appears to be the main responsible of the cytolytic effect. Since lactate <strong>and</strong> ATP are<br />

severely <strong>and</strong> rapidly depressed following ascorbate/menadione exposure, we suggest that the<br />

major intracellular event is related to the impairment of glycolysis [Verrax et al, 2005].<br />

Indeed, NAD+ is rapidly consumed, related with a strong Poly(ADP-ribose) polymerase<br />

activation, thus explaining the glycolysis inhibition. The profile of cell death do not<br />

correspond to apoptosis (no caspase-3 nor PARP processing, DNA str<strong>and</strong> breaks with spread<br />

pattern) but is rather close to necrosis [Verrax et al, 2004]. Strong morphological events occur<br />

during the cytolytic process including the formation of cytoplasmic pieces, organelles-free.<br />

On these morphological basis, such a cell death has been called autoschizis, which appears as<br />

a new kind of necrotic cell death [Jamison et al, 2002]. Due to the high energetic dependence<br />

of cancer cells towards glycolysis, the impairment of such an essential pathway may explain<br />

the effectiveness of this association on many tumor cells.<br />

This work was financed by grant from the Belgian Fonds National de la Recherche<br />

Scientifique (FNRS-FRSM Grant 3.4.594.04.F)<br />

- 434 -


Session X : Cell death in cancer Poster X, 102<br />

Influence on Caspase-3 <strong>and</strong> –9 activity by Survivin splice variants in vitro<br />

J Wagener, HE Gabbert, C Mahotka<br />

Institute of Pathology, Heinrich Heine-University, Moorenstr.5, 40225 Düsseldorf,<br />

Germany, jessica.wagener@gmx.de<br />

Objective: One important hallmark of cancer cells is the resistance to apoptosis. Survivin is a<br />

structurally <strong>and</strong> functional unique member of the inhibitor of apoptosis protein (IAP) family<br />

that acts at the interface between apoptosis <strong>and</strong> cell cycle control. In many human cancers<br />

aberrant expression of Survivin correlates with poor prognosis <strong>and</strong> resistance to<br />

chemotherapy. Moreover, one of the main impacts of Survivin is the inhibition of caspases.<br />

Therefore, we analysed the inhibitory potential of the three Survivin splice variants, i.e.<br />

Survivin-deltaEx3, Survivin-2B, <strong>and</strong> Survivin-3B in vitro.<br />

Methods: Transfection of HEK293 cells (with GFP-Survivin, GFP-Survivin-deltaEx3, GFP-<br />

Survivin-2B <strong>and</strong> GFP-Survivin-3B), Western Blotting, Caspase-Assays with purified<br />

recombinant Survivin, XIAP, <strong>and</strong> Caspase-3 <strong>and</strong> –9, <strong>and</strong> cell extracts of transfected HEK293<br />

cells.<br />

Results: 1. Survivin inhibits recombinant Caspase-3 as well as Caspase-9 in a concentration<br />

dependent manner. 2. XIAP enhances the ability of Survivin to inhibit Caspase-3 activity. 3.<br />

In contrast, Survivin does not increase the potential of XIAP to suppress Caspase-9 activity.<br />

4. After transfection with GFP-tagged Survivin, HEK293 cells extracts inhibit recombinant<br />

Caspase-9 but not recombinant Caspase-3. 5. HEK293 cell extracts transfected with GFP-<br />

Survivin-deltaEx3, GFP-Survivin-2B <strong>and</strong> GFP-Survivin-3B fail to inhibit both Caspase-3 <strong>and</strong><br />

-9 activity.<br />

Conclusions: IAPs contribute to the lack of ability of many tumors to undergo apoptosis. Our<br />

findings show that – in contrast to Survivin <strong>and</strong> XIAP – the mainly carboxy-terminal<br />

modified splice variants Survivin-deltaEx3, Survivin-2B, <strong>and</strong> Survivin-3B fail to inhibit<br />

caspases-3 <strong>and</strong> -9 activity in vitro. Therefore, there is a striking evidence for the<br />

carboxyterminal region of Survivin to be involved in caspase inhibition.<br />

- 435 -


Session X : Cell death in cancer Poster X, 103<br />

Dual action of roscovitine on human MCF-7 breast cancer cells: induction of G2 arrest<br />

via inhibition of CDKs <strong>and</strong> initiation of apoptosis by activation of site-specific<br />

phosphorylation of wt p53 protein<br />

Józefa W*sierska-G+dek, Carmen Ranftler <strong>and</strong> Marieta Gueorguieva<br />

Cell Cycle Regulation Group, Division: Institute of Cancer Research, Department of<br />

Medicine I, Medical University of Vienna, Borschkegasse 8 a, A-1090 Vienna, Austria.<br />

E-mail: Jozefa.Gadek-Wesierski@meduniwien.ac.at<br />

<strong>Expo</strong>sure of MCF-7 cells to roscovitine (ROSC), a potent CDK inhibitor, resulted in a strong<br />

inhibition of cell proliferation. Detailed analysis revealed that ROSC arrested MCF-7 cells in<br />

G2 phase of the cell cycle <strong>and</strong> induced apoptosis. Cell cycle arrest preceded the main wave of<br />

apoptosis. The cell cycle block was attributable to the inactivation of CDK2 <strong>and</strong> CDK1. Postincubation<br />

of G2 arrested cells for 24h in a drug-free medium did not diminish the number of<br />

the G2 cell population indicating that ROSC-mediated cell cycle arrest was prolonged. ROSC<br />

induced wt p53 tumor suppressor protein in MCF-7 cells in a time- <strong>and</strong> dose-dependent<br />

manner. ROSC increased p53 steady-state. The half-life of wt p53 protein increased<br />

approximately forty-fold after exposure of MCF-7 cells to ROSC for 15h. At 4h after ROSC<br />

administration a strong phosphorylation of serine at position 46 was observed. The onset of<br />

Ser-46-p53 phosphorylation preceded the induction of apoptosis. P-Ser-46-p53<br />

transcriptionally upregulated p53AIP1 protein, a component of mitochondria. Surprisingly,<br />

the reconstitution with human caspase-3 did not sensitize MCF-7 cells to the action of ROSC.<br />

Our results unequivocally show that ROSC simultaneously inhibits CDK2 <strong>and</strong> CDK1 <strong>and</strong><br />

activates a kinase catalyzing the phosphorylation of p53 protein at Ser46.<br />

- 436 -


Session X : Cell death in cancer Poster X, 104<br />

Protein profiling of different human breast cancer cells lines in response to cisplatin<br />

treatment <strong>and</strong> validation of anti-apoptotic Bcl-2 as a target for enhancing cisplatin<br />

sensitivity<br />

Christina Westmose Yde <strong>and</strong> Olaf-Georg Issinger<br />

Institute of Biochemistry <strong>and</strong> Molecular Biology, University of Southern Denmark,<br />

Campusvej 55, 5230 Odense M, Denmark. E-mail: chriswy@bmb.sdu.dk<br />

Clinical problems in the current treatment of breast cancer patients include severe dosedependent<br />

side effects of the compounds used <strong>and</strong> development of resistance to the treatment.<br />

Therefore, in prospect of improving the current therapy of breast cancer patients it is<br />

important to study the mechanisms behind drug responsiveness. Five different human breast<br />

carcinoma cancer cell lines, MCF-7, MDA-MB-231, MDA-MB-435, HCC-1937 <strong>and</strong> CAL-<br />

148, were analyzed in respect to sensitivity to the chemotherapeutic compound cisplatin <strong>and</strong><br />

dose-dependent drug-induced cell death was measured by reduction in cell viability (WST-1<br />

test) <strong>and</strong> by poly(ADP)ribose polymerase (PARP) cleavage. While high doses of cisplatin<br />

were required in order to induce cell death in MCF-7 <strong>and</strong> MDA-MB-231 the cell lines, MDA-<br />

MB-435, HCC-1937 <strong>and</strong> CAL-148, were sensitive to cisplatin at much lower doses. In order<br />

to identify proteins involved in sensitizing the cells to cisplatin the breast cancer cell lines<br />

were profiled in respect to protein expression levels of a variety <strong>signaling</strong> proteins. In all cell<br />

lines an increase in cyclin E <strong>and</strong> a decrease in cyclin D1 was found after cisplatin treatment.<br />

Furthermore, the mitogen-activated protein kinases, ERK <strong>and</strong> p38, were phosphorylated at<br />

their activating residues when cell were treated with cisplatin. Neither expression of wild type<br />

p53, estrogen receptor status nor activated Akt seemed to be of primary importance in respect<br />

to cisplatin sensitivity, however, an inverse correlation was found between expression of the<br />

anti-apoptotic protein Bcl-2 in the breast cancer cell lines <strong>and</strong> cisplatin sensitivity. Therefore,<br />

RNA interference was used in order to knock down Bcl-2 protein in MCF-7, which expressed<br />

the highest amount of Bcl-2, <strong>and</strong> it was found that Bcl-2 siRNA significantly sensitized MCF-<br />

7 to cisplatin.<br />

Hence, <strong>our</strong> results confirm the anti-apoptotic Bcl-2 as a possible target for increasing the<br />

effect of chemotherapeutic compounds in breast cancer.<br />

- 437 -


Session X : Cell death in cancer Poster X, 105<br />

Significant co-expression of GLUT-1, Bcl-xl <strong>and</strong> Bax in colorectal cancer.<br />

Andrzej Wincewicz, Mariola Sulkowska, Mariusz Koda, Luiza Kanczuga-Koda,<br />

Stanislaw Sulkowski.<br />

Department of Pathology, Medical University of Bialystok, Waszyngtona 13, 15-269<br />

Bialystok, Pol<strong>and</strong>. E-mail: sulek@zeus.amb.edu.pl<br />

Hypoxic cancer cells overexpress GLUT-1 (Glucose transporter 1) to accelerate glucose<br />

intake mainly for low effective, anaerobic respiration, so that they wouldn’t die of oxygen<br />

deficiency. Ischemic cell injury triggers apoptosis. Regulators of cell suicide like Bax <strong>and</strong><br />

Bcl-xl combine their functions to cause apoptosis or to rescue cells from death. GLUT-1, Bax<br />

<strong>and</strong> Bcl-xl are of prognostic significance in colorectal cancer but they have not been<br />

compared, yet. Thus, we aimed to determine eventual correlations between GLUT-1, Bax <strong>and</strong><br />

Bcl-xl in association with different clinicopathological features of colorectal cancer patients.<br />

Expressions of the proteins were evaluated in specimens of 150 colorectal patients by<br />

immunohistochemistry with appliance of specific antibodies. The levels of tissue expressions<br />

were statistically analyzed with Spearman’s correlation test. Similarly to all the patients,<br />

GLUT-1 matched Bcl-xl <strong>and</strong> Bax in statistically significant manner regardless node<br />

involvement, grade of histological differentiation, histopathological type, tumor site, sex <strong>and</strong><br />

age of patients. GLUT-1 correlated highly with Bcl-xl in both T2 <strong>and</strong> T3 tumors<br />

(p


Session X : Cell death in cancer Poster X, 106<br />

Impact of Rb knock down on the modulation erufosine signal transduction in human<br />

leukemia cells<br />

Maya M. Zaharieva1,2, Milen Kirilov2, Minqiang Chai2, Stefan Berger3, Spiro M.<br />

Konstantinov1,2, Martin R. Berger2<br />

1Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str, 1000 Sofia, Bulgaria<br />

2German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg,<br />

Germany<br />

3Central Institute for Mental Help, Department of Molecular Biology, J5, 68 159<br />

Mannheim<br />

The retinoblastoma gene Rb1 is a prototypical tum<strong>our</strong> supressor. Consistent with its tom<strong>our</strong>supressor<br />

function Rb is known to inhibit proliferation. Recent studies indicate its role in<br />

suppressing apoptosis, too. Loss of Rb sensitizes cells to apoptosis, but loss of Rb can only<br />

contribute to tumor development under conditions in which apoptosis response is already<br />

compromised. The new alkylphosphocholine erufosine is an inductor of Rb in leukemic cells.<br />

Therefore, we decided to suppress the expression of RB using conventional phosphorotioate<br />

antisense oligonucleotides <strong>and</strong> siRNAs. Unfortunately, both types of oligonucleotides used<br />

didn’t cause stable <strong>and</strong> reproducible knock-down <strong>and</strong> we initiated viral transfection<br />

experiments for stable Rb knock-down in SKW-3, BV-173 <strong>and</strong> K-562 cells. The antisense<br />

sequence was firstly cloned in the pSUPER vector <strong>and</strong> tested on HEK 293T cells. For<br />

normalizing the transfection efficiency an EGFP-expressing vector was used in addition. To<br />

realize the transfer of the H1-shRNA cassette directed to the Rb-mRNA, the cells were<br />

infected with LentiLox virus which co-express an EGFP-protein. The resulting levels of target<br />

mRNA were measured by real-time RT-PCR after excluding of non-fluorescent cells by flow<br />

cytometry <strong>and</strong> cell sorting. The highest decrease of the Rb-mRNA (about 86%) was found in<br />

SKW-3 cells. A concomitent knock-down at the protein level was evidenced by Western<br />

blotting. The proliferation activity of the infected cells was estimated by colorimetric MTTdye<br />

reduction assay before <strong>and</strong> after erufosine treatment. Comparison of Rb knock-down <strong>and</strong><br />

wild type leukemic cells revealed the presence of significant differences in cell cycle duration<br />

<strong>and</strong> sensitivity to erufosine, thus confirming the impact of Rb modulation for the mode of<br />

action of alkylphosphocholines which are known as promising antineoplastic signal<br />

transduction modulators.<br />

- 439 -


Session X : Cell death in cancer Poster X, 107<br />

Upregulation <strong>and</strong> phosphorylation of p53 <strong>and</strong> its related proteins after treatment with<br />

valproic acid in MOLT-4 leukemia cells<br />

Darina Zá)kodová1, Martina ,ezá(ová1, Ji-ina Vávrová J2, Doris Vokurková3, Ale)<br />

Tich.2<br />

1Inst. of Medical Biochemistry <strong>and</strong> 3Inst. of Clin. Immunology <strong>and</strong> Allergology, Univ.<br />

Hospital <strong>and</strong> Faculty of Medicine in Hradec Králové, Charles Univ. in Prague, Czech<br />

Republic, 2Dept. of Radiobiology, Faculty of Military Health Sciences Hradec Králové,<br />

Univ. of Defense Brno, Czech Republic, E-mail: zaskodovad@lfhk.cuni.cz<br />

Objective: Our work consists in evaluating changes of protein expression in MOLT-4<br />

leukemia cells after treatment with valproic acid, a histone deacetylases inhibitor. Inhibitors<br />

of HDAC significantly affect transcription of genes involved in the regulation of cell cycle,<br />

apoptosis <strong>and</strong> DNA synthesis by changes of histone acetylation <strong>and</strong> therefore by changes of<br />

tertiary structure of DNA. The cell cycle arrest or apoptosis can be induced by tumor supresor<br />

protein p53. Activated p53 binds to DNA <strong>and</strong> induces transcription of appropriates genes<br />

including p21, bax, MDM2 <strong>and</strong> cyclin G. Materials <strong>and</strong> methods: Apoptosis has been<br />

measured by flow-cytometry. Western blotting has been used for determination of histone<br />

acetylation <strong>and</strong> expression of p53, MDM2 <strong>and</strong> p21. Results: VA in low concentrations (2 <strong>and</strong><br />

4 mM) causes elevated expression of p53 <strong>and</strong> p53 phosphorylation at serin 392 in a period<br />

from 2 to 4 h<strong>our</strong>s of treatment. This is followed (in the case of 2mM VA) by the expression of<br />

protein p21. Concurrently with p53 upregulation occurs the MDM2 phosphorylation at serine<br />

166. The cleavege of lamin B as an apoptosis control proceeds from 24 h<strong>our</strong>s after 4mM VA<br />

treatment. When the cells were incubated three days with VA, EC50 determined by<br />

clonogenic survival assay was 1.76 mM. In the case of continuous exposure (14 days) to VA<br />

EC50 decreased to 0.625 mM. The percentage of apoptic cells determined with flowcytometric<br />

analysis (subG1 peak) increased to 37% after 72 h<strong>our</strong>s of treatment with 4 mM<br />

VA. Conclusion: Our results show that after treatement by valproic acid the amount of active<br />

p53 increases, which is related to increased inactivation of MDM2. VA in 2 mM<br />

concentration causes primarily the cell cycle arrest via p21, but in 4 mM concentration leads<br />

the cells directly to apoptosis. This work was supported by The Ministry of Education of<br />

Czech Republic, project MSM 0021620820<br />

- 440 -


Session X : Cell death in cancer Poster X, 108<br />

Targeting the cell cycle <strong>and</strong> the PI3K pathway: a universal strategy to reactivate innate<br />

tumor suppressor programmes in cancer cells?<br />

Thérèse David-Pfeuty1*, Michel Legraverend2 , Odile Ludwig2, <strong>and</strong> David S. Grierson2<br />

Institut Curie-Recherche, 1UMR 146 <strong>and</strong> 2UMR 176 du CNRS, Bâtiment 110, Centre<br />

Universitaire, 91405 Orsay Cedex, France<br />

A rationale for why corruption of the Rb <strong>and</strong> p53 modules facilitates tumorigenesis is because<br />

it lends oncogene-bearing cells a surfeit of Cdk activity <strong>and</strong> growth enabling them to<br />

elaborate strategies to evade tumor suppressor mechanisms <strong>and</strong> inappropriately divide. Thus,<br />

a potentially universal means to palliate their deficiency in cancer cells would be to target<br />

both the activity of Cdks <strong>and</strong> their PI3K module that is a central regulator of cell growth. This<br />

hypothesis is supported by <strong>our</strong> observation that the killing efficacy of Cdk inhibitors,<br />

roscovitine <strong>and</strong> 16 other purine derivatives, decreased with increasing corruption of the Rb<br />

<strong>and</strong> p53 modules. Notably, the two most resistant tumor cell lines were (Rb-/p53-) Saos-2,<br />

followed by HBL100 (in which Rb <strong>and</strong> p53 are inactivated by binding to the large T antigen<br />

of SV40); conversely, the most sensitive cell lines were (Rb+/p53*) HaCaT <strong>and</strong> A431 that<br />

display normal differentiation features in culture, suggestive of a robust Rb pathway, followed<br />

by (Rb+/p53+) MCF-7 <strong>and</strong> SCC15 tumor cell lines that carry a faulty Rb pathway. A further<br />

case has been provided by the observation that the potentiation of roscovitine-induced<br />

apoptosis by the PI3K inhibitor, LY294002, also decreased with increasing corruption of the<br />

Rb <strong>and</strong> p53 modules. Another important finding of <strong>our</strong> work is that purines differing by a<br />

single substitution, which exerted little lethal effects on distant cell types (differing by their<br />

Rb/p53 status <strong>and</strong> tumorigenic potential) grown on a rich medium, could display widelydiffering<br />

cytotoxicity profiles toward the same cell types grown on a poor medium. This<br />

highlights that structurally related compounds targeting the same Cdks may also target unique<br />

proteins or Cdks that may control unique biological functions or compete for the interaction<br />

between the compounds <strong>and</strong> their therapeutically relevant Cdk targets. The range <strong>and</strong><br />

concentration of such crossreacting targets would depend on the cell translational capacity<br />

that, in turn, would depend on the cell genotype. In the perspective of clinical development in<br />

association with inhibitors of the PI3K pathway, it might be advised, then, to select tumor cell<br />

type-selective Cdk inhibitors on the basis of their cytotoxicity in cell-based assays performed<br />

at a limiting serum concentration, sufficient to suppress their interaction with undesirable<br />

crossreacting targets.<br />

- 441 -


Notes<br />

- 442 -


Notes<br />

Notes<br />

- 443 -


- 444 -


Session XI : Cell death <strong>and</strong> cardiovascular diseases<br />

- 445 -


Session XI : Cell death <strong>and</strong> cardiovascular diseases Poster XI, 1<br />

Role of phospho-p42/44 MAPK in the altered reactivity of vascular smooth muscle cells<br />

during experimental cirrhosis.<br />

Antonia Alcaraz, David Iyu, Noemí M. Atucha, Joaquín García-Estañ, María C. Ortiz.<br />

Dpto. Fisiología, Fac. Medicina, Universidad de Murcia, Spain. E-mail: clara@um.es<br />

High levels of nitric oxide (NO) <strong>and</strong> oxidative stress (OS) appear as important contributors in<br />

the altered vascular function of liver cirrhosis. In fact, administration of NO synthesis<br />

inhibitors or antioxidants improves or corrects this alteration although the underlying<br />

mechanisms by which that happen are not yet clear. NO <strong>and</strong> OS derived products are<br />

regulatory molecules in <strong>signaling</strong> pathways influencing the contractile function of vascular<br />

smooth muscle cells (VSMC). OS can activate the MAPK (mitogen-activated protein kinase)<br />

<strong>signaling</strong> pathway, which can be interdependent with other pathways (PKB, NF-kB)<br />

implicated in the intracellular regulation of NO levels. Thus, in this study we evaluated the<br />

role of phospho-p42/44 MAPK in the altered reactivity of VSMC in cirrhotic rats by chronic<br />

bile duct ligation (CBDL; 3 weeks). For that, we examined the effect of the antioxidant<br />

vitamin E (VitE; 5000 IU/kg diet) or/<strong>and</strong> the NO synthesis inhibitor, L-NAME (10-4 M,<br />

acutely), on the vasoconstrictor responses to phenylephrine (PE) in aortic rings of CBDL rats.<br />

Besides, we studied the activation of p42/44 MAPK in primary culture of aortic VSMC of<br />

CBDL <strong>and</strong> control rats (3-5 pass), before or after adding a NO-donor with sodium<br />

nitroprusside (SNP; 200-1000 µM) or stimulating OS with deoxycholic acid (DOXCA; 50-<br />

200 µM). To assess the role of NO <strong>and</strong> OS on MAPK activation, we pre-incubated the cells<br />

with L-NAME or VitE (300-1000 µM). Results: aortic rings of CBDL rats (treated or not with<br />

VitE) responded less to PE than controls <strong>and</strong> the addition of L-NAME abolished this<br />

hyporesponsiveness. Aortic phospho-p42/44 MAPK expression tended to increase in CBDLs<br />

(100±10, 117±7 <strong>and</strong> 133±23 %, in control, CBDL <strong>and</strong> CBDL+VitE, respectively) without<br />

difference between groups. Cultured VSMC from CBDLs showed higher activation of p42/44<br />

MAPK than controls <strong>and</strong> L-NAME <strong>and</strong> VitE significantly decreased it. SNP <strong>and</strong> DOXCA<br />

increased dose-dependently the activation of p42/44 MAPK in both groups of cells, but at<br />

higher degree in controls at lower doses. In conclusion, VSMC from CBDL rats showed a<br />

MAPK activation sensitive to NO <strong>and</strong> OS. This altered intracellular <strong>signaling</strong> might<br />

contribute to specific functional changes in VSMC <strong>and</strong> thus to the abnormal vascular<br />

response in cirrhosis.<br />

- 446 -


Session XI: Cell death <strong>and</strong> cardiovascular diseases Poster XI, 2<br />

Greater storage calcium capacity but lower capacitative calcium entry in platelets of<br />

rats with biliary cirrhosis.<br />

Noemí M. Atucha, F. Ramírez, Esther García, Dolores Robles, David Iyú, Antonia<br />

Alcaraz, María C. Ortiz, Joaquín García-Estañ.<br />

Dpto de Fisiología, Facultad de Medicina, 30100 Murcia, Spain. E-mail: ntma@um.es<br />

It is known that liver cirrhosis shows a tendency to bleeding, which is in part due to a<br />

functional alteration of platelets. Platelet stimulation with physiological agonists such as ADP<br />

produces an increase in cytoplasmic calcium levels ([Ca 2+ ] c ), due to the release from internal<br />

stores <strong>and</strong> to the entry stimulated by its depletion (capacitative entry). In platelets there are<br />

two types of calcium stores <strong>and</strong> they can be separetely studied by analyzing their sensitivity to<br />

calcium pump inhibitors, thapsigargin (TG) <strong>and</strong> 2,5-di-(tert-butyl)-1,4-hydroquinone<br />

(TBHQ). In this work, we have analyzed the role of these two types of stores in the mediation<br />

of the changes in [Ca 2+ ] c produced by capacitative calcium entry. Methods: the experiments<br />

have been performed in washed platelets, loaded with fura-2, obtained from cirrhotic (bile<br />

duct ligation) or control rats. Fluorescence has been measured in a spectrofluorimeter,<br />

following a well established method. Capacitative calcium entry was studied, after the<br />

depletion of internal stores in zero-calcium conditions, by adding 0.5 mM external calcium or<br />

by fura-2 quenching in the presence of extracellular Mn 2+ , measured at 360 nm. Results: the<br />

elevations in extracellular calcium were greater with TBHQ than with TG, which indicates<br />

that the stores sensitive to TBHQ are bigger than those sensitive to TG. In both cases,<br />

platelets of BDL rats showed greater calcium elevations than those of the controls, suggesting<br />

a greater storage capacity in these rats. A bigger calcium storage was also confirmed with<br />

ADP or thrombin which, in the absence of external calcium, produced greater calcium<br />

elevations in platelets from BDL rats. After partial depletion with TBHQ or total depletion<br />

with TBHQ+TG, capacitative calcium entry was always significantly lower in platelets from<br />

BDL rats. Platelet aggregation in response to ADP was also lower in BDL rats. These results<br />

indicate that capacitative calcium entry, in response to store depletion, is altered in platelets<br />

from rats with experimental cirrhosis. This altered response can produce lower calcium levels<br />

in response to agonists acting through the depletion of internal calcium stores. This alteration<br />

can be related to the aggregation defects showed by the platelets of the cirrhotic rats.<br />

- 447 -


Session XI: Cell death <strong>and</strong> cardiovascular diseases Poster XI, 3<br />

Role of urocortin in early myocardial pre- <strong>and</strong> postconditioning<br />

Barbara Cserepes, Boglarka Racz, Balazs Gasz, Andrea Ferencz, Maria Kurthy, Janos<br />

Lantos, Gabor Jancso, Erzsebet Roth<br />

Department of Surgical Research <strong>and</strong> Techniques, Medical Faculty, University of Pecs,<br />

Hungary. E-mail: barbicska@yahoo.co.uk<br />

Pre- <strong>and</strong> postconditioning are powerful endogenous adaptive phenomenon of the organism<br />

whereby different stimuli (hypoxia, drugs, hypotermia) enhance the tolerance against various<br />

types of stress. Urocortin, member of the corticotrophin-releasing factor family has potent<br />

effects on the cardiovascular system including vasodilation, increases in coronary blood flow<br />

<strong>and</strong> cardiac contractility, <strong>and</strong> induces protection against ischaemia/reperfusion damage.<br />

Recent studies reported that urocortin has similar cardioprotective effects as the ischaemic<br />

preconditioning in both early <strong>and</strong> delayed phases. The aim of this study was to investigate the<br />

effect of urocortin on cultured cardiomyocytes in the process of pre– <strong>and</strong> postconditioning.<br />

Isolated neonatal rat ventricular myocytes were preconditioned with adenosine, simulated<br />

ischaemia <strong>and</strong> urocortin (10 minutes treatment followed by 10 minutes<br />

reperfusion/resolution). For detecting the effect of alternative types of preconditioning,<br />

necrosis enzyme (LDH) release, vital staining (trypan blue) <strong>and</strong> ratio of apoptosis/necrosis<br />

were examined after cardiac cells were exposed to 3 h<strong>our</strong>s sustained ischaemia <strong>and</strong> 2 h<strong>our</strong>s<br />

reperfusion. Same parameters were measured in the postconditioned groups (30 or 60 minutes<br />

ischaemia followed by postconditioning with 10 min ischaemic stimulus or urocortin <strong>and</strong> 2<br />

h<strong>our</strong>s reperfusion). Cells exposed to 3 h ischaemia followed by 2 h reperfusion were shown as<br />

control.<br />

Our results show, that LDH release <strong>and</strong> the ratio of necrotised cells was decreased, but<br />

proportion of alive/dead myocytes was increased in all preconditioned groups compared with<br />

control group. In postconditioned groups LDH content of culture medium, number of trypan<br />

blue-stained cells was reduced, rate of apoptotic/necrotic cells was raised contrasted with non<br />

postconditioned group, <strong>and</strong> the difference was more expressed in urocortin treated group after<br />

60 min ischaemia.<br />

We can conclude that urocortin might play an important role in myocardial cytoprotection.<br />

Preconditioning with urocortin induced such a powerful cell protective effect as adenosine<br />

<strong>and</strong> ischaemia. Furthermore, postconditioning with urocortin after 60 min ischaemia was<br />

more cardioprotective than ischaemic postconditioning. Supported by the Hungarian<br />

Scientific Research Foundation (OTKA) T-048851, T-038035, F046593.<br />

- 448 -


Session XI: Cell death <strong>and</strong> cardiovascular diseases Poster XI, 4<br />

MAPKs are activated via cPLA2 during ischaemia/reperfusion induced injury in<br />

neonatal cardiomyocytes.<br />

Anna-Mart Engelbrecht <strong>and</strong> Am<strong>and</strong>a Lochner.<br />

Departments of Physiological Sciences <strong>and</strong> Medical Physiology, University of<br />

Stellenbosch, Stellenbosch, 7600, South Africa. E-mail: ame@sun.ac.za<br />

Myocardial ischaemia/reperfusion (I/R) injury induces death of a significant number of<br />

cardiomyocytes <strong>and</strong> contributes to mortality. Although it has become increasingly clear that<br />

myocardial I/R result in the death of myocytes through apoptosis, the molecular basis of this<br />

process remains to be elucidated. Therefore, the aim of this study was to investigate the role<br />

of cPLA2 in MAPK phosphorylation <strong>and</strong> caspase-3 cleavage in I/R-induced apoptosis in<br />

neonatal cardiomyocytes.<br />

Cultured neonatal cardiomyocytes were exposed to various inhibitors for 30 minutes before<br />

the onset of 60 minutes of simulated ischaemia (induced with deoxyglucose <strong>and</strong> KCN)<br />

followed by 30 minutes of reperfusion. Samples were analysed by Western blotting with<br />

phospho-specific antibodies recognizing total- <strong>and</strong> phosphorylated p38 MAPK, ERK <strong>and</strong><br />

cPLA2. Apoptosis was determined by poly(ADP-ribose)polymerase cleavage <strong>and</strong> caspase-3<br />

activation. Hoechst 33342 stain was used to view the morphological features of apoptosis by<br />

fluorescence microscopy. Cell viability was measured by the MTT assay.<br />

Inhibition of cPLA2 with AACOCF3 significantly improved cell viability during SI/R (60.17<br />

± 1.77 to 80.17 ± 1.97%, p


Session XI: Cell death <strong>and</strong> cardiovascular diseases Poster XI, 5<br />

The effect of different angiotensin converting enzyme inhibitors on subcellular toxicity<br />

of paraquat in isolated rat liver mitochondria<br />

Mahmoud Ghazi-Khansari, Mir Jamal Hossieni , Ali Mohammadi- bardbori<br />

Department of Pharmacology, School of Medicine, Tehran University of Medical<br />

Sciences, Tehran, I.R. Iran. Email: ghazikha@sina.tums.ac.ir<br />

Widespread use of the herbicide paraquat in the recent years has necessitated further studies<br />

on the mechanisms of its toxicity <strong>and</strong> metabolism. The defect in electron transfer chain of<br />

mitochondria by paraquat is linked to free radical formation. In the present study we<br />

compared the abilities of different angiotensin converting enzyme inhibitor, captopril (a thiol<br />

ACEi), enalapril, <strong>and</strong> lisinopril (two nonthiol ACEi) on mitochondria toxicity due to paraquat.<br />

The rat liver mitochondria were first isolated by centrifuge (at 4°C at speed of 7000g) in a<br />

mixture of 0.25M saccharose solution <strong>and</strong> 0.05M Triss buffer. Various concentrations of<br />

paraquat (1, 5, 10µM), enalapril (0.05, 0.5, 0.0.25µM), lisinopril (0.05, 0.01, 0.1µM) <strong>and</strong><br />

captopril (0.5, 0.08, 0.1µM) on the mitochondria isolated from the liver with respect to time<br />

were investigated. Paraquat at concentration of 5µM was determined to be significantly<br />

different compared to control (p


Session XI: Cell death <strong>and</strong> cardiovascular diseases Poster XI, 6<br />

SMAD <strong>and</strong> AP1 are crucial for apoptosis induction in cardiomyocytes<br />

J. Heger, D. Schneiders, HM Piper, G Euler-Taimor<br />

Physiologisches Institut, Justus-Liebig Universität, Gießen, Germany E-mail:<br />

jacqueline.heger@physiologie.med.uni-giessen.de<br />

In adult cardiomyocytes both apoptosis <strong>and</strong> hypertrophy are mediated by the transcription<br />

factor AP-1. SMAD proteins that are only activated by TGF# under apoptotic conditions are<br />

able to interact with AP-1 <strong>and</strong> may be c<strong>and</strong>idates which direct AP-1 activation towards<br />

apoptosis. To test this hypothesis we infected isolated ventricular cardiomyocytes (CM) with<br />

adenovirus encoding Smad4 (AdSmad4) <strong>and</strong> analyzed its impact on apoptosis induction in<br />

comparison to TGF#.<br />

TGF# enlarged apoptosis (11,3 ± 0,9 % vs. controls 7,3 ± 0,7 % (n=10, p


Session XI: Cell death <strong>and</strong> cardiovascular diseases Poster XI, 7<br />

Atrial appendages transcriptional profile in atrial fibrillation patients with structural<br />

heart diseases.<br />

Maria S. Kharlap, Angelica V.Timofeeva, Lyudmila E. Goryunova, George L.<br />

Khaspekov, Andrey V. Skamrov, Sergey L. Dzemeshkevich, Renat S. Akchurin, Sergey<br />

P.Golitsyn <strong>and</strong> Robert Sh. Beabealashvilli.<br />

Department of Clinical Electrophysiology/Laboratory of Genetic Engineering, Russian<br />

Cardiology Research <strong>and</strong> Development Complex, 3rd Cherepkovskaya str. 15a, 121552,<br />

Moscow, Russia. E-mail: kharlapmaria@yahoo.com<br />

During the last few years several investigations devoted to gene expression analysis in human<br />

atria in patients with atrial fibrillation (AF) have been performed. The subject of those studies<br />

was atrial tissue from the patients undergoing open heart surgery with sinus rhythm <strong>and</strong> with<br />

AF. Such kind of investigations were limited by the presence of structural changes associated<br />

with underlying heart disease in control atrial tissue samples.<br />

In this regard, it seemed to be reasonable to compare the atrial tissue samples from AF<br />

patients with those from subjects without structural heart diseases. For this purpose, we<br />

employed cDNA microarray technique. Tissue samples of right atrial appendages (RAA)<br />

were obtained from 12 AF patients undergoing open heart surgery with valve disease,<br />

myxoma <strong>and</strong> coronary artery disease. Control group included 11 autopsy RAA tissue samples<br />

from people with no signs of cardiovascular diseases died during the accidents. Gene<br />

expression was analyzed using Human cDNA Expression Arrays from Clontech (total 4704<br />

genes). The results were verified by reverse transcription polymerase chain reaction. 15 genes<br />

were found to be down-regulated in AF patients independently of the type of the structural<br />

heart disease <strong>and</strong> ranged from 1.2 to 28 fold (p


Session XI: Cell death <strong>and</strong> cardiovascular diseases Poster XI, 8<br />

Effects of selenium deficiency on the protein expression of tissues of the cardiovascular<br />

system<br />

A. Kyriakopoulos., A. Richter., C. Wolf., A. Plotnikov., I. Grbavac., D. Behne<br />

Hahn-Meitner-Institut, Dept. “Molecular Trace Element Research in the Life Sciences”<br />

Glienicker Str. 100, D-14109 Berlin, Germany.<br />

Organs of the cardiovascular system such as Heart <strong>and</strong> aorta contain a lot of mitochondria<br />

(powerhouse of the cell) in which the reactive oxygen species (ROS) are generated. ROS are<br />

damaging the cell. Antioxidantive system protects the cell against the aggressive ROS <strong>and</strong><br />

inflammation. Some components of the antioxidative defence system are also metalloenzymes<br />

<strong>and</strong> selenoenzymes. In order to examine the expression of the heart <strong>and</strong> aorta proteins (part of<br />

them) in the homogenate of selenium deficient (Se-) <strong>and</strong> selenium control rats (Se+), 2 D<br />

electrophoresis was used by means of giga gels (30x35 cm). After the electrophoresis the<br />

protein-expression pattern of the (Se-)-gel <strong>and</strong> (Se+)-gel were compared. The evaluation of<br />

the protein difference was implemented by means of a computer program suitable for the<br />

analysis of protein separated by the two-dimensional electrophoresis. In this way more than<br />

2000 proteins a gel (heart) were detected <strong>and</strong> more than 1900 protein spots were detected in<br />

the aorta fraction. Ten significant differences were found between the gel of (Se+) <strong>and</strong> (Se-)-<br />

heart of the rat <strong>and</strong> more than 15 significant differences between the gel of (Se+) <strong>and</strong> (Se-) of<br />

the aorta. By means of MALDI-MS/ESI-MS some of these proteins with the different<br />

expression level were determined until now. Of those three proteins were detected as the<br />

alpha myosin heavy chain (alpha-MHC), myosin light chain 1 <strong>and</strong> 2 (MLC 1 <strong>and</strong> 2) <strong>and</strong> the<br />

mitochondrial enzyme creatinin Kinase. First results suggest that selenium deficiency effects<br />

myocardial energy metabolism <strong>and</strong> contractile proteins.<br />

- 453 -


Session XI: Cell death <strong>and</strong> cardiovascular diseases Poster XI, 9<br />

Transient <strong>and</strong> sustained oxidative stress cause differential JNK/SAPK <strong>and</strong> c-Jun<br />

activation in rat cardiac myocytes<br />

+nastasia Pechtelidou, Catherine Gaitanaki, <strong>and</strong> Isidoros Beis<br />

Department of Animal <strong>and</strong> Human Physiology, School of Biology, Faculty of Sciences,<br />

University of Athens, Panepistimioupoli, Athens, 157 84, Greece<br />

email: ibeis@biol.uoa.gr<br />

The aim of the present study was to examine the hypothesis that oxidative stress induces cell<br />

death in H9c2 cell line (rat embryonic cardiac myocytes), <strong>and</strong> to determine whether signalling<br />

via JNKs/SAPKs may be involved. We examined the activation of JNKs/SAPKs in response<br />

to increasing concentrations of H2O2 (0.04-1 mM), both for the transient <strong>and</strong> for the<br />

sustained oxidative stress. Sustained H2O2 stimulation (0.4 mM) significantly induces<br />

maximal phosphorylation of JNKs at 2h, while transient H2O2 stimulation for 5 min induces<br />

a maximum phosphorylation of JNKs at 15min after withdrawal. At 2 h<strong>our</strong>s of sustained<br />

stimulation peak phosphorylation of the transcription factor c-Jun is also observed, while at<br />

the same time significant levels of nuclear activated JNKs are detected. SP600125, the JNK<br />

inhibitor II (0.025 mM), abolishes JNK activation induced by 0.4 mM H2O2, as shown by the<br />

absence of phosphorylated c-Jun at 2 h<strong>our</strong>s of exposure. For the purpose of measuring cell<br />

viability, we used the MTT method. Our results reveal that sustained H2O2 stimulation<br />

significantly decreases cell viability, while transient stress does not. Moreover, from the<br />

antioxidants tested, only catalase inhibits cell death induced by oxidative stress. Preliminary<br />

experiments showed increased Hoechst staining after 24 h<strong>our</strong>s of sustained exposure, but<br />

normal nuclear morphology after 5min of stress <strong>and</strong> 24h withdrawal. In addition, the same<br />

pattern of cell morphology is observed under the optical microscope. These results<br />

demonstrate an apoptotic type of death under oxidative stress. Based on the above results, we<br />

conclude that in H9c2 cells the transient <strong>and</strong> sustained activation of the JNKs/SAPKs<br />

pathway may be differentially involved in cellular signalling during oxidative stress-induced<br />

apoptosis.<br />

(This study was funded by O.P.E.I.V.T II)<br />

- 454 -


Session XI: Cell death <strong>and</strong> cardiovascular diseases Poster XI, 10<br />

RoY a Novel Synthetic Peptide with Angiogenic <strong>and</strong> anti Apoptotic properties<br />

1Annat Raiter, 1 Chana Weiss, 1 Orly Kudasi, 2Alex<strong>and</strong>er Battler <strong>and</strong> 1 Britta Hardy.<br />

1Felsenstein Medical Research Center, Tel-Aviv University School of Medicine,<br />

2Cardiology Dept., Rabin Medical Center, Petah-Tikva, Israel.<br />

Email: bhardy@post.tau.ac.il<br />

Background: We have identified a novel 12 amino-acid synthetic peptide, RoY, selected<br />

from a phage display peptide library by screening against endothelial cells under hypoxia.<br />

RoY peptide specifically binds endothelial cells, induces angiogenesis in vitro manifested by<br />

proliferation, migration <strong>and</strong> tube formation.<br />

Objectives: to evaluate the therapeutic potential of the synthetic RoY peptide in restoring<br />

perfusion in a mouse hind limb ischemic model <strong>and</strong> to elucidate the mechanism of RoY<br />

peptide angiogenic activity under hypoxia by gene expression.<br />

Results: Ischemia was created by ligation <strong>and</strong> excision of the femoral artery in one hind limb<br />

of C57BL mice. Seven days after operation, perfusion in the ischemic leg was reduced to<br />

50% in control PBS group, to 65% in RoY peptide group <strong>and</strong> to 70% in the VEGF group.<br />

However, while RoY peptide increased perfusion to 99% at 21 days, perfusion was reduced to<br />

80% with VEGF <strong>and</strong> remained unchanged with PBS. Gene array, confirmed by RT-PCR,<br />

revealed up-regulation of angiogenesis growth factors Angiopoitin-2 <strong>and</strong> bFGF, Upregulation<br />

of chemokine CCL20 <strong>and</strong> of the anti-apoptotic gene TNF-RSF10D. In contrast,<br />

VEGF gene expression in endothelial cells incubated with RoY peptide for 3 or 24 h<strong>our</strong>s was<br />

not up-regulated. The percent of apoptosis, measured by FACS analysis, of endothelial cells<br />

incubated under hypoxic conditions was inhibited by the addition of RoY peptide from 62%<br />

to 33%.<br />

Conclusion: RoY is a synthetic peptide that exhibits angiogenic <strong>and</strong> anti-apoptotic properties<br />

under hypoxia by up regulation of growth factors, cytokines <strong>and</strong> anti apoptotic genes. RoY<br />

peptide activity is VEGF independent thus suggests a way to bypass classical VEGF<br />

angiogenic system. Treatment of ischemic leg with RoY peptide improved perfusion <strong>and</strong><br />

walking <strong>and</strong> climbing function in the mouse. This novel peptide with pro-angiogenic<br />

properties may be used as a new therapeutic modality to hypo-vascular diseases.<br />

- 455 -


Session XI: Cell death <strong>and</strong> cardiovascular diseases Poster XI, 11<br />

Gene expression profiling in peripheral blood leukocytes from patients with arterial<br />

hypertension<br />

Angelica V.Timofeeva, Lyudmila E. Goryunova, George L. Khaspekov, Dmitrii A.<br />

Kovalevskii, Andrey V. Skamrov, Olga S. Bulkina <strong>and</strong> Robert Sh. Beabealashvilli.<br />

Russian Cardiology Reseach <strong>and</strong> Development Complex, 3rd Cherepkovskaya str. 15a,<br />

121552, Moscow, Russia. E-mail: Angelica_T@cardio.ru<br />

The pathogenesis of hypertension is a multifactorial process that involves the interaction of<br />

genetic <strong>and</strong> environmental factors. In varying degrees, abnormalities of volume regulation,<br />

enhanced vasoconstriction, <strong>and</strong> remodeling of the arterial wall contribute to the development<br />

of hypertension. A wide variety of interdependent physiologic systems have been found to<br />

influence blood pressure. Among these systems are baroreceptors, natriuretic peptides, the<br />

renin-angiotensin-aldosterone system, the kinin-kallikrein system, the adrenergic receptor<br />

system, <strong>and</strong> factors produced by blood vessels that cause vasodilation or contraction. As these<br />

physiologic systems interact in complex fashion, it is difficult to establish the primary<br />

abnormalities underlying blood pressure elevation for each studied patient. Since blood can be<br />

considered as a connecting-link between all these systems, we hypothesized that circulating<br />

blood cells may carry disease-specific information because of alterations in their local<br />

environment. In this regard, we employed cDNA microarray technology to reveal differences<br />

in gene expression in peripheral blood leukocytes from patients with arterial hypertension<br />

(AH) comparing with normal individuals. Microarray results were confirmed by semiquantitative<br />

RT-PCR. We have identified a gene expression pattern that accurately<br />

distinguished AH patients from normal volunteers <strong>and</strong> was the same among AH patients<br />

independently of the AH stage <strong>and</strong> the presence or absence of pharmacotherapy. We could<br />

identify 13 AH-specific genes significantly activated compared with control group <strong>and</strong> 3<br />

genes expressed at reduced level in AH. These genes are reffered to the cystein protease,<br />

tyrosine phosphatase, thiol-disulfide oxidoreductase <strong>and</strong> chemokine receptor families,<br />

transcription factors <strong>and</strong> heat shock proteins. Unfortunately, we cannot conclude from <strong>our</strong><br />

findings whether the changes in peripheral leukocytes gene expression are cause or<br />

consequence of the pressure elevation in the AH, but we can speculate about the possible<br />

implications of selected group of genes in vascular pathophysiology occurring in AH.<br />

- 456 -


Notes<br />

- 457 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases<br />

- 458 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 1<br />

MDMX is a Critical Regulator of Neuronal Apoptosis induced by APP Stimulat Session<br />

Samir Benosman1, Koji Okamoto2, Christian Gaiddon3 <strong>and</strong> Jean-Philippe Loeffler4<br />

1,3,4 INSERM U692, School of Medicine, Louis Pasteur University; 11 Humann St.,<br />

Strasb<strong>our</strong>g, France.<br />

2 National Cancer Center; Tokyo, Japan<br />

Emails: 1 benosmans@neurochem.u-strasbg.fr ; 3 gaiddon@neurochem.u-strasbg.fr ;<br />

4 loeffler@neurochem.u-strasbg.fr<br />

MDMX is an analogue of MDM2 that has been shown to modulate p53 function in various<br />

cell lines. In the case of neurons, data regarding the implication of MDMX in the p53<br />

apoptotic pathway are still lacking. We investigated the role of MDMX in primary immature<br />

<strong>and</strong> mature Cerebellar Granule Neurons undergoing different stresses leading to cell death. To<br />

that aim, we used DNA damaging agents including Neocarzinostatin (NCS) <strong>and</strong> Cisplatin<br />

(CIS), <strong>and</strong> neuron-specific stresses such as low K+ (LK) <strong>and</strong> APP stimulation. In All<br />

conditions we obtained a caspase-activating apoptosis. p53 protein was stabilized in all<br />

conditions except LK. Furthermore, apoptosis caused by DNA damage, but not by LK, was<br />

suppressed by the dominant-negative mutant of p53, indicating that cell death caused by LK<br />

is p53-independent. In contrast, E2F-1, another pro-apoptotic factor known to interact with<br />

MDMX, was stabilized in response to LK <strong>and</strong> poorly in the remaining conditions.<br />

Concomitantly, MDMX protein levels decreased in all conditions, LK included. The loss of<br />

MDMX was not due to transcriptional repression since MDMX mRNA levels remained stable<br />

in the former conditions. On the other h<strong>and</strong>, protease inhibitors reversed the loss of MDMX<br />

suggesting a regulation of protein stability during neuronal insults. Furthermore, overexpression<br />

of MDMX inhibited the transcriptional activity of both p53 <strong>and</strong> E2F-1, <strong>and</strong><br />

partially restored neuronal viability following apoptotic treatments. Taken together, <strong>our</strong> data<br />

show that MDMX is an antiapoptotic factor in neurons in which MDMX degradation is<br />

induced by multiple stress signals, allowing activation of p53 <strong>and</strong> E2F-1 during neuronal<br />

apoptosis.<br />

Supported by Association Francaise contre les Myopathies (AFM) <strong>and</strong> Association p<strong>our</strong> la<br />

Recherche sur le Cancer (ARC)<br />

- 459 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 2<br />

Age-related decline of synaptic turnover as an early event in neurodegeneration<br />

Carlo Bertoni-Freddari, Patrizia Fattoretti, Belinda Giorgetti, Yessica Grossi, Marta<br />

Balietti, Tiziana Casoli, Giuseppina Di Stefano, *Gemma Perretta<br />

Neurobiology of Aging Laboratory, INRCA Research Department, Via Birarelli 8,<br />

60121 Ancona, Italy Email: c.bertoni@inrca.it <strong>and</strong> *Istituto di Neurobiologia e<br />

Medicina Molecolare-CNR, S.p Anguillarese km 1.3, 00060 Roma, Italy<br />

The changes of synaptic ultrastructure in the frontal (FC) <strong>and</strong> temporal (TC) cortex were<br />

investigated in adult <strong>and</strong> aged monkeys (Macaca fascicularis), by means of quantitative<br />

morphometry, in order to assess the potential role of age-related synaptic deterioration in<br />

neurodegeneration. The average synaptic size (S), the synaptic numeric density (Nv: number<br />

of synapses/cubic micron of tissue), the synaptic surface density (Sv: overall area of synaptic<br />

junctional zones/cubic micron of tissue) <strong>and</strong> the number of synapses/neuron (Syn/Neur) were<br />

semiautomatically measured by a computer-assisted image analysis system. The results of FC<br />

evaluation revealed no significant differences of Nv <strong>and</strong> Sv between adult <strong>and</strong> old monkeys,<br />

while S was significantly increased in the aged animals. In TC, Sv did not show changes due<br />

to age, while Nv was significantly decreased <strong>and</strong> S was significantly increased in aged<br />

monkeys. The fraction (%) of perforated junctions was similar in FC <strong>and</strong> TC of the adult<br />

animals, while it was decreased by 22.6% in TC vs. FC in the old monkeys. A percent<br />

distribution of S showed that the fraction of enlarged synapses (>0.20 square micron) was<br />

higher in TC than in FC regardless of the age of the animals (21.3% vs.16.9% in adult <strong>and</strong><br />

33.9% vs. 26.0% in aged monkeys, respectively). The comparison between data from adult<br />

<strong>and</strong> old TC revealed that the increase was markedly higher in aged monkeys (i.e. 33.9% vs.<br />

21.3%). In these animals, Syn/Neur was significantly decreased by 14.1% in TC, whereas it<br />

was not significantly reduced in FC (- 4.4%). Sv, Nv, S <strong>and</strong> Syn/Neur are morphometric<br />

parameters currently adopted to estimate the ongoing rearrangements of synaptic<br />

ultrastructure to react to environmental stimuli. Our findings provide evidence of an agerelated<br />

decline of synaptic plasticity in the brain of aged monkeys that is significant in TC.<br />

According to current literature data on synaptic structural dynamics, this decay might<br />

represent an early <strong>and</strong> subtle alteration able to trigger the development of senile plaques <strong>and</strong><br />

neurodegenerative events.<br />

- 460 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 3<br />

Early ultrastructural alterations of synaptic mitochondria in ischemic delayed neuronal<br />

death<br />

Carlo Bertoni-Freddari, Patrizia Fattoretti, Tiziana Casoli, Giuseppina Di Stefano,<br />

Moreno Solazzi, *Elisa Perna, *Clara De Angelis<br />

Neurobiology of Aging Laboratory, INRCA Research Department, Via Birarelli 8,<br />

60121 Ancona, Italy. Email: c.bertoni@inrca.it, *Morphometry Laboratory, Sigma-Tau,<br />

00040 Pomezia, Italy<br />

The effect of transient global ischemia (20 minutes) on the ultrastructural features of synaptic<br />

mitochondria at the distal dendrites of CA1 hippocampal neurons was investigated in 3month-old<br />

rats. Sham surgery was carried out on animals of the same age used as controls.<br />

The number of mitochondria/cubic micron of neuropil (Nv: numeric density) was measured<br />

by means of the disector counting method. On the same organelles sampled by the disector we<br />

measured: the mitochondrial average size (average volume: V), the mitochondrial longer<br />

diameter (Fmax) <strong>and</strong> the overall fraction of neuropil occupied by mitochondria (volume<br />

density: Vv). In ischemic rats, a 10% not significant decrease of Nv was found, V increased<br />

not significantly by 11% <strong>and</strong> Fmax increased not significantly by 5% vs. controls. No<br />

significant difference was found between the two groups of rats as regards Vv. The percent<br />

distribution of V showed that in ischemic animals the population of CA1 synaptic<br />

mitochondria was composed by an increased fraction of oversized organelles while the<br />

percent distribution of Fmax showed that this enlargement was due to an increased percent of<br />

elongated organelles. According to current literature data, the increase in mitochondrial size<br />

may represent a physiological compensatory response to balance the numeric loss of<br />

organelles, however, it remains to be proven whether this compensation is of functional<br />

significance. Although not significant, because of the high plasticity of mitochondrial<br />

ultrastructure in the young CNS, the alterations found by us may play an early <strong>and</strong> subtle role<br />

in triggering necrotic/apoptotic processes reported to affect selectively CA1 neurons<br />

following ischemia. Mild metabolic impairments <strong>and</strong> the peculiar rake-like vasculature of the<br />

hippocampal formation may be reasonably supposed to be responsible for the marked<br />

vulnerability of these neurons that are also characterized by extended dendritic trees.<br />

- 461 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 4<br />

Preservation of mitochondrial volume homeostasis at the early stages of age-related<br />

synaptic deterioration<br />

Carlo Bertoni-Freddari, Patrizia Fattoretti, Belinda Giorgetti, Yessica Grossi, Marta<br />

Balietti, Tiziana Casoli, Giuseppina Di Stefano, *Gemma Perretta<br />

Neurobiology of Aging Laboratory, INRCA Research Department, Via Birarelli 8,<br />

60121 Ancona, Italy Email: c.bertoni@inrca.it <strong>and</strong> *Istituto di Neurobiologia e<br />

Medicina Molecolare-CNR, S.p Anguillarese km 1.3, 00060 Roma, Italy<br />

A morphometric study on synaptic mitochondria was performed in the frontal (FC) <strong>and</strong><br />

temporal (TC) cortex of adult (mean age: 10.6 years) <strong>and</strong> aged (mean age: 20.8 years)<br />

monkeys (Macaca fascicularis). In order to identify ultrastructural alterations due to age, the<br />

average mitochondrial size (average volume: V), the overall volume covered by mitochondria<br />

(volume density: Vv) <strong>and</strong> the number of mitochondria per cubic micron of tissue (numeric<br />

density: Nv) were measured. Either in FC <strong>and</strong> TC no significant age-related differences were<br />

revealed for any of the above mentioned morphometric parameters. Namely, in FC of aged<br />

monkeys a decrease of Nv (6%) <strong>and</strong> Vv (2%) was observed, whereas V showed an increase<br />

by 5%. In TC of aged animals, both Nv <strong>and</strong> Vv increased by 7% <strong>and</strong> V was decreased by 2%.<br />

While the observed changes in FC are in general agreement with data previously reported on<br />

age-related changes in mitochondrial ultrastructure with reference to TC an opposite trend<br />

was shown. Mitochondria are very plastic organelles capable of consistent rearrangements of<br />

their morphology in reaction to different environmental stimuli. Accordingly, V, Nv <strong>and</strong> Vv<br />

account for changes in single aspects of mitochondrial ultrastructure, nonetheless, when<br />

considered together per experimental group, they might represent an index of the structural<br />

rearrangements occurring on discrete pools of organelles (in this study, those located at the<br />

synaptic terminals). On the basis of these assumptions, the present findings document a<br />

preservation of the mitocondrial volume homeostasis in the brain of aged monkeys. Since <strong>our</strong><br />

data from a previous investigation on the same animals showed early signs of synaptic<br />

deterioration in FC <strong>and</strong> TC during aging, this seems to be in contrast with the results obtained<br />

in the present study. However, an age-related preservation of the mitochondrial potential for<br />

structural dynamics may be interpreted as a reactive response to an altered synaptic function.<br />

We suggest that mitochondria might represent sensitive targets for therapeutic interventions<br />

aimed at counteracting early events of synaptic critical alterations able to trigger the<br />

pathogenesis of neurodegenerative diseases.<br />

- 462 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 5<br />

Immunohistochemical evidence for impaired neuregulin-1 <strong>signaling</strong> in the prefrontal<br />

cortex in schizophrenia but not in affective disorders<br />

Iris Bertram1, Hans-Gert Bernstein1, Uwe Lendeckel2, Alicja Bukowska2, Henrik<br />

Dobrowolny1, Gerburg Keilhoff3, Christian Mawrin4 ,Peter Falkai5 <strong>and</strong> Bernhard<br />

Bogerts1<br />

1 Exp. Psychiatry Lab., Psychiatry, University of Magdeburg, Magdeburg, Germany;<br />

2 Exp. Int. Med. lab., Internal Medicine, University of Magdeburg, Magdeburg,<br />

Germany; 3 Cell Tissue Lab., Med. Neurobiology, University of Magdeburg,<br />

Magdeburg, Germany; 4 Exp. Neuropathology Lab., Neuropathology, University of<br />

Magdeburg, Magdeburg, Germany; 5 Department of Psychiatry, University of<br />

Saarl<strong>and</strong>, Homburg; E-mail: Hans-Gert.Bernstein@medizin.uni-magdeburg.de<br />

In the CNS neuregulin-1 (NRG-1) proteins function in neuronal migration, differentiation <strong>and</strong><br />

survival of oligodendrocytes. The NRG-1 gene codes for at least 15 different isoforms, which<br />

may be classified on the basis of their molecular structure. At least two different haplotypes of<br />

the NRG-1 gene may be associated with schizophrenia. An abnormal expression pattern of<br />

NRG-1 mRNA was found in the prefrontal cortex of schizophrenic patients in comparison to<br />

controls. We here show that the NRG-1alpha isoform is significantly reduced in white matter<br />

of the prefrontal cortex in schizophrenia but not in affective disorders. Brains were obtained<br />

from pathologists in accordance rules outlined by German law <strong>and</strong> the local ethics<br />

commission. We studied brains of 22 schizophrenics, 12 patients with affective disorders (7<br />

unipolar <strong>and</strong> 5 bipolar) <strong>and</strong> 22 matched controls. NRG-1alpha immunoreactive material was<br />

detected with a polyclonal antiserum against the synthetic peptide from alpha-type EGF-like<br />

domain of human neuregulin (Ab-3, Neomarkers). When stereologically analysing the<br />

number of NRG-1alpha expressing neurons we found a statistically significant reduction of<br />

these cells in the white, but not in the gray, cortical matter in schizophrenia but not in<br />

depression. The demonstrated decreased number of NRG-1alpha immunoreactive neurons in<br />

brains of schizophrenics point to an important role of this NRG-1 splice variant in<br />

neuropsychiatric disorders. The diminished expression of NRG-1alpha in interstitial white<br />

matter neurons strongly supports an early neurodevelopmental component to schizophrenia.<br />

Supported by NBL-3 of the Ministry of Research of Germany, <strong>and</strong> Stanley Foundation.<br />

- 463 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 6<br />

Contribution of RhoGTPases to calcium dependent activation of p38 <strong>and</strong> excitotoxic cell<br />

death<br />

Jiong Cao, Maria M. Semenova, Anu M.J. Mäki-Hokkonen <strong>and</strong> Michael J. C<strong>our</strong>tney/<br />

Molecular Signalling Lab, A. I. Virtanen Institute, University of Kuopio, Kuopio FIN<br />

70211, Finl<strong>and</strong>. E-mail : c<strong>our</strong>tney@messi.uku.fi /<br />

Excitotoxic neuronal death contributes to many neurological disorders. It results from<br />

excessive stimulation of glutamate receptors <strong>and</strong> involves both calcium influx <strong>and</strong> stressactivated<br />

protein kinases (SAPKs), though the relationship between these events is poorly<br />

defined. RhoGTPases are major regulators of SAPK activation but their contribution to<br />

excitotoxic cell death has not been reported. Indirect evidence implicates Rac/cdc42<br />

RhoGTPases in neuronal death subsequent to NGF withdrawal, whereas RhoA is involved in<br />

inhibition of neurite regeneration <strong>and</strong> release of amyloidogenic A#42 peptide. We find that<br />

stimulation of glutamate receptors leads to activation of RhoGTPases in primary cultured<br />

neurons cultured neurons. This activation is calcium-dependent <strong>and</strong> contributes to the rapid<br />

glutamate-induced activation of p38 <strong>and</strong> the ensuing neuronal death that we find is dependent<br />

on this kinase. However, RhoGTPases alone are not sufficient to induced cell death, revealing<br />

that requirements in addition to the GTPase-mediated p38 activation exist for excitotoxic cell<br />

death to proceed. These observations reveal RhoGTPases as novel <strong>and</strong> essential components<br />

of the excitotoxic cell death pathway.<br />

- 464 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 7<br />

Protein domain interactions as a target for inhibition of excitotoxic p38 stress-activated<br />

protein kinase activation <strong>and</strong> neuronal cell death: the PSD95–nNOS interface./<br />

Jiong Cao, Lotta E. Parviainen, Anu M.J. Mäki-Hokkonen, Jenni I. Viholainen <strong>and</strong><br />

Michael J. C<strong>our</strong>tney/<br />

Molecular Signalling Lab, A. I. Virtanen Institute, University of Kuopio, Kuopio FIN<br />

70211, Finl<strong>and</strong>. E-mail : c<strong>our</strong>tney@messi.uku.fi /<br />

The stress-activated protein kinase p38 <strong>and</strong> nitric oxide (NO) are proposed downstream<br />

mediators of excitotoxicity, a mechanism of neuronal cell death common to both acute<br />

conditions such as cerebral ischaemia <strong>and</strong> chronic neurodegenerative diseases. The<br />

postsynaptic density protein PSD95 can recruit a wide range of effectors, such as the calciumdependent<br />

neuronal NO synthase (nNOS), to the calcium-permeable channel of the NMDA<br />

subtype of glutamate receptor. Depletion <strong>and</strong> displacement of PSD95 from NMDA receptors<br />

reportedly inhibits excitotoxicity, but the multifunctional nature of the PSD95 interactome<br />

makes it hard to deduce from this the precise mechanism of neuroprotection provided by<br />

targeting PSD95. The possibility that selective uncoupling of nNOS from PSD95 might be<br />

neuroprotective has not been previously explored. The relationship between excitotoxic<br />

stress–generated NO <strong>and</strong> activation of p38, <strong>and</strong> the significance of the PSD95–nNOS<br />

interaction to p38 activation also remain unclear. We find that NOS inhibitors reduce both<br />

glutamate-induced p38 activation <strong>and</strong> the resulting neuronal death, whereas NO donor has<br />

effects consistent with NO as an upstream regulator of p38 in glutamate-induced cell death.<br />

Experiments using a panel of decoy constructs targeting interactions between NMDA<br />

receptors, PSD95 <strong>and</strong> nNOS suggest that the PSD95–nNOS interaction <strong>and</strong> subsequent NO<br />

production are critical for glutamate-induced p38 activation <strong>and</strong> the ensuing cell death, <strong>and</strong><br />

demonstrate that the PSD95–nNOS interface provides a genuine possibility for design of<br />

neuroprotective drugs with increased selectivity.<br />

- 465 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 8<br />

Release of ß-amyloid from high-density platelets: implications for Alzheimer’s disease<br />

pathology<br />

Tiziana Casoli, Giuseppina Di Stefano, Belinda Giorgetti, Yessica Grossi, Marta Balietti,<br />

Patrizia Fattoretti, Carlo Bertoni-Freddari<br />

Neurobiology of Aging Laboratory, INRCA Research Department, Via Birarelli 8,<br />

60121 Ancona, Italy. Email: t.casoli@inrca.it<br />

The main component of Alzheimer’s disease senile plaques in the brain is amyloid-beta<br />

peptide (Abeta), a proteolytic fragment of the amyloid precursor protein (APP). Platelets<br />

contain both APP <strong>and</strong> amyloid-beta peptide <strong>and</strong> many evidence suggest that these cells may<br />

represent a useful tool to study both amyloidogenic <strong>and</strong> non-amyloidogenic pathways of APP<br />

processing. It has been demonstrated that platelets activated by physiological agonists such as<br />

thrombin <strong>and</strong> collagen specifically secrete amyloid-beta peptide ending at residue 40. To<br />

verify whether APP beta-processing could be observed also in an in vitro system of highly<br />

concentrated platelets, we measured the Abeta released in the incubation media of 5x109<br />

platelets/ml by enzyme-linked immunosorbent assay. The activation status of platelets was<br />

investigated by ultrastructural analysis. We found that Abeta40 levels were significantly<br />

higher in incubation media of 5x109/ml platelets in comparison with 108/ml platelets<br />

(normalized values), while Abeta42 levels were not affected by cell density. The<br />

ultrastructural analysis showed platelets at different phases of activation: some platelets were<br />

at earlier stage, characterized by granule swelling <strong>and</strong> dilution, others had granules<br />

concentrated in a compact mass in the cell centres within constricted rings of circumferential<br />

microtubules (later stage). Normally concentrated cells had the characteristic morphology of<br />

resting platelets. Our data suggest that high-density platelets undergo activation likely by<br />

increased frequency of platelet-platelet collisions. This, in turn, determines the activation of<br />

APP beta-processing with consequent release of Abeta40. Investigating the biochemical<br />

pathways triggering amyloid-beta peptide secretion in platelets could provide important<br />

informations for developing tools to modulate this phenomenon in AD brains.<br />

- 466 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 9<br />

c-Jun N-terminal protein kinase signalling mediates lovastatin-induced neuroblasts<br />

apoptosis.<br />

María I. Cerezo-Guisado1, Luis J. García-Marín2, Alberto Álvarez-Barrientos3, Eva<br />

Pérez-Lorenzo1, Ricardo Argent1, María J. Bragado1 <strong>and</strong> María J. Lorenzo1.<br />

1Departamento de Bioquímica y Biología Molecular y Genética, 3Departamento de<br />

Fisiología, Facultad de Veterinaria, Universidad de Extremadura, Cáceres. 2Centro<br />

Nacional de Investigaciones Cardiovasculares, Madrid. Spain. E-mail:<br />

mjlorenzo@unex.es<br />

We have previously shown that lovastatin, a competitive HMG-CoA reductase inhibitor,<br />

induces apoptosis in spontaneously immortalized rat brain neuroblasts. Many studies have<br />

implicated the c-Jun N-terminal protein kinase (JNK) pathway as a key regulator of apoptosis.<br />

The aim of this study was to investigate the role for JNK in neuroblasts apoptosis induced by<br />

lovastatin. Treatment of neuroblasts with lovastatin increased JNK activity in a concentration-<br />

<strong>and</strong> time-dependent manner. The activation of JNK preceded the induction of apoptosis, <strong>and</strong><br />

JNK activity remained elevated for at least 24 hr. Lovastatin also increased c-Jun<br />

phosphorylation <strong>and</strong> AP-1 DNA-binding activity in a concentration- <strong>and</strong> time-dependent<br />

manner. Lovastatin effects were fully prevented by mevalonate, the final product of HMG-<br />

CoA reductase. To examine whether the activation of JNK is involved in the process of<br />

lovastatin-induced neuroblasts apoptosis, we utilized JNK inhibitor I, a specific inhibitor for<br />

JNK. JNK inhibition prevented the morphological changes, the appearance of<br />

internucleosomal DNA fragmentation, the increase in the percentage of apoptotic neuroblasts<br />

<strong>and</strong> the amount of the activated form of caspase-3 elicited by lovastatin. In all the<br />

experiments tested, JNK inhibitor effects were concentration-dependent. Taken together,<br />

these data suggest that lovastatin may induce neuroblasts apoptosis by activating the JNKdependent<br />

cell death pathway.<br />

This work has been supported by Grants, SAF 2001-0154 (MCYT) <strong>and</strong> 2PR01B007 (JUEX).<br />

M. I. Cerezo-Guisado is supported by a doctoral fellowship from Junta de Extremadura.<br />

- 467 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 10<br />

Attenuation of A#-induced apoptosis of plant extract (Shaengshik) mediated by the<br />

inhibition of mitochondrial dysfunction <strong>and</strong> anti-oxidative effect<br />

Chu-Yue Chen 1 , Jung-Hee Jang 1 , Mi Hyun Park 2 , Sung Joo Hwang 2 , Young-Joon Surh 1 ,<br />

Ock Jin Park 3<br />

1 College of Pharmacy, Seoul National University, Seoul, Korea<br />

2 Eromlife R &D center Eromlife Cooporation, Korea<br />

3 Department of Food <strong>and</strong> Nutrition, Hannam University, 133 Ojeong-dong Daedeok-gu,<br />

Daejeon 306-791, Korea, E-mail: ojpark@hannam.ac.kr<br />

Recently, considerable attention has been focused on dietary manipulation of oxidative <strong>and</strong>/or<br />

nitrosative damage on neuronal cells. In this study, a neuroprotective effect of plant<br />

(Shaengshik) extracts was investigated. Rat pheochromocytoma (PC12), cells treated with<br />

A#-amyloid underwent apoptotic death as determined by positive in situ terminal endlabeling<br />

(TUNEL staining), decreased mitochondrial transmembrane potential, <strong>and</strong> elevated<br />

caspase-3 activity co-occurring with enhanced MDA accumulation <strong>and</strong> the reduction of GSH<br />

levels. Shaengshik pretreatment attenuated A#-amyloid-induced apoptosis in PC12 cells<br />

possibly by inhibiting mitochondrial dysfunction <strong>and</strong> exerting antioxidant properties.<br />

Shaengshik pretreatment inhibited the loss of mitochondrial membrane potentials <strong>and</strong> reduced<br />

the activation of caspase-3. The in vitro antioxidant activities of Shaengshik extracts were<br />

verified by the DPPH method <strong>and</strong> superoxide dismutase (SOD) mimetic activity. In A#amyloid-challenged<br />

PC 12 cells, Shaengshik prevented the production of ROS, decreased the<br />

level of MDA, <strong>and</strong> elevated GSH. The potential of Shaengshik as one of the neuroprotective<br />

regimens has been suggested through this study, <strong>and</strong> the combination with defined<br />

pharmaceuticals or other dietary antioxidants may provide a better therapeutic or preventive<br />

advantage for the management of Alzheimer diseases.<br />

- 468 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 11<br />

Selenium supplementation acting through the induction of specific selenoproteins<br />

protects microglial cells from damage by hydrogen peroxide<br />

Lisa Dalla Puppa1, Nicolai E. Savaskan2, Anja U. Bräuer3, Dietrich Behne1 <strong>and</strong><br />

Antonios Kyriakopoulos1.<br />

1Hahn-Meitner-Institute, Department of Molecular Trace Element Research in the Life<br />

Sciences, Glienicker Str. 100, 14109 Berlin, Germany (e-mail: dallapuppa@hmi.de)<br />

2Division of Cellular Biochemistry, The Netherl<strong>and</strong>s Cancer Institute (NKI),<br />

Plesmanlaan 121, 1066 CX Amsterdam, The Netherl<strong>and</strong>s<br />

3Institute of Cell Biology <strong>and</strong> Neurobiology, Center for Anatomy Charité-University<br />

Medical School Berlin, Phillipstr. 12, 10115 Berlin, Germany<br />

Activation of glia has been observed in numerous central nervous system pathologies, like<br />

brain infections, neurodegenerative diseases, inflammation, ischemia <strong>and</strong> normal aging. A<br />

hallmark of such pathologies is the activation of microglia cells (the resident macrophage of<br />

the brain) that produce neurotoxic factors such as cytokines, reactive oxygen species (ROS),<br />

nitric oxide, which contribute to neuronal injury.<br />

In the present study, we demonstrate that the trace element selenium, added as sodium<br />

selenite, repressed the damage induced by hydrogen peroxide (H2O2) on microglial BV2<br />

cells. Selenium is known to play a critical role in the central nervous system in addition to<br />

acting as an essential nutrient for general body functions. Selenium is specifically<br />

incorporated as the amino acid selenocysteine into numerous selenoproteins which are mainly<br />

involved in antioxidant processes <strong>and</strong> redox status. It could therefore be assumed that<br />

selenium, added as selenite, inhibits microglial activation via the expression of specific<br />

selenoproteins. Here we report that selenium has a protective effect on cell viability of BV2<br />

treated with H2O2. It reduces also the intracellular ROS production <strong>and</strong> inhibits the induced<br />

apoptosis. We further applied 75Se-selenite in order to assess the expression of selenoproteins<br />

<strong>and</strong> we used siRNA technology to identify protective selenoproteins in microglia.<br />

This project was supported by the German Research Council DFG: Priority Program<br />

Selenoproteins 1087.<br />

- 469 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 12<br />

Altered subcellular distribution of the Alzheimer’s Amyloid Precursor Protein under<br />

stress conditions<br />

Sara C. T. S. Domingues, Ana Gabriela Henriques, Edgar F. da Cruz e Silva <strong>and</strong> Odete<br />

A. B. da Cruz e Silva<br />

Center for Cell Biology, University of Aveiro, 3810-193 Aveiro, Portugal (E-mail:<br />

odetecs@bio.ua.pt)<br />

Altered metabolism of the Alzheimer’s Amyloid Precursor Protein (APP) appears to be a key<br />

event in the pathogenesis of Alzheimer’s Disease <strong>and</strong> both altered phosphorylation <strong>and</strong><br />

oxidative stress appear to affect the production of the toxic Abeta fragment. Our results show<br />

that altered processing of APP was observed under conditions of stress induced by sodium<br />

azide in the presence of 2-deoxy-D-glucose (2DG). As previously reported, the production of<br />

sAPP (the secreted fragment of APP) was inhibited. Using APP-GFP fusion proteins, we<br />

show that 2DG causes the accumulation/delay of APP in the ER/Golgi. This effect was<br />

augmented in the presence of sodium azide. APP subcellular distribution was also affected<br />

<strong>and</strong> changes could also be monitored at the plasma membrane. The involvement of protein<br />

phosphorylation in APP subcellular localization was reinforced by the effect of drugs such as<br />

PMA (phorbol 12-myristate 13-acetate), since in the presence of 2DG <strong>and</strong> PMA APP was<br />

completely absent from the membrane. Thus, the hypothesis that APP is processed in a<br />

phosphorylation-dependent manner <strong>and</strong> that this may be of clinical relevance appears to hold<br />

true even under stress conditions. Our results provide evidence for a role of protein<br />

phosphorylation in APP trafficking under stress conditions <strong>and</strong> contribute to the<br />

underst<strong>and</strong>ing of the molecular basis of Alzheimer’s Disease.<br />

Supported by the EU VI Framework Program, FCT <strong>and</strong> CBC.<br />

- 470 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 13<br />

Mitochondrial metabolic competence in the early stages of neuronal cell death<br />

Patrizia Fattoretti, Carlo Bertoni-Freddari, *Rina Recchioni, Belinda Giorgetti, Marta<br />

Balietti, Yessica Grossi, Moreno Solazzi, Tiziana Casoli, Giuseppina Di Stefano,<br />

*Fiorella Marcheselli<br />

Neurobiology of Aging Laboratory <strong>and</strong> *Centre of Cytology, INRCA Research<br />

Department, Via Birarelli 8, 60121 Ancona, Italy Email: p.fattoretti@inrca.it<br />

A quantitative morphometric study has been carried out in human neuroblastoma SKNBE<br />

cells to evaluate the ultrastructural features <strong>and</strong> the metabolic efficiency of mitochondria<br />

involved in the early steps of apoptosis. In mitochondria from control <strong>and</strong> apoptotic cells<br />

cytochrome oxidase (COX) activity was estimated by preferential cytochemistry. Number of<br />

mitochondria (numeric density: Nv) <strong>and</strong> volume fraction occupied by mitochondria/cubic<br />

micron of cytoplasm (volume density: Vv) <strong>and</strong> average mitochondrial volume (V) were<br />

calculated on both COX positive <strong>and</strong> negative organelles. The ratio (R) of the cytochemical<br />

precipitate area (CPA) to the overall area of each mitochondrion (MA) was evaluated on COX<br />

positive organelles to estimate the inner mitochondrial membrane fraction actively involved<br />

in cellular respiration. Following apoptotic stimulus, the whole mitochondrial population<br />

showed a significant increase of Nv <strong>and</strong> Vv, while V was significantly decreased. In COX<br />

positive organelles higher values of Nv were found, V appeared significantly reduced <strong>and</strong> Vv<br />

was unchanged. R was increased at a not significant extent in apoptotic cells. COX positive<br />

mitochondria accounted for 21% <strong>and</strong> 35% of the whole population in control <strong>and</strong> in apoptotic<br />

cells, respectively. These findings document that in the early stages of apoptosis the increased<br />

numeric density of mitochondria of small size provides an adequate amount of ATP for<br />

progression of the programmed cell death process. In turn, an increased fraction of small <strong>and</strong><br />

more efficient mitochondria appears to represent a reactive response to the loss of<br />

metabolically-impaired organelles. A better underst<strong>and</strong>ing of the mitochondrial role in<br />

neuronal apoptosis may suggest potential interventions to prevent the extensive nerve cell<br />

death typical of neurodegenerative diseases.<br />

- 471 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 14<br />

The role <strong>and</strong> regulation of BH3-only proteins in arsenite-induced apoptosis of cortical<br />

neurons – a dominant role for Puma<br />

Michael Fricker, Hon K. Wong, Aviva M. Tolkovsky<br />

Department of Biochemistry, University of Cambridge, Tennis C<strong>our</strong>t Road, Cambridge,<br />

CB2 1QW, UK<br />

Email: mf309@cam.ac.uk; <strong>and</strong>uswong@brain.riken.jp; amt@mole.bio.cam.ac.uk<br />

Increasing evidence points towards apoptosis as a major mechanism of developmental <strong>and</strong><br />

neurodegenerative cell death in the central nervous system. Proapoptotic BH3-only Bcl-2<br />

family members are thought to act as sentinels of the cell, with different BH3-only proteins<br />

recruited by discrete signalling pathways. We previously demonstrated that sodium arsenite<br />

caused apoptosis of cortical neurons <strong>and</strong> induced expression of Bim - via a JNK/cJundependent<br />

mechanism - as well as Puma <strong>and</strong> Noxa - via a partially p53-dependent<br />

mechanism.<br />

Using neurons cultured from knockout (KO) mice we investigated the relative importance of<br />

three BH3-only proteins, Bim, Noxa <strong>and</strong> Puma, in causing arsenite-induced apoptosis. Whilst<br />

genetic ablation of Bim <strong>and</strong> Noxa provided little protection against arsenite-induced<br />

apoptosis, deletion of Puma imparted complete resistance to arsenite treatment despite<br />

continued upregulation of Bim <strong>and</strong> Noxa in these neurons. Puma deletion also protected<br />

cortical neurons from numerous DNA damaging agents. As Puma has been previously shown<br />

to be regulated at the transcriptional level by p53, we examined the effect of p53 deletion on<br />

arsenite-induced death <strong>and</strong> Puma upregulation. Although p53 KO neurons were completely<br />

protected against DNA damaging agents, the ability of arsenite to induce apoptosis was only<br />

partially impaired in the absence of p53, as was its ability to cause upregulation of both Puma<br />

mRNA <strong>and</strong> protein levels. Potential c<strong>and</strong>idates for p53-independent Puma upregulation<br />

include the p53 homologue p73. A number of p53/p73 target genes were upregulated in the<br />

absence of p53 including CHOP/gadd153, a proapoptotic transcription factor that has also<br />

been linked to Puma regulation. In summary, <strong>our</strong> data establish Puma as an important <strong>and</strong><br />

dominant inducer of apoptosis in cortical neurons, <strong>and</strong> we show that at least in the case of<br />

arsenite, p53-independent mechanisms of Puma upregulation are active.<br />

- 472 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 15<br />

Involvement of cathepsin S in amyloid precursor protein-mediated neuronal cell death<br />

Carole HONIGMANN, Corinne MBEBI, Luc DUPUIS, Jean-Philippe LOEFFLER <strong>and</strong><br />

Yves LARMET.<br />

INSERM U-692, Laboratoire de Signalisations Moléculaires et Neurodégénérescence,<br />

Université Louis Pasteur, Faculté de Médecine, Strasb<strong>our</strong>g, France<br />

E-mails : honigmann@neurochem.u-strasbg.fr, mbebi@neurochem.u-strasbg.fr,<br />

larmet@neurochem.u-strasbg.fr, loeffler@neurochem.u-strasbg.fr,<br />

The Amyloid Precursor Protein (APP), the precursor of beta-amyloid, plays a central role in<br />

Alzheimer's disease (AD) since mutations in the APP gene are associated with early onset<br />

AD. The biological function of APP is however still not elucidated. APP is a transmembrane<br />

proteine <strong>and</strong> there is now accumulating evidence that APP operates as a transduction unit. We<br />

have previously reported that antibody binding to the cell surface APP (APP-Ab) causes<br />

neuronal cell death via a GTP binding protein G0 that forms complexes with the APP<br />

cytoplasmic domain (Mbebi et al., 2002). Anatomopathological studies revealed increased<br />

cathepsins levels in Alzheimer brain tissues <strong>and</strong> preliminary studies from <strong>our</strong> laboratory<br />

showed that APP-Ab are sufficient to increase cathepsins in primary culture of cortical<br />

neurones.<br />

In the study presented here, we delineate the relationship between the APP cytotoxic<br />

functions <strong>and</strong> cathepsin S expression. We provide evidence that acting directly <strong>and</strong><br />

specifically upon neuronal APP through APP-Ab binding, stimulates the synthesis of<br />

cathepsin S. In addition, a Jun-Kinase inhibitor could reverse cathepsin S activation. Finally,<br />

following APP-Ab treatment, the addition of a specific inhibitor of cathepsin S reverse APP-<br />

Ab dependent cell death. Taken together, <strong>our</strong> results show that cathepsin S is a crucial<br />

executor of APP neurotoxicity. These findings raise the possibility that the lysosomal<br />

pathway may contribute to neuronal cell death in AD.<br />

Supported by Alsace Alzheimer 68.<br />

- 473 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 16<br />

Transduction pathway in oligodendrocyte death induced by TNF<br />

Anna Jurewicz, Mariola Matysiak, Krzysztof Tybor <strong>and</strong> Krzysztof Selmaj<br />

Department of Neurology <strong>and</strong> Department of Neurosurgery, Medical University of<br />

Lodz, Lodz, Pol<strong>and</strong>. E-mail: ajur@afazja.am.lodz.pl<br />

Tumor necrosis factor (TNF) induces apoptosis-like death of oligodendrocytes (OLs), the<br />

putative cell target in multiple sclerosis. We defined that the intracellular transduction<br />

pathway involved in TNF-induced death of human OLs (hOLs) is dependent on c-jun NH2terminal<br />

kinase-3 (JNK3) activation, change of mitochondrial membrane permeability <strong>and</strong><br />

intranuclear apoptosis inducing factor (AIF) translocation. TNF-induced death of hOLs is<br />

non-caspase dependent, as evidenced by: lack of generation of caspase-8, -1 <strong>and</strong> -3 active<br />

subunits; lack of cleavage of caspase-1 <strong>and</strong> -3 fluorogenic substrates; <strong>and</strong> lack of hOL death<br />

inhibition by the general caspase inhibitor, ZVAD.FMK. JNK activation, measured by c-jun<br />

phosphorylation <strong>and</strong> induction of the phosphorylated form of JNK, was enhanced, prolonged<br />

<strong>and</strong> correlated with cell death in hOLs exposed to TNF. Comparative autoradiographic<br />

analysis revealed that JNK-3, but not JNK-1 or JNK-2, is responsible for prolonged JNK<br />

activation in TNF exposed hOLs. Expression of a dominant-negative mutant of JNK upstream<br />

kinase, MKK4/SEK1, inhibited apoptosis induced by TNF, whereas expression of a<br />

constitutive active mutant of MEKK1, an upstream kinase to JNK, accelerates TNF-induced<br />

apoptosis. JNK activation occurred prior to changes of mitochondrial membrane potential in<br />

hOLs exposed to TNF. The co-localization experiments showed that upon exposure to TNF<br />

AIF translocated into the nucleus <strong>and</strong> electrophoresis of TNF-exposed hOLs DNA revealed<br />

large scale DNA fragmentation characteristic of apoptosis-inducing factor (AIF)-mediated<br />

cell death. AIF depletion by an antisense strategy prevented TNF-induced hOL death. These<br />

results demonstrate that TNF-induced death in adult hOLs depends on prolonged JNK-3<br />

activation, requires the mitochondrial dysfunction that occurs after JNK activation <strong>and</strong><br />

requires AIF translocation into nucleus. This is the first evidence that a JNK-3 isoform <strong>and</strong><br />

AIF are involved in oligodendrocyte death <strong>and</strong> might have significant importance in<br />

designing new molecules to protect hOLs demise in multiple sclerosis.<br />

- 474 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 17<br />

GALANTAMINE PREVENTS THE NEUROTOXICITY INDUCED BY BETA-<br />

AMYLOID PEPTIDE<br />

Joana B Melo 1,2, Carla Sousa 1, Pedro Garção1, Paula Agostinho1,3, Catarina R<br />

Oliveira1,3<br />

1Center for Neurosciences <strong>and</strong> Cell Biology, 2Institute of Medical Biology <strong>and</strong> 3Institute<br />

of Biochemistry, Faculty of Medicine, University of Coimbra, Portugal<br />

jbmelo@cnc.cj.uc.pt<br />

Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by the formation of<br />

senile plaques in patients’ brain, composed by amyloid #-peptide (A#). Several reports<br />

indicate that the degree of #-aggregation seems to be particularly important for the<br />

neurotoxicity of this amyloidogenic peptide. Cholinergic abnormalities, such as severe loss of<br />

nicotinic acetylcholine receptors (nAChRs) are also found in AD brains. Several studies on<br />

brains displaying AD lesions have shown changes in the expression <strong>and</strong> distribution of<br />

acetylcholinesterase (AChE). Although AChE activity is lost in specific regions of the AD<br />

brain, an increase of AChE activity has also been found to co-localize with A# deposits.<br />

Galantamine, a drug currently approved for the symptomatic treatment of AD, is an inhibitor<br />

of AChE <strong>and</strong> has an additional activity as an allosteric modulator of nAChRs.<br />

The goal of the current study, using cortical cells as a neuronal model, was to evaluate the<br />

effect of several concentrations of galantamine in the neurotoxicity induced by the synthetic<br />

peptide A#1-40 correlating these results with the state of aggregation of this peptide. We<br />

observed that galantamine prevents, in a concentration dependent manner, the increase of<br />

AChE activity <strong>and</strong> the neuronal injury induced by A#1-40. As expected, amyloid deposition,<br />

evaluated by Congo Red dye, that positively stains #-sheet conformation <strong>and</strong> Thioflavin S<br />

dye, is increased in the presence of A#1-40 <strong>and</strong> the treatment of cells with galantamine<br />

decreases this deposition. However, in a cell-free system, galantamine did not revert amyloid<br />

fibril formation. These results suggest that the neuroprotective role of galantamine could also<br />

be correlated with an effect on the aggregation state of amyloid beta-peptide present in the<br />

patients’ brains.<br />

(Supported by Janssen -Cilag <strong>and</strong> GAI-Faculty Medicine of University of Coimbra)<br />

- 475 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 18<br />

Intensive remodelling of Purkinje cells spines after climbing fibres deafferentation does<br />

not involve MAPKs <strong>and</strong> AKT activation<br />

Jelena M. Mila)in1,2, Annalisa Buffo1, Daniela Carulli1, Piergiorgio Strata 1,3<br />

1 Rita Levi Montalcini Brain Repair Center, University of Turin, Turin, Italy<br />

2 School of Dentistry, University of Belgrade, Belgrade, Serbia <strong>and</strong> Montenegro, E-mail:<br />

jelena_milasin@yahoo.com<br />

3 Fondazione Santa Lucia, Rome, Italy<br />

Subtotal lesion of the inferior olive (IO) achieved by treating experimental animals with 3<br />

acetylpyridine (3AP) induces partial Purkinje cells (PC) deafferentation that leads to PC<br />

hyperactivity <strong>and</strong> new spine formation. Coincidently, the olivary terminals belonging to the<br />

few survived olivary neurons undergo an extensive collateral sprouting resulting in<br />

reinnervation of the neighb<strong>our</strong>ing denervated PCs. We obtained chemical deafferentation of<br />

PCs in adult rats (body weight, 120-170 gm; age, 35-40 d) by a single intraperitoneal injection<br />

of 3-AP (65 mg/kg body weight) <strong>and</strong> as early as three days after 3AP treatment, important<br />

morphological changes could be observed on PCs. MAPK cascades <strong>and</strong> more specifically<br />

ERK1/2 play a critical role in the <strong>signaling</strong> events underlying synaptic plasticity. For instance<br />

LTD in the adult hippocampus <strong>and</strong> LTP in cerebellum both involve ERK activation. Since<br />

PCs deprived of their CF afferents initiate an intensive remodelling of the spines <strong>and</strong> rapid<br />

recall of the remaining CFs, it prompted us to see whether the observed phenomena correlated<br />

with ERK activation. Immunohistochemistry <strong>and</strong> western blotting were done at various time<br />

points after 3AP application (from 24h to 6 days), as the exact dynamics of CF loss is not<br />

precisely known. As judged by western-blotting, there was no increase of activated ERK in<br />

the cerebellum. However, immunohistochemistry revealed increased ERK phosphorylation in<br />

the „pinceaux“ of basket cells in 3AP animals. Similarly, SAPK/JNK, p38 MAPK <strong>and</strong> Akt<br />

activation were also studied by means of western-blotting <strong>and</strong> immunohistochemistry. In the<br />

c<strong>our</strong>se of IO destruction, PCs <strong>and</strong> CFs sprouting following 3AP treatment no changes in<br />

phosphorylation status could be seen in the different kinases subjected to analysis. Our results<br />

suggest that activation of MAPK <strong>and</strong> Akt cascades is not essential in this model of neuronal<br />

plasticity.<br />

- 476 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 19<br />

Amyloid precursor protein <strong>and</strong> Presenilin 1 interaction in human H4cells studied by<br />

FLIM <strong>and</strong> FRET.<br />

Mario Nizzari1, Valentina Venezia1, Paolo Bianchini2, Valentina Caorsi2, Alberto<br />

Diaspro2, Emanuela Repetto1, Stefano Thellung1, Aless<strong>and</strong>ro Corsaro1, Gennaro<br />

Schettini1, Pia Carlo1, Tullio Florio1 <strong>and</strong> Claudio Russo3.<br />

1Pharmacology, Dept. Oncology, Biology <strong>and</strong> Genetics, 2LAMBS-MicroscoBio, Dept<br />

Physics, Univ Genova, Italy, 3Dept. of Health Sciences Univ Molise. e-mail:<br />

mario.nizzari@unige.it<br />

The pathologic hallmarks of Alzheimer’s disease (AD) are senile plaque <strong>and</strong> neurofibrillary<br />

tangles. Senile plaque are primilary made up of deposits of amyloid-beta protein, a proteolytic<br />

product derived from the amyloid precursor protein (APP). APP is a transmembrane protein<br />

inserted into the endoplasmic reticulum, transported to the Golgi apparatus, to the cell surface,<br />

<strong>and</strong> recycled by endocytosis to endosomes. Proteolytic processing, lead at the formation of<br />

amyloid-beta protein, <strong>and</strong> a C-terminal fragments (CTFs). It’s not fully understood where<br />

amyloid-beta is generated. However the identification of presenilins (PS), a component of<br />

gamma secretase complex that cleave CTFs, leaving 40 or 42 amino acids amyloid-beta<br />

peptides <strong>and</strong> 58 or 56 amino acids intracellular domains (AICD), provides a opportunity to<br />

study APP-Presenilin interaction in specific cell compartments. In <strong>our</strong> study we used two<br />

biophysical assays of protein proximity: fluorescence resonance energy transfer (FRET), <strong>and</strong><br />

fluorescence lifetime immaging microscopy (FLIM), that can provide information about<br />

molecular interactions when two proteins are in the close proximity (of


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 20<br />

Study of staurosporine-induced apoptotic response in a cybrid cell line carrying the<br />

A8344G MERRF mutation in the mitochondrial DNA.<br />

Guillaume Rommelaere, Ludovic Mercy, Andrée Houbion, Catherine Demazy, Noelle<br />

Ninane, José Remacle, Martine Raes, Thierry Arnould <strong>and</strong> Patricia Renard<br />

Laboratory of Biochemistry <strong>and</strong> Cellular Biology, University of Namur (F.U.N.D.P.),<br />

Rue de Bruxelles, 61, 5000 Namur, Belgium. Email : patsy.renard@fundp.ac.be<br />

The Myoclonic Epilepsy with Ragged Red Fibers (MERRF) is a neurodegenerative<br />

mitochondrial disease characterized by ataxia, myoclonic epilepsy <strong>and</strong> progressive muscular<br />

weakness. It is caused by the single base pair substitution A8344G in the mitochondrial<br />

tRNALys gene. This mutation causes a 70% decrease in the mitochondrial protein synthesis,<br />

leading to impaired respiratory complexes activities <strong>and</strong> a strong decrease in ATP synthesis<br />

by OXPHOS. In high energy consuming tissues like brain <strong>and</strong> muscles, apoptotic features<br />

have been reported in biopsies (Mirabella et al, Brain (2000), 123, 93). Hypothesizing an<br />

apoptotic origin of the degenerative aspect of the pathology, we are studying the apoptotic<br />

response of cybrid cells carrying the A8344G mutation .<br />

In a preliminary study, we have shown that a cybrid cell line derived from 143B osteosarcoma<br />

cells <strong>and</strong> carrying the A8344G mutation (mutated cybrids) is more sensitive to staurosporineinduced<br />

apoptosis than a parental cybrid cell line carrying the wild type mtDNA (wild type<br />

cybrids). Indeed the staurosporine-induced DNA fragmentation <strong>and</strong> caspase-3 activity, the<br />

main protease involved in programmed cell death, are higher in the mutated cybrids.<br />

Using a DNA microarray dedicated to apoptosis studies, we analysed the gene expression<br />

variation in the two cybrid cell lines in response to staurosporine treatment. Among others,<br />

two genes, BIRC2 <strong>and</strong> BIRC3 are upregulated in the wild type cybrids in response to<br />

staurosporine, as compared to mutated cybrids. As they code for cIAP-1 <strong>and</strong> cIAP-2, two<br />

antiapoptotic proteins, this upregulation could explain the resistance of wild type cybrids to<br />

staurosporine-induced apoptosis. This hypothesis is still under investigation.<br />

This study will help to reveal the molecular bases for an enhanced apoptotic response of cells<br />

presenting a strong mitochondrial impairment due to a defective mitochondrial tRNALys<br />

gene.<br />

G. Rommelaere is a fellow of the FRIA (Fonds p<strong>our</strong> la Recherche dans l’Industrie et<br />

l’Agriculture, Belgium). T. Arnould is a Research Assistant of FNRS (Fonds National de la<br />

Recherche Scientifique, Belgium).<br />

- 478 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 21<br />

ERK1/2 phosphorylation, I-NOS induction <strong>and</strong> chemokine secretion are three events<br />

involved in the complex <strong>signaling</strong> of prion protein fragment 90-231 in microglial cells.<br />

1Stefano Thellung, 1Aless<strong>and</strong>ro Corsaro, 1Valentina Villa, 1Valentina Venezia, 1Mario<br />

Nizzari, 1Michela Bisaglia, 2Claudio Russo, 1Gennaro Schettini, 3Antonio Aceto <strong>and</strong><br />

1Tullio Florio<br />

1Pharmacology Dept. of Oncology, Biology & Genetics, University of Genova, Italy.<br />

2Dept. Health Sciences, University of Molise, Campobasso Italy. 3Section of<br />

Biochemistry, Dept. Biomedical Sciences, University G. D’Annunzio of Chieti, Italy.<br />

E-mail: thellung@yahoo.com.<br />

We show that PrP90-231, a neurotoxic prion protein fragment, activates the murine microglial<br />

cell line N9 <strong>and</strong> describe the transduction mechanisms involved. PrP90-231 induced<br />

phosphorylation of MAP kinase ERK1/2 <strong>and</strong> the expression of the inducible isoform of nitric<br />

oxide synthase (I-NOS). Time c<strong>our</strong>se experiments, show that ERK1/2 activation is detectable<br />

after 15 minutes of treatment, reaches a maximum at 25 minutes <strong>and</strong> is sustained up to 60<br />

minutes of cell exposure to the peptide. Importantly, late onset <strong>and</strong> sustained ERK1/2<br />

activation are typical events involved in cell differentiation rather than proliferation. Indeed,<br />

we have observed by both MTT <strong>and</strong> [3H]-tymidine uptake assays, that PrP90-231 blocked N9<br />

cell proliferation without inducing cell death. I-NOS induction was detectable after 6 h<strong>our</strong>s of<br />

cell exposure to PrP90-231 <strong>and</strong> reached its maximal value after 24 h<strong>our</strong>s of treatment. We<br />

excluded that I-NOS expression was dependent on ERK1/2 activation, since cell pretreatment<br />

with the MEK inhibitor PD98059 did not prevent I-NOS activation by PrP90-231. Beside<br />

nitric oxide, microglial cells produce <strong>and</strong> release several cytokines in response to<br />

proinflammatory inputs. We observed that after 24 h<strong>our</strong>s of stimulation with PrP90-231,<br />

RANTES release is enhanced. By culturing N9 in chambers separated by a microporous<br />

barrier we have observed that ERK1/2 activation <strong>and</strong> I-NOS expression were elicited by<br />

PrP90-231 only if cells were directly exposed to the peptide. We conclude that these events<br />

are not dependent on autocrine/paracrine loop triggered by the peptide <strong>and</strong> sustained by<br />

diffusible mediators Rather, we suggest that a sustained physical interaction of PrP90-231<br />

with the cell is required to keep microglia in an activated state.<br />

- 479 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 22<br />

Cas III-ia Induces Autophagy <strong>and</strong> Apoptotic Death in Glioma C6 Cells In Vitro <strong>and</strong> In<br />

Vivo.<br />

Cristina Trejo-Solís1, Guadalupe Palencia1, Lucrecia Márquez-Rosado2, Dolores<br />

Jiménez-Farfán3, Aurora Sánchez1, Arturo Cruz-Salgado1, Isabel Gracia-Mora4, Lena<br />

Ruiz-Ramírez4 <strong>and</strong> Julio Sotelo1.<br />

1Departamento de Neuroinmunología, Instituto Nacional de Neurología y Neurocirugía<br />

“MVS”, México, DF. 2Departamento de Biología Celular, Centro de investigación y<br />

Estudios Avanzados del IPN, México, DF. 3 Departamento de Neuroinmunología,<br />

Instituto de Odontología, UNAM, México, DF. 4Departamento de Química y Medicina<br />

Nuclear, UNAM, México, DF.<br />

In this reaserch, we investigated the effects of casiopeina III-ia (Cas III-ia)- a new copper<br />

compound that exhibits antineoplastic activity- on glioma C6 cells under both in vitro <strong>and</strong> in<br />

vivo conditions, in an attempt to identify potential therapeutic agents against maliganant<br />

glioma. The exposure of C6 to Cas III-ia significantly inhibited cell proliferation, increased<br />

reactive oxygen species (ROS) formation, <strong>and</strong> induced cell death. In cultured C6 cells, Cas<br />

III-ia caused the accumulation of autophagic vacuoles, formation of LC3-II, <strong>and</strong> nuclear<br />

translocation of AIF <strong>and</strong> Endonuclease G in all tested concentrations. At higher concentration<br />

were observed, loss of mitochondrial transmembrane potential, over-expression of Beclin 1,<br />

LAMP3, Bax <strong>and</strong> Bid, phosphorylation of JNK <strong>and</strong> ERK, <strong>and</strong> nuclear translocation of AP-1.<br />

Administration of N-acetyl-L—cystein (an antioxidant) resulted in significative inhibition of<br />

anti-neoplastic effect of Cas III-ia on C6 glioma cells. These results suggest that Cas III-ia<br />

induced death cell by autophagy <strong>and</strong> caspase independient apoptosis. In addition, treatment<br />

of glioma C6- positive rats with Cas III-ia reduced tumor volumen as well as mitotic <strong>and</strong> cell<br />

proliferation indexes, while it increased the apoptotic index. Our findings support the use of<br />

cas III-ia for the treatment of malignant gliomas.<br />

This work was supported by CONACYT grant U41997-MA1.<br />

- 480 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 23<br />

Targeting the IGF-IR for Therapy in Metastatic Human Neuroblastoma<br />

Cynthia M. van Golen, Ph.D. 1, Tracy S. Schwab, Ph.D. 1, Bhumsoo Kim, Ph.D.1, Mary<br />

Soules1, Sang Su Oh1, Kevin Fung, M.D. 2, Kenneth L van Golen, Ph.D. 3 <strong>and</strong> Eva L<br />

Feldman, M.D., Ph.D. 1.<br />

1Neurology, University of Michigan, Ann Arbor, MI, 48109, 2Otorhinolaryngology,<br />

University of Michigan, Ann Arbor, MI, 48109, <strong>and</strong> 3Internal Medicine, Hematology<br />

<strong>and</strong> Oncology Division, University of Michigan, Ann Arbor, MI, 48109.<br />

Neuroblastoma (NBL), the second most common pediatric extracranial tumor, produces<br />

metastases in bone resulting in a survival rate of less than 7%. Therefore, underst<strong>and</strong>ing how<br />

bony metastases form is critical for improving patient survival. Our laboratory has shown<br />

that Type I insulin-like growth factor receptor (IGF-IR) expression <strong>and</strong> activation are present<br />

in advanced stage NBL tumors <strong>and</strong> regulate NBL cell proliferation, motility, invasion, <strong>and</strong><br />

survival. Bone expresses large amounts of IGF lig<strong>and</strong>s, <strong>and</strong> the IGF system is required for<br />

normal bone physiology. Therefore, we addressed in the current study the role of the IGF<br />

system in NBL metastasis to bone. Upon reaching the bone marrow through the circulation,<br />

NBL cells must adhere to the bone marrow endothelium, extravasate into the bone<br />

microenvironment, <strong>and</strong> destroy bone tissue to allow for tumor growth. We show high-IGF-<br />

IR-expressing NBL cells migrate across a human bone marrow endothelial cell layer toward<br />

bone stromal cells, which produce IGF-I, <strong>and</strong> then adhere tightly to these stromal cells.<br />

Transendothelial migration is blocked by both a small molecule inhibitor <strong>and</strong> a monoclonal<br />

neutralizing antibody against the IGF-IR. Intratibial injection of human NBL cells expressing<br />

high levels of the IGF-IR, either endogenously or through transfection of an IGF-IR<br />

expression construct, leads to tumor formation, osteolytic lesions, <strong>and</strong> additional secondary<br />

tumors in other organs. Intraventricular injection of high-IGF-IR-expressing NBL cells also<br />

leads to tumor formation within bone <strong>and</strong> osteolysis. Osteolytic lesions form when<br />

osteoclasts differentiate <strong>and</strong> become activated in response to tumor cells. When high IGF-IR<br />

expressing NBL cells are injected intratibially, increased numbers of multinucleated tartrateresistant<br />

acid phosphatase (TRAP) positive cells are detected, suggesting an increase in<br />

osteoclast differentiation. In vivo, micro-CT analysis of the bone demonstrates that high-IGF-<br />

IR expressing NBL cells promote osteolysis of 81% of trabecular bone by 3 weeks <strong>and</strong> over<br />

97% of trabecular bone by 4 weeks, with resorption pits also evident within the cortical bone.<br />

Conditioned media from high IGF-IR-expressing NBL cells are also capable of supporting<br />

osteoclast differentiation in vitro within 10 d. These data suggest that IGF-IR expression in<br />

NBL cells in part determines their ability to form osteolytic metastatic tumors within bone;<br />

therefore, targeted treatment against the receptor may prove efficacious in treating advanced<br />

stage NBL. Targeting the IGF-IR for therapy has recently gained attention in numerous<br />

tumors, leading to pharmaceutical production of a number of inhibitors. We are currently<br />

testing several of these inhibitors for both in vitro <strong>and</strong> in vivo efficacy. This work was<br />

supported by the Program for Underst<strong>and</strong>ing Neurological Diseases (PFUND), the Juvenile<br />

Diabetes Research Foundation Center for the Study of Complications in Diabetes, NIH RO1<br />

NS36778, <strong>and</strong> NIH RO1 NS38849.<br />

- 481 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 24<br />

Amyloid precursor protein modulates ERK1,2 <strong>signaling</strong> in glial cells.<br />

Valentina Venezia1, Mario Nizzari1, Emanuela Repetto1, Elisabetta Violani1,<br />

Aless<strong>and</strong>ro Corsaro1, Stefano Thellung1, Valentina Villa1, Pia Carlo1, Gennaro<br />

Schettini1, Tullio Florio1 <strong>and</strong> Claudio Russo2.<br />

1Section of Pharmacology, Dept. Oncology, Biology <strong>and</strong> Genetics, University of Genova,<br />

Italy. 2Dept. Health Sciences, University of Molise, Campobasso, Italy. e-mail:<br />

venezia@unige.it<br />

The amyloid precursor protein (APP) is a transmembrane protein with a short cytoplasmic<br />

tail. Its location <strong>and</strong> structural features are characteristics of a receptor for extracellular<br />

lig<strong>and</strong>. Yet, the physiological function of APP is unclear, although it is well documented that<br />

APP’s proteolytic processing, could influence the development of Alzheimer’s disease (AD),<br />

through the formation of membrane-bound C-terminal fragments (CTFs) <strong>and</strong> of beta-amyloid<br />

peptides (Ab). We have recently shown that tyrosine-phosphorylated APP <strong>and</strong> CTFs may<br />

interact with Grb2 <strong>and</strong> ShcA adaptor proteins <strong>and</strong> that this coupling occurs at a higher extent<br />

in AD subjects only.<br />

To study the interaction between APP or CTFs <strong>and</strong> ShcA/Grb2 <strong>and</strong> to investigate their<br />

molecular target we have used as experimental model two different glial cell lines: H4 human<br />

neuroglioma cells <strong>and</strong> APP/APLP null mouse embryonal fibroblast cells (MEF). Here we<br />

show that in H4 cells APP interacts with Grb2; conversely in APP/APLP null MEF cells this<br />

interaction is possible only after the reintroduction of human APP by transfection. We have<br />

also shown that in MEF cells the transfection of a plasmid encoding for human APP wt<br />

enhances the phosphorylation of ERK 1,2 as revealed by western blotting <strong>and</strong><br />

immunofluorescence experiments. Finally, also in H4 cells the overexpression of APP<br />

upregulates the levels of phospho-ERK1,2.<br />

In summary <strong>our</strong> data suggest that APP may influence phospho-ERK 1,2 <strong>signaling</strong> through its<br />

binding with Grb2 <strong>and</strong> ShcA adaptors. The meaning of this event is not clear, but APP<br />

interaction with these adaptors could be relevant to regulate either the physiological function<br />

of APP or to sustain glial proliferation in Alzheimer Disease pathology.<br />

- 482 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 25<br />

Characterization of the pro-apoptotic intracellular mechanisms induced by a toxic<br />

conformer of the recombinant human prion protein fragment 90-231<br />

Valentina Villa1, Aless<strong>and</strong>ro Corsaro1, Stefano Thellung1, Domenico Paludi2, Katia<br />

Chiovitti3, Valentina Venezia1, Mario Nizzari1, Claudio Russo4, Gennaro Schettini1,<br />

Antonio Aceto3 <strong>and</strong> Tullio Florio1<br />

1Section of Pharmacology, Dept. Oncology Biology <strong>and</strong> Genetics, University of Genova,<br />

Italy. 2Dept. Scienze degli Alimenti, Veterinary School, University of Teramo, Italy.<br />

3Section of Biochemistry, Dept. Biomedical Sciences, University G. D’Annunzio of<br />

Chieti, Italy. 4Dept. Health Sciences, University of Molise, Campobasso Italy.<br />

Prion diseases comprise a group of fatal neurodegenerative disorders that affect both animals<br />

<strong>and</strong> humans. The transition of the prion protein (PrP) from a mainly alpha structured isoform<br />

(PrPC) to a prevalent beta sheet-containing protein (PrPSc) is believed to represent a major<br />

pathogenetic mechanism in prion diseases. To investigate the linkage between PrP<br />

neurotoxicity <strong>and</strong> its conformation, we used a recombinant prion protein fragment<br />

corresponding to the amino acidic sequence 90-231 of human prion protein (hPrP90-231).<br />

Using thermal denaturation, we set up an experimental model to induce the process of<br />

conversion from PrPC to PrPSc .We report that partial thermal denaturation converts hPrP90-<br />

231 into a beta sheet-rich isoform, displaying a temperature <strong>and</strong> time-dependent conversion<br />

into oligomeric structures that share some physico-chemical characteristics with brain PrPSc.<br />

SH-SY5Y cells were chosen to characterize the potential neurotoxic effect of hPrP90-231 in<br />

its different structural conformations. We demonstrated that hPrP90-231 in beta<br />

conformation, but not when alpha structured, powerfully affected the survival of this cells.<br />

hPrP90-231 beta structured caused DNA fragmentation <strong>and</strong> a significant increase in caspase-3<br />

proteolytic activity (maximal effects +170%), suggesting the occurrence of apoptotic cell<br />

death. Finally we investigated the involvement of MAP kinases in the regulation of beta<br />

hPrP90-231 dependent apoptosis. We observed that the p38 MAP kinase blocker SB203580<br />

prevented the apoptotic cell death evoked by hPrP90-231 <strong>and</strong> Western blot analysis revealed<br />

that the exposure of the cells to the peptide induced p38 phosphorylation. In conclusion, we<br />

demonstrate that the hPrP90-231 pro-apoptotic activity is mediated by p38 <strong>and</strong> caspase-3<br />

activation. (grants by MIUR PRIN 2004 <strong>and</strong> FIRB 2001 to TF)<br />

- 483 -


Session XII : Cell death <strong>and</strong> neurodegenerative diseases Poster XII, 26<br />

MEKK1 controls neurite regrowth after injury via ERK1/2 <strong>and</strong> JNK2 <strong>signaling</strong><br />

Vicki Waetzig <strong>and</strong> Thomas Herdegen<br />

Institute of Pharmacology, Hospitalstrasse 4, UKSH, 24106 Kiel, Germany, E-mail:<br />

vicki.waetzig@pharmakologie.uni-kiel.de<br />

After injury, peripheral neuronal cells initiate complex <strong>signaling</strong> cascades to promote survival<br />

<strong>and</strong> regeneration. We have identified the mitogen-activated protein kinase (MAPK) isoforms<br />

which are necessary for nerve growth factor (NGF)-induced neurite regrowth after injury of<br />

differentiated PC12 cells. Extracellular signal-regulated kinases 1 <strong>and</strong> 2 (ERK1/2) <strong>and</strong> the<br />

otherwise apoptotic c-Jun N-terminal kinase 2 (JNK2) are crucial for neurite regrowth, while<br />

p38 plays no role in this context. Surprisingly, the MEK1 inhibitors PD98059 <strong>and</strong> U0126<br />

blocked ERK1/2 <strong>and</strong> JNK phosphorylation, indicating a novel form of balancing MAPK<br />

cascade crosstalk. We identified the upstream kinase MEKK1 as an activator of both the<br />

ERK1/2 <strong>and</strong> JNK2 pathways, whereby the ERK1/2 kinase MEK1 <strong>and</strong> the JNK kinase MKK7<br />

bound to MEKK1 in a competing fashion. The use of siRNAs against ERK1/2 excluded a<br />

direct interaction between ERK1/2 <strong>and</strong> JNKs. In JNK knockout mice, we have confirmed the<br />

importance of JNKs for facial nerve regeneration.<br />

- 484 -


Notes<br />

- 485 -


Notes<br />

Notes<br />

- 486 -


- 487 -


Session XIII : Cell <strong>signaling</strong> pathways leading to regulated<br />

chromatin modifications<br />

- 488 -


Session XIII : Cell <strong>signaling</strong> pathways leading to regulated chromatin modifications<br />

Poster XIII, 1<br />

ICBP90, a methyl binding protein, as potential gene suppressor for p73 expression in<br />

Jurkat cells<br />

Abdul-Qader Abbady, Michaël Jeanblanc, Marie Schöller-Guinard, Jean-Paul Klein,<br />

Ermanno C<strong>and</strong>olfi <strong>and</strong> Marc Mousli.<br />

Institut National de la Santé et de la Recherche Médicale, UMR-S 392/E.A. 3950, Faculté<br />

de Médecine, 3 rue Koeberlé, 67000 Strasb<strong>our</strong>g, France. E-mail:<br />

marc.mousli@medecine.u-strasbg.fr<br />

T cell activation involves induction of genes mediating antigen-induced cell death (AICD)<br />

through T cell receptor activation. AICD controls the expansion of antigen-activated T cells<br />

after an immune response <strong>and</strong> deletes self-reactive T cells by negative selection. This occurs<br />

from late G1 cell cycle checkpoint that is dependent on E2F-1 <strong>and</strong> p73. Methylation at CpG<br />

dinucleotides, the most abundant epigenetic modification in vertebrate genomes, plays an<br />

essential part in the control of gene expression. Inverted-CCAAT-box-binding protein of 90<br />

kDa (ICBP90) has been recently identified as a novel methyl-CpG-binding protein. Since<br />

methylation of p73 gene plays an important role in its transcriptional activity, we<br />

hypothesized that ICBP90 might be involved in the regulation of p73 gene expression. In the<br />

present study, we analyzed the expression of ICBP90 in Jurkat <strong>and</strong> CCRF-CEM lymphocytes<br />

<strong>and</strong> in THP-1 monocytes as control after triggering by phytohemagglutinin (PHA). We found<br />

that activation of Jurkat <strong>and</strong> CCRF-CEM cells with PHA decreased the number of living cells<br />

<strong>and</strong> increased the number of apoptotic cells in a significant manner while these remained<br />

unchanged in THP-1 cells. We also observed a significant decrease in ICBP90 expression<br />

when compared to unstimulated lymphocytes <strong>and</strong> THP-1 control cells. Moreover, the<br />

promoter activity of ICBP90 gene, measured using the luciferase reporter gene, was also<br />

significantly inhibited after triggering Jurkat cells by PHA whereas no change was observed<br />

in THP-1 cells. Finally, ICBP90 down expression was accompanied with an increase of p73<br />

expression in activated Jurkat cells whereas p73 was almost undetectable in THP-1 cells. In<br />

conclusion, <strong>our</strong> results suggest that down regulation of ICBP90 as a methyl binding protein,<br />

may play a key role in p73 gene activation in AICD <strong>and</strong> T cell clonal expansion.<br />

- 489 -


Session XIII : Cell <strong>signaling</strong> pathways leading to regulated chromatin modifications<br />

Poster XIII, 2<br />

Interdependency or Exclusion? Investigation of the phosphorylation state of histone H3<br />

at serine 10 <strong>and</strong> threonine 11 during mitosis<br />

Anne Conradi <strong>and</strong> Karl Heinz Scheidtmann<br />

Institute of Genetics, University of Bonn, Roemerstr. 164, D-53117 Bonn, Germany<br />

E-mail: kh.scheidtmann@uni-bonn.de<br />

Histones are subject to numerous post-translational modifications. During mitosis histone H3<br />

becomes heavily phosphorylated at serine 10 by Aurora-B kinase, as well as at threonine 11<br />

probably by Dlk/ZIP kinase. Phosphorylation at serine 10 occurs along the entire<br />

chormosomal arms, but not at centromeres. It coincides with chromatin condensation <strong>and</strong> may<br />

be involved in initiation of this process. In contrast, phosphorylation at threonine 11 appears<br />

to be centromere-specific <strong>and</strong> may be involved in kinetochore assembly <strong>and</strong>/or function.<br />

These phosphorylation events seem to occur in an exclusive manner which is also supported<br />

by in vitro phosphorylation data. We investigated a possible interrelationship between<br />

Aurora-B <strong>and</strong> Dlk/ZIP kinase. In vitro kinase reactions with purified Aurora-B <strong>and</strong> Dlk/ZIP<br />

kinase revealed no crossphosphorylation between the two kinases. In vivo Aurora-B-specific<br />

kinase inhibitors revealed loss of H3 phosphorylation at serine 10 but not at threonine 11. Our<br />

data suggest that both phosphorylations occur independently.<br />

Acknowledgements:<br />

This work was supported by Deutsche Forschungsgemeinschaft grant Sche 246/16-1.<br />

- 490 -


Session XIII : Cell <strong>signaling</strong> pathways leading to regulated chromatin modifications<br />

Poster XIII, 3<br />

Pharmacological restoration of ROS impaired steroid function: the role of HDAC-2<br />

Paul A. Kirkham, Koremu Meja, Gillian Spooner <strong>and</strong> John A. Marwick.<br />

Novartis Institutes for Biomedical Research, Horsham, West Sussex, RH12 5AB, UK.<br />

Email: paul.kirkham@novartis.com<br />

HDAC-2 activity plays a major role in regulating steroid function <strong>and</strong> inflammation. COPD<br />

patients, severe asthmatics <strong>and</strong> mild asthmatics who smoke are all insensitive to steroid<br />

treatment, as a result of exposure to oxidative stress. Moreover, oxidative stress has a direct<br />

inhibitory effect on HDAC-2 activity through post-translational modification. We<br />

hypothesised that upregulating HDAC activity after exposure to oxidative stress should<br />

restore steroid function. Oxidative stress reduced HDAC-2 but not HDAC-1 activity. Both<br />

theophylline <strong>and</strong> curcumin specifically restored HDAC-2 activity in pre-ROS stressed cells<br />

only, in both a time <strong>and</strong> dose dependant manner. Similar effects on restoring HDAC activity<br />

after ROS stress were observed using either JNK or Akt inhibitors, but not p38, cAMP or<br />

calcium modulators. ROS induced reduction of HDAC-2 activity correlated with a decrease<br />

in steroid responsiveness. Co-incubation or theophylline or curcumin with budesonide (1nM)<br />

before stimulation with LPS (10ng/ml) for 16 h<strong>our</strong>s restored steroid responsiveness in ROS<br />

treated cells as assessed by TNFalpha release. Interestingly, neither theophyline or curcumin<br />

alone (upto 10uM) were anti-inflammatory in LPS stimulated pre-ROS stressed U397 cells.<br />

Moreover, theophylline (1µM) also restored the ability of dexamethasone to reduce<br />

inflammatory gene associated histone acetylation in pre-ROS stressed U937 cells as assessed<br />

by chromatin immunoprecipitation (ChIP) assay. Serine phosphorylation on HDAC-2 is<br />

reduced by oxidative stress. However, treatment with curcumin/theophyline (1µM) post ROS<br />

stress increases HDAC-2 serine phosphorylation. In conclusion, <strong>our</strong> data suggests that<br />

restoring/upregulating HDAC activity <strong>and</strong> thereby steroid function after prior reduction by<br />

oxidative stress can be achieved through pharmacological intervention. The mechanism of<br />

action for theophyline <strong>and</strong> curcumin in this process is unclear at present. However, inhibition<br />

of JNK or Akt <strong>signaling</strong> pathways may play a role. It is therefore likely that both theophyline<br />

<strong>and</strong> curcumin act on signalling pathways that regulate HDAC protein post-translational status<br />

<strong>and</strong> hence activity.<br />

- 491 -


Session XIII : Cell <strong>signaling</strong> pathways leading to regulated chromatin modifications<br />

Poster XIII, 4<br />

Two states of the nuclear matrix-bound p53 in HEK293 cells<br />

Maria A. Lapshina, Igor I. Parkhomenko <strong>and</strong> Alexei A. Terentiev.<br />

Molecular Biology Laboratory, Department of Kinetics of Chemical <strong>and</strong> Biological<br />

Processes, Institute of Problems of Chemical Physics RAS, Chernogolovka, Moscow<br />

Region, 142432, Russia. E-mail: lapshina@iap.ac.ru<br />

The tumor suppressor protein p53 is a transcription factor playing important roles in<br />

regulation of the cell cycle progression <strong>and</strong> the cell death in response to a vide variety of<br />

stressors, such as DNA damaging factors, oncogene activation, oxidation, etc. Like many<br />

other transcription factors, p53 protein is found to associate with the nuclear matrix (NM), the<br />

filamentous protein network of the cell nucleus that contributes to the higher order chromatin<br />

structures <strong>and</strong> to most of nuclear processes including transcription, replication, DNA repair,<br />

RNA processing <strong>and</strong> molecular transport. We studied the intranuclear distribution of the p53<br />

protein in HEK293 cells with use of various techniques of NM preparation. P53 is found to be<br />

associated with NM independently on the methods used, but its appearance in non-matrix<br />

fractions depends strongly on the order of the extraction steps: approximately 40% of nuclear<br />

p53 is observed in high salt fraction before DNase I digestion, <strong>and</strong> almost no p53 is extracted<br />

in high salt buffer after nuclease treatment. The matrix-bound p53 is found to be modified,<br />

most probably ubiquitinated, as it is observed as higher molecular weight b<strong>and</strong>s. Further<br />

extraction of matrix-bound proteins in alkaline buffers causes partial release of p53 in a<br />

soluble fraction, <strong>and</strong> the higher molecular weight forms of p53 are extracted completely under<br />

alkaline conditions. In acidic buffers, p53 protein is also partially extracted from the matrix,<br />

but the higher molecular weight forms of p53 retain their association with NM. To study if<br />

these two forms of matrix-bound p53 could be functionally different, we activated p53 in<br />

MCF-7 cells with actinomycin D <strong>and</strong> carried out the same extractions of NM proteins. The<br />

ActD-activated p53 is found to represent only one of two forms observed in HEK293 cells: it<br />

is completely extracted from NM under alkaline conditions <strong>and</strong> resistant to acidic extractions.<br />

Thus, p53 protein is found in the nuclear matrix in different states (forms). These forms of<br />

matrix-bound p53 are differentially sensitive to alkaline <strong>and</strong> acidic extractions <strong>and</strong> might<br />

differ in either protein-protein interactions (with separate NM proteins) or charges (e.g. as the<br />

consequence of differential phosphorylation <strong>and</strong>/or ubiquitination).<br />

- 492 -


Session XIII : Cell <strong>signaling</strong> pathways leading to regulated chromatin modifications<br />

Poster XIII, 5<br />

MARK/Par-1 kinases, EMK <strong>and</strong> C-TAK1 regulate localization <strong>and</strong> activity of class IIa<br />

HDACs through hierarchical phosphorylation<br />

Maud Martin, Nathalie Mari, Julia Von Blume, Didier Vertommen, Emily Lecomte,<br />

Sabine Ruidant, Marie-France Heinen, Malte Bachmann, Jean-Claude Twizere, Chris<br />

Huang, Mark H. Rider, Helen Piwnica-Worms, Thomas Seufferlein, Richard Kettmann<br />

<strong>and</strong> Franck Dequiedt<br />

Cellular <strong>and</strong> Molecular Biology Unit, Faculty of Agronomy, B-5030, Gembloux,<br />

Belgium. E-mail : dequiedt.f@fsagx.ac.be<br />

Class IIa histone deacetylases (HDACs) are found both in the cytoplasm <strong>and</strong> in the nucleus<br />

where they repress genes involved in several developmental programs. In response to specific<br />

signals, the repressive activity of class IIa HDACs is neutralized through their<br />

phosphorylation on multiple N-terminal serine residues <strong>and</strong> 14-3-3 mediated nuclear<br />

exclusion. Here, we demonstrate that class IIa HDACs are subjected to signal-independent<br />

nuclear export that relies on their constitutive phosphorylation. We identify EMK <strong>and</strong> C-<br />

TAK1, two members of the MARK/Par-1 family, as regulator of this process. We further<br />

show that EMK <strong>and</strong> C-TAK1 phophorylate class IIa HDACs on one of their multiple 14-3-3<br />

binding sites <strong>and</strong> alter their subcellular localization <strong>and</strong> repressive function. Using HDAC7 as<br />

a paradigm, we extended these findings by demonstrating that signal-independent<br />

phosphorylation of class IIa HDACS conforms to a hierarchical pattern in which<br />

phosphorylation of the MARK/Par-1 site is a prerequisite for the phosphorylation of the other<br />

14-3-3 sites. We propose that this multisite hierarchical phosphorylation by a variety of<br />

kinases allows for sophisticated regulation of class IIa HDACs function.<br />

- 493 -


Session XIII : Cell <strong>signaling</strong> pathways leading to regulated chromatin modifications<br />

Poster XIII, 6<br />

Combinatorial histone deacetylase inhibitors effect to the differentiation of human<br />

promyelocytic leukemia HL-60 cell<br />

Rasa Merzvinskyte1, Grazina Treigyte1, Jurate Savickiene1, Karl-Eric Magnusson2<br />

<strong>and</strong> Ruta Navakauskiene1<br />

1Department of Developmental Biology, Institute of Biochemistry, LT-08662 Vilnius,<br />

Lithuania. E-mail: rasa_merzvinskyte@yahoo.com<br />

grazina.treigyte@bchi.lt savickiene@bchi.lt ruta.navakauskiene@bchi.lt<br />

2Division of Medical Microbiology, Department of Molecular <strong>and</strong> Clinical Medicine,<br />

Linköping University, SE-581 85 Linköping, Sweden. E-mail: karma@imk.liu.se<br />

Recently, a novel strategy for the treatment of leukemias through the modulation of chromatin<br />

structure has been applied. In this study, we conducted an analysis of anti-leukemic efects of<br />

the histone deacetylase (HDAC) inhibitors, phenyl butyrate (PB) <strong>and</strong> vitamin B3, <strong>and</strong> in<br />

combination with the differentiation agent, retinoic acid (RA), on the promyelocytic leukemia<br />

cell line HL-60. We found that the HDACI combinations exert different effects on cell cycle<br />

arrest, differentiation <strong>and</strong> apoptosis. We also assessed the expression of the early granulocytic<br />

differentiation marker CD11b on the cells treated with different combination of HDACI <strong>and</strong><br />

RA. The most promising treatment for differentiation therapy was defined using a 6-h<br />

pretreatment with phenyl butyrate <strong>and</strong> vitamin B3 before the combined exposition to RA with<br />

vitamin B3. This significantly accelerated <strong>and</strong> increased cell differentiation (up to 95%)<br />

during the 48-h treatment. The examined HDAC inhibitors, in combination with the<br />

differentiation agent caused rapid histone H3 <strong>and</strong> H4 modifications in HL-60 cells. The<br />

increased level of histone H4 acetylation was associated with the initiation <strong>and</strong> maturation<br />

stages of HL-60 cell differentiation. Our results suggest that the chromatin remodeling using<br />

HDAC inhibitors provides a rationale for the treatment of acute promyelocytic leukemia.<br />

- 494 -


Session XIII : Cell <strong>signaling</strong> pathways leading to regulated chromatin modifications<br />

Poster XIII, 7<br />

Histone acetylation-targeted differential responses of leukemia cells HL-60 <strong>and</strong> NB4 to<br />

histone deacetylase inhibitor FK228<br />

Jurate Savickiene1, Grazina Treigyte1, Ruta Navakauskiene1 <strong>and</strong> Karl-Eric<br />

Magnusson2<br />

1Department of Developmental Biology, Institute of Biochemistry, LT-2600 Vilnius,<br />

Lithuania. E-mail: grazina.treigyte@bchi.lt savickiene@bchi.lt<br />

ruta.navakauskiene@bchi.lt<br />

2Division of Medical Microbiology, Department of Molecular <strong>and</strong> Clinical Medicine,<br />

Linköping University, SE-58185 Linköping, Sweden. E-mail: karma@imk.liu.se<br />

The histone deacetylase inhibitor (HDACI), depsipeptide (FK228), was shown previously as a<br />

chemopreventive agent for the treatment of adult T-cell lymphomas. In this report, we<br />

investigated the in vitro antileukemic activity of FK228 alone, <strong>and</strong> in combination with alltrans<br />

retinoic acid (RA), on the human leukemia cell lines, NB4 <strong>and</strong> HL-60. The results show<br />

that FK228 induced a dose-dependent (0.2-1 ng/ml) cell growth arrest <strong>and</strong> death by apoptosis<br />

in NB4 <strong>and</strong> HL-60. The combined treatment with RA accelerated <strong>and</strong> enhanced granulocytic<br />

differentiation in both cell lines distinctly; the 6 h - pretreatment with HDACI had an additive<br />

differentiating effect in HL-60 cells only. These effects were accompanied by a time- <strong>and</strong><br />

dose-dependent histone H4 hyper-acetylation <strong>and</strong> histone H3 dephosphorylation, occuring<br />

after 2-8 h exposure to HDACI. FK228 up-regulated NF-!B binding to the FasL promoter,<br />

<strong>and</strong> in combination with RA it altered the DNA binding of NF-!B linking the findings to cell<br />

death <strong>and</strong> differentiation. Pifithrin-" (PFT), an inhibitor of p53 transcriptional activity,<br />

protected from apoptosis only in NB4 cells with functional p53, i.e. when used 4 h before<br />

treatment with FK228 only or together with HDACI. In NB4 cells, PFT inhibited p53 binding<br />

to the p21 (Waf1/Cip1) promoter <strong>and</strong> induced DNA binding of NF-!B leading to enhanced<br />

cell survival. Thus, FK228 may be a promising antileukemic agent in combination with RA,<br />

since it exerts antiproliferative, differentiating <strong>and</strong> apoptotic effects in leukemia cells without<br />

<strong>and</strong> with functional p53, acting via histone modifications <strong>and</strong> selective involvement of<br />

transcription factors, like NF-!B <strong>and</strong> p53.<br />

- 495 -


Session XIII : Cell <strong>signaling</strong> pathways leading to regulated chromatin modifications<br />

Poster XIII, 8<br />

A role for Class I histone deacetylases in proliferation of tumor cells<br />

Silvia Senese 1, Katrin Zaragoza-Dorr 1, Simone Minardi 2, Loris Bernard 2, Giulio F.<br />

Draetta 3, Myriam Alcalay 2, Christian Seiser 4 <strong>and</strong> Susanna Chiocca 1*<br />

1European Institute of Oncology, Department of Experimental Oncology, 20141 Milan,<br />

Italy,<br />

2 IFOM-IEO Campus, Via Adamello 16, 20139 Milan, Italy, 3 Cancer Research, Merck<br />

Research Laboratories, Basic Research, 33 Avenue Louis Pasteur, Boston, MA 02115,<br />

USA, 4 Department of Medical Biochemistry, Division of Molecular Biology,Medical<br />

University of Vienna, Vienna Biocenter, Dr. Bohr-Gasse 9/2, A-1030 Vienna, Austria<br />

* Corresponding author. Mailing address: European Institute of Oncology, Department<br />

of Experimental Oncology, Via Ripamonti, 435, 20141 Milan, Italy.<br />

Histone deacetylases (HDACs) inhibitors are currently tested in clinical trials as anti-cancer<br />

drugs. Previous studies point towards HDAC1 as one of the possible targets for these tumor<br />

drugs. Therefore, the role of individual Class I HDACs in the regulation of cancer cell<br />

proliferation was investigated using RNAi-mediated protein knockdown. We show here that<br />

ablating HDAC1 <strong>and</strong> HDAC3 protein expression results in the inhibition of U2OS cell<br />

proliferation <strong>and</strong> an increase in the percentage of apoptotic cells. On the contrary HDAC2<br />

knockdown shows no effect, unless we concurrently knockdown both HDAC1 <strong>and</strong> HDAC2.<br />

Moreover, RNAi against HDAC1 alone or in combination with HDAC2 increases the<br />

expression of p21 protein, a cyclin-dependent kinase inhibitor. We also observe that only in<br />

the absence of both HDAC1 <strong>and</strong> HDAC2 histones H3 <strong>and</strong> H4 are hyperacetylated. In<br />

addition, HDAC1 knockdown abolishes the ability of cells to reach M phase. Our results<br />

demonstrate that HDAC1 <strong>and</strong> HDAC3 play a major role in proliferation <strong>and</strong> survival of tumor<br />

cells <strong>and</strong> that these HDACs might impair cell cycle progression not only by affecting the<br />

transcription of specific target genes (p21) but also through effects on other important<br />

biological processes.<br />

- 496 -


Session XIII : Cell <strong>signaling</strong> pathways leading to regulated chromatin modifications<br />

Poster XIII, 9<br />

Modulation of epigenetic processes in HRAS-transformed cells<br />

Christine Sers1, Karen Weißhaupt1, Thomas Mikeska2, Diana Jammas2, Xiaohua<br />

Chen1, Ralf-Jürgen Kuban3, Ute Ungethüm3, Ulf Krapfenbauer1, Hans-Peter Herzel4,<br />

Reinhold Schäfer1,3, Jörn Walter2, Per Lund1.<br />

1Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité, Berlin,<br />

Schumannstr. 20/21, D-10117 Berlin, Germany; 2Department of Natural Sciences –<br />

Technical Faculty III FR 8.3, Biological Sciences, Institute of Genetics/Epigenetics,<br />

University of Saarl<strong>and</strong>, Saarbrücken, Germany; 3Laboratory of Functional Genome<br />

Research, Charité, Berlin, Schumannstr. 20/21, D-10117 Berlin, Germany; 4Institute for<br />

Theoretical Biology, Humboldt University Berlin, Invalidenstrasse 43, D-10115 Berlin,<br />

Germany;<br />

Silencing of tum<strong>our</strong> suppressor genes is often caused by promoter hypermethylation, but little<br />

is known about the impact of oncogenic signalling pathways on methylation. The aim of this<br />

study was to investigate the role of signalling pathways downstream of RAS involved in<br />

methylation-dependent down-regulation of genes. Immortalised 208F rat fibroblasts <strong>and</strong><br />

HRAS(V12)-transformed rat fibroblasts (FE-8) were treated with the de-methylating agent 5aza-deoxycytide<br />

(5-aza-CdR) <strong>and</strong> with the HDAC-inhibitor Trichostatin A (TSA). The effect<br />

of 5-aza-CdR on the expression of genes down-regulated in the HRAS-transformed cells was<br />

investigated by conventional northern blot analysis (74 genes) <strong>and</strong> by microarray<br />

hybridisation (7186 sequences). Fifty-three genes analysed by Northern blot analysis <strong>and</strong> 10<br />

genes analysed by microarray hybridsation were re-expressed in FE-8 cells after treatment<br />

with 5-aza-CdR. Among these genes, Clusterin, Mama, MMP2, TIMP2 <strong>and</strong><br />

Thrombospondin-1 were also re-expressed after inhibition of the MEK/ERK pathway.<br />

Hypermethylation of putative regulatory elements in two independent HRAS-transforemd cell<br />

lines <strong>and</strong> in cells harb<strong>our</strong>ing an inducible HRAS, was detected within a CpG-isl<strong>and</strong> 14.5 kb<br />

upstream of clusterin, within the clusterin promoter <strong>and</strong> within a CpG-isl<strong>and</strong> of the Mmp2promoter<br />

by bisulphite sequencing. No significant hypermethylation was detected within the<br />

promoter regions of Timp2 <strong>and</strong> syndecan 4. Our study shows that RAS oncogene-dependent<br />

suppression <strong>and</strong> methylation-dependent silencing of genes are connected <strong>and</strong> impinge in part<br />

on the same target genes.<br />

- 497 -


Session XIII : Cell <strong>signaling</strong> pathways leading to regulated chromatin modifications<br />

Poster XIII, 10<br />

Signaling pathways converting extra-cellular cues into chromatin modifications at<br />

muscle specific loci during myogenesis<br />

Cristiano Simone1,2 , Sonia V. Forcales3,<br />

Lucia Latella3, Pier Lorenzo Puri3<br />

1 Sbarro Institute for Cancer Research <strong>and</strong> Molecular Medicine, Department of<br />

Biology, College of Science <strong>and</strong> Technology Temple University, Philadelphia PA 19122,<br />

USA. 2 Division of Medical Genetics, Department of Biomedicine in Childhood,<br />

University of Bari, Bari 70124, Italy. 3 DTI at Fondazione A. Cisalpino, ICBTE, Rome<br />

00128, Italy; <strong>and</strong> The Burnham Institute, La Jolla, San Diego, CA 92093, USA<br />

Myogenic differentiation is a stepwise process influenced by external cues, which are<br />

converted by cytoplasmic cascades into specific chromatin modifications, to coordinate the<br />

transcription of muscle specific genes. For instance, two distinct cytoplasmic cascades, the<br />

p38 <strong>and</strong> IGF1-mediated PI3K/Akt pathways, transmit to the nucleus pro-differentiation cues<br />

to activate the myogenic program.<br />

We have investigated the individual contribution of these pathways in the regulation of the<br />

assembly of the chromatin-modifying enzymes on muscle-specific promoters. Sequential<br />

recruitment of muscle regulating factors (MRFs), histone acetyltransferases (HATs) <strong>and</strong><br />

remodeling complexes (SWI/SNFs) on muscle enhancers/promoters was differently affected<br />

by specific inhibition of PI3K (by LY294002) <strong>and</strong> p38 (by SB203580), resulting in distinct<br />

patterns of chromatin modifications. While pharmacological blockade of PI3K inhibited<br />

HATs recruitment, thereby preventing acetylation of both histones <strong>and</strong> MyoD <strong>and</strong> reducing<br />

the levels of chromatin-bound MyoD, SB203580 (SB) selectively affected SWI/SNF<br />

chromatin binding <strong>and</strong> remodeling. In both cases, transcription of muscle-specific genes was<br />

inhibited.<br />

This evidence establishes a functional interdependence between IGF-1/PI3K/Akt <strong>and</strong> p38<br />

pathways in coordinating the assembly of the myogenic transcriptosome, <strong>and</strong> suggests that<br />

hyperacetylation of muscle enhancers/promoters in myoblasts exposed to IGF-1 precedes<br />

chromatin remodeling, which is induced by p38. The dissection of the events leading to<br />

muscle-specific transcription by pharmacological targeting of individual <strong>signaling</strong> cascades<br />

could reveal the rationale for manipulating skeletal myogenesis in vivo.<br />

- 498 -


Session XIII : Cell <strong>signaling</strong> pathways leading to regulated chromatin modifications<br />

Poster XIII, 11<br />

The role of p300/CBP complex in the differential regulation of apoptosis induced by<br />

diverse types of cytotoxic stress<br />

G. Xenaki1, M. Krstic-Demonacos2, C. Demonacos1<br />

University of Manchester 1. School of Pharmacy, Coupl<strong>and</strong> 3 building, <strong>and</strong><br />

2. Faculty of Life Sciences, Michael Smith building, Oxford Road, Manchester, M13<br />

9PL<br />

The underlying mechanisms triggering apoptosis have been unravelled in recent years <strong>and</strong> it<br />

is accepted that diverse apoptotic stimuli (anti-cancer drugs, $- or UV-irradiation, cytokines,<br />

hypoxia, deprivation of survival factors) converge on a common cascade of events leading to<br />

apoptosis. The Bcl-2 family members play a central role in these apoptotic events. Protein<br />

complexes that include transcription factors <strong>and</strong> coactivators (p300, P/CAF HATs) or<br />

repressors (HDACs) frequently govern the expression of this specific set of genes in order to<br />

delicately balance the protein levels of anti- <strong>and</strong> pro-apoptotic members of this family <strong>and</strong><br />

eventually determine the cell fate. The main focus of <strong>our</strong> work is to dissect the regulatory<br />

pathways determining the cellular fate under diverse stress conditions. We have observed that<br />

differential composition of the transcription co-activator complexes under diverse stress<br />

conditions fine tune the protein levels of pro- <strong>and</strong> anti apoptotic members of the Bcl-2 family<br />

determining whether the cell will survive or die through apoptosis<br />

. In particular, we have studied the expression of Bid, a pro-apoptotic member of the Bcl-2<br />

family, under conditions known to activate the p53 <strong>and</strong>/or Hif-1" transcription factors<br />

(binding sites for both transcription factors have been identified in the Bid promoter region).<br />

We have identified this way the cellular components regulating the mRNA synthesis of Bid<br />

under diverse stress conditions. In this context <strong>our</strong> studies have provided evidence that two<br />

components of the p300/CBP complex namely Strap <strong>and</strong> P/CAF directly or indirectly mediate<br />

critical signalling events that regulate both p53 <strong>and</strong> Hif-1" transcriptional activities under<br />

hypoxia.<br />

- 499 -


Notes<br />

- 500 -


Notes<br />

Notes<br />

- 501 -


- 502 -


Session XIV : Transcriptional <strong>and</strong> translational control<br />

- 503 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 1<br />

MAPKs <strong>signaling</strong> cascades-mediated transcriptional regulation in H9c2 cardiac cells<br />

exposed to oxidative stress<br />

Ioanna-Katerina Aggeli, Catherine Gaitanaki <strong>and</strong> Isidoros Beis<br />

Department of Animal <strong>and</strong> Human Physiology, School of Biology, Faculty of Sciences,<br />

University of Athens, Panepistimioupolis, Athens 157 84, GREECE, E-mail :<br />

iageli@hotmail.com<br />

One of the most important insults that cardiomyocytes experience in vivo is an increase in the<br />

levels of reactive oxygen species (ROS) i.e. during ischemia, reperfusion, as well as, in the<br />

failing myocardium. MAPKs have been found to play a pivotal role in transducing the<br />

oxidative stress-signal in cardiomyocytes. Accordingly, we found that all three subfamilies<br />

(ERKs, JNKs <strong>and</strong> p38) were phosphorylated thus activated maximally at 5-15 min of H2O2<br />

treatment (200µ)), declining thereafter (immunoblotting). In an effort to define possible<br />

substrates of the MAPKs activated, the phosphorylation of ATF2 as well as c-Jun was studied<br />

(immunoblotting). Both transcription factors were equally induced after 30min <strong>and</strong> 1h,<br />

respectively, a result which indicates their involvement in the observed response. In order to<br />

decipher the <strong>signaling</strong> mechanism triggered under these conditions a number of<br />

pharmacological inhibitors of kinase pathways was used. What is more, since ATF2 <strong>and</strong> c-Jun<br />

are known AP1 components, the transcriptional regulation of possible target genes with a<br />

CRE/AP-1 element in their promoter region was examined (RT-PCR). Further studies are<br />

however required so as to establish the physiological role of these cascades’ components<br />

promoting either cell survival or apoptosis.<br />

This work was funded by PYTHAGORAS 1 grant (70/3/7399).<br />

- 504 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 2<br />

Bile acids modulate p53 transcriptional targeting in primary rat hepatocytes<br />

Joana D. Amaral, Rui E. Castro, Susana Solá, Cecília M.P. Rodrigues<br />

Centro de Patogénese Molecular, Faculty of Pharmacy, University of Lisbon, 1600-083<br />

Lisbon, Portugal. E-mail: jamaral@ff.ul.pt<br />

Ursodeoxycholic acid (UDCA) is a potent inhibitor of classical mitochondrial pathways of<br />

apoptosis. Further, UDCA prevents transforming growth factor beta1 (TGF-beta1)-induced<br />

hepatocyte apoptosis by modulating the E2F-1/p53/Bax pathway. However, the<br />

mechanism(s) by which UDCA regulates gene transcription remains to be clarified. The aim<br />

of this study was to evaluate the modulation of a specific pro-apoptotic transcriptional target<br />

of p53 by several bile acids. Primary rat hepatocytes were cotransfected with plasmid DNA<br />

encoding wild-type or mutant p53 <strong>and</strong> a reporter gene construct that utilized the bax gene<br />

promoter to drive transcription of chloramphenicol acetyltransferase (CAT). A luciferase<br />

reporter construct was used to assess transfection efficiencies. Hepatocytes were transfected<br />

<strong>and</strong> simultaneously treated with either vehicle or 100 µM of several bile acids. Twelve h<strong>our</strong>s<br />

later, 1 nM TGF-beta1 was included in the cultures. After an additional 36 h, hepatocytes<br />

were harvested for CAT ELISA <strong>and</strong> luciferase assays. Total protein extracts were also<br />

prepared <strong>and</strong> evaluated by immunoblotting. Cultures were scored for apoptotic cells by<br />

Hoechst staining. Electrophoretic mobility shift assays (EMSA) were performed in nuclear<br />

extracts from COS-7 cells, producing high levels of endogenous p53. Our results confirmed<br />

that UDCA abrogated TGF-beta1- <strong>and</strong> p53-induced hepatocyte apoptosis. DNA-binding<br />

activity of p53 was not altered by the bile acid. Nevertheless, in functional studies,<br />

hepatocytes transfected with wild-type p53 <strong>and</strong> treated with UDCA showed a marked<br />

decrease in bax transcriptional activation. When the mutant form of p53 was used, UDCA no<br />

longer exerted its protective effect. Similar results were obtained after incubation of cells<br />

with both taurine- <strong>and</strong> glycine-conjugated analogues of UDCA. However, when cells were<br />

treated with lithocholic, deoxycholic, or chenodeoxycholic acids, the pro-apoptotic gene was<br />

further activated at the transcription level, revealing that bile acids are able to differentially<br />

modulate transcription of p53-driven bax. Taken together, these data suggest that proapoptotic<br />

p53 is a specific molecular target of UDCA, further clarifying the role of bile acids<br />

at modulating apoptosis. (Supported by POCTI/SAU-FCF/62479/04 from FCT, Portugal).<br />

- 505 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 3<br />

Liver steatosis alters the expression of nuclear receptors <strong>and</strong> genes involved in bile acid<br />

metabolism<br />

Patricia Aspichueta, Oscar Briz, L<strong>our</strong>des Palacios, Ainara Cano, Xabier Buqué, Jose J.<br />

Marín, Begoña Ochoa<br />

Department of Physiology, University of the Basque Country, Faculty of Medicine <strong>and</strong><br />

Dentistry, Leioa, Spain. Email: patricia.aspichueta@ehu.es<br />

Accumulation of fat within hepatocytes is a simple steatosis <strong>and</strong> a sign of lipotoxicity that is<br />

frequently associated with changes in bile acid metabolism <strong>and</strong> cholestasis. The aim of this<br />

work was to analyze whether transport proteins <strong>and</strong> cytochromes involved in bile acid<br />

synthesis <strong>and</strong> secretion as well as some regulatory nuclear receptors are altered in obesityrelated<br />

steatosis at the mRNA expression level. For this purpose, we i) isolated hepatocytes<br />

from obese Zucker rats, an established model for non-alcoholic fatty liver disease, of 6, 9 <strong>and</strong><br />

12 weeks of age -hence, in three stages of steatosis- <strong>and</strong> from their lean littermates, ii)<br />

extracted the RNA from hepatocytes, <strong>and</strong> iii) determined specific transcript levels by realtime<br />

PCR. Our results showed that the mRNA levels of Cyp7a1 were 60% <strong>and</strong> 40% decreased<br />

in the 6- <strong>and</strong> 12-week old steatotic animal while those of Cyp27a1 were similar to lean rat<br />

hepatocytes. There was a dramatic decrease (of ,98%) in ABCG8 mRNA levels in the three<br />

steatotic groups, accompanied by an important, though less marked decrease in ABCG5<br />

expression. No changes were found in the mRNA levels of the transporters Bsep, mdr2, mrp2<br />

<strong>and</strong> Ntcp. Discordant changes were observed between several major nuclear receptors <strong>and</strong><br />

their regulated target genes. Particularly, the transcription factors studied involved in Cyp7a1<br />

expression regulation do not seem to be responsible for the changes observed in this study.<br />

The mRNA levels of farnesoid X receptor (FXR) were modified in none of the obesity-related<br />

steatotic groups, of the small heterodimer partner (SHP) were doubled only in 9 week-old<br />

obese rat hepatocytes <strong>and</strong> of the alpha-fetoprotein transcription factor (FTF) were increased in<br />

the three stages of liver steatosis. In conclusion, <strong>our</strong> results suggest that repression of Cyp7a1<br />

in obesity-related steatosis may be linked to down-regulation of ABCG5, ABCG8 <strong>and</strong> LXR,<br />

rather than to the mediation of mechanisms controlled by the FXR/SHP/FTF cascade.<br />

Supported by Grant G03/015 from the Ministerio de Sanidad y Consumo<br />

- 506 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 4<br />

Proteomic analysis for Helicobacter pylori - infected human gastric mucosa: oxidative<br />

stress - related proteome changes<br />

Hye Yeon BAEK, Joo Weon LIM, <strong>and</strong> Hyeyoung KIM*<br />

Department of Pharmacology <strong>and</strong> Brain Korea 21 Project for Medical Science, Yonsei<br />

University College of Medicine, Seoul 120-752, Korea, E-mail: ehrehrwh@hanmail.net<br />

Helicobacter pylori (H. pylori) infection leads to gastroduodenal inflammation, peptic<br />

ulceration <strong>and</strong> gastric carcinoma. Proteomic analysis for human gastric mucosa of the patients<br />

with erosive gastritis or peptic ulcer, which were either infected or non-infected with H.<br />

pylori, was used to determine the differentially expressed proteins by H. pylori in human<br />

gastric mucosa to investigate the pathogenic mechanism of H. pylori-induced gastric diseases.<br />

Mass spectrometry analysis (MALDI-TOF) of tryptic fragment <strong>and</strong> data search allowed<br />

identification of increased f<strong>our</strong> proteins (78kD glucose-regulated protein precursor,<br />

endoplasmin precursor, aldehyde dehydrogenase 2, L-lactate dehydrogenase B chain) <strong>and</strong><br />

decreased f<strong>our</strong> proteins (intracellular chloride channel protein 1, glutathione-s-transferase,<br />

heat shock protein 60, cytokeratin 8) caused by H. pylori infection in gastric mucosa. These<br />

proteins are related to cell proliferaton, carcinogenesis, cytoskeletal function, <strong>and</strong> cellular<br />

defensive mechanism. Common feature is that these proteins are related to oxidative stressmediated<br />

cell damage. In conclusion, the established gastric mucosal proteome map could be<br />

useful for the detection of diseases-related protein changes. H. pylori-induced alterations in<br />

protein expression demonstrates the involvement of oxidative stress in the pathogenesis of H.<br />

pylori-induced gastric diseases including inflammation.<br />

- 507 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 5<br />

Gene expression modulation in A549 human lung cells in response to combustiongenerated<br />

nano-sized particles.<br />

Christa Baumstark-Khan, Andrea Arenz, Christine E. Hellweg <strong>and</strong> Horst-Henning<br />

Grotheer.<br />

Cellular Biodiagnostics, Department of Radiobiology, Institute of Aerospace Medicine,<br />

German Aerospace Center, 51170 Köln, Germany. E-mail : christa.baumstarkkhan@dlr.de<br />

Cell response to different kinds of environmental pollution is complex <strong>and</strong> involves the<br />

participation of different classes of genes for different cellular outcomes (DNA repair, cell<br />

cycle control, signal transduction, inflammation, apoptosis <strong>and</strong> oncogenesis). Ambient<br />

particulate matter (PM) is a complex mixture of chemicals <strong>and</strong> particles that may be<br />

compositionally diverse depending on geography <strong>and</strong> season. High levels of ambient air<br />

pollution are associated with aggravation of asthma, respiratory morbidity, <strong>and</strong><br />

cardiopulmonary morbidity; long-term exposures to PM have been linked to possible<br />

increases in lung cancer risk, chronic respiratory disease, <strong>and</strong> death rates. Nanometer-sized<br />

particles (diameter 1.5 to 5 nm, mass numbers ranging from 1000 to 40.000 amu) which are<br />

generated by combustion processes as soot precursors show surprising features, such as water<br />

solubility <strong>and</strong> transparency, in contrast to the better known soot particles. The damaging<br />

effects of nanoparticles are attributed to the fact that they are respirable <strong>and</strong> water soluble <strong>and</strong><br />

can penetrate lung mucosa. These particles are less readily cleared than fine particles, thereby<br />

prolonging interaction with the lung epithelium <strong>and</strong> potentiating cellular damage. Particulate<br />

matter has been shown to invoke inflammatory responses after exposure in animal models. Invitro<br />

experiments have revealed that diesel exhaust <strong>and</strong> PM10 are capable to induce the<br />

release of proinflammatory cytokines such as IL-6 <strong>and</strong> IL-8 from bronchial epithelial cells<br />

mediated by the transcription factor NF-!B through a mechanism partially involving TNF- ".<br />

Thus, an important aspect of <strong>our</strong> work was to determine whether nanoparticles cause<br />

activation of NF-!B <strong>and</strong> expression of NF-!B dependent genes in pulmonary epithelial cells.<br />

Recombinant A549 lung cells (A549-NF-!B-EGFP) were incubated with different<br />

concentrations of condensation water samples collected from model flames (combustion of<br />

gaseous fuels propane <strong>and</strong> ethylene under laboratory conditions). TNF- " treated cells were<br />

used for comparison. RNA was extracted from exposed cells after various recovery times <strong>and</strong><br />

a real-time QRT-PCR assay was applied, which employs relative quantification of c<strong>and</strong>idate<br />

mRNA biomarkers. The expressions of different DNA damage inducible genes (GADD45#,<br />

p21) <strong>and</strong> NF-kB dependent genes (NFkBIA, IL-6, “!B-EGFP”) were analysed. The results<br />

show a reproducible up-regulation for the NF-kB dependent genes IL-6 (32x) <strong>and</strong> NF!BIA<br />

(13x) with maximal values for 1 <strong>and</strong> 2 hrs treatment with TNF-", while the DNA damage<br />

inducible genes are not induced. For nanoparticle treated cells, a down-regulation of NF-!B<br />

dependent genes, especially for IL-6, could be shown for high particle concentrations<br />

(Dilution ratios 1:8 to 1:20). For intermediate particle concentrations (Dilution ratio 1:100),<br />

the DNA damage inducible gene GADD45 # is up-regulated (~5x) for up to 4 hrs <strong>and</strong> for<br />

lower particle concentrations (Dilution ratio 1:200) the maximal induction value for<br />

GADD45# is about 3. NF-!B dependent gene expression is down-regulated for the first h<strong>our</strong>s<br />

of nanoparticle incubation for NF!BIA (-3x, 4 hrs). For IL-6 a first down-regulation (-6.5, 30<br />

min) is followed by a significant up-regulation for incubation times of 2 <strong>and</strong> 4 hrs.<br />

- 508 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 6<br />

p38 Mitogen-Activated Protein Kinase Pathway Regulates the Transcription of Skeletal<br />

Muscle Late Genes .<br />

Eyal Bengal1, Bennett H Penn2 <strong>and</strong> Stephen J Tapscott2.<br />

1Department of Biochemistry, Rappaport Institute for Research in the Medical<br />

Sciences,Faculty of Medicine. Technion-Israel Institute of Technology. P.O. Box 9649.<br />

Haifa 31096, Israel. email: bengal@tx.technion.ac.il <strong>and</strong> 2 Division of Human Biology,<br />

Fred Hutchinson Cancer Research Center, Seattle, Washington 98109-1024 USA.<br />

The p38 mitogen-activated protein kinase (MAPK) pathway is induced during the<br />

differentiation of proliferating myoblasts to multinucleated myotubes <strong>and</strong> is absolutely<br />

essential for this process to occur in cell cultures.<br />

Previous work showed that the p38 pathway augmented the activity of transcription factors<br />

from the MEF2 <strong>and</strong> MyoD families. Here, we present data suggesting that only a small<br />

subgroup of muscle-specific genes is regulated by p38 MAPK. A DNA microarray analysis<br />

performed on muscle cells expressing an activated MKK6 protein revealed that the p38<br />

MAPK pathway was involved in the expression of several muscle-structural genes.<br />

Expression of these late-activated genes was shifted to the early stages of differentiation by<br />

precocious activation of p38 <strong>and</strong> expression of Mef2D.<br />

To underst<strong>and</strong> how p38 MAPK regulates the transcription of these genes, we performed a<br />

chromatin immunoprecipitation (ChIP) analysis. We show that p38 activity facilitates MyoD<br />

<strong>and</strong> Mef2 binding at a subset of late-activated promoters, <strong>and</strong> the binding of Mef2D recruits<br />

Pol II.<br />

This demonstrates that a MyoD-generated feed-forward regulatory circuit, wherein factors<br />

induced by MyoD feed-forward to regulate MyoD activity at subsequent target genes, acts to<br />

temporally pattern the relative timing of gene expression during skeletal myogenesis.<br />

- 509 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 7<br />

Holger Bertelsmann a, Harald Sieme b, Dietrich Behne a <strong>and</strong> Antonios Kyriakopoulos a<br />

Selenium <strong>and</strong> zinc in equine spermatozoa: Correlations between the element<br />

concentrations in sperm nuclei <strong>and</strong> spermatozoa<br />

a Hahn- Meitner Institut, Department “Trace Element Research in the Life Science”,<br />

Glienickerstr.100, 14109 Berlin, Germany<br />

E- mail: bertelsmann@hmi.de<br />

b Hanoverian State Stud Celle, Spoerckenstr. 10, D-29221 Celle, Germany<br />

E- mail: stallions.celle@t-online.de<br />

In the sperm nuclei of mammalian species selenium has been found only in the form of sperm<br />

nuclei glutathione peroxidase (snGPx) where it is most likely bound to the chromatin of<br />

spermatozoa. Over 80 % of selenium in sperm is bound to the selenoprotein phospholipid<br />

hydroperoxide glutathione (PHGPx) peroxidase in the mid- piece of sperm. Zinc in sperm is<br />

mainly contained in the outer dense fiber (ODF) proteins of the flagella of spermatozoa. In the<br />

sperm nuclei zinc is predominately located in the chromatin to the protamine proteins. In<br />

order to investigate if the insertion of zinc <strong>and</strong> selenium in sperm chromatin is regulated the<br />

element concentrations were determined in equine spermatozoa <strong>and</strong> purified sperm nuclei.<br />

We found a significant positive correlation between the selenium concentration in<br />

spermatozoa <strong>and</strong> sperm nuclei. The same finding was obtained for the zinc concentration in<br />

spermatozoa <strong>and</strong> sperm nuclei. The results assume that the distribution of selenium <strong>and</strong> zinc<br />

in equine spermatozoa is regulated by a signal transduction pathway <strong>and</strong> in this way<br />

determining the selenium <strong>and</strong> zinc amount in the chromatin of spermatozoa.<br />

- 510 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 8<br />

Protein folding information in nucleic acids which is not present in the genetic code<br />

Jan C. Biro<br />

Homulus Foundation; 88 Howard, #1205; San Francisco, 94195 CA, USA.<br />

jan.biro@sbcglobal.net; www.janbiro,com<br />

Background: All the information necessary for protein folding is supposed to be present in the<br />

amino acid sequence. It is still not possible to provide specific ab initio structure predictions<br />

by bioinformatical methods. It is suspected that additional folding information is present in<br />

protein coding nucleic acid sequences, which is not represented by the known genetic code.<br />

Results: Nucleic acid subsequences comprising the 1st <strong>and</strong>/or 3rd codon residues in mRNAs<br />

express significantly higher free folding energy (FFE) than the subsequence containing only<br />

the 2nd residues (p


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 9<br />

Reconstitution of human hypoxia inducible factor HIF-1 in yeast cells: a simple in vivo<br />

system for identification of HIF-1 inhibitors.<br />

Georgia Braliou, Emmanouel Venieris, Alkmini Kalousi <strong>and</strong> George Simos<br />

Laboratory of Biochemistry, School of Medicine, University of Thessaly, Larissa,<br />

Greece. E-mail: simos@med.uth.gr<br />

Hypoxia inducible factor 1 (HIF-1) is the master regulator of the genes that are activated<br />

under hypoxic conditions. HIF-1 is also involved in many diseases, including cancer, <strong>and</strong> is<br />

an important target for drug development. The control of HIF-1 itself is complex <strong>and</strong> involves<br />

many post-transcriptional events triggered by hypoxia <strong>and</strong> by various <strong>signaling</strong> <strong>and</strong> oncogenic<br />

pathways that operate in metazoan cells. In order to study the basic function of human HIF-1<br />

in a simple <strong>and</strong> genetically tractable in vivo system we attempted to express it in the yeast S.<br />

cerevisiae that lacks the components of the mammalian hypoxia response pathway. We show<br />

here that inducible expression of both human HIF-1 subunits (HIF-1alpha <strong>and</strong> ARNT) in<br />

yeast is possible <strong>and</strong> leads to the formation of a transcriptional active heterodimer, as shown<br />

by hypoxia response element (HRE) - dependent production of a reporter enzyme. The<br />

activity of HIF-1 in yeast is impaired by two Hsp90 inhibitors, geldanamycin A <strong>and</strong> radicicol,<br />

<strong>and</strong> by mutations in Hsp90 co-chaperones, suggesting that the Hsp90 chaperone system is<br />

important for HIF-1 integrity. We conclude that the expression of human HIF-1 in yeast<br />

provides a model system for both functional studies <strong>and</strong> identification of compounds that can<br />

act as direct HIF-1 inhibitors.<br />

- 512 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 10<br />

Translational control of pancreatic beta cell survival : PKB-activation inhibits apoptosis<br />

through stimulation of mTOR<br />

Ying Cai, Qidi Wang, Zhidong Ling, Daniel Pipeleers, Harry Heimberg <strong>and</strong> Mark Van<br />

de Casteele<br />

(Diabetes Research Center, Brussels Free University - VUB. Laarbeeklaan 103, B-1090<br />

Brussels, Belgium. E-mail : mvdcaste@vub.ac.be).<br />

Prolonged exposure of primary beta cells to low glucose levels induces their apoptosis [1].<br />

This effect has been attributed to: a) reduced synthesis of anti-apoptotic proteins [1], b)<br />

sustained activation of AMP-activated protein kinase (AMPK) [2]. Since AMPK activators<br />

AICA-riboside <strong>and</strong> Metformin inhibit protein biosynthesis before inducing apoptosis of the<br />

cells [3], we investigated whether this inhibition could be responsible for the apoptotic effect.<br />

It has been reported that AMPK negatively regulates mTOR <strong>signaling</strong> in various cell types<br />

<strong>and</strong> thus inhibits protein synthesis [4]. We therefore examined whether AMPK activation<br />

decreases mTOR <strong>signaling</strong> in beta cells, <strong>and</strong> whether PKB, an upstream activator of mTOR,<br />

can protect the cells against apoptosis.<br />

Our data demonstrate that culture in low glucose, AICAR, or metformin, decreases<br />

phosphorylation of both ribosomal S6 protein <strong>and</strong> 4EBP protein, but not of PKB, which is<br />

similar to the effect of the specific mTOR inhibitor rapamycin. Expression of constitutively<br />

active AMPK mimicked these effects. Adenoviruses expressing constitutively active PKB<br />

(CA-PKB) decreased both low glucose <strong>and</strong> AICAR induced apoptosis, while increasing<br />

mTOR-activity in beta cells. In AICAR or low glucose, CA-PKB restored partially the mTOR<br />

<strong>signaling</strong> without influencing the phosporylation of AMPK. Inhibition of mTOR activity by<br />

rapamycin, curtailed the antiapoptotic effect of PKB. We conclude that sustained AMPKactivation<br />

decreases mTOR activity in beta cells leading to inactivation of translation factors ;<br />

this effect can be counteracted by activated PKB resulting in rescue of beta cells from<br />

apoptosis by mTOR-stimulation.<br />

- 513 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 11<br />

JAK-STAT - mediated cytokine expression in Helicobacter pylori-infected gastric<br />

epithelial cells<br />

BORAM CHA, KYUNG HWAN KIM, <strong>and</strong> HYEYOUNG KIM<br />

Department of Pharmacology, Brain Korea 21 Project for Medical Science, Yonsei<br />

University College of Medicine, Seoul 120-752, Korea. Email:<br />

chaboram@yumc.yonsei.ac.kr<br />

The JAK-STAT(Janus kinase-signal transducers <strong>and</strong> activators of transcription) cascade is<br />

known as an essential inflammatory <strong>signaling</strong> pathway. Peroxisome proliferators-activated<br />

receptor gamma (PPAR gamma) lig<strong>and</strong>, 15Deoxy- 12,14 Prosta-gl<strong>and</strong>in J2 (15d-PGJ2)is<br />

reported as an anti-inflammatory agent in rheumatoid arthritis. We previously demonstrated<br />

that H. pylori in a Korean isolate (HP99) induced proinflammatory cytokines in gastric<br />

epithelial cells. The purpose of the present study is to determine whether 1) HP99 induces<br />

proinflammatory cytokines, IL-8 <strong>and</strong> RANTES, <strong>and</strong> the activation of JAK-STAT in human<br />

gastric epithelial AGS cells, 2) 15d-PGJ2 inhibits HP99-induced IL-8 <strong>and</strong> RANTES<br />

expression in AGS cells, <strong>and</strong> 3) 15d-PGJ2 inhibits the phosphorylation of STAT-JAK in<br />

HP99-infected AGS cells. As a result, HP99 induced the expression of IL-8 <strong>and</strong> RANTES as<br />

well as the phosphorylation of JAK <strong>and</strong> STAT3 in AGS cells. PPAR gamma lig<strong>and</strong>, 15d-<br />

PGJ2 attenuated HP99-induced IL-8 <strong>and</strong> RANTES expression by inhibiting the activation of<br />

JAK-STAT in gastric epithelial AGS cells. In conclusion, PPAR gamma lig<strong>and</strong>, 15d-PGJ2<br />

might be beneficial for treatment of Helicobacter pylori- induced gastric inflammation.<br />

- 514 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 12<br />

Mechanisms of hypoxia inducible factor HIF-1alpha regulation in airway smooth muscle<br />

cells<br />

Georgia Chachami1, 2, George Simos2, Apostolia Hatziefthimiou1, Panagiotis Liakos2,<br />

Sophia Bonanou2, Paschalis-Adam Molyvdas1 <strong>and</strong> Efrosyni Paraskeva1<br />

1Laboratory of Physiology <strong>and</strong> 2Laboratory of Biochemistry, Department of Medicine,<br />

University of Thessaly, Larissa, Greece. E-mail:fparaskeva@med.uth.gr<br />

Hypoxia inducible factor 1alpha (HIF-1alpha) is the regulatory subunit of HIF-1, the key<br />

mediator of the cellular response to hypoxia. HIF-1alpha is induced by hypoxia but also at<br />

normoxic conditions by exposure to heavy metals, treatment with growth factors or oncogenic<br />

transformation. We investigate the role of HIF-1 in the physiology of the respiratory tract by<br />

the use of primary cultures of airway smooth muscle (ASM) cells derived from rabbit trachea.<br />

Smooth muscle cells are not terminally differentiated <strong>and</strong> dynamically exhibit distinct<br />

contractile <strong>and</strong> proliferative phenotypes with unique morphological, biochemical, functional<br />

<strong>and</strong> gene expression characteristics. ASM cells in primary cultures maintained in full growth<br />

medium that contains fetal bovine serum (FBS) have the tendency to acquire the proliferative<br />

phenotype. However, prolonged serum deprivation allows a subset of cells to re-acquire the<br />

morphological <strong>and</strong> functional characteristics of contractile cells within intact tissues.<br />

<strong>Expo</strong>sure of “proliferative” ASM cells to low oxygen concentration as well as to cobalt can<br />

cause a rapid increase of the intracellular levels of HIF-1alpha. The use of specific inhibitors<br />

reveals that induction of HIF-1alpha by cobalt is due to an increase of both HIF-1alpha<br />

stabilization <strong>and</strong> active protein synthesis <strong>and</strong> involves the phosphatidylinositol 3-kinase<br />

(PI3K) pathway <strong>and</strong> the production of reactive oxygen species.<br />

We are currently investigating the induction of HIF-1alpha in serum-deprived quiescent<br />

“contractile” cells <strong>and</strong> the cellular pathways involved. Cobalt induces HIF-1alpha in<br />

“contractile” cells by affecting predominantly its protein stability. On the other h<strong>and</strong>, readdition<br />

of serum to these cells also causes HIF-1alpha induction, which is not due to<br />

increased protein stability, but rather requires on-going transcription, active protein synthesis<br />

<strong>and</strong> is PI3K-dependent. Interestingly, the simultaneous incubation of “contractile” cells with<br />

cobalt <strong>and</strong> serum results in an additive induction of HIF-1alpha protein levels <strong>and</strong> more<br />

efficient nuclear localization. Moreover, simultaneous incubation of rabbit trachea strips with<br />

cobalt <strong>and</strong> serum changes their contractile properties by increasing their responsiveness to<br />

acetylcholine.<br />

- 515 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 13<br />

Transcriptional regulation of cyclooxygenase-2 in response to proteasome inhibitors<br />

involves reactive oxygen species-mediated <strong>signaling</strong> pathway <strong>and</strong> recruitment of<br />

CCAAT/enhancer binding protein-delta <strong>and</strong> CREB-binding protein<br />

Jun-Jie Chen, Wei-Chien Huang <strong>and</strong> Ching-Chow Chen<br />

Department of Pharmacology, College of Medicine, National Taiwan University,<br />

Taipei 10018, Taiwan. E-Mail: ccchen@ha.mc.ntu.edu.tw<br />

Inhibition of ubiquitin-proteasome pathway has been shown to be a promising strategy for the<br />

treatment of inflammation <strong>and</strong> cancer. Here, we show that proteasome inhibitors MG132,<br />

PSI-1 <strong>and</strong> lactacystin induce COX-2 expression via enhancing gene transcription rather than<br />

preventing protein degradation in the human alveolar NCI-H292 <strong>and</strong> A549, <strong>and</strong> gastric AGS<br />

epithelial cells. NF-IL6 <strong>and</strong> CRE, but not NF-!B elements on the COX-2 promoter were<br />

involved in the gene transcription event. The binding of CCAAT/enhancer binding protein<br />

(C/EBP)-beta <strong>and</strong> C/EBP-delta to the CRE <strong>and</strong> NF-IL6 elements, as well as the recruitment of<br />

CBP <strong>and</strong> the enhancement of histone H3 <strong>and</strong> H4 acetylation on the COX-2 promoter was<br />

enhanced by MG132. However, it did not affect the total protein levels of C/EBP-beta <strong>and</strong><br />

C/EBP-delta. MG132-induced DNA-binding activity of C/EBP-delta but not C/EBP beta<br />

was regulated by p38, PI3K, Src <strong>and</strong> PKC. Small interfering RNA of C/EBP-delta suppressed<br />

COX-2 expression, further strengthening the role of C/EBP-delta in COX-2 gene<br />

transcription. In addition, the generation of intracellular reactive oxygen species (ROS) in<br />

response to MG132 contributed to the activation of MAPKs <strong>and</strong> Akt. These findings reveal<br />

that the induction of COX-2 transcription induced by proteasome inhibitors requires ROSdependent<br />

protein kinases activation <strong>and</strong> the subsequent recruitments of C/EBP-delta <strong>and</strong><br />

CBP.<br />

- 516 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 14<br />

Signal transduction on the expression of inflammatory enzymes in Helicobacter pylori -<br />

infected gastric epithelial cells<br />

Soon Ok Cho, Hyeyoung Kim, Kyung Hwan Kim<br />

Department of Pharmacology <strong>and</strong> Brain Korea 21 Project for Medical Science<br />

Yonsei University College of Medicine, Seoul 120-752, Korea<br />

E-mail:ehrehrwh@hanmail.net<br />

Cyclooxygenase-2 (COX-2) <strong>and</strong> inducible nitric oxide synthase (iNOS) are important<br />

enzymes that mediate inflammatory processes. Oxygen radicals are important regulators in<br />

Helicobacter pylori (H. pylori)-induced gastric ulceration <strong>and</strong> carcinogenesis, COX-2 <strong>and</strong><br />

iNOS may be regulated by oxidant-sensitive transcription factors, NF-kB <strong>and</strong> AP-1. In<br />

addition, the binding sites for NF-kB <strong>and</strong> AP-1 are found in the promoter region of COX-2<br />

<strong>and</strong> iNOS gene. Present study aims to investigate whether H. pylori-induced expressions of<br />

COX-2 <strong>and</strong> iNOS are regulated by NF-kB <strong>and</strong> AP-1 in gastric epithelial AGS cells, <strong>and</strong><br />

whether the transcriptional regulation of COX-2 <strong>and</strong> iNOS are inhibited by transfection with<br />

mutant genes for Ras (ras N-17), c-jun (TAM67), <strong>and</strong> IkBa (MAD3). As a result, H. pylori<br />

induced the expression of mRNA <strong>and</strong> protein for COX-2 <strong>and</strong> iNOS via activation of NF-kB<br />

<strong>and</strong> AP-1. Transfection with mutant genes for Ras (ras N-17), c-jun (TAM67), <strong>and</strong> IkBa<br />

(MAD3) inhibited H. pylori-induced COX-2 <strong>and</strong> iNOS expression in AGS cells. In<br />

conclusion, H. pylori induced activation NF-kB <strong>and</strong> AP-1 <strong>and</strong> thus COX-2 <strong>and</strong> iNOS<br />

expression in gastric epithelial cells. Either suppression of NF-kB or AP-1 may inhibit H.<br />

pylori-induced COX-2 <strong>and</strong> iNOS expression in gastric epithelial cells.<br />

- 517 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 15<br />

Serine/threonine protein phosphatases <strong>and</strong> MCP-1 in Helicobacter pylori–infected<br />

gastric epithelial cells<br />

Hae-Yun Chung, Boram Cha, Ji Hye Seo, Kyung Hwan Kim, <strong>and</strong> Hyeyoung Kim<br />

Department of Pharmacology, Brain Korea 21 Project for Medical Science, Yonsei<br />

University College of Medicine, Seoul 120-752, Korea. E-mail :<br />

hchung02@yumc.yonsei.ac.kr<br />

The phosphorylation of proteins controlled by protein kinases <strong>and</strong> phosphatases is a major<br />

mechanism that regulates cellular processes such as inflammation. It has been reported that<br />

the activity of at least 30% of all proteins can be regulated by phosphorylation in eukaryotes.<br />

Among these proteins, MAPK <strong>and</strong> some transcription factors play a pivotal role in<br />

inflammation. We previously demonstrated that Helicobacter pylori in a Korean isolate<br />

(HP99) induced chemokine expression by activating MAPK <strong>and</strong> transcription factors, NF-kB<br />

<strong>and</strong> AP-1 in gastric epithelial AGS cells. To determine the role of phosphorylation /<br />

dephosphorylation in HP99-induced inflammation, we analyzed the expression of<br />

phosphatases, the activation of MAPK <strong>and</strong> AP-1 <strong>and</strong> the expression of chemokine, MCP-1 in<br />

AGS cells stimulated with HP99 <strong>and</strong> cultured in the presence or absence of a serine/threonine<br />

phosphatase inhibitor, okadaic acid (OA). As a result, HP99 induced the expression of protein<br />

phosphatases, PP1 <strong>and</strong> PP2A as well as the activation of MAPK <strong>and</strong> AP-1, <strong>and</strong> the induction<br />

of MCP-1 in AGS cells, which was augmented by OA. In conclusion, gastric epithelial cells<br />

induced the expression of PP1 <strong>and</strong> PP2A in response to HP99 as a defense mechanism against<br />

inflammatory chemokine expression by inhibiting the activation of MAPK <strong>and</strong> AP-1.<br />

- 518 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 16<br />

Regulation of Vascular Endothelial Growth Factor (VEGF) expression by Human<br />

Papillomavirus 18 E6 oncoprotein in HeLa cells<br />

Nicolas Clere1, Laurent Bermont1, Sylvie Fauconnet1, Isabelle Lascombe1, Evelyne<br />

Chezy2 <strong>and</strong> Christiane Mougin1,2<br />

(1) University of Franche-Comté, EA 3181–IFR133, UFR des Sciences Médicales et<br />

Pharmaceutiques, Besançon, France, (2) Laboratoire de Biologie Cellulaire et<br />

Moléculaire, Centre Hospitalier Universitaire, Besançon, France<br />

Human papillomaviruses (HPVs) are a large group of small DNA viruses which infect<br />

cutaneous or mucosal epithelia. High risk HPVs are the primary cause of cervical cancer, the<br />

second most prevalent cancer in women worldwide, with the continuous expression of early<br />

viral oncogene E6 secondary to viral DNA integration into the host genome. E6 is able to<br />

immortalize cells <strong>and</strong> induces malignant transformation by inactivating p53 protein. However,<br />

this expression is not sufficient for the progression of solid tumor. Tumor growth <strong>and</strong><br />

metastasis are often influenced by the production of VEGF, a key mediator of angiogenesis.<br />

In solid tumor, angiogenesis is associated with VEGF expression by tumor cells. VEGF exists<br />

as five isoforms of 121, 145, 165, 189 <strong>and</strong> 206 amino-acids. In cervical cancer, regulation of<br />

VEGF expression is complex <strong>and</strong> poorly described. Thus, we addressed the question of<br />

whether E6 oncoprotein could modulate the expression of VEGF in HPV 18 cervical derived<br />

cancer cells. We performed a quantitation of VEGF by real time RT-PCR from HeLa cells<br />

harboring HPV18 <strong>and</strong> a wild type p53. Three transcripts encoding VEGF 121, 165 <strong>and</strong> 189<br />

were expressed. Synthetic siRNA targeting E6 were also applied to inhibit oncogene<br />

expression <strong>and</strong> to evaluate the role of the E6 oncoprotein on VEGF expression. Among the<br />

three VEGF transcripts expressed, those encoding VEGF121 were majority <strong>and</strong> represented<br />

90% of all isoforms. The silencing of E6 reduced by 50% the level of these transcripts when<br />

compared to cells transfected with control siRNA. In the same way, a decrease of<br />

approximatively 30% of secreted VEGF was observed in transfected cells. According to <strong>our</strong><br />

results, we may raise the hypothesis that E6 induced VEGF expression in a p53-independent<br />

manner. Additionnal data will be presented at the meeting.<br />

- 519 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 17<br />

Lipid metabolism alterations induced by mitochondrial dysfunction in preadipocytes: a<br />

new role for the transcription factor CREB<br />

Aurélia De Pauw 1 , Sebastien Vankoningsloo 1 , Andrée Houbion 1 , Silvia Tejerina 1 ,<br />

Catherine Demazy 1 , Françoise de Longueville 2 , Vincent Bertholet 2 , Patricia Renard 1 ,<br />

José Remacle 1,2 , Martine Raes 1 <strong>and</strong> Thierry Arnould 1<br />

1<br />

Laboratory of Biochemistry <strong>and</strong> Cellular Biology, University of Namur (F.U.N.D.P.),<br />

Rue de Bruxelles, 61, 5000 Namur, Belgium. Email : HYPERLINK<br />

"mailto:thierry.arnould@fundp.ac.be" thierry.arnould@fundp.ac.be,<br />

2<br />

Eppendorf<br />

Array Technologies, Rue du Séminaire, 12, 5000 Namur, Belgium.<br />

The role of mitochondria in cellular lipid homeostasis, adipogenesis <strong>and</strong> insulin sensitivity is<br />

now evidenced in various pathologies resulting from genetic alterations or chronical exposure<br />

to molecules that impair mitochondrial activity. However, the molecular mechanisms<br />

underlying abnormal fat distribution induced by mitochondrial dysfunction remain poorly<br />

understood. In this study, we showed that respiratory complex III inhibition by antimycin A<br />

as well as mitochondrial protein synthesis inhibition trigger the accumulation of triglyceride<br />

vesicles in 3T3-L1 fibroblasts. We also found that antimycin A treatment triggers CREB<br />

activation in these cells. To delineate how mitochondrial dysfunction induces triglyceride<br />

accumulation in preadipocytes, we developed a low-density DNA microarray that contains 89<br />

probes allowing gene expression analysis for major effectors <strong>and</strong>/or markers of adipogenesis.<br />

We thus determined gene expression profiles in 3T3-L1 incubated with antimycin A <strong>and</strong><br />

compared the patterns obtained with differentially expressed genes during the c<strong>our</strong>se of in<br />

vitro adipogenesis induced by a st<strong>and</strong>ard proadipogenic cocktail. After a 8-day treatment, a<br />

set of 39 genes was found to be differentially expressed in antimycin A-treated cells among<br />

which CHOP-10 (C/EBP homologous protein-10), GPDmit (mitochondrial glycerol-<br />

3phosphate dehydrogenase), <strong>and</strong> SCD1 (stearoyl-CoA desaturase 1). We also demonstrated<br />

that the overexpression of dominant negative mutants of CREB (K-CREB <strong>and</strong> M1-CREB) as<br />

well as siRNA transfection that respectively disrupt the factor activity <strong>and</strong> expression inhibit<br />

antimycin A-induced triglyceride accumulation. These results highlight a new role for CREB<br />

in the control of triglyceride metabolism during the adaptative response of preadipocytes to<br />

mitochondrial dysfunction.<br />

T. Arnould <strong>and</strong> A. De Pauw are respectively Research Associate <strong>and</strong> Research Assistant of<br />

FNRS (Fonds National de la Recherche Scientifique, Brussels, Belgium).S. Tejerina is a<br />

recipient of a CUD (Coopération Universitaire au Développement) fellowship.<br />

- 520 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 18<br />

Transcriptional regulation of cell proliferation signalling pathways by the transcription<br />

factor E2F2 during liver regeneration<br />

Igotz Delgado, Yuri Rueda, Olatz Fresnedo <strong>and</strong> Begoña Ochoa.<br />

Department of Physiology, University of the Basque Country Medical School, PO Box<br />

699, Bilbao, Spain. E-mail: igotzdelgado@yahoo.es<br />

E2F transcription factors are key regulators of cell cycle progression; they are fundamental<br />

for crossing the G1/S checkpoint as they regulate the expression of genes required for DNA<br />

replication. During the G1 phase, pocket proteins such as pRb (retinoblastoma protein) are<br />

blocking E2Fs activity. Cyclin dependent kinases phosphorylate pRbs so that E2Fs are<br />

released. When associated with pRb family members, the E2Fs function as transcriptional<br />

repressors, whereas free E2F activates transcription. The pRb-E2F pathway is a downstream<br />

target of mitogenic signalling pathways. E2F2 transcription factors are activator members of<br />

E2F family as they induce cell cycle entry in quiescent cells. Liver regeneration is considered<br />

an appropriate model to analyse factors related to cell cycle because, following an hepatic<br />

tissue resection, 95% of hepatic cells (which normally stay quiescent) enter cell cycle to start<br />

regeneration while maintaining their metabolic functions. We have addressed the role of E2F2<br />

in liver regeneration using the well characterised paradigm of 70% partial hepatectomy (PH)<br />

in wild-type <strong>and</strong> E2F2-/- mice <strong>and</strong> high-density oligonucleotide arrays to identify genes in<br />

which the change in expression in response to hepatectomy differed. Measurements were<br />

performed at 48 h<strong>our</strong>s post-hepatectomy, as hepatocyte replication peaked at this time in the<br />

regenerating mouse. E2F2-/- mice exhibited a delayed liver regeneration, showing the<br />

importance of this transcription factor in the regenerative process. The microarray analysis<br />

showed that genes belonging to antiproliferative <strong>signaling</strong> pathways such as Rras, dual<br />

specific phosphatases 6 <strong>and</strong> 16 or Map kinase-activated protein kinase 2 were upregulated in<br />

E2F2-/- mice during the regenerative process, this explaining, at least in fact, that liver<br />

regeneration is retarded in the absence of E2F2. In conclusion, regardless of its action on<br />

proliferation, the transcription factor E2F2 enhances the expression of some genes involved in<br />

antiproliferative signalling pathways of the liver.<br />

Supported by the Basque Government (Etortek02, IE019)<br />

- 521 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 19<br />

Transcriptional Regulation of Human Plasminogen Activator Inhibitor-1 Gene<br />

Expression by IGF-1 <strong>and</strong> Insulin: Role of HIF-1 <strong>and</strong> Forkhead transcription factor<br />

Elitsa Y. Dimova, Daniela Fluegel <strong>and</strong> Thomas Kietzmann<br />

Department of Biochemistry, Faculty of Chemistry, University of Kaiserslautern, D-<br />

67663 Kaiserslautern, Germany. E-mail: edimova@gwdg.de<br />

One risk factor for pathological conditions associated with hypoxia or hyperinsulinemia is<br />

plasminogen activator inhibitor-1 (PAI-1). IGF-1 <strong>and</strong> insulin have been shown to induce PAI-<br />

1 expression but the molecular mechanisms behind these effects have not been fully<br />

elucidated. Therefore, we searched for putative IGF-1- <strong>and</strong> insulin-responsive element(s),<br />

transcription factor(s) <strong>and</strong> signalling components mediating the IGF-1 <strong>and</strong> insulin-dependent<br />

human PAI-1 expression, using HepG2 cells as a model system. IGF-1 <strong>and</strong> insulin treatment<br />

enhanced PAI-1 expression <strong>and</strong> promoter activity. Mutation of the hypoxia responsive<br />

element (HRE), which could bind hypoxia-inducible factor-1 (HIF-1), nearly abolished the<br />

induction by IGF-1 <strong>and</strong> insulin. Mutation of E-boxes E4 <strong>and</strong> E5 did not affect the IGF-1- <strong>and</strong><br />

insulin-dependent activation of the PAI-1 promoter constructs under normoxia but abolished<br />

the effects of IGF-1 <strong>and</strong> insulin under hypoxia. Additionally, mutation of a putative Forkhead<br />

binding site abolished the insulin-dependent activation under normoxia, but not under<br />

hypoxia. Overexpression of Forkhead transcription factor resulted in suppression of the<br />

hypoxia–mediated response of the human PAI-1 promoter constructs, but not in abolishment<br />

of the insulin effect. Further the IGF-1 <strong>and</strong> insulin-induced up-regulation of PAI-1 was<br />

associated with activation of HIF-1alpha IGF-1 enhanced HIF-1alpha protein levels <strong>and</strong><br />

HIF-1 DNA-binding to each HRE, E4 <strong>and</strong> E5 as shown by EMSAs. Inhibition of the PI(3)K<br />

<strong>and</strong> MAPK pathways by specific inhibitors or by overexpression of the key enzymes e.g.<br />

dominant-negative PDK1, TRB3 which inhibits Akt/PKB or dominant-negative Raf-1<br />

revealed that IGF-1 activates human PAI-1 gene expression through activation of PI(3)K <strong>and</strong><br />

ERK1/2 whereas insulin-induced PAI-1 gene expression occurred mainly via the MAPK<br />

pathway.<br />

- 522 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 20<br />

Daily swimming diminishes the isolation-induced changes on the level of glucocorticoid<br />

receptor <strong>and</strong> heat shock protein 70 in rat brain<br />

Filipovi' M. Dragana, Gavrilovi' Ljubica, Dronjak Sladjana <strong>and</strong> Radoj(i' B. Marija<br />

Institute of Nuclear Sciences «Vin(a», Laboratory of Molecular Biology <strong>and</strong><br />

Endocrinology,11000 Belgrade, Serbia <strong>and</strong> Montenegro. E-mail : dragana@vin.bg.ac.yu<br />

Animals chronically exposed to a particular stressor display an exaggerated response of<br />

hypothalamo-pituitary adrenal axis to various novel stressors, as judged by plasma level of<br />

glucocorticoids <strong>and</strong> adrenocorticotropic hormone (ACTH). The molecular mechanism of such<br />

observation is believed to involve the action of glucocorticoid receptor (GR) <strong>and</strong> its nuclear<br />

transporter heat shock protein 70 (Hsp70). The main aim of the present study was to define<br />

the change on the level of GR <strong>and</strong> Hsp70 in a cytosol of hippocampus <strong>and</strong> brain cortex of<br />

Wistar rat males exposed to 21 daily isolation or isolation plus 15 minute/daily swimming as<br />

chronic stressors, sole or in combination with 2h acute stress of immobilization or cold (40C).<br />

The level of GR <strong>and</strong> Hsp70 were quantified by Western immunoblotting. Plasma ACTH <strong>and</strong><br />

corticosterone (CORT) were measured by chemiluminescent method <strong>and</strong> RIA, respectively. A<br />

significant decrease of both proteins was observed in acutely stressed rats. Chronic stress<br />

conditions led to moderate decreases in both cytosol GR <strong>and</strong> Hsp70 level. Isolation as chronic<br />

stressor caused reduced responsiveness to a novel acute stressors, judged by the cytosol GR<br />

<strong>and</strong> Hsp70. This was not observed when isolation plus swimming preceeded the application<br />

of the acute stressors. The only exeption was GR level when isolation plus swimming were<br />

followed by immobilization. The obtained results led to the conclusion that 15 minute/daily<br />

swimming may diminish chronic social isolation-induced changes on the levels of<br />

glucocorticoid receptor <strong>and</strong> heat shock protein Hsp70 in cytosol of hippocampus <strong>and</strong> brain<br />

cortex of rats.<br />

- 523 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 21<br />

Nemo-Like Kinase Inhibits Androgen Receptor Transcriptional Activity in Prostate<br />

Cancer Cells<br />

Katayoon H. Emami, Lisha G. Brown, Tiffany EM Pitts, <strong>and</strong> Eva Corey<br />

GU Cancer Research Laboratory, Department of Urology, University of Washington,<br />

Seattle, WA 98195, USA. Email: ecorey@u.washington.edu<br />

Androgen receptor (AR) <strong>signaling</strong> <strong>and</strong> its interactions with other <strong>signaling</strong> pathways play<br />

important roles in growth <strong>and</strong> differentiation of prostate <strong>and</strong> prostate cancer (CaP) cells.<br />

Nemo-like kinase (NLK), a MAP kinase, has been reported to affect the activity of various<br />

transcriptional factors via its interactions with co-activators #-catenin <strong>and</strong> CBP/p300. Because<br />

MAP Kinases have been implicated in regulation of the AR <strong>signaling</strong> pathway <strong>and</strong> AR<br />

<strong>signaling</strong> utilizes both #-catenin <strong>and</strong> CBP/p300 as coactivators, y we examined the effects of<br />

NLK on AR <strong>signaling</strong> in CaP cells.<br />

Our data show that inhibition of NLK expression by siRNA upregulates AR promoter<br />

activity, while overexpression of NLK dramatically inhibits the activation of both an artificial<br />

ARE <strong>and</strong> the PSA promoter in LNCaP prostate cancer cells. A kinase-dead NLK(K155M)<br />

inhibited only ~50% of the AR transcriptional activity in LNCaP cells, implying that the<br />

kinase activity of NLK is important for this inhibition but that other mechanisms are also<br />

involved. Our data show that the mechanisms involved in the reduced transcriptional activity<br />

of AR include disruption of interactions of AR with #-catenin <strong>and</strong> p300, decreased AR-DNA<br />

binding, <strong>and</strong> alteration of the subcellular localization of AR. We undertook further studies to<br />

investigate the biological effects associated with the inhibitory role of NLK in AR-directed<br />

transcription <strong>and</strong> showed that NLK increases the rate of apoptosis in these cells.<br />

Further studies of the roles of NLK in regulation of prostate cancer are warranted, since<br />

underst<strong>and</strong>ing the molecular mechanism of regulation of AR transcriptional activity in CaP<br />

cells is essential for identification of new targets to control this disease <strong>and</strong> may have a<br />

dramatic impact on the therapeutic strategies adopted for the prevention <strong>and</strong> treatment of CaP.<br />

- 524 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 22<br />

The effects of dexamethasone on the expression of alpha- <strong>and</strong> beta- isoforms of<br />

glucocorticoid receptor in cultured human myoblasts <strong>and</strong> myotubes<br />

Dragana M. Filipovi'1, Katarina Mi)2, Toma0 Mar)2 <strong>and</strong> Zoran Grubi( 2.<br />

1Institute of Nuclear Sciences “Vin(a”, Laboratory of Molecular Biology <strong>and</strong><br />

Endocrinology, 11000 Belgrade, Serbia <strong>and</strong> Montenegro, 2Institute of Pathophysiology,<br />

School of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia. E-mail :<br />

dragana@vin.bg.ac.yu<br />

Critical illness myopathy is one of the major problems in intensive care units. High doses of<br />

glucocorticoids (GCs) received by such patients are assumed to play essential role in the<br />

etiology of this disorder, however, the precise pathophysiologic mechanisms underlying this<br />

GC action are mostly unknown. Recovery is often incomplete in these patients <strong>and</strong> the final<br />

outcome depends at least partly on muscle regeneration which might also be affected by<br />

glucocorticoids. In order to approach this problem systematically, we first studied the<br />

expression of glucocorticoid receptor (GR) isoforms, GR alpha <strong>and</strong> GR beta differentially in<br />

human myoblasts <strong>and</strong> myotubes which are the precursors of muscle regeneration. We also<br />

studied the effects of synthetic GC Dexamethasone (Dex) on the GR alpha <strong>and</strong> GR beta<br />

expression since this is the major regulatory mechanism modulating GC sensitivity in<br />

peripheral tissues including muscle. Cultured human myoblasts <strong>and</strong> myotubes were treated by<br />

Dex (0.2µM <strong>and</strong> 1µM) for 24h. Expression pattern of the GR isoforms in control <strong>and</strong> Dextreated<br />

cells were followed at both, mRNA <strong>and</strong> protein levels using RT-PCR <strong>and</strong> Western<br />

blot, respectively. Up to now we found both receptor subtypes expressed already at the<br />

earliest (myoblast) as well as at the myotube stage, however we found no differential response<br />

between these two stages with regard to the Dex – regulated expression of GR subtypes. This<br />

finding suggests that response to GCs at the receptor level is established very early in the<br />

myogenesis of the human muscle <strong>and</strong> remains practically unchanged afterwards. We will<br />

continue this study by approaching other mechanisms underlying GC effects on muscle<br />

regeneration.<br />

- 525 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 23<br />

Antagonistic function of S100 proteins regulates AP-1 dependent gene expression during<br />

tumor development<br />

Christoffer Gebhardt,1,4 Michiel van der S<strong>and</strong>en,1 Lars Hummerich,2 Kai Breuhahn,3<br />

Julia Németh,1 Meinhard Hahn,2 Alex<strong>and</strong>er Enk,4 Gerhard Fuerstenberger,5 Cornelia<br />

Mauch,6 Peter Schirmacher,3 Peter Lichter,2 Peter Angel1 <strong>and</strong> Jochen Hess1<br />

1Division of Signal Transduction <strong>and</strong> Growth Control; 2Division of Molecular Genetics;<br />

5Division of Eicosanoids <strong>and</strong> Tumor Development, Deutsches Krebsforschungszentrum,<br />

D-69120 Heidelberg, Germany; 3Department of Pathology; 4Department of<br />

Dermatology, University Hospital Heidelberg, 69120 Heidelberg, 6Department of<br />

Dermatology, University of Cologne, Joseph-Stelzmann-Str. 9, D-50924 Köln, Germany<br />

Email: c.gebhardt@dkfz.de<br />

Despite compelling data demonstrating a direct link between altered expression of S100<br />

proteins located on human chromosome 1q21 <strong>and</strong> common epithelial malignancies, the<br />

knowledge of their function <strong>and</strong> mode of action in epithelial cells <strong>and</strong> in the evolution or<br />

progression of cancer is largely unknown. Here, we have identified a novel <strong>signaling</strong> pathway<br />

in epithelial cells initiated by extracellular S100A8/A9 heterodimers resulting in the<br />

activation of AP-1-dependent gene expression. Importantly, co-expression of S100A3 inhibits<br />

S100A8/A9 mediated AP-1 activation, which is in line with repression of this gene during<br />

chemically induced skin carcinogenesis in mice suggesting a negative role for S100A3 in<br />

epithelial malignancy. We found elevated levels of MMP2 <strong>and</strong> MMP9, two well-known AP-1<br />

regulated genes, <strong>and</strong> identified S100A6 as an additional target gene of S100A8/A9 <strong>signaling</strong><br />

in epithelial cells. Using tissue-microarrays, significant co-expression of S100A8 <strong>and</strong> S100A9<br />

in concert with phosphorylation of c-Jun <strong>and</strong> elevated levels of S100A6 protein was evident<br />

in cutaneous squamous cell carcinomas of patients. Altogether, <strong>our</strong> data suggest that targeting<br />

the net activity of S100 induced <strong>signaling</strong> represents an auspicious strategy for innovative<br />

cancer prevention <strong>and</strong>/or therapy.<br />

- 526 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 24<br />

Study of a molecular retrograde communication that involves SREBP-1 <strong>and</strong> SREBP-2 in<br />

143 B cell responding to a lysosomal sucrose accumulation<br />

Sophie Goblet1, Andrée Houbion2, Isabelle Hamer1, Laetitia Payen2, Marie-Jeanne<br />

Vertez1, Michel Jadot1 <strong>and</strong> Thierry Arnould2<br />

1. URPhyM : Unité de Recherche en Physiologie Moléculaire, Faculté de Médecine<br />

(F.U.N.D.P.)<br />

2. URBC : Unité de Recherche en Biologie Cellulaire, Faculté des Sciences (F.U.N.D.P)<br />

61 rue de Bruxelles, 5000 Namur, Belgium. e-mail : thierry.arnould@fundp.ac.be<br />

While numerous lysosomal diseases have been described to result from mutations in genes<br />

encoding lysosomal hydrolases leading to lysosomal enzyme activity deficiency <strong>and</strong>/or<br />

accumulation of non-hydrolysable substrates, cell <strong>signaling</strong> initiated by lysosomal storage<br />

disorder is still poorly understood. We thus intended to identify new <strong>signaling</strong> pathway(s)<br />

activated by a lysosomal storage of sucrose in human osteosarcoma cells (143B).<br />

We found that these cells overexpress lysosomal markers in response to sucrose <strong>and</strong> we<br />

characterized some aspects of the cell phenotype during sucrose accumulation. Among<br />

several transcription factors studied, we next showed that members of SREBP (sterolresponsive<br />

element binding protein) transcription factors are activated in response to a sucrose<br />

lysosomal storage as luciferase reporter constructs driven by the FAS (fatty acid synthase),<br />

HMG-CoA reductase, LDLR (low density lipoprotein receptor) promoters are found to be<br />

activated in response to a sucrose incubation. We found that both SREBP-1 <strong>and</strong> SREBP-2<br />

protein accumulate in the nucleus of the sucrose-treated cells. These data are supported by<br />

findings showing that SREBP activation is correlated with a modification in cholesterol<br />

distribution in the sucrose-treated cells as observed after a filipin staining. Furthermore, 25hydroxycholesterol<br />

completely inhibits SREBP activation <strong>and</strong> dramatically reduces SREBP-2<br />

abundance in the nucleus of sucrose-treated cells. We also investigated <strong>signaling</strong> pathways<br />

that could lead to the activation or nuclear translocation of SREBP <strong>and</strong> found that SREBP<br />

activation is sensitive to PI3-K <strong>and</strong> MEK1/2 inhibitors. Taken together, these data suggest<br />

that lysosomal accumulation of a non-hydrolysable substrate such as sucrose, triggers the<br />

activation of a cell <strong>signaling</strong> pathway leading to SREBP activation.<br />

T. Arnould is a Research Associate of FNRS (Fonds National de la Recherche Scientifique,<br />

Brussels, Belgium). S. Goblet is a recipient of a FRIA fellowship. We thank the governement<br />

of the french-speaking community (Belgium) for financial support through an ARC<br />

(Action de Recherche Concertée) programme.<br />

- 527 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 25<br />

Effect of NF-kB inhibition on the development of endometriosis in a nude mouse model<br />

Reinaldo González Ramos1, Anne Van Langendonckt1, Sylvie Defrère1, Marcel<br />

Mettlen2 <strong>and</strong> Jacques Donnez1<br />

1 Gynecology Unit, 2 Cell Unit, Université Catholique de Louvain, 1200 Brussels,<br />

Belgium. E-mail: donnez@gyne.ucl.ac.be<br />

Introduction: Endometriosis is a gynecological disorder characterized by the ectopic growth<br />

of endometrial tissue, in which inflammation plays an important role. The transcription factor<br />

nuclear factor-kappaB (NF-kB), has a key function in the transduction of proinflammatory<br />

signals. The NF-kB pathway is activated in response to TNF" in human endometrial <strong>and</strong><br />

endometriotic stromal cells.<br />

The purpose of this study is to test BAY 11-7085 (an inhibitor of IkB phosphorylation) <strong>and</strong><br />

SN-50 (inhibitor of NF-kB nuclear translocation) on the development of endometriosis <strong>and</strong><br />

the proliferation activity of ectopic endometrium in a murine model of endometriosis.<br />

Material <strong>and</strong> Methods: Endometriosis was induced in nude mice by direct intraperitoneal<br />

injection of minced human menstrual endometrium (200µl), labelled with CFDA-SE (a<br />

fluorescent tracker). Mice were injected on days 1, 3 <strong>and</strong> 5, either with 200 µl PBS (control<br />

mice), BAY 11-7085 (5 mg/kg in 200 µl of PBS) or SN-50 (5 mg/kg in 200 µl of PBS).<br />

Five days after endometrial injection, lesions were identified <strong>and</strong> dissected under a<br />

fluorescence microscope. Endometriosis development was evaluated using three methods:<br />

weight, fluorimetry <strong>and</strong> morphometry. Wet weight was measured for each lesion <strong>and</strong> the<br />

results were pooled per mouse. Fluorimetry involves the quantification of the fluorescence of<br />

lesions by means of a Kodak 2000MM image station. Morphometry measures the surface of<br />

endometriotic tissue on the largest section of a lesion immunolabelled with CK-22 (an<br />

epithelial marker) <strong>and</strong> CD-10 (a stromal marker) antibodies.<br />

NF-kB activation <strong>and</strong> epithelial cell proliferation were assessed after immunostaining with<br />

NF-kB <strong>and</strong> Ki-67 antibodies, respectively.<br />

Results: Six series of experiments were performed for BAY 11-7085 versus control mice <strong>and</strong><br />

three series for SN-50 versus control mice. 25 lesions were recovered in a total of 15 mice<br />

operated. There was a significant reduction in lesion weight, fluorimetry (p


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 26<br />

Heavy metals <strong>and</strong> thermal stress induce pro- or anti-apoptotic events via the p38-MAPK<br />

signal transduction pathway in Mytilus galloprovincialis<br />

Eleni G<strong>our</strong>gou, Eirini Kefaloyianni, Catherine Gaitanaki <strong>and</strong> Isidoros Beis<br />

Department of Animal & Human Physiology, School of Biology, Faculty of Sciences,<br />

University of Athens, Panepistimioupolis, Athens 15784, Greece<br />

In the present study, a number of heavy metals (copper, zinc <strong>and</strong> cadmium) as well as thermal<br />

stress was used, in order to investigate their possible effect on the p38-MAPK <strong>signaling</strong><br />

pathway, in Mytilus galloprovincialis mantle <strong>and</strong> gill tissues. All the stressful stimuli were<br />

applied to the whole organism <strong>and</strong> both mantle <strong>and</strong> gill tissues of the same animal were used<br />

for protein detection. In the mantle tissue, 1 uM Cu2+ induced a sustained p38-MAPK<br />

activation, 50 uM Zn2+ induced a transient activation <strong>and</strong> 1 uM Cd2+ a biphasic profile with<br />

maximal values at 15 <strong>and</strong> 120 min of treatment, respectively. Furthermore, 1 uM SB203580<br />

abolished the Cu2+-induced kinase phosphorylation. In gills, both Cu2+ <strong>and</strong> Zn2+, induced a<br />

considerably higher p38-MAPK activation which sustained for at least 2 h<strong>our</strong>s, whereas,<br />

Cd2+ induced a maximal kinase activation within 60 min of treatment. Hypothermia (4oC)<br />

induced a moderate p38-MAPK phosphorylation (maximised at 60 min), whereas<br />

hyperthermia (30°C) induced a rapid (within 15 min) kinase phosphorylation that sustained<br />

considerably above basal levels for at least 2 h<strong>our</strong>s. As far as it concerns the possible<br />

synergistic effects of hyperthermia <strong>and</strong> Cu2+, it was revealed that these two stressful stimuli<br />

act co-operatively, inducing an almost double p38-MAPK activation. Furthermore,<br />

investigating the possibility of pro-apoptotic or anti-apoptotic events occurring as a result of<br />

the p38-MAPK activation, it was revealed that identical stimuli may lead to pro-apoptotic<br />

events in the mantle tissue via the caspase-3 activation <strong>and</strong> to anti-apoptotic events possibly<br />

via the induction of Hsp70 over-expression in the gill tissue, indicating a tissue-specific<br />

physiological reaction.<br />

Acknowledgements: This study was funded by the Empeirikion Foundation of Athens <strong>and</strong> by<br />

G.S.R.T. (PENED 01ED5).<br />

- 529 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 27<br />

Arylhydrocarbon Receptor Repressor (AhRR) controls AhR regulated genes: clues for<br />

recruitment of HDACs<br />

Thomas Haarmann-Stemmann, Hanno Bothe, Ellen Fritsche, Josef Abel<br />

Institut für umweltmedizinische Forschung, Auf’m Hennekamp 50, 40225 Düsseldorf,<br />

Germany; E-mail: haarmann@uni-duesseldorf.de<br />

The AhRR is a recently discovered new member of the bHLH-PAS superfamily of<br />

transcription factors, closely related to the arylhydrocarbon receptor (AhR) <strong>and</strong> ARNT. These<br />

proteins are key regulators of drug metabolizing enzymes in response to xenobiotics like<br />

benzo(a)pyrene (B(a)P) or 3-methylcholanthrene (3-MC). Here we report about the regulation<br />

<strong>and</strong> function of the AhRR in different human cell lines, varying in expression <strong>and</strong> inducibility<br />

of the AhRR: the responsive HepG2 cells, the low-responsive A549 cells, <strong>and</strong> the nonresponsive<br />

human primary fibroblast <strong>and</strong> HeLa cells. Expression analyses revealed a high<br />

expression of the AhRR in fibroblast <strong>and</strong> HeLa cells <strong>and</strong> low expression in HepG2 <strong>and</strong> A549<br />

cells. Short term treatment with 10µM B(a)P enhanced the AhRR mRNA only in HepG2<br />

cells. No significant increases were found in A549, fibroblast <strong>and</strong> HeLa cells. In order to test<br />

whether methylation or acetylation status of the AhRR promoter account for the cell-specific<br />

response of the AhRR, non-responsive cell lines were treated with either Azacytidine (Aza) or<br />

Na-butyrate with or without B(a)P. Aza-treatment did not change the constitutive expression<br />

of the AhRR, whereas co-treatment with Aza <strong>and</strong> B(a)P enhanced the AhRR mRNA only in<br />

A549 cells. Bisulfit sequencing revealed no remarkable differences in methylation status of<br />

AhRR promoter between responsive <strong>and</strong> non-responsive cell lines. Studies with the HDACinhibitor<br />

indicate the importance of acetylation status in the regulation of AhRR <strong>and</strong> CYP1A1<br />

expression. Chromatin immunoprecipitation (ChIP) revealed a strong XRE-binding of AhRR<br />

in the non-responsive cell lines which disappears after Na-butyrate treatment. Coimmunoprecpitation<br />

analyses imply an AhRR-HDAC interaction. These findings indicate that<br />

the AhRR recruits HDACs <strong>and</strong> thereby controls expression of AhR-regulated genes. This<br />

model is confirmed by siRNA analyses showing that with decreasing amount of AhRR the<br />

expression of CYP1A1 increased. In summary, <strong>our</strong> data provide evidence that the AhRR is an<br />

important regulator of transcription of AhR regulated genes via its ability to recruit HDACs.<br />

From <strong>our</strong> study we suggest that the AhRR exerts a nuclear co-repressor function, but details<br />

of this mechanism remain to be elucidated.<br />

- 530 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 28<br />

Activation of NF-kappaB by different agents – influence of culture conditions.<br />

Christine E. Hellweg, Andrea Arenz, Susanne Bogner, Claudia Schmitz <strong>and</strong> Christa<br />

Baumstark-Khan.<br />

Cellular Biodiagnostics, Department of Radiobiology, Institute of Aerospace Medicine,<br />

German Aerospace Center, 51170 Köln, Germany. E-mail : christine.hellweg@dlr.de<br />

The transcription factor NF-kappaB or other components of this pathway have been identified<br />

as possible therapeutic targets in inflammatory processes, cancer <strong>and</strong> autoimmune diseases. In<br />

order to clarify the role of NF-kappaB in epithelial cells in the response to different stressors,<br />

a cell-based screening assay for activation of NF-kappaB dependent gene transcription in<br />

human embryonic kidney cells (HEK/293) was developed (J. Biomol. Screen. 2003, 8, 511-<br />

521). This assay allows detection of NF-kappaB activation by measurement of the<br />

fluorescence of the reporter protein destabilized Enhanced Green Fluorescent Protein<br />

(d2EGFP). HEK/293 cells are stably transfected with a vector carrying the d2EGFP gene<br />

under control of a synthetic promoter consisting of f<strong>our</strong> kappa B elements <strong>and</strong> the minimal<br />

thymidine kinase promoter. Treatment of the recombinant HEK-pNF-kB-d2EGFP/Neo cells<br />

with the known NF-kappaB activator tum<strong>our</strong> necrosis factor alpha (TNF-alpha) results in<br />

strong induction of NF-kappa B dependent gene expression in up to 90 % of the cells.<br />

Involvement of the p65 subunit in this response was shown using an oligonucleotide-assay.<br />

For a better characterisation of the cell-based assay, activation of the pathway by other agents,<br />

e.g. interleukin-1beta (IL-1beta), lipopolysaccharide (LPS), camptothecin, <strong>and</strong> phorbol ester<br />

(PMA), the influence of the culture conditions on NF-kappa activation by TNF-alpha, <strong>and</strong> the<br />

expression of possible target genes were examined. NF-kappa B was activated by TNF-alpha,<br />

IL-1beta <strong>and</strong> PMA in a dose-dependent manner, but not by camptothecin or LPS. TNF-alpha<br />

results in the strongest induction of NF-kappa B dependent gene expression. However, this<br />

response fluctuated from 30 to 90 %. Activation of NF-kappaB by TNF-alpha was maximal<br />

within 48 h<strong>our</strong>s after seeding of cells. With increasing confluence of the cells, the activation<br />

potential decreased. In a confluent cell layer, only 30-50 % of the cell population showed<br />

d2EGFP expression. This effect was also observed when cells were seeded in different cell<br />

densities <strong>and</strong> treated at the same time point; the higher the cell density, the lower the d2EGFP<br />

expression was after 20 h<strong>our</strong>s incubation with TNF-alpha. The underlying mechanism to this<br />

phenomenon can be the production of soluble factors by the cells inhibiting the NF-kappaB<br />

activation or the direct communication via gap junctions in the cell layer diminishing the<br />

TNF-alpha response. The quantification of the transcripts of several possible NF-kappaB<br />

target genes by quantitative PCR after treatment of cells with TNF-alpha revealed a strong<br />

induction of IkappaB alpha <strong>and</strong> IL-6. In conclusion, the sequence of events from liberation of<br />

NF-kappa B subunit p65 <strong>and</strong> its binding to DNA, the initiation of transcription <strong>and</strong> translation<br />

in response to the NF-kappaB binding have been shown for the cell-based NF-kappaB assay.<br />

The experiment conditions of this assay have to be very well controlled since cell density <strong>and</strong><br />

growth duration influence the NF-kappa B activation.<br />

- 531 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 29<br />

The transcription factor Nrf2 controls the expression of heme oxygenase-1 <strong>and</strong> Phase IIrelated<br />

genes in cells exposed to aqueous extracts of cigarette smoke<br />

Arnd Hengstermann1, Constanze Knörr-Wittmann1, Stephan Gebel1, Jawed Alam2,<br />

Thomas Müller1.<br />

1PHILIP MORRIS Research Laboratories GmbH, Cologne, Germany. 2Department of<br />

Molecular Genetics, Alton Ochsner Clinic Foundation, New Orleans, LA, U.S.A. E-mail:<br />

Arnd.Hengstermann@pmintl.com<br />

<strong>Expo</strong>sure of cells <strong>and</strong> tissues to cigarette smoke (CS) triggers a pronounced anti-oxidant<br />

response, which is hallmarked by the transcriptional up-regulation of heme oxygenase-1<br />

(hmox1), initiating a self-protection mechanism resulting in the formation of endogenous<br />

antioxidant molecules, i.e., biliverdin <strong>and</strong> bilirubin from intracellular heme moieties. To<br />

characterize the regulatory elements involved in CS-mediated hmox1 expression, we studied<br />

the expression of various hmox1 promoter/luciferase reporter constructs in NIH3T3 cells<br />

exposed to aqueous extracts of CS. The results showed that the CS-dependent expression of<br />

hmox1 is governed primarily by the distal enhancers 1 <strong>and</strong> 2, both of which contain three<br />

canonical anti-oxidant responsive element (ARE)-like stress responsive elements. These sites<br />

are potentially addressed by the transcription-factor Nrf2, a principal inducer of antioxidant<br />

<strong>and</strong> Phase II-related genes. As shown by Western-blot analysis, Nrf2 was strongly stabilized<br />

<strong>and</strong> became detectable in nuclear extracts in cells exposed to aqueous extracts of CS.<br />

Furthermore, nuclear localization of Nrf2 coincided with increased DNA binding of a putative<br />

Nrf2/MafK heterodimer to ARE, as determined by EMSA. Notably, siRNA-mediated knockdown<br />

of Nrf2 expression significantly compromised both CS-induced hmox1 promoter<br />

activation <strong>and</strong> HO-1 expression. Finally, by using this approach CS-induced expression of<br />

Phase II-related genes NAD(P)H:quinone oxidoreductase <strong>and</strong> glutamate-cysteine ligase,<br />

catalytic subunit was shown to be completely abrogated, as determined by Real Time<br />

quantitative PCR.<br />

Taken together, these results add to the underst<strong>and</strong>ing of the central role of Nrf2 in the<br />

context of CS-mediated oxidative stress by orchestrating an efficient transcriptional response<br />

aimed at resolving the stressing conditions.<br />

- 532 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 30<br />

Expression <strong>and</strong> Regulation of various tumor suppressor genes in Gastrointestinal<br />

Stromal Tumor<br />

Keun Hur, Hyuk-Joon Lee, Han-Kwang Yang.<br />

Cancer Research Institute Seoul National University College of Medicine 28 Yongon-<br />

Dong, Chongno-Gu, Seoul, Korea 110-744. E-mail: blue2001@snu.ac.kr (alternate email:<br />

longcoat@hanmail.net )<br />

Gastrointestinal stromal tumors (GISTs) are Kit/CD117 expressing mesenchymal tumors.<br />

Aberrant hypermethylation of promoter CpG isl<strong>and</strong>s is an important mechanism for the<br />

inactivation of tumor suppressor genes. CpG isl<strong>and</strong> hypermethylation occurs in relation to<br />

tumorigenesis in many human tumors. Moreover, we have previously reported that<br />

transcriptional repression of Cox-2 is caused by hyper-methylation of the Cox-2 promoter<br />

region CpG isl<strong>and</strong> in human gastric carcinoma (BBRC., 310(3), 844-851, 2003). The<br />

pathogenesis of GISTs involves a gain-of-function mutation in the KIT proto-oncogene,<br />

leading to lig<strong>and</strong>-independent constitutive activation of the KIT receptor. KIT-wild-type<br />

GISTs have shown mutually exclusive platelet-derived growth factor receptor (PDGFR)<br />

mutation <strong>and</strong> activation. However, the data on the methylation status of various tumor<br />

suppressor genes <strong>and</strong> mutation status of KIT <strong>and</strong> PDGFR in GISTs has been very limited. We<br />

attempted to determine the methylation status of 12 genes (APC, COX-2, GSTP1, MGMT,<br />

p14, p16, RUNX-3, THBS-1, TIMP-3, DAP-kinase, E-cadherin, RASSF1A), in 30 human<br />

gastrointestinal stromal tumor tissues. In addition, we confirmed the mutation status of KIT<br />

<strong>and</strong> PDGFR genes in 30 human gastrointestinal stromal tumor tissues. Thirty c-kit-positive<br />

Gastrointestinal stromal tumors were selected for this study. Genomic DNAs <strong>and</strong> RNAs from<br />

human Gastrointestinal stromal tumors were analyzed for the relationship between expression<br />

<strong>and</strong> methylation status by cDNA microarray <strong>and</strong> methylation specific PCR (MSP). CpG<br />

isl<strong>and</strong> hyper-methylation was found in 40% for APC, 46% for COX-2, 16% for GSTP1, 26%<br />

for MGMT, 60% for p14, 30% for p16, 16% for RUNX-3, 23% for THBS-1, 10% for TIMP-<br />

3, 20% for DAP-kinase, 36% for E-cadherin, 10% for RASSF1A. Our results suggest that the<br />

DNA methylation-mediated transcriptional silencing of various tumor suppressor genes is a<br />

predominant mechanism for the down-regulation of various tumor suppressor genes<br />

expression in human gastrointestinal stromal tumors.<br />

- 533 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 31<br />

Multiple isoforms of delta TA p73 transcripts accumulate in liver fluke related<br />

cholangiocarcinoma<br />

Patcharee Jearanaikoon1,5, Cheuypratoom P 1 Temduang Limpaiboon1,5 Banchob<br />

Sripa2,5 Leelayuwat C,3,5,Vajarabhongsa Bhudhisawasdi4,5<br />

1Department of Clinical Chemistry, Centre for Research <strong>and</strong> Development of Medical<br />

Diagnostic Laboratories, Faculty of Associated Medical Sciences, 2Department of<br />

clinical imunology , Faculty of Associated Medical Sciences , 3Pathology Department of<br />

Pathology, 4Department of Surgery, 5Liver Fluke <strong>and</strong> Cholangiocarcinoma Research<br />

Center, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thail<strong>and</strong>.<br />

E-mail: patjea@kku.acth<br />

Intrahepatic cholangiocarcinoma (CCA) is usually a fatal malignant neoplasm originating<br />

from bile duct epithelium. It is the highest incidence cancer in Northeast Thail<strong>and</strong> which is<br />

caused by liver fluke (Opisthorchis viverrrini) infection. No data describing the presence of<br />

oncogenic delta transactivation p73 transcripts( &TAp73) which lacking N terminal in<br />

correlation to the clinicopathological parameter has been reported in liver fluke related CCA.<br />

We examined the p73 expression in 88 tumors using IHC staining with two monoclonal<br />

antibodies specific to N <strong>and</strong> C terminal of TP73 in combination. Nearly to 80% (79/88cases)<br />

reveal no p73 expression indicating down regulation during tumor development. About 15%<br />

(13/88cases) of the IHC positive cases , demonstrate oncogenic( &TAp73) protein. To<br />

improve the lower sensitivity for the oncogenic p73detection , Nested RT –PCR was firstly<br />

established to investigate five TP73 (TAp73, p73 &ex2 , p73 &ex2/3, &Np73 <strong>and</strong> &N’p73)<br />

in 23 frozen tumor specimen. The frequency of f<strong>our</strong> p73 transcripts can be observed in wild<br />

type TA p73, &Np73,p73 &ex2 <strong>and</strong> p73 &ex2/3 with 74%,74 %,26 %<strong>and</strong> 26% ,respectively.<br />

The &Np73 oncogenic transcript is generated from the second promoter usage of p73 gene<br />

whereas p73 &ex2 <strong>and</strong> p73 &ex2/3 created by the alternate splicing of the common promoter.<br />

Our results demonstrate the accumulation of oncogenic isoforms simultaneously generating<br />

from both p73 promoters in 4/23 cases . Although, no statistical significance has been shown<br />

between the oncogenic transcript <strong>and</strong> patient survival. This might be depended on the reason<br />

that most of patients were in advanced stage.<br />

- 534 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 32<br />

Aromatase expression was not detected by immunohistochemistry in endometrial cancer<br />

Yong-Tark Jeon, Jae Weon Kim, Noh-Hyun Park, Soon-Beom Kang, Hyo-Pyo Lee,<br />

Yong-Sang Song<br />

Department of Obstetrics <strong>and</strong> Gynecology, Cancer Research Institute, College of<br />

Medicine, Seoul National University, Seoul, Korea, E-mail: yssong@snu.ac.kr<br />

Several studies suggested that aromatase could play an important role in tumor progression<br />

<strong>and</strong> prognosis in endometrial cancer because <strong>and</strong>rostenedione is converted to estrogen by the<br />

enzyme. For better underst<strong>and</strong>ing of the aromatase expression in endometrial cancer <strong>and</strong> its<br />

relation to diverse clinicopathological parameters, we conducted this study. This study was<br />

carried out with 141 endometrial cancer patients, all of whom had undergone operations in<br />

<strong>our</strong> institution from 1993 to 2002. Paraffin-embedded tissue blocks were sectioned <strong>and</strong><br />

immunostained with monoclonal anti-aromatase antibody using human placental tissue as<br />

positive control. Clinicopathological variables of all patients were also reviewed. Despite of<br />

quite high aromatase expression in positive control, there was no endometrial cancer<br />

specimen showing the enzyme expression. Our result, although need further investigation on<br />

the cause of the difference from other studies, suggested that aromatase might not have<br />

important role in endometrial cancer.<br />

- 535 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 33<br />

NF-kB, AP-1 <strong>and</strong> mitogen-activated protein kinase in chemokine expression in<br />

pancreatic Acinar cells.<br />

Kyung Don Ju, Ji Hoon Yu, Kyung Hwan Kim <strong>and</strong> Hyeyoung Kim*<br />

Department of Pharmacology, Brain Korea 21 Project for Medical Science, Yonsei<br />

University College of Medicine, Seoul 120-752, Korea,E-mail:clock1375@paran.com<br />

Cholecystokine(CCK) analogue cerulein causes pathophysiological, morphological <strong>and</strong><br />

biochemical similarities to various aspects of human pancreatitis. Doses of CCK or cerulein<br />

beyond those that cause the maximum pancreatic secretion of amylase <strong>and</strong> lipase results in<br />

pancreatitis, which is characterized by a dysregulation of the digestive enzyme production <strong>and</strong><br />

cytoplasmic vacuolization <strong>and</strong> the death of acinar cells, edema formation, <strong>and</strong> an infiltration<br />

of inflammatory cells into the pancreas. Present study aims to investigate whether cerulein<br />

induced NF-kB <strong>and</strong> AP-1 activation, MAPK phosphorylation <strong>and</strong> the expression of MCP-1 in<br />

pancreatic acinar cells, <strong>and</strong> whether these alterations were inhibited by transfection of I-kB<br />

mutant, c-jun dominant negative <strong>and</strong> inhibitors of MAPK. As a result, cerulein induced NFkB,<br />

AP-1 activation, MAPK phosphorylation <strong>and</strong> induction of chemokines(IL-8, MCP-1)<br />

expression in the cells. Both transfection of I-kB mutant, c-jun dominant negative <strong>and</strong><br />

inhibitors of MAPK inhibited cerulein-induced chemokines in acinar cells. In conclusion,<br />

cerulein activates NF-kB, AP-1 <strong>and</strong> MAPK resulting in upregulation of inflammatory MCP-1<br />

expression in acinar cells. Inhibition of NF-kB, AP-1 <strong>and</strong> MAPK might alleviate the<br />

inflammatory response in pancreatic acinar cells by suppressing chemokine expression.<br />

- 536 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 34<br />

NF!B <strong>and</strong> MAPK signalling in response to oxidative stress in skeletal myoblasts<br />

Eirini Kefaloyianni, Catherine Gaitanaki <strong>and</strong> Isidoros Beis<br />

Department of Animal <strong>and</strong> Human Physiology, School of Biology, Faculty of Sciences,<br />

University of Athens, Panepistimioupolis, Athens 15784, Greece<br />

e-mail: ibeis@biol.uoa.gr<br />

Skeletal muscle cells are continuously subjected to oxidative stress during exercise due to an<br />

increased production of reactive oxygen species (ROS). Oxidative stress has been also<br />

associated with skeletal muscle atrophy <strong>and</strong> damage in many diseases, as well as, during<br />

aging. However, skeletal muscle cells are capable of modifying their function in order to limit<br />

ROS <strong>and</strong> protect themselves <strong>and</strong> the surrounding tissues. Main signalling pathways that are<br />

activated during ROS accumulation are the MAPKs <strong>and</strong> NFkB ones, as it has been<br />

demonstrated in various cell types. Nevertheless, the precise mechanisms involved in their<br />

activation <strong>and</strong>/or function remain obscure. In this study, we examined whether NFkB<br />

participates in the response of skeletal myoblasts to oxidative stress as exemplified by H2O2<br />

<strong>and</strong> whether there is a cross talk with the MAPK signal transduction pathways. H2O2 induced<br />

a mild translocation of the transcription factor to the nucleus, in a dose <strong>and</strong> time dependent<br />

profile, as well as a moderate phosphorylation of its endogenous cytoplasmic inhibitor IkB (at<br />

Ser32/36), without any significant decrease in IkB total levels. Moreover, oxidative stress<br />

induced a strong phosphorylation of the p65 subunit of NFkB by a mechanism involving the<br />

Src kinases <strong>and</strong>, at least in part, EGFR. The Src pathway also mediated the activation of<br />

ERKs <strong>and</strong> JNKs by H2O2. Nevertheless, inhibition of these two MAPK pathways by<br />

selective inhibitors did not appear to affect this H2O2-induced phosphorylation of p65. In<br />

addition, a strong phosphorylation of p65 at Ser276 was also induced by H2O2. This<br />

phosphorylation was found to be mediated by MSK1, a known substrate of ERKs <strong>and</strong> p38-<br />

MAPK <strong>and</strong> to be Src-dependent. In conclusion, it seems that IkB degradation is crucial for<br />

NFkB translocation to the nucleus <strong>and</strong> probably these two procedures are not related with the<br />

activation of MAPKs. On the other h<strong>and</strong>, p65 phosphorylations, which are in part mediated<br />

by MAPKs pathways, are probably related to signal specificity, possibly through diverse<br />

interaction with other signalling factors.<br />

This study was funded by G.S.R.T. (PENED 01ED5).<br />

- 537 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 35<br />

Finding crossroads of signal transduction pathways targeting promoters of “disease”<br />

genes involved in pathological calcification.<br />

Alex<strong>and</strong>er Kel1, Nico Voss1, Olga Kel-Margoulis1, Evelin Katona3, Gerd Schmitz3 <strong>and</strong><br />

Edgar Wingender1,4<br />

1 BIOBASE GmbH, Halchtersche Str. 33, D-38304 Wolfenbüttel, Germany; 3 Institute<br />

for Clinical Chemistry <strong>and</strong> Laboratory Medicine University Hospital Regensburg<br />

Franz-Josef-Strauss-Allee 11, D-93053 Regensburg, Germany; 4 Dept. Bioinformatics,<br />

UKG/Univ. Göttingen, Goldschmidstr. 1, 37077 Göttigne, Germany<br />

E-mail : alex<strong>and</strong>er.kel@biobase-international.com<br />

Different signal transduction pathways converge at key molecules that master the regulation<br />

of certain cellular processes. Such crossroads of signal networks are often their “Achilles<br />

Heels” causing a disease when not functioning properly. We have developed a computational<br />

method for analysis microarray gene expression data <strong>and</strong> identification the key transcription<br />

factors <strong>and</strong> “upstream” <strong>signaling</strong> molecules that might be the causes of the observed change<br />

in gene expression. There are two steps of analysis (using TRANSFAC® <strong>and</strong><br />

TRANSPATH® databases): 1) CMFinder analyzes 5’-upstream regions of coexpressed<br />

genes <strong>and</strong> applies a genetic algorithm to reveal composite modules (CMs) consisting of cooccurring<br />

single TF binding sites <strong>and</strong> composite elements; 2) ArrayAnalyzer is a fast<br />

network search engine that analyzes signal transduction networks controlling the activities of<br />

the corresponding TFs <strong>and</strong> finds the key <strong>signaling</strong> molecules. The method was applied to<br />

micrarray data on Pseudoxanthoma elasticum (PXE) (a disease linked to the monogenic<br />

defects of ABCC6 transporter <strong>and</strong> characterized by alterations of elastic fibers <strong>and</strong> dystrophic<br />

calcification). We provided evidence that the SOX9-related transcriptional pathway <strong>and</strong> the<br />

IL-1#-linked <strong>signaling</strong> route are defective in PXE. We have identified PKAc kinase <strong>and</strong><br />

TRAF-6 as important key nodes in the <strong>signaling</strong> pathways that are pathologically altered.<br />

They can be considered as prospective drug targets. We hypothesized that impaired activity of<br />

the transporter may change a lipid balance in extracellular matrix resulting in activation of<br />

certain signal transduction mechanism leading to the disease symptoms.<br />

- 538 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 36<br />

15-Deoxy-&12,14-prostagl<strong>and</strong>in J2-mediated upregulation of heme oxygenase-1 <strong>and</strong><br />

vascular endothelial growth factor in human breast cancer cells : a potential role of Nrf2<br />

Eun-Hee Kim, Hye-Kyung Na <strong>and</strong> Young-Joon Surh<br />

National Research Laboratory of Molecular Carcinogenesis <strong>and</strong> Chemoprevention,<br />

College of Pharmacy, Seoul National University, Seoul 151-742, South Korea<br />

Elevated expression or activity of heme oxygenase-1 (HO-1), a ubiquitous stress-responsive<br />

enzyme, has been reported to stimulate proliferation <strong>and</strong> to accelerate angiogenesis in several<br />

types of tumor cells. 15-Deoxy-&12,14-prostagl<strong>and</strong>in J2 (15d-PGJ2), an endogenous lig<strong>and</strong><br />

of peroxisome proliferator-activated receptor $, has been known to induce HO-1 in some cell<br />

lines. In the present work, we found that treatment of human breast cancer (MCF-7) cells<br />

with nontoxic doses of 15d-PGJ2 led to concentration- <strong>and</strong> time-dependent increases in the<br />

expression <strong>and</strong> activity of HO-1. The induction of HO-1 expression preceded the<br />

upregulation of vascular endothelial growth factor (VEGF) in MCF-7 cells stimulated with<br />

15d-PGJ2. The upregulation of VEGF production by 15d-PGJ2 was abrogated by the HO-1<br />

inhibitor, ZnPP. Moreover, ZnPP decreased the in vitro capillary formation induced by 15d-<br />

PGJ2 in human umbilical vein endothelial cells. Likewise, ZnPP treatment mitigated the<br />

manifestation of migrative phenotype of 15d-PGJ2-treated MCF-7 cells as determined by the<br />

wound migration assay. Nrf2, a basic-leucine zipper transcription factor, plays a key role in<br />

regulating the antioxidant response element (ARE)-mediated expression of HO-1 <strong>and</strong> other<br />

antioxidant enzymes. 15d-PGJ2 increased nuclear translocation <strong>and</strong> subsequent ARE binding<br />

of Nrf2. MCF-7 cells transfected with dominant negative Nrf2 exhibited reduced expression<br />

of HO-1 <strong>and</strong> VEGF in response to 15d-PGJ2 treatment. Taken together, these results suggest<br />

that 15d-PGJ2-induced expression of HO-1 via the Nrf2 <strong>signaling</strong> is implicated in<br />

angiogenesis in human breast cancer.<br />

- 539 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 37<br />

Inhibition of leptin receptor <strong>signaling</strong> by SOCS proteins<br />

Holger Knobelspies, Julia Zeidler, Simone Bamberg-Lemper <strong>and</strong> Walter Becker<br />

Department of Pharmacology, RWTH Aachen University, Wendlingweg 2, 52074<br />

Aachen, Germany; E-mail: hknobelspies@ukaachen.de<br />

The murine leptin receptor (LR) comprises three conserved intracellular tyrosine residues<br />

(Y985, Y1077, Y1138) that are phosphorylated upon lig<strong>and</strong> binding. Phosphorylated (p)<br />

Y1138 <strong>and</strong> pY1077 have been shown to form binding sites for the signal transducers <strong>and</strong><br />

activators of transcription (STAT)3 <strong>and</strong> STAT5. pY985 recruits suppressor of cytokine<br />

<strong>signaling</strong> (SOCS)3, a negative feedback regulator of LR signal transduction. We used point<br />

mutants of the LR, lacking either Y985 (designated FYY), Y1077 (YFY) or both tyrosines<br />

(FFY) to elucidate interactions between the intracellular tyrosines of the LR <strong>and</strong> SOCS1 or<br />

SOCS3. A pancreatic beta-cell line from rat (RINm5F) stably expressing the LR showed<br />

desensitization during leptin stimulation of 2h or longer, as we assessed by Western blotting<br />

using specific antibodies against pY-STAT3. Binding studies revealed that this effect was not<br />

due to internalization of LR, since the surface expression was reduced by only 22±16%<br />

(mean±S.D.;n=8) after 2h of stimulation. The time-c<strong>our</strong>se of leptin-induced pY-STAT3 levels<br />

was similar for the wild-type (WT) LR <strong>and</strong> for the FYY mutant, approaching background<br />

levels after 4h of stimulation. In contrast, the FFY double mutant showed a decrease by only<br />

14%. This suggests the involvement of Y1077 <strong>and</strong> not only Y985 in the desensitization.<br />

Reporter gene assays in HepG2 cells with a STAT3-responsive promoter showed that<br />

overexpressed SOCS3 decreased the <strong>signaling</strong> by any YF mutant, but the receptors with<br />

functional Y985 (WT, YFY) were stronger affected than the mutants lacking Y985 (decrease<br />

to 6%, 59%, 4% <strong>and</strong> 23% for WT, FYY, YFY <strong>and</strong> FFY receptors, respectively). In SOCS3<br />

knockdown experiments, the FFY mutant showed no effect, whereas the WT receptor<br />

increased the reporter gene expression to 508 % <strong>and</strong> the Y985F to 278%. This supports <strong>our</strong><br />

hypothesis of SOCS3 being able to bind to Y985 or Y1077 <strong>and</strong> thereby inhibiting LR<br />

<strong>signaling</strong>. Overexpression of SOCS1, which is not induced by leptin in RINm5F LR cells,<br />

dose-dependently inhibited either receptor mutant. Again, the receptors with functional Y985<br />

showed stronger effects (WT 14%, FYY: 55%, YFY: 14%, FFY: 60%). Knockdown of<br />

SOCS1 showed an increase in <strong>signaling</strong> of WT <strong>and</strong> either single mutant (WT: 58%, FYY:<br />

47%, YFY: 50%) whereas the knockdown had no effect on the FFY mutant. We conclude<br />

hereof that SOCS1 inhibits LR <strong>signaling</strong> by interaction with Y985 or Y1077 in <strong>our</strong> system.<br />

- 540 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 38<br />

SOX3 expression is driven by multiple CCAAT control elements<br />

Aleks<strong>and</strong>ar Krstic, Milena Stevanovic<br />

Laboratory for human molecular genetics, Institute of Molecular Genetics <strong>and</strong> Genetic<br />

Engineering, Vojvode Stepe 444a, 11010 Belgrade, Serbia <strong>and</strong> Montenegro,<br />

E-mail : hmgbox@eunet.yu<br />

Early neurogenesis <strong>and</strong> neuronal differentiation are precisely controlled by a series of genes.<br />

Sox3 is expressed in the brain from initial stages of development <strong>and</strong> it is considered to be<br />

one of the earliest markers in vertebrates playing the role in specifying neuronal fate. In order<br />

to elucidate molecular mechanisms underlying the regulation of the human SOX3 gene<br />

expression, computer prediction software was used to search the matrix database <strong>and</strong> to<br />

identify potential transcription binding sites within the human SOX3 promoter. Among the<br />

number of putative consensus binding sites three evolutionary conserved CCAAT boxes,<br />

representing the putative binding sites for the general transcription factor NF-Y, were<br />

identified at positions –101 bp to –105 bp, -246 bp to -250 bp <strong>and</strong> -326 bp to -330 bp. EMSA<br />

<strong>and</strong> “supershift” experiments, for fragments containing each of the identified CCAAT boxes,<br />

were performed to prove the specificity of the NF-Y binding. To examine whether the<br />

putative CCAAT boxes are functional, site directed mutagenesis was performed. The ability<br />

of the single, double <strong>and</strong> triple site mutants <strong>and</strong> its wild-type counterpart to drive expression<br />

of the cat reporter gene was examined in stem <strong>and</strong> RA induced NT2/D1 cells. We have<br />

shown that mutagenesis of all three CCAAT boxes affects the expression of the reporter gene,<br />

compared to the wild-type counterpart. Results obtained with double NF-Y site mutants<br />

suggest functional/spatial interaction of these regulatory elements. Triple NF-Y site mutant<br />

reduced reporter construct activity by almost 20 fold. All results strongly suggest -that all<br />

three CCAAT box motifs present in the SOX3 promoter play functional role as transcriptional<br />

activators of this gene.<br />

- 541 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 39<br />

Runx2 Transcription Factor Controls F promoter Activity of Human ER alpha Gene<br />

in Osteoblasts<br />

Elisabetta Lambertini, Elisa Tavanti, Letizia Penolazzi, Roberto Gambari <strong>and</strong> Roberta<br />

Piva.<br />

Department of Biochemistry <strong>and</strong> Molecular Biology, University of Ferrara, Italy. Email:<br />

piv@unife.it<br />

Bone growth, development <strong>and</strong> maintenance is a highly regulated process. The balance<br />

between bone formation <strong>and</strong> resorption involves two cell types: bone forming osteoblasts<br />

(OBs) <strong>and</strong> bone-resorbing osteoclasts (OCs). OBs-dependent bone formation varies with age,<br />

metabolic disease states or with pharmacological intervention. OBs formation <strong>and</strong> maturation<br />

are under control of a specific transcription factor, Runx2, that regulates the expression of<br />

bone-specific genes. Another important transcription factor playing a central role in bone is<br />

the estrogen receptor alpha (ER) that mediates estrogen-induced effects on gene expression.<br />

Considering the potent gene regulatory activity of Runx2 <strong>and</strong> ER we investigated the role of<br />

Runx2 on the regulation of ER expression valuating its interaction with F promoter of ER<br />

gene. F promoter is one of the multiple promoters of human ER gene <strong>and</strong> is the only active<br />

promoter in bone. In this promoter three Runx2 sites (A), (B), <strong>and</strong> (C) are present. To<br />

underst<strong>and</strong> if they are involved in influencing F promoter activity, different promoter-reporter<br />

deletion <strong>and</strong> mutation constructs were assayed for function after transient transfection into the<br />

Runx2-producing SaOS-2 human osteoblastic cells. The comparison of luciferase activities<br />

allowed the identification of a negative role of a sequence context, within the F promoter<br />

containing the three Runx2 binding sites. In addition, by using electrophoretic mobility shift<br />

assay (EMSA) in combination with supershift we demonstrated that a significant supershifted<br />

complex was formed only with the Runx2 (A) site. In order to demonstrate the<br />

possible association of the Runx2 protein to the F promoter “ in vivo” chromatin<br />

immunoprecipitation assay (ChIP) was performed. The results demonstrate that Runx2<br />

interacts “ in vivo” with the region of the F promoter within the sequence context containing<br />

the Runx2 binding sites in SaOS-2 cells indicating that Runx2 may be considered one of the<br />

factors controlling specific expression of human ER alpha gene in osteoblasts.<br />

- 542 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 40<br />

Differential regulation of vascular endothelial growth factor expression by peroxisome<br />

proliferator-activated receptors via adipocyte-fatty acid binding protein in bladder<br />

cancer cells<br />

Isabelle Lascombe, Guillaume Boiteux, Hugues Bittard <strong>and</strong> Sylvie Fauconnet<br />

Université de Franche-Comté, EA 3181 “Carcinogenèse épithéliale”, IFR133, UFR<br />

Médecine et Pharmacie, 19 rue Ambroise Paré, 25000 Besançon, France. E-mail :<br />

isabelle.lascombe@voila.fr<br />

Vascular endothelial growth factor (VEGF) plays a prominent role in vesical tumor<br />

angiogenesis regulation (Lascombe et al., 2005, PBCR review). Peroxisome proliferatoractivated<br />

receptors (PPAR), which are transcription factors, are involved in angiogenesis<br />

process. Recently, we reported for the first time that, in two different human bladder cancer<br />

cell lines RT4 (derived from grade I tumor) <strong>and</strong> T24 (derived from grade III tumor), VEGF is<br />

differentially up-regulated by the three PPAR isotypes. Its expression is increased by<br />

PPARalpha, # <strong>and</strong> in RT4 cells <strong>and</strong> only by PPAR# in T24 cells via a transcriptional<br />

activation of the VEGF promoter through an indirect mechanism (Fauconnet et al., J Biol<br />

Chem, 2002). Our purpose was to elucidate the molecular mechanism involved in PPARinduced<br />

VEGF expression. We hypothesized that in high grade T24 cells, the inactivity of<br />

PPARalpha <strong>and</strong> $ could be due to the absence of adipocyte-fatty acid binding protein (A-<br />

FABP) expression or to the loss of the protein functionality. Indeed, loss of A-FABP is<br />

associated with progression in bladder transitional cell carcinomas. Furthermore, recent<br />

studies establish that FABP govern the transcriptional activities of their lig<strong>and</strong>s by targeting<br />

them to PPAR in the nucleus, thereby enabling PPAR to exert their biological functions. Our<br />

first results suggest that A-FABP is present in cytoplasm of both RT4 <strong>and</strong> T24 cells but does<br />

not locate to the nucleus of the T24 cells. Preliminary data obtained with siRNA targeting A-<br />

FABP seem to confirm the role of this protein in PPAR-induced VEGF expression. These<br />

data contribute to a better underst<strong>and</strong>ing of the mechanisms by which PPARs regulate VEGF<br />

expression in non aggressive bladder tumors. This could lead to a new therapeutic approach<br />

for human bladder cancer in which excessive angiogenesis is a negative prognostic factor.<br />

- 543 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 41<br />

Role of VEGF-D <strong>and</strong> AT2 receptor on cytokine expression in pancreatic acinar cells<br />

JANGWON LEE, SHIN YOUNG YUN, JOO WEON LIM, KYUNG HWAN KIM, <strong>and</strong><br />

HYEYOUNG KIM<br />

Department of Pharmacology, Brain Korea 21 Project for Medical Science, Yonsei<br />

University College of Medicine, Seoul 120-752, Korea. E-mail :honorjw@hotmail.com<br />

Cerulein pancreatitis is similar to human edematous pancreatitis with dysregulation of the<br />

digestive enzyme production, the death of acinar cells, <strong>and</strong> an infiltration of inflammatory<br />

cells into the pancreas. Our previous proteomic analysis showed several differentially<br />

expressed proteins in cerulein-treated pancreatic acinar cells, which are related to cellular<br />

stress such as reactive oxygen species. We hypothesized that membrane proteins may be<br />

altered as the early event during the induction of acute pancreatitis. The present study was<br />

designed to elucidate firstly whether or not VEGF receptor <strong>and</strong> AT2 receptor be involved in<br />

controlling cytokine expression. Secondly, the role of VEGF-D <strong>and</strong> AT2 receptor in<br />

pancreatic acinar cells <strong>and</strong> their regulation by acute pancreatitis are also investigated. The<br />

differentially expressed membrane proteins by cerulein will provide valuable information to<br />

underst<strong>and</strong> pathophysiologic mechanism of acute pancreatitis <strong>and</strong> may be useful for<br />

prognostic indices of acute pancreatitis. AP-1 <strong>and</strong> NF-kB regulate cytokine gene expression<br />

in pancreatic acinar cells treated with cerulein. AT2 receptor is associated with VEGF-D to<br />

induce IL-6 expression in AR42J cells. Cerulein-induced VEGF-D expression <strong>and</strong> AP-1/NFkB<br />

activation were inhibited in the cells transfected with VEGF-D antisense oligonucleotide,<br />

indicating the involvement of VEGF-D in regulating the cytokine expression during acute<br />

pancreatitis.<br />

- 544 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 42<br />

Nuclear loss of Ku DNA repair protein in cells exposed to oxidative stress<br />

Jong Hwa Lee, Ji Yeon Song, Ji Hoon Yu, Kyung Hwan Kim, Hyeyoung Kim*<br />

Department of Pharmacology, Brain Korea 21 Project for Medical Science, Yonsei<br />

University College of Medicine, Seoul 120-752, Korea. E-mail : pico798@naver.com<br />

Recent studies show that NF-kB plays a critical role in protecting the cells from apoptotic<br />

cell death. We previously demonstrated that oxidative stress – induced cell death is caused by<br />

nuclear loss of DNA repair protein Ku proteins in pancreatic acinar AR42J cells. This study<br />

aims to investigate the role of NF- kB on nuclear translocation of Ku proteins <strong>and</strong> apoptotic<br />

cell death, induced by oxidative stress in AR42J cells. We examined Ku expression, cell<br />

viability, <strong>and</strong> apoptosis of the cells treated with or without H2O2, which is continuously<br />

generated by G/GO <strong>and</strong> in the caspase-3 inhibitor. Wild-type cells as well as the cells<br />

transfected with Ku dominant negative gene or I-kB alpha mutant gene was used. As a result,<br />

G/GO induced decrease in nuclear Ku70 <strong>and</strong> Ku80 time-<strong>and</strong> concentration-dependently.<br />

G/GO induced alterations in Ku expression were inhibited, in part, by caspase-3 inhibitor.<br />

G/GO induced apoptosis, which was in parallel with loss of nuclear Ku70 <strong>and</strong> Ku80 in the<br />

cells transfected with the control pcDNA3 vector. G/GO increased apoptosis in those<br />

transfected with Ku dominant negative mutant or I-kB alpha mutant gene. Nuclear level of Ku<br />

proteins were decreased in the cells transfected with I-kB alpha mutant gene. Conclusively,<br />

nuclear translocation of Ku proteins may be mediated by NF-kB in the cells. Decrease in<br />

nuclear translocation of Ku proteins may result in apoptotic cell death in AR42J cells.<br />

- 545 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 43<br />

DNA hypomethylation of CAGE Promoters in Squamous Cell Carcinoma of Uterine<br />

Cervix<br />

Taek Sang Lee a,b, Jae Weon Kima,b,c,*, Gyeong Hoon Kangb,d Noh Hyun Parka,b,<br />

Yong Sang Songa,b, Soon Beom Kanga,b, Hyo Pyo Leea,b<br />

aDepartment of Obstetrics <strong>and</strong> Gynecology, bCancer Research Institute, <strong>and</strong> cHuman<br />

Genome Research Institute, dDepartment of Pathology, College of Medicine, Seoul<br />

National University, Seoul, 110-744, Korea<br />

E-mail: kjwksh@snu.ac.kr<br />

This study was performed to determine whether promoter hypomethylation of CAGE is<br />

involved in cervical carcinogenesis. The surgical specimens from 40 cervical squamous cell<br />

carcinoma patients who treated at Seoul National University Hospital <strong>and</strong> from 48 healthy<br />

controls were used with informed consent. By methylation specific PCR (MSP) using<br />

unmethylation specific primer, the promoter hypomethylation status of CAGE was<br />

investigated. We found that hypomethylation of CAGE promoter was present at frequencies<br />

of nearly one hundred percent in cervical squamous cell carcinomas (38/40, 95%), but less<br />

than 4% in controls (p


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 44<br />

Identification in rat of a yeast She3 homolog as interaction partner of AATF<br />

Peter Leister, Sven Burgdorf, Andrea Felten, Lutz Uhlmann, <strong>and</strong> Karl Heinz<br />

Scheidtmann<br />

Institute of Genetics, University of Bonn<br />

Roemerstr. 164, D-53117 Bonn<br />

peterleister@uni-bonn.de<br />

AATF, or Che-1, is a coactivator of several transcription factors including steroid hormone<br />

receptors, E2F, <strong>and</strong> SP1. In search of novel interaction partners of AATF we identified a so<br />

far unknown protein from rat with high degree of homology to She3p from budding yeast.<br />

She3p is an adapter protein for myosinV type motor protein Myo4p <strong>and</strong> is involved in<br />

selective transport of mRNA <strong>and</strong> ER vesicles. The novel protein was termed SHIA for She3<br />

Homolog Interacting with AATF. A single transcript of SHIA mRNA of 1050 bp was highly<br />

expressed in brain, heart, spleen, liver, testis <strong>and</strong> kidney <strong>and</strong> to a lower extent in lung,<br />

whereas it was undetectable in skeletal muscle. However, sequence alignments with database<br />

sequences revealed that human, mouse <strong>and</strong> rat orthologs exist in two or three isoforms<br />

generated by alternative splicing or alternative usage of start codons, giving rize to<br />

polypeptides of 142 <strong>and</strong> 99 residues, respectively. Strikingly, besides some variations at their<br />

N-termini SHIA proteins are extremely conserved. A core sequence between residues 20 <strong>and</strong><br />

140 is strictly conserved within all vertebrates represented in the database, <strong>and</strong> certain<br />

sequence motifs are found even in insects <strong>and</strong> C. elegans <strong>and</strong> yeast. This high degree of<br />

conservation suggests that SHIA plays an essential role in all eukaryotes.<br />

The rat SHIA clone isolated appears to contain an extra exon (termed exon 2b) resulting in a<br />

protein of 168 amino acids with MW of 20 kDa. SHIA contains a She3p homology region at<br />

its N-terminus <strong>and</strong> a leucine zipper at its C-terminus, which mediates interaction with AATF.<br />

When expressed as GFP fusion protein, GFP-SHIA was localized in the cytoplasm,<br />

preferentially nuclear near structures <strong>and</strong> the actin cytoskeleton. A small fraction of GFP-<br />

SHIA was also observed within the nucleus. Overexpression of SHIA resulted in disturbed<br />

actin fibers, <strong>and</strong> coexpression of SHIA <strong>and</strong> AATF resulted in partial relocalisation of AATF<br />

from the nucleus to the cytoplasm. Interestingly, SHIA enhanced reporter gene activty in<br />

<strong>and</strong>rogen receptor based transactivation assays, thus, SHIA behaved like a transcriptional coactivator.<br />

- 547 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 45<br />

The MECP2 gene mutation screening in Rett syndrome patients from Croatia<br />

Tanja Matijevi'1, Jelena Kne0evi'1, Ingeborg Bari)i'2, Vida 1uli'3 <strong>and</strong> Jasminka<br />

Paveli'1.<br />

1 Laboratory of Molecular Oncology, Division of Molecular Medicine, Rudjer Bo)kovi'<br />

Institute, 10002 Zagreb, Croatia. E-mail : jpavelic@irb.hr<br />

2 Department of Pediatrics, Children’s Hospital Zagreb, University of Zagreb, Medical<br />

School, 10000 Zagreb, Croatia<br />

3 Department of Medical Genetics, Pediatric Clinic, Clinical Hospital Split, 21000 Split,<br />

Croatia<br />

Rett syndrome is an X-linked dominant neurodevelopmental disorder almost exclusively<br />

affecting females <strong>and</strong> is usually sporadic. Mutations in MECP2 gene have been found in more<br />

than 80% of females with typical features of Rett syndrome. In this study, we analyzed 14<br />

sporadic cases of Rett syndrome. In 6 of 14 patients (43%), we detected pathogenic<br />

mutations in the coding parts of MECP2. We found one missense (T158M), two nonsense<br />

(R168X, R270X), two frameshift mutations (P217fs <strong>and</strong> a double deletion of 28-bp at 1132-<br />

1159 <strong>and</strong> 10-bp at 1167-1176) <strong>and</strong> one in-frame deletion (L383_E392del10). According to<br />

<strong>our</strong> knowledge, the last two mutations have not been reported yet. We also detected one<br />

previously described polymorphism (S194S). In conclusion, these results show that f<strong>our</strong>th<br />

exon should be the first one analyzed because it harbors most of the known mutations.<br />

Moreover, mutation-negative cases should be further analyzed for gross rearrangements. This<br />

is the first study of this kind in Croatia <strong>and</strong> it enabled us to give the patients an early<br />

confirmation of Rett syndrome diagnosis.<br />

- 548 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 46<br />

Identification of a prostagl<strong>and</strong>in-responsive element in the Na,K-ATPase beta1 subunit<br />

promoter which is regulated by cAMP <strong>and</strong> Ca2+ : Evidence for an interactive role of<br />

CREB <strong>and</strong> Sp1<br />

Keikantse Matlhagela, Maryann Borsick, Trivikram Rajkhowa, <strong>and</strong> Mary Taub*<br />

Biochemistry Dept, 140 Farber Hall, State University of New York at Buffalo, Buffalo,<br />

NY 14214, U.S.A., Email: *biochtau@buffalo.edu<br />

The Na,K-ATPase, is a transmembrane protein responsible for maintaining electrochemical<br />

gradients across the plasma membrane in all mammalian cells, a process which is subject to<br />

regulation at the transcriptional <strong>and</strong> post-transcriptional levels. Included amongst physiologic<br />

regulators in the kidney are prostagl<strong>and</strong>ins. Previously, we presented evidence that<br />

transcription of the Na,K-ATPase ß1 subunit is stimulated by PGE1, an effect mediated<br />

through both cAMP <strong>and</strong> Ca2+ pathways. Transient transfection studies using 5’ deletion<br />

mutants in the human ß1 subunit promoter indicate that a region located between –92 to –100<br />

containing the sequence AGTCCCTGC (a Prostagl<strong>and</strong>in Response Element, or PGRE) is<br />

required for eliciting the stimulatory effects of PGE1, 8Br-cAMP, Phorbol 12-myristate 13acetate<br />

(TPA) <strong>and</strong> okadaic acid. Electrophoretic Mobility Shift Assays (EMSAs) indicate that<br />

both the cAMP Regulatory Element Binding Protein (CREB) <strong>and</strong> Sp1 bind to this PGRE. The<br />

involvement of the PGRE <strong>and</strong> Sp1 sites in regulation by PGE1 was further confirmed by<br />

studies with a heterologous, <strong>and</strong> mutant promoters. The PGE1 stimulation was reduced when<br />

the 2 GC boxes adjacent to the PGRE were translocated further upstream from the PGRE.<br />

Also consistent with an interaction between CREB <strong>and</strong> Sp1 are <strong>our</strong> immunoprecipitation <strong>and</strong><br />

GST-pull down studies. In summary <strong>our</strong> results indicate the PGE1 induction involves<br />

interactions between Sp1 <strong>and</strong> CREB which occur during the binding of these transcription<br />

factors to a novel regulatory element, a PGRE, <strong>and</strong> an adjacent Sp1 site on the Na,K-ATPase<br />

ß1 subunit promoter.<br />

- 549 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 47<br />

Process simulation in a mechatronic bioreactor device with speed regulated motors for<br />

growing of three-dimensional cell cultures<br />

M. Mihailova1, V. Trenev2, P. Genova2, S. Konstantinov3<br />

1IPF of TU-Sofia, 8800 Sliven, 59 B<strong>our</strong>gasko chaussee Str, Bulgaria;<br />

e-mail: mina-todorova@abv.bg<br />

2CLMI, Bulgarian Academy of Sciences, 1 Acad. G. Bonchev Str, 1113 Sofia, Bulgaria;<br />

e-mail: vtrenev@clmi.bas.bg; Pgen@TU-Sofia.bg<br />

3Lab for Experimental Chemotherapy, Dept. of Pharmacology, Faculty of Pharmacy,<br />

Medical University of Sofia, 1000 Sofia, 2 Dunav Str, Bulgaria;<br />

e-mail: Konstantinov.spiromihaylov@gmail.com<br />

Tissue engineering is a new scientific research field that allows the establishment of tissue<br />

equivalents rising from isolated cells in combination with biocompatible materials <strong>and</strong><br />

cultivation in more or less sophisticated bioreactor systems. Such systems gave the unique<br />

opportunity to perform in vitro investigations of transcription <strong>and</strong> translation, cell growth,<br />

biochemistry <strong>and</strong> mechanics of healthy normal organs as well as those affected by malignant<br />

tum<strong>our</strong>s, infections <strong>and</strong> immune deficiency under controlled conditions. In rotating vessel<br />

bioreactors under microgravity <strong>and</strong> defined medium content cells proliferate, stay abundant to<br />

each other <strong>and</strong> form three-dimensional structures, assigned as spheroids. Such spheroids<br />

might be grown on micro carriers. A wide spectrum of different cell culture experiments<br />

involving normal <strong>and</strong> transformed human cells indicate that: in a rotating bioreactor system<br />

miniPERM no complete lack of gravity could be reached; a great part of the seeded cell<br />

material do not proliferate at the beginning <strong>and</strong> the appearance of bigger spheroids is rather<br />

r<strong>and</strong>om. We describe the acquisition of spheroids in HD-MY-Z <strong>and</strong> Neuro-2A tum<strong>our</strong> cell<br />

lines as well as in bone marrow fibroblasts from acute myeloid leukaemia patients. Spheroids<br />

of 100 <strong>and</strong> more cells were obtained from HD-MY-Z <strong>and</strong> Neuro-2A cells. Interestingly,<br />

chronic myeloid leukaemia LAMA-84 cells did not form any cell clumps <strong>and</strong> they kept a<br />

completely undifferentiated phenotype despite their semi-adherent growth manner under<br />

conventional conditions. A detailed theoretical <strong>and</strong> virtual simulation study of the influence of<br />

every component of gravitation, inertia <strong>and</strong> hydrodynamic force fields was performed.<br />

Thereby, a new concept for mechatronic bioreactor device with active electronic control was<br />

developed <strong>and</strong> virtually tested.<br />

- 550 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 48<br />

Estrogen receptor-mediated transcriptional regulation may involve different cross-talk<br />

interaction between estrogen receptor <strong>and</strong> xenobiotic nuclear receptors depending on<br />

the estrogen target cell types<br />

Gyesik Min<br />

Department of Microbiological Engineering, Jinju National University, Jinju,<br />

Gyeongsangnam-Do, 660-758 South Korea E-mail: g-min@jinju.ac.kr<br />

The purpose of this study was to examine the effects of xenobiotic nuclear receptors, CAR,<br />

SXR, <strong>and</strong> PPARgamma on the transcriptional activity of estrogen receptor in human breast<br />

cancer cell lines <strong>and</strong> compare with those in human hepatoma cell line. Three different breast<br />

cancer cell lines, MCF-7 with ER expression, MDA-MB-231 without ER expression, <strong>and</strong><br />

MCF-7K3 with estrogen-independent growth were cultured <strong>and</strong> effects of CAR, SXR, <strong>and</strong><br />

PPARgamma on the ER-mediated transcriptional activation of synthetic (4ERE)-tk-luciferase<br />

reporter gene were analyzed. Consistent with the previous report, CAR significantly inhibited<br />

ER-mediated transactivation <strong>and</strong> SXR repressed modestly whereas the PPARgamma did not<br />

repress the ER-mediated transactivation in Hep G2 cells. However, in breast cancer cells<br />

neither of the xenobiotic receptors repressed the ER-mediated transactivation. Instead, they<br />

tend to increase the transactivation depending on the cell type <strong>and</strong> xenobiotic nuclear<br />

receptors. In MCF-7, SXR but not CAR or PPARgamma slightly increased ER-mediated<br />

transactivation whereas in MDA-MB-231, CAR <strong>and</strong> PPARgamma but not SXR tend to<br />

increase the transactivation of the reporter gene. In addition, in MCF-7K3 only high dose of<br />

CAR slightly increased estrogen dependent ER transactivation. These results indicate that the<br />

effects of cross-talk in ER-mediated transactivation between estrogen <strong>and</strong> the xenobiotic<br />

nuclear receptors, CAR, SXR, PPARgamma, are different in breast cancer cells from<br />

hepatoma cells. Different responses to each xenobiotic receptor from the three breast cancer<br />

cell lines may suggest diverse signal transduction pathways for the receptors, CAR, SXR,<br />

PPARgamma, present in different breast cancer cell lines. In conclusion, the transcriptional<br />

regulation by estrogen in ER-mediated transactivation can involve different cross-talk<br />

interaction between estrogen receptor <strong>and</strong> xenobiotic nuclear receptors depending on the<br />

estrogen target cell types.<br />

- 551 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 49<br />

Differential transactivation by constitutive <strong>and</strong>rostane receptor of Phenobarbital<br />

Responsive Unit(PBRU) of CYP2B gene between human hepatoma cell line Hep G2 <strong>and</strong><br />

monkey kidney epithelial derived cell line COS-7<br />

Gyesik Min<br />

Department of Microbiological Engineering, Jinju National University, Jinju,<br />

Gyeongsangnam-Do, 660-758 South Korea E-mail: g-min@jinju.ac.kr<br />

The objective of this study was to examine if transient transfection of CAR can transactivate<br />

CYP2B1 PBRU reporter gene in COS-7 cells in which the endogenous CYP2B1 gene is not<br />

induced by PB. In non-transfeced cells of both Hep G2 <strong>and</strong> COS-7, the endogenous<br />

expression of CAR was not detected by antibody against CAR. When cultured cells were<br />

transfected with CAR expression plasmid, mCAR1-GFP, both cell types expressed high<br />

levels of CAR protein <strong>and</strong> could allow to examine the effect of CAR in PBRU transactivation.<br />

Both cell types expressed endogenous RXR <strong>and</strong> transfection of RXR expression plasmid<br />

dramatically increased its protein expression. Whereas CAR transactivated<br />

PBRU2C1Luciferase about 12 fold as compared to 2C1Luciferase in Hep G2 cells, it did not<br />

stimulate the luciferase activity of the PBRU reporter gene in COS-7 cells. These results<br />

indicate that Hep G2 cells can respond to CAR differently from COS-7 cells, <strong>and</strong> suggest that<br />

factors other than CAR <strong>and</strong> RXR may be required in inducing PBRU activation <strong>and</strong> the<br />

expression of these factors may be different between liver <strong>and</strong> kidney.<br />

- 552 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 50<br />

Regulation of 2-Deoxy Glucose Transport, Lactate Metabolism <strong>and</strong> MMP-2 Secretion by<br />

the Hypoxia Mimetic Cobalt Chloride in Articular Chondrocytes<br />

Ali Mobasheri1 Nicola Platt1, Colin Thorpe1 <strong>and</strong> Mehdi Shakibaei2<br />

1Faculty of Veterinary Science, University of Liverpool, Liverpool, L69 7ZJ, United<br />

Kingdom; 2Musculoskeletal Research Group, Institute of Anatomy, Ludwig-<br />

Maximilian-University Munich, Pettenkoferstrasse 11, 80336 Munich, Germany.<br />

E-mail: a.mobasheri@liverpool.ac.uk, mehdi.shakibaei@med.uni-muenchen.de<br />

Articular cartilage is avascular <strong>and</strong> this significantly reduces the availability of oxygen <strong>and</strong><br />

nutrients. Therefore, chondrocyte survival <strong>and</strong> cartilage homeostasis require effective<br />

mechanisms for oxygen <strong>and</strong> nutrient <strong>signaling</strong>. To gain a better underst<strong>and</strong>ing of the<br />

mechanisms responsible for oxygen <strong>and</strong> nutrient sensing in chondrocytes we investigated the<br />

effects of hypoxic simulation induced by cobalt chloride treatment (a hypoxia mimetic) on<br />

glucose uptake <strong>and</strong> lactate production in chondrocytes. We also studied the effects of cobalt<br />

chloride <strong>and</strong> glucose deprivation on the expression <strong>and</strong> secretion of active MMP-2.<br />

Primary cultures of articular chondrocytes were either maintained in 20% O2 (normoxia) or<br />

exposed to the hypoxia mimetic cobalt chloride for up to 24 h at the following concentrations:<br />

15 µM, 37.5, µM <strong>and</strong> 75 µM. Glucose transport was determined by measuring the net uptake<br />

of non-metabolizable 2-deoxy-D-[2, 6-3H] glucose into chondrocytes. Expression of GLUT1<br />

<strong>and</strong> GLUT3 was determined by western blotting <strong>and</strong> immunohistochemistry. Active MMP-2<br />

secretion was assayed by gelatin zymography. Lactic acid production was assayed using a<br />

lactate kit.<br />

The hypoxia-responsive GLUT1 <strong>and</strong> GLUT3 proteins were expressed in chondrocytes.<br />

<strong>Expo</strong>sure to cobalt chloride significantly increased the uptake of 2-deoxy-D-[2, 6-3H] glucose<br />

<strong>and</strong> the production of lactate. Glucose deprivation <strong>and</strong> cobalt chloride treatment increased<br />

levels of active MMP-2 in the culture medium. Our results suggest that these metabolic<br />

alterations are important events during adaptation to hypoxia. Upregulation of MMP-2 <strong>and</strong><br />

the build-up of lactic acid will have detrimental effects on the extracellular matrix <strong>and</strong> may<br />

contribute to the pathogenesis <strong>and</strong> progression of osteoarthritis.<br />

- 553 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 51<br />

Microtubule interfering agents affect <strong>signaling</strong> pathways involved in the regulation of<br />

cytochrome P450 genes expression.<br />

Martin Modriansk., Jitka Ulrichová, <strong>and</strong> Zden2k Dvo-ák<br />

Institute of Medical Chemistry <strong>and</strong> Biochemistry, Faculty of Medicine, Palacky<br />

University, Hnevotinska 3, 775 15 Olomouc, Czech Republic. Email:<br />

oregon@tunw.upol.cz<br />

Microtubule interfering agents, such as vincristine, vinblastine, taxol (paclitaxel) etc., are used<br />

in human pharmacotherapy as anti-neoplastic drugs. Use of colchicine, a substance displaying<br />

the same microtubule disrupting properties, is limited to the treatment of acute gout attacks<br />

<strong>and</strong> familial mediterranean fever. Besides the common feature of tubulin binding these<br />

substances share metabolism by cytochromes P450 (CYP). This is the basis for investigations<br />

into potential drug-drug interactions on the level of CYP genes expression. All of these<br />

microtubule interfering agents restrict the expression of CYP genes regulated by<br />

glucocorticoid (GR) <strong>and</strong> aryl hydrocarbon (AhR) receptors. While this activity may be<br />

associated with cell cycle phase, we show that it is independent of the cell cycle status.<br />

Therefore microtubule integrity is important for GR <strong>and</strong> AhR transcriptional activity.<br />

ACKNOWLEDGEMENT<br />

This research is supported by grant MSM 6198959216 from the Ministry of Education, Youth<br />

<strong>and</strong> Sports of the Czech Republic <strong>and</strong> grant GACR 303/04/P074 from the Grant Agency of<br />

the Czech Republic.<br />

- 554 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 52<br />

Immunomorphological analysis of RAGE-receptor expression <strong>and</strong> NFkB-activation in<br />

tissue samples from normal <strong>and</strong> degenerated intervertebral discs of various ages<br />

A. Nerlich (1), B. Bachmeier (2), E. Schleicher (3), H. Rohrbach (1), G. Paesold (4), N.<br />

Boos (4)<br />

Dept. of Pathology, Academic Hospital Munich-Bogenhausen, Germany,<br />

<strong>and</strong>reas.nerlich@extern.lrz-muenchen.de<br />

Dept. of Clinical Chemistry Clin. Biochemistry, Univ. Munich, Germany<br />

Dept. Pathobiochemistry, Univ. Tübingen, Germany<br />

Spinal Surgery Unit, Orthopaedic Clinic, Univ. Zürich, Switzerl<strong>and</strong><br />

Recent studies provided circumstantial evidence that the activation of the RAGE-receptor by<br />

the oxidation-mediated amino acid modification carboxymethyl-lysine (CML) leads to NFkBactivation<br />

<strong>and</strong> translocation into the nucleus. This leads to the up-regulation of the synthesis<br />

of extracellular matrix molecules along with matrix degrading enzymes. In consequence, the<br />

structure <strong>and</strong> function of tissue changes. Recent extensive analyses provided good evidence<br />

that the age-associated degeneration of the intervertebral disc is closely associated with both<br />

the alteration of the extracellular disc matrix <strong>and</strong> the accumulation of oxidation-mediated<br />

CML modification of proteins. Accordingly, pervious own studies have shown significantly<br />

enhanced levels of CML associated with disc degeneration. We therefore investigated the<br />

pattern of RAGE-expression <strong>and</strong> NFkB-translocation into the nucleus.<br />

For this study, 43 complete cross-sections of complete human lumbar intervertebral discs had<br />

been prepared for the immunohistochemical localization of the RAGE-receptor <strong>and</strong> NFkBexpression<br />

(both antibodies: Santa Cruz Biotech., CA, USA). The samples covered an age<br />

between fetal (35th weeks of gestation) to senile age (85 years) with well defined macro- <strong>and</strong><br />

micromorphological features. The amount of positively labelled cells (RAGE) or nuclei<br />

(activated NFkB) were evaluated morphometrically by light microscopy in the inner <strong>and</strong> outer<br />

anterior <strong>and</strong> posterior annulus fibrosus <strong>and</strong> nucleus pulposus.<br />

No significant expression of RAGE <strong>and</strong> no obvious activation of NFkB (by translocation into<br />

the nucleus) were seen in fetal, juvenile <strong>and</strong> young adolescent discs (until age 13). In senile<br />

discs (age 77 – 85 years) RAGE-expression remained elevated, but NFkB activation was<br />

absent. In between, variably high numbers of labelled cells/ nuclei were seen with highest<br />

values ranging between 25 – c. 50% of cells in the nucleus pulposus for RAGE, <strong>and</strong> 15 – 60<br />

% of nuclei for NFkB with significant correlation between both parameters. In the anterior<br />

<strong>and</strong> posterior annulus significantly lower values were seen again for both parameters with<br />

more pronounced levels in the inner than the outer annulus.<br />

This study is the first that described activation of the NFkB system in human klumbar<br />

intervertebral discs in vivo. The close correlation between the expression of RAGE <strong>and</strong><br />

NFkB-activation suggests that the stimulation of RAGE (e.g. by CML) may induce<br />

transcription factor activation in this system. The association between expression rates for<br />

both parameters <strong>and</strong> increasing age indicates a potential link between transcription activation<br />

<strong>and</strong> disc degeneration. This may represent an important mechanism inducing changes in both<br />

cellular <strong>and</strong> extracellular structures of the disc, potentially prone to therapeutic intervention.<br />

- 555 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 53<br />

The nuclear scaffold protein NIPP1 directly binds to EZH2 <strong>and</strong> is essential for the<br />

trimethylation of Histone H3 on Lysine 27<br />

Mieke Nuytten1, Nivedita Roy1, Aleyde Van Eynde1, Lijs Beke1, Monique Buellens1,<br />

Gerald Thiel2, <strong>and</strong> Mathieu Bollen1<br />

1Division of Biochemistry, Department of Molecular Cell Biology, Faculty of Medecine,<br />

KULeuven, B-3000 Leuven, Belgium <strong>and</strong> 2Department of Medical Biochemistry <strong>and</strong><br />

Molecular Biology, University of Saarl<strong>and</strong> Medical Center, Homburg, Germany.<br />

NIPP1 is a nuclear protein that is ubiquitously expressed in metazoans <strong>and</strong> plants but not in<br />

yeast. The knockout of NIPP1 in mice is embryonic lethal before gastrulation <strong>and</strong> NIPP1-/-<br />

cell lines are not viable. NIPP1 binds to protein Ser/Thr kinase MELK <strong>and</strong> protein Ser/Thr<br />

phosphatase-1 but also interacts with nucleic acids <strong>and</strong> the essential pre-mRNA splicing<br />

factors CDC5L <strong>and</strong> SAP155.<br />

EZH2 is part of the Polycomb Repressive complex 2 (PRC2), which also contains EED <strong>and</strong><br />

SUZ12 as core elements. PRC2 is involved in the initiation of gene silencing. EZH2 is a<br />

methyltransferase that methylates Lys27 of Histone H3 (H3K27). Trimethylated H3K27<br />

serves as a docking site for the chromodomain-containing Polycomb protein, a component of<br />

PRC1. The recruitment of PRC1 maintains gene silencing by mechanisms that are not yet<br />

completely understood.<br />

We show that NIPP1 is associated with H3K27-trimethylated chromatin <strong>and</strong> interacts directly<br />

with both EED <strong>and</strong> EZH2. Moreover, NIPP1 acts as a transcriptional repressor of a reporter<br />

gene <strong>and</strong> this activity requires both EED <strong>and</strong> catalytically active EZH2. NIPP1-/-mouse<br />

blastocyst outgrowths <strong>and</strong> cultured cells with a siRNA-induced knockdown of NIPP1 are<br />

severely deficient in trimethylation of histone H3 on Lys27 but are normally trimethylated on<br />

Lys9. Our data show that the spliceosomal protein NIPP1 is required for trimethylation of<br />

histone H3 by EZH2, possibly by its ability to recruit PRC2 to its target loci.<br />

- 556 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 54<br />

A dynamic equilibrium of IkB metabolism confers NF-kB responsiveness to UV<br />

irradiation<br />

Ellen O’Dea <strong>and</strong> Alex<strong>and</strong>er Hoffmann<br />

Signaling Systems Laboratory, Department of Chemistry <strong>and</strong> Biochemistry, University<br />

of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0375, USA<br />

Email: eodea@ucsd.edu; ahoffmann@ucsd.edu<br />

The transcription factor NF-kB (nuclear factor-kappaB) is activated by a variety of stimuli,<br />

the majority of which signal to the IKK (IkappaB kinase) complex that triggers stimulusresponsive<br />

degradation of IkB proteins. Ultraviolet (UV) irradiation activates NF-kB through<br />

a distinctly different mechanism. Previous work reveals that UV-induced NF-kB activation<br />

involves translational inhibition through an ER-stress mediated pathway that induces<br />

phosphorylation of the eukaryotic initiation factor eIF2a.<br />

Even a modest suppression of protein synthesis may result in a disruption of the homeostasis<br />

of the cell <strong>and</strong> its <strong>signaling</strong> components. In the context of the NF-kB <strong>signaling</strong> module, we<br />

utilize a mathematical model to guide <strong>our</strong> experimental studies of the mechanisms that allow<br />

translational inhibition to result in NF-kB activation. Interestingly, the protein half-lives of<br />

IkB-bound to NF-kB <strong>and</strong> of the free form are regulated by distinct degradation pathways. We<br />

find that even in unstimulated cells, bound IkB is degraded by the well-characterized IKKmediated<br />

mechanism that is dependent on constitutive IKK activity. Indeed, computational<br />

simulations <strong>and</strong> experimental results demonstrate that genetic alterations in constitutive IKK<br />

activity or in IkB-responsiveness to IKK alter UV inducibility of NF-kB. In contrast, free<br />

IkB exists as a small pool in the cell that undergoes rapid degradation by a poorly understood<br />

IKK-independent mechanism. Similarly integrated computational/experimental studies reveal<br />

that short half-life control of free IkB is critical to UV responsiveness. Finally, we present<br />

evidence that cells are capable of altering this dynamic equilibrium of rapid IkB degradation<br />

balanced by constitutive synthesis thereby either attenuating or enhancing UV-induced NF-kB<br />

activation. Potential <strong>signaling</strong> crosstalk mechanisms will be discussed.<br />

- 557 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 55<br />

Identification <strong>and</strong> characterization of <strong>signaling</strong> pathways to NF-kB in lymphocytes<br />

Andrea Oeckinghaus1, Elmar Wegener1, Alex<strong>and</strong>er Beck2, Claus Scheidereit1, Daniel<br />

Krappmann1,3<br />

1 Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin,<br />

Germany<br />

2 University Hospital Tübingen, Internal Medicine IV, Otfried-Müller-Str. 10, 72076<br />

Tübingen<br />

3 GSF - Research Center for Environment <strong>and</strong> Health, Institute of Toxicology,<br />

Ingolstädter<br />

L<strong>and</strong>str.1, 85764 Neuherberg, Germany<br />

Activation, differentiation <strong>and</strong> clonal expansion of T-lymphocytes are initiated upon the<br />

concerted action of the T-cell receptor (TCR) <strong>and</strong> the CD28 co-stimulatory receptor.<br />

TCR/CD28 engagement initiates canonical NF-kB <strong>signaling</strong>, which involves IKK (IkB<br />

kinase) dependent phosphorylation <strong>and</strong> degradation of inhibitors of NF-kB (IkBs) enabling<br />

nuclear translocation of NF-kB. Genetic ablation in mice has identified PKCq, Carma1, Malt1<br />

<strong>and</strong> Bcl10 as essential mediators of antigen receptor induced IKK activation. Formation of a<br />

Carma1/Bcl10/Malt1 (CBM) complex represents a crucial step. But the composition of the<br />

regulatory CBM complex, that forms after T cell stimulation in vivo, <strong>and</strong> the mechanism of<br />

signal transduction from the TCR to the IKK complex are not understood to date.<br />

Using gel filtration analysis, GST pull down experiments, t<strong>and</strong>em affinity purifications <strong>and</strong><br />

antibody immunoprecipitations of <strong>signaling</strong> mediators, we have analyzed the composition <strong>and</strong><br />

the dynamics of the cellular CBM complex. We find that Bcl10-Malt1 associates with IKK#<br />

in unstimulated T cells <strong>and</strong> is rapidly recruited to Carma1 in response to TCR <strong>signaling</strong>.<br />

Sustained TCR/CD28 <strong>signaling</strong> leads to a disengagement of the CBM complex <strong>and</strong><br />

abrogation of downstream <strong>signaling</strong>. Further, we find that antigen induced phosphorylation of<br />

Bcl10 requires the putative downstream kinase IKKb. Mapping <strong>and</strong> functional studies of<br />

phospho-acceptor sites on Bcl10 reveal, that IKKb triggered Bcl10 phosphorylation serves to<br />

balance TCR induced lymphocyte activation. Our studies indicate that the formation of large<br />

<strong>signaling</strong> clusters upon T cell activation initiates a network of protein interactions <strong>and</strong><br />

modifications <strong>and</strong> thereby facilitates a non-linear order of <strong>signaling</strong> to IKK/NF-kB.<br />

- 558 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 56<br />

Characterization <strong>and</strong> functional analysis of human SOX18 promoter<br />

Isidora Petrovic, Milena Stevanovic.<br />

Laboratory for human molecular genetics, Institute of Molecular Genetics <strong>and</strong> Genetic<br />

Engineering, Vojvode Stepe 444a, 11010 Belgrade, Serbia <strong>and</strong> Montenegro,<br />

E-mail: hmgbox@eunet.yu<br />

Sox/SOX constitute a large family of genes encoding transcription factors, involved in the<br />

decision of cell fates during development <strong>and</strong> implicated in the control of diverse<br />

development processes. Human SOX18 gene is expressed in fetal brain as well as in a wide<br />

range of fetal <strong>and</strong> adult tissues indicating its function is not restricted to early development.<br />

Now it is clear that human SOX18 gene is involved in vascular cell growth <strong>and</strong> suggest that<br />

this transcription factor may play role in atherosclerosis. The aim of <strong>our</strong> study has been to<br />

determine <strong>and</strong> characterize the promoter of the human SOX18 gene <strong>and</strong> to elucidate<br />

molecular mechanisms underlying the regulation of its expression. We have isolated <strong>and</strong><br />

performed the first characterization of human SOX18 promoter. We have identified the<br />

transcription start point <strong>and</strong> carried out the structural <strong>and</strong> functional analysis of the regulatory<br />

region responsible for SOX18 expression in HeLa cell line. Using promoter-reporter<br />

constructs, we have determined the minimal SOX18 promoter region that shows basal<br />

promoter activity. Further, we have investigated functional properties of GC-rich motif within<br />

the core promoter sequence. In silico analysis has shown that this motif has several putative<br />

binding sites for transcription factor Sp1 (specificity protein 1). SP1 is characteristic<br />

transcription factor that regulates gene expression within TATA-less promoters, <strong>and</strong> SOX18<br />

promoter has no TATA box or any other conserved element commonly present in TATA-less<br />

promoter. Our experiments confirmed binding of Sp1 transcription factor to proximal GC-rich<br />

motif. Finally, functional analysis revealed that over expression of Sp1 transcription factor in<br />

HeLa cell line positively regulates the activity of SOX18 promoter.<br />

- 559 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 57<br />

Expression of the E2F family of transcription factors <strong>and</strong> its clinical relevance in a test<br />

set of 78 ovarian cancer patients.<br />

Daniel Reimer, Susann Sadr, Annemarie Wiedemair, Nicole Concin, Gerda Hofstetter,<br />

Christian Marth <strong>and</strong> Alain G. Zeimet<br />

Department of Obstetrics <strong>and</strong> Gynaecology, Medical University Innsbruck, Austria<br />

Email: daniel.reimer@uibk.ac.at<br />

The E2F family of transcription factors, firstly discovered as a cellular activity that could bind<br />

to <strong>and</strong> activate the adenoviral E2 gene promoter, plays a pivotal role in the regulation of<br />

cellular proliferation by controlling the expression of genes required for both cell cycle<br />

progression <strong>and</strong> apoptosis. Based upon sequence homology <strong>and</strong> function, eight distinct<br />

members of E2F transcription factors have been distinguished until to date. E2F family is<br />

roughly subdivided into proliferation-promoting (E2F1, E2F2, E2F3a) <strong>and</strong> –inhibiting<br />

(E2F3b, E2F4, E2F5, E2F6) transcription factors. E2F7 <strong>and</strong> the recently described E2F8,<br />

represent a distinct subgroup through their unique structure predominantly inhibiting E2Fdriven<br />

gene promoters. Whereas, E2F family members generally require binding to its<br />

dimerization partner DP for transcriptional activity, E2F7 <strong>and</strong> E2F8 lack the dimerization<br />

domain.<br />

The regulation of E2F transcription factors is closely associated with the function of the<br />

retinoblastoma family of tum<strong>our</strong> suppressors, the retinoblastoma binding protein (pRB), p107<br />

<strong>and</strong> p130. In the last decade various alterations of distinct components of the pRB-E2F<br />

pathway were found to be associated with tum<strong>our</strong> progression through increase of<br />

proliferation or inhibition of apoptosis. However, no data on the role of E2F family members<br />

are available in tum<strong>our</strong> biology of ovarian cancer. Here we describe an expression study of<br />

E2F transcription factors in various human ovarian cancer cell lines <strong>and</strong> its clinical relevance<br />

was examined in a training set of 78 ovarian cancer patients.<br />

Expression levels of E2F1, E2F2 <strong>and</strong> E2F8 were elevated in all the ovarian cancer cell lines<br />

studied when compared with peritoneal mesothelial cells (PMC) <strong>and</strong> ovarian surface<br />

epithelial cells (OSE). Interestingly, interferon-gamma treatment showed a time-dependent<br />

reduction of the activating transcription factors E2F1 <strong>and</strong> E2F2 in HTB-77 cells, indicating<br />

that the antiproliferative effect of interferon-gamma is mediated at least in part through downregulation<br />

of proliferation-promoting E2F members. High expression of E2F1, E2F2 <strong>and</strong><br />

E2F8 was found to be associated with poor overall survival <strong>and</strong> large residual disease after<br />

initial debulking surgery in ovarian cancer patients, whereas high expression levels of E2F4, a<br />

repressive transcription factor, was associated with fav<strong>our</strong>able overall survival.<br />

Taken together, these data suggest that E2F transcription factors, mainly E2F1, E2F2, E2F8<br />

<strong>and</strong> E2F4, play a pivotal role in tum<strong>our</strong> biology of ovarian cancer <strong>and</strong> may be c<strong>and</strong>idates for<br />

specific therapeutic targets.<br />

- 560 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 58<br />

Identification <strong>and</strong> Functional Analysis of genes related to the malignant transformation<br />

induced by the Polyomavirus Middle T oncoprotein<br />

Leonardo de O. Rodrigues.; Fern<strong>and</strong>a Festa.; Sheila Maria B. Winnischofer; Karin<br />

Krogh; Christian Colin; Mari Cleide Sogayar<br />

University of São Paulo, Chemistry Institute, Biochemistry Department, CP26077, São<br />

Paulo, SP, Brazil, E-mail: leonardo@iq.usp.br<br />

Polyomavirus (Py), a virus associated with the development of animal tumors, has been<br />

widely used as a model for studies of malignant transformation, cell <strong>signaling</strong> pathways <strong>and</strong><br />

cell proliferation control. Polyomavirus Middle T antigen (MT) plays a central role in cell<br />

transformation by binding to <strong>and</strong> activating several cytoplasmic proteins, which participate in<br />

the transduction of growth factor-induced mitogenic signals to the nucleus. To uncover the<br />

molecular basis of cell transformation induced by the MT antigen we used three different<br />

approaches. Firstly, commercially available cDNA arrays were hybridized with cDNA probes<br />

synthesized from mRNA extracted from Balb/c 3T3 cell lines transformed with a retroviral<br />

vector containing the wild type MT-antigen (MTWT) or with the empty vector (PLJ). In the<br />

second approach, cDNA libraries for MT-induced or repressed genes were constructed using<br />

the RDA (Representational Difference Analysis) cDNA subtraction methodology, followed<br />

by immobilization onto nylon membranes to generate DNA macroarrays, which were<br />

screened for induced or repressed genes. The third approach involved a commercially<br />

available gene array (CodeLink bioarrays) consisting of 10,000 mouse genes. Upon<br />

identification of differentially expressed cDNA c<strong>and</strong>idates, their differential expression was<br />

confirmed by quantitative Real-Time PCR analysis. After confirmation of their differential<br />

expression, two genes displaying a complete inhibition upon MT overexpression were<br />

selected for further functional analysis. Isolation <strong>and</strong> characterization of Py-MT-regulated<br />

genes may contribute to better underst<strong>and</strong>ing of the molecular mechanisms underlying not<br />

only malignant transformation, but also the cell cycle control. Support: FAPESP, CNPq,<br />

FINEP <strong>and</strong> PRP-USP<br />

- 561 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 59<br />

Endoglin modify wound healing related dermal fibroblasts properties<br />

Alicia Rodríguez-Barbero, Miguel Pericacho, Marta Prieto, José M. López-Novoa<br />

Instituto Reina Sofía de Investigación Nefrológica. Departamento de Fisiología y<br />

Farmacología. Universidad de Salamanca. Campus Unamuno, Edificio Departamental.<br />

Avda. Campo Charro s/n. 37007. Salamanca. Spain. E-mail: barberoa@usal.es<br />

Endoglin is a transmembrane glycoprotein involved in pathophysiological processes such a<br />

vascular remodeling, angiogenesis <strong>and</strong> fibrosis, but its contribution in wound healing has not<br />

been study. Endoglin is an accessory receptor of the transforming growth factor beta (TGF-#).<br />

As TGF-# is a cytokine involve in wound healing <strong>and</strong> endoglin modulate some cellular<br />

responses to TGF-#, we hypothesize that endoglin could modulate processes involved in<br />

wound healing. The aim of the present study was to evaluate the role of endoglin on cellular<br />

properties implicated in wound healing such as cell proliferation, migration <strong>and</strong> extracellular<br />

matrix synthesis. We generated an in vitro model of primary cultured dermal fibroblasts from<br />

Eng+/- C57BL/6 <strong>and</strong> wild type mice. Western blot analysis revealed that dermal fibroblasts<br />

expressed endoglin <strong>and</strong> that its expression is reduced by 50% in Eng+/- fibroblasts compared<br />

to control cells. The rate of cell growth was faster in Eng+/- than in control fibroblasts. Cell<br />

motility was measured by a multi-channel wounding system such that repair into the denuded<br />

area was due entirely to cell migration <strong>and</strong> by using the Boyden camera. Migration was higher<br />

in Eng+/- fibroblasts than in control cells. In addition, fibronectin synthesis, assessed by<br />

Western blot, was higher in Eng+/- fibroblasts than in control cells. These data reveal the<br />

importance of endoglin in modulating proliferation, migration, <strong>and</strong> extracellular matrix<br />

synthesis in dermal fibroblasts.<br />

- 562 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 60<br />

Isolation <strong>and</strong> characterization of the rat SND p102 gene promoter region<br />

Lorena Rodríguez, Nerea Bartolomé, Itsaso García-Arcos, Begoña Ochoa, María J.<br />

Martínez<br />

Department of Physiology, Faculty of Medicine <strong>and</strong> Dentistry, University of the Basque<br />

Country, P. Box 669, Bilbao, Bizkaia, Spain.<br />

ofbronol@lg.ehu.es<br />

In this work we describe the isolation <strong>and</strong> characterization of the promoter region of the rat<br />

endoplasmic reticulum cholesteryl ester hydrolase gene, named as SND p102 in GenBank<br />

according to the structural properties <strong>and</strong> molecular weight of the protein. A region of 1900<br />

bases upstream the ATG start codon has been isolated by a double PCR-based method. The<br />

transcription initiation site has been localised 216 bases upstream the ATG codon by RLM-<br />

RACE. Bioinformatic analysis of the isolated sequence has revealed the presence of putative<br />

binding sites for many transcription factors implicated in both basal <strong>and</strong> regulated processes,<br />

although no TATA box has been found. Electrophoretic mobility shift <strong>and</strong> supershift assays<br />

(EMSA) using nuclear extracts from HepG2 cells have demonstrated the specific binding of<br />

the nuclear factor-Y (NF-Y), a heterotrimeric transcriptional activator which binds to CCAAT<br />

boxes, to regions [-257, -253], [-290, -286] <strong>and</strong> [-370, -366] of the promoter sequence. The<br />

absence of TATA box <strong>and</strong> the situation of the binding sites for NF-Y suggest a role for this<br />

transcription factor in the transcriptional regulation of the rat SND p102 gene.<br />

This work was supported by DGICYT (BMC2001-0067).<br />

- 563 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 61<br />

Synergy of p38 MAPK <strong>and</strong> Stat1 in Transcription<br />

Iwona Sadzak, Barbara Schaljo <strong>and</strong> Pavel Kovarik<br />

Max F.Perutz Laboratories, Department of Microbiology <strong>and</strong> Immunobiology,<br />

University of Vienna, A-1030 Vienna, Austria<br />

Both interferon (IFN) <strong>and</strong> stress signals are required for full activation of immune responses.<br />

We <strong>and</strong> others have shown that the stress-activated p38 MAPK increases transcription by<br />

Stat1, the key transcription factor of interferon <strong>signaling</strong>. Thus, the two signals synergistically<br />

increase transcriptional responses under conditions of inflammation. We show here that<br />

synergy of p38 MAPK with Stat1 generates unique gene expression patterns that are specific<br />

for inflammatory conditions <strong>and</strong> allow identification of novel interferon-regulated genes. The<br />

molecular basis for the synergy is unknown, but it does not depend on phosphorylation of<br />

Stat1 by p38 MAPK.<br />

We propose a working model for the synergy <strong>and</strong> describe the proteomic approach that we<br />

employ to unravel the molecular mechanisms. We focus on the analysis of Stat1-containing<br />

transcriptional complexes <strong>and</strong> p38-mediated modifications of their composition <strong>and</strong>/or<br />

activity. We show that the amount of Stat1 protein on the promoters of target genes does not<br />

change upon synchronous activation of IFNg <strong>and</strong> p38 MAPK pathway.Further we observe<br />

that the association of CBP/p300 with Stat1 is unaffected by p38 MAPK activation.<br />

- 564 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 62<br />

JUNB-DEPENDENT VEGF TRANSCRIPTION AND TUMOR ANGIOGENESIS<br />

Dirk Schmidt1, Oliver Pein1, Björn Textor1, Alex<strong>and</strong>er Licht1, Norbert E. Fusenig2,<br />

Peter Angel1 <strong>and</strong> Marina Schorpp-Kistner1<br />

1,2Deutsches Krebsforschungszentrum (DKFZ), 1Division of Signal Transduction <strong>and</strong><br />

Growth Control <strong>and</strong> 2Division of Differentiation <strong>and</strong> Carcinogenesis, Im Neuenheimer<br />

Feld 280, D-69120 Heidelberg, Germany. E-mail: marina.schorpp@dkfz.de<br />

The JunB subunit of the AP-1 transcription factor complex mediates gene regulation in<br />

response to a plethora of extracellular stimuli. Previously, we showed that the complete<br />

ablation of JunB resulted in an early embryonic lethal phenotype due to vascular defects. In<br />

order to elucidate the underlying molecular mechanisms we analyzed various JunB-deficient<br />

cell systems <strong>and</strong> targeted deletion of the junB gene to endothelial cells. Endothelial-cell<br />

specific ablation of junB resulted in a similar embryonic lethal phenotype <strong>and</strong> confirmed the<br />

absolute requirement of JunB for vasculogenic <strong>and</strong> angiogenic processes in the developing<br />

embryo.<br />

Analysis of JunB-deficient embryonic stem cells, endothelioma cells <strong>and</strong> fibroblasts revealed<br />

that JunB itself is upregulated in response to hypoxia. This induction is independent of HIF<br />

<strong>and</strong> MAPK <strong>signaling</strong> but requires NF-kB, as junB mRNA induction is lost in fibroblasts with<br />

suppressed NF-kB activity. JunB, in turn, is directly required for basal <strong>and</strong> hypoxia-induced<br />

transcription of VEGF as demonstrated by independent experimental approaches such as<br />

QRT-PCR, EMSA <strong>and</strong> coexpression studies. VEGF regulation by JunB is also of<br />

physiological relevance in tumor angiogenesis, as teratocarcinomas derived from junB-/- ES<br />

cells exhibit a pale <strong>and</strong> growth retarded phenotype with significantly reduced amounts of<br />

midsize <strong>and</strong> large blood vessels. The failure of host-derived vessels to recolonise the tumor<br />

tissue correlates with a reduced capacity of JunB null tumor cells to release the angiogenic<br />

factor VEGF due to the absence of JunB. Yet, other typically hypoxia-induced genes as such<br />

of the glycolytic pathway are not governed by JunB. Consistently, JunB is dispensable for the<br />

early maximal induction of VEGF in response to hypoglycemia which requires the JunB<br />

sibling c-Jun.<br />

- 565 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 63<br />

IKKalphalpha controls NF-kappaB at the skp2 gene promoter to regulate G1 to S phase<br />

progression in pancreatic cancer cells<br />

Günter Schneider, Dieter Saur, Jens T. Siveke, Ralph Fritsch <strong>and</strong> Rol<strong>and</strong> M. Schmid<br />

II. Department of Internal Medicine, Technical University of Munich, Ismaningerstr.<br />

22, 81675 Munich, Germany. E-mail: guenter.schneider@lrz.tum.de<br />

The IkappaB inducing kinase (IKK) consists of the catalytical subunits IKKalphalpha <strong>and</strong><br />

IKK#eta <strong>and</strong> the regulatory subunit IKK amma. IKK regulated <strong>signaling</strong> pathways are<br />

believed to promote proliferation of normal cells <strong>and</strong> aberrant proliferation of cancer cells.<br />

The molecular mechanisms linking IKK components to the cell cycle machinery are not<br />

entirely understood. To study the functions of the catalytical subunits of the IKK complex we<br />

used pancreatic cancer cells, because of their known constitutive IKK activity. We show that<br />

the G1 Phase of the cell cycle is selectively regulated by the IKKalphalpha subunit.<br />

IKKalphalpha regulates protein stability of the cyclin-dependent kinase inhibitor p27Kip1.<br />

Increased levels of p27Kip1 after the transfection of IKKalphalpha specific siRNAs are due to<br />

the downregulation of the F-box protein S-phase-kinase associated protein 2 (skp2). We<br />

further demonstrate, that IKKalphalpha <strong>signaling</strong> regulates transcription of the skp2 gene by<br />

controlling a NF-kappaB complex at the skp2 gene promoter. Together, this work defines a<br />

novel IKKalphalpha regulated growth pathway important for pancreatic cancer cell biology.<br />

- 566 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 64<br />

The cAMP Responsive Unit of the human Insulin-like Growth Factor Binding Protein-1<br />

(hIGFBP-1) gene promoter comprises a functional Insulin Response Element<br />

Ghislaine Schweizer-Groyer 1, Guillaume Fallot 3, Françoise Cadepond 1 <strong>and</strong> André<br />

Groyer 2<br />

1 Inserm U.488, Lab hormones, 94276, Le Kremlin-Bicêtre Cédex, France ; 2 Inserm<br />

U.683 <strong>and</strong> 3 Inserm U.481, Faculté de Médecine Xavier Bichat, 75870, Paris Cédex 18,<br />

France. E-mail: groyer@bichat.inserm.fr<br />

Insulin-like Growth Factor-Binding Protein-1 (IGFBP-1) is one of the genes involved in<br />

glucose homeostasis. In vivo, its level is increased by glucocorticoids <strong>and</strong> glucagon (via<br />

cAMP) <strong>and</strong> decreased by insulin, these variations being primarily correlated with IGFBP-1<br />

gene transcription. A functional Insulin Response Element (IRE), localised immediately 5’ to<br />

the Glucocorticoid Response Element (GRE), has already been shown to mediate insulin<br />

inhibition of basal <strong>and</strong> glucocorticoid-induced stimulation of IGFBP-1 promoter activity.<br />

In this work, using the human hepatoma HepG2 cells as a model system <strong>and</strong> transient<br />

transfection, we showed : 1) that both basal <strong>and</strong> cAMP-induced hIGFBP-1 promoter (nt-1 to -<br />

341) activity are inhibited by insulin; 2) that Forkhead Box class O (Foxo) transcription<br />

factors enhance constitutive hIGFBP-1 promoter activity without interfering with stimulatory<br />

effect of cAMP; 3) that PI-3’ kinase <strong>signaling</strong> is involved in the inhibition of both constitutive<br />

<strong>and</strong> cAMP-induced promoter activity by insulin; 4) that the Foxos mediate the inhibitory<br />

effect of insulin on transcription from the whole IGFBP-1 promoter; 5) that the cAMP<br />

Responsive Unit (CRU) composed of a cAMP Responsive Element (CRE) <strong>and</strong> of a putative<br />

IRE, is functional in mediating both cAMP stimulation <strong>and</strong> insulin inhibition of a<br />

heterologous promoter <strong>and</strong> 6) that the inhibitory effects of insulin on the isolated CRU are<br />

mediated by the Foxos.<br />

- 567 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 65<br />

Role of Proteinase-Activated Receptor-2 on Cyclooxygenase-2 induction in H. pylori -<br />

Infected Gastric Epithelial Cells.<br />

Ji Hye Seo, Joo Weon Lim, Kyung Hwan Kim <strong>and</strong> Hyeyoung Kim*<br />

Department of Pharmacology, Brain Korea 21 Project for Medical Science, Yonsei<br />

University College of Medicine, Seoul 120-752, Korea. E-mail:jhseo@yumc.yonsei.ac.kr<br />

Proteinase-activated receptor-2(PAR2) belongs to a novel subfamily of G-protein-coupled<br />

receptors with seven-transmembrane domains. PAR2 is activated by serine proteases such as<br />

trypsin, mast cell tryptase, <strong>and</strong> allergic or bacterial proteases. The presence of trypsin has<br />

been shown in human stomach. Cyclooxygenase-2(COX-2) is induced by inflammatory<br />

cytokines, growth factors, gastrin <strong>and</strong> reactive oxygen species in gastric epithelial cells which<br />

may led to mutagenesis <strong>and</strong> subsequent metaplasia, dysplasia, <strong>and</strong> cancer formation. We<br />

investigated whether PAR-2 is activated in H. pylori-infected cells, which is related to<br />

induction of COX-2 in gastric epithelial cells. After treatment of H. pylori to AGS cells at the<br />

ratio of 100:1, we determine the expression <strong>and</strong> activation of PAR-2 <strong>and</strong> induction of COX-2.<br />

The same experiments were performed in the cells treated with PAR2 agonist peptide or<br />

PAR2 antisense oligonucleotide. mRNA <strong>and</strong> protein expression were determined by RT-PCR<br />

<strong>and</strong> Western blotting. PAR-2 activation was assessed by increase in intracellular calcium<br />

level. As a result, H. pylori induced the activation <strong>and</strong> expression of PAR-2 as well as COX-2<br />

expression, which were inhibited in the cells treated with PAR2 antisense oligonucleotide.<br />

PAR2 agonist peptide increased the expression of COX-2 in H. pylori-infected AGS cells. In<br />

conclusion, PAR-2 mediates H. pylori- induced COX-2 expression in gastric epithelial cells.<br />

- 568 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 66<br />

IRF-7: new role in the regulation of genes involved in the adaptive immunity<br />

Marco Sgarbanti, Giulia Marsili, Anna Lisa Remoli, Roberto Orsatti<br />

<strong>and</strong> Angela Battistini<br />

Department of infectious, Parasitic <strong>and</strong> immunomediated Diseases, Istituto Superiore di<br />

Sanità, viale Regina Elena, 299-Rome 00161, Italy. E-mail: battist@iss.it<br />

The interferon regulatory factor 7 (IRF-7) a member of the IRF family of transcription factors<br />

is a key player in the innate immune response against viral infections. Constitutive expression<br />

of IRF-7 is limited to peripheral blood lymphocytes <strong>and</strong> dendritic cells while in other cell<br />

types its expression can be induced by type I Interferon. IRF-7 is sequestered in the cytoplasm<br />

of uninfected cells <strong>and</strong> following viral infection, double str<strong>and</strong>ed RNA (dsRNA) or Toll-like<br />

receptor (TLR) signalling it becames phosphorylated by TBK <strong>and</strong> IKK-i kinases.<br />

Phosphorylated IRF-7 migrates in the nucleus where it can activate IFN-alpha genes <strong>and</strong> other<br />

Interferon stimulated genes (ISGs). Here we report that the over-expression of a constitutively<br />

active form of IRF-7 positively regulates the promoter of Interferon regulatory factor 1 (IRF-<br />

1) <strong>and</strong> of the latent membrane protein 2 (LMP-2), two proteins, which play an important role<br />

in adaptive immunity. We previously showed that in T cells, the HIV-1 trans-activator Tat<br />

protein is able to modulate the expression of LMP2 through regulation of IRF-1 expression.<br />

Experiments are, therefore, ongoing to determine whether the up-regulated expression of IRF-<br />

1 <strong>and</strong> LMP-2 is mediated by the Tat-induced IRF-7 activation through the engagement of<br />

TBK-1 <strong>and</strong> IKK-i kinases. Our data point to IRF-7 as a mediator that bridges innate <strong>and</strong><br />

adaptive immunity <strong>and</strong> a potential target of the HIV-1 Tat-mediated immune-modulation.<br />

- 569 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 67<br />

Hodgkin/Reed-Sternberg cells as a Model System for Studying the Canonical <strong>and</strong> Novel<br />

NF-kB pathways<br />

Michael Stillmann, Yoshiaki Sunami, Michael Hinz, Meike Brömer, Jan Ebert <strong>and</strong><br />

Claus Scheidereit.<br />

Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13092 Berlin,<br />

Germany. E-mail : y.sunami@mdc-berlin.de<br />

Proteins of the NF-kB/Rel transcription factor family are involved in the regulation of a<br />

variety of physiological processes including adaptive <strong>and</strong> innate immune responses,<br />

inflammation, cellular survival <strong>and</strong> proliferation. Constitutive NF-kB activity is hallmark of<br />

malignant Hodgkin/Reed-Sternberg cells (H/RS) in Hodgkin lymphoma. In some H/RS cell<br />

types aberrant NF-!B activation could be traced back to inactivating mutations in the IkBa<br />

gene. Moreover, H/RS cell types exhibit enduring activation of the canonical pathway, due to<br />

a not yet identified molecular aberration. Employing a combination of classical biochemical<br />

techniques <strong>and</strong> RNAi we want to analyze the signal transduction through canonical <strong>and</strong> novel<br />

<strong>signaling</strong> pathways in H/RS cells.<br />

We have observed an enhanced kinase activity of the IKK complex <strong>and</strong> high expression of<br />

p100/p52 resulting in a highly increased NF-kB DNA-binding <strong>and</strong> hence target gene<br />

activation. Moreover, phosphorylation of p100 which is prerequisite for further processing to<br />

p52, suggests ongoing <strong>signaling</strong> through the novel pathway. Pulse chase analysis in H/RS<br />

cells demonstrated direct generation of p52 from de novo translated endogenous p100,<br />

suggesting a co-translational mechanism.<br />

Several groups proposed the aberrant overexpression of TNF family receptors CD40 <strong>and</strong><br />

CD30 in H/RS cells as initial cause for constitutive NF-kB activation. However, silencing of<br />

CD40 <strong>and</strong> CD30 expression by RNAi neither affected NF-kB activity nor p100 processing. At<br />

the moment, a possible contribution of a range of NF-kB regulators is analyzed using this<br />

approach. Alternatively, we are applying comparative proteomic methods to define<br />

differential protein-protein interaction patterns of NF-kB key regulators in Hodgkin <strong>and</strong> Non-<br />

Hodgkin cells.<br />

- 570 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 68<br />

Induction of PAI-1 Expression by TNFalpha in Endothelial Cells is Mediated by its<br />

Responsive Element Located in the 4G/5G Site.<br />

Maria Swiatkowska1, Janusz Szemraj 2, Czeslaw S. Cierniewski1,3<br />

Department of Molecular <strong>and</strong> Medical Biophysics1, Department of Biochemistry2,<br />

Medical University in Lodz, Center of Medical Biology3, Polish Academy of Sciences,<br />

Lodz, Pol<strong>and</strong><br />

Plasminogen activator inhibitor type 1 (PAI-1) is induced by many proinflammatory <strong>and</strong> prooxidant<br />

factors. Among them, tumor necrosis factor alpha (TNFalpha), a pivotal early<br />

mediator that regulates <strong>and</strong> amplifies the development of inflammation, is one of the strongest<br />

PAI-1 synthesis activators. Location of the TNFalpha response element in the PAI-1 promoter<br />

is still ambiguous. In this study, we attempted to evaluate the significance of the element<br />

located in the 4G/5G site of the PAI-1 promoter in the TNFalpha stimulation of PAI-1<br />

expression in endothelial cells. PAI-1 expression was monitored at: (a) the level of mRNA<br />

using real-time PCR, (b) PAI-1 gene transcription by transfection reporter assays, <strong>and</strong> (c)<br />

protein synthesis using the enzyme immunoassay. NF-kB activity was monitored using the<br />

electrophoretic mobility shift assay. Its activity was modified by either antisense<br />

oligonucleotides or transfection of endothelial cells with the wild-type or mutated IkBalpha.<br />

We have shown that TNFalpha-induced expression <strong>and</strong> gene transcription of PAI-1 involves a<br />

regulatory region present in segment -664 ⁄-680 of the PAI-1 promoter. This reaction involves<br />

the TNFalpha-induced generation of superoxide leading to activation of NF-kB, <strong>and</strong> can be<br />

abolished by antioxidants <strong>and</strong> by overexpression of a super-suppressor phosphorylationresistant<br />

IkBalpha. Stimulation of PAI-1 under these conditions involves the motif of the PAI-<br />

1 promoter adjacent to the 4G⁄5G site, which can directly interact with NF-kB. We show that<br />

activation of PAI-1 gene by TNFalpha <strong>and</strong> reactive oxygen species is mediated by interaction<br />

of NF-kB with the cis-acting element located in the -675 4G⁄5G insertion ⁄ deletion in the<br />

PAI-1 promoter.<br />

- 571 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 69<br />

Monitoring of dynamics NF-kB activation <strong>and</strong> correlation with mitochondrial <strong>and</strong> RE<br />

Ca2+ in cftr-deficient airway cells<br />

Olivier Tabary1,2, Emilie Boncoeur1, Rainer de Martin3, Rainer Pepperkok2, Annick<br />

Clément1, Carsten Schultz2, Jacky Jacquot1<br />

Inserm, U719, Université Pierre et Marie Curie, Hôpital Saint-Antoine, 75012, Paris,<br />

France ; Cell Biology/Cell Biophysics <strong>and</strong> Gene Expression Program, EMBL,<br />

Meyerhosfstr. 1,D-69117, Heidelberg, Germany;<br />

Department of Vascular Biology <strong>and</strong> Thrombosis Research, Medical University of<br />

Vienna, Austria.<br />

Dysregulation of nuclear factor kappa B (NF-kB) <strong>and</strong> increased Ca2+ signals have been<br />

reported in airway epithelial cells of CF patients. The hypothesis that Ca2+ signalling may<br />

regulate NF-kB activation was tested in a CF bronchial epithelial cell line (IB3-1, CFTR<br />

genotype DF508/W1282X) <strong>and</strong> compared to the CFTR-corrected epithelial cell line S9 using<br />

fluorescence microscopy to monitored in situ dynamics of NF-kB activation at the single cell<br />

level. We examined the change of [Ca2+]i levels using Fluo-3, Rhod-FF <strong>and</strong> NF-kB<br />

activation was monitored by fluorescence microscopy (FRET) with YFP-p65 <strong>and</strong> IkBalpha-<br />

CFP fluorescent fusion proteins in response to stimulation with 20 ng/ml of IL-1b. Upon<br />

stimulation with IL-1b, we first observed, a slow but prolonged [Ca2+]i increase (up to 10<br />

min) in IB3-1 cells compared to S9 cells. During the same time we observed a rapid <strong>and</strong> large<br />

discharge of mitochondrial Ca2+ in CFTR-corrected S9 cells whereas in CFTR-deficient IB3-<br />

1 cells we observed a relative increase to baseline followed by a slow decrease. The IL-1binduced<br />

[Ca2+]i response was further accompanied by an activation of NF-kB in IB3-1 but<br />

not in S9 cells. Pretreatment of IB3-1 cells with the ER Ca2+ pump inhibitor thapsigargin<br />

inhibited the IL-1b-induced [Ca2+]i response. Treatment with either the calcium chelator<br />

BAPTA or an inhibitor of IkBalpha phosphorylation (digitoxin) led to a drastic [Ca2+]i<br />

decrease accompanied by an inhibition of NF-kB activation of IL-1beta-stimulated IB3-1<br />

cells in comparison to untreated cells. In IB3-1 cells cultured at low temperature (26°C)<br />

during 16h, the IL-1b-induced [Ca2+]i response was inhibited <strong>and</strong> no significant NF-kB<br />

activation was observed. To <strong>our</strong> knowledge, this is the first report of visualization of the<br />

Ca2+-mediated activation of NF-kB in individual living airway epithelial cells. Our results<br />

support the concept that [Ca2+]i is a key regulator of NF-kB activation in CF airway<br />

epithelial cells.<br />

Tabary O, Boncoeur E, de Martin R, Pepperkok R, Clement A, Schultz C, Jacquot J. Calciumdependent<br />

regulation of NF-(kappa)B activation in cystic fibrosis airway epithelial cells. Cell<br />

Signal. 2005<br />

- 572 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 70<br />

Characterization of 3T3-L1 adipocytes dedifferentiation induced by a mitochondrial<br />

dysfunction<br />

Silvia Tejerina1, Aurélia De Pauw1, Sebastien Vankoningsloo1, Andrée Houbion1,<br />

Catherine Demazy1, Françoise de Longueville2, Vincent Bertholet2, Patricia Renard1,<br />

José Remacle1,2, Martine Raes1 <strong>and</strong> Thierry Arnould1<br />

1Laboratory of Biochemistry <strong>and</strong> Cellular Biology, University of Namur (F.U.N.D.P.),<br />

Rue de Bruxelles, 61, 5000 Namur, Belgium. Email : thierry.arnould@fundp.ac.be,<br />

2Eppendorf Array Technologies, Rue du Séminaire, 12, 5000 Namur, Belgium.<br />

Studies of several lipid disorders have evidenced a strong link between mitochondrial<br />

dysfunction <strong>and</strong> fat storage abnormalities <strong>and</strong> it is now well accepted that a mitochondrial<br />

dysfunction affects lipid-metabolizing tissues. However, molecular mechanisms involved in<br />

the adipocyte dedifferentiation programme are still poorly understood. In this study, we set up<br />

an experimental model to characterize the dedifferentiation of 3T3-L1 adipocytes induced by<br />

a mitochondrial dysfunction triggered by FCCP, an uncoupling molecule. Differentiating<br />

adipocytes incubated for several days with FCCP show modifications in their phenotype <strong>and</strong><br />

are characterized by a decrease in the triglyceride content accompanied by an increase in<br />

glycerol release (a marker of fatty acid mobilization). However, mechanisms <strong>and</strong> cell<br />

<strong>signaling</strong> seem to be totally different than the ones triggered by TNFalpha! a cytokine<br />

known to stimulate adipocyte dedifferentiation, as PKA <strong>and</strong> MEK1/2 inhibitors do not modify<br />

the triglyceride content in FCCP-treated adipocytes. Using a low density cDNA microarray<br />

allowing gene expression analysis for numerous adipogenic markers, we also found a<br />

significant decrease in the expression of genes encoding key transcription factors involved in<br />

adipogenesis (PPARgamma <strong>and</strong> C/EBPalpha) in adipocytes with impaired mitochondrial<br />

activity. Decrease in the activity of these transcriptional regulators has also been confirmed by<br />

DNA-binding activity assays. Finally, among the genes found to be differentially expressed in<br />

differentiating adipocytes, CHOP-10/GADD153, a natural dominant negative mutant of<br />

C/EBPbeta is specifically up-regulated in adipocytes with uncoupled mitochondria. These<br />

results highlight some new effectors potentially involved in adipocyte dedifferentiation in<br />

response to a mitochondrial dysfunction.<br />

T. Arnould <strong>and</strong> A. De Pauw are respectively Research Associate <strong>and</strong> Research Assistant of<br />

FNRS (Fonds National de la Recherche Scientifique, Brussels, Belgium).S. Tejerina is a<br />

recipient of a CUD (Coopération Universitaire au Développement) fellowship.<br />

- 573 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 71<br />

Ferric citrate abolishes the induction of the hypoxia inducible factor HIF-1alpha<br />

expression <strong>and</strong> restores the inhibition of cell proliferation produced by the flavonoid<br />

quercetin<br />

Anastasia Triantafyllou1, Panagiotis Liakos1, Andreas Tsakalof1, Georgia<br />

Chachami1,2, Efrosyni Paraskeva2, Ilias Athanasiadis1, Paschalis-Adam Molyvdas2,<br />

Eleni Georgatsou1, George Simos1 <strong>and</strong> Sophia Bonanou1<br />

1Laboratory of Biochemistry <strong>and</strong> 2Laboratory of Physiology, Department of Medicine,<br />

School of Health Sciences, University of Thessaly, Larissa, Greece.<br />

E-mail: atsakal@med.uth.gr<br />

Hypoxia induces the expression of the regulatory alpha subunit of the Hypoxia-Inducible<br />

Factor-1 (HIF-1), by preventing its hydroxylation by O2 - <strong>and</strong> Fe2+-dependent prolyl<br />

hydroxylases (PHDs), which target it for proteosomal degradation. Quercetin, a bioflavonoid<br />

with anti-oxidant, metal-chelating, kinase-modulating <strong>and</strong> anti-proliferative properties, can<br />

induce the expression of HIF-1alpha in normoxia, but its mechanism of action has not been<br />

determined. In this study we characterized the induction of HIF-1alpha expression <strong>and</strong><br />

inhibition of proliferation in HeLa <strong>and</strong> ASM (airway smooth muscle) cells by quercetin <strong>and</strong><br />

examined the effect of iron on these processes. We also investigated the cellular uptake <strong>and</strong><br />

biotransformations of quercetin <strong>and</strong> its relevance for HIF-1alpha induction <strong>and</strong> growth<br />

inhibition. Our data demonstrate that addition of excess Fe(III) can abrogate the quercetininduced<br />

stabilization of HIF-1alpha in both cell systems. Moreover, the inhibition of DNA<br />

synthesis, cell proliferation <strong>and</strong> cycle progression caused by quercetin can also be reversed by<br />

excess iron. We propose that iron chelation is the key event in the induction of HIF-1alpha as<br />

well as in the inhibition of cell proliferation produced by quercetin.<br />

- 574 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 72<br />

Glutamate-induced synapse-to-nucleus shuttling of ERK <strong>and</strong> the transcription factor<br />

Elk-1 in striatal neurons requires vesicular transport.<br />

Trifilieff P1,2; Lavaur J1 ; Brami-Cherrier K1 ; Pagès C1; Micheau J2 ; Caboche J1<br />

<strong>and</strong> Vanhoutte P1<br />

1Lab. Signalisation Neuronale et Régulations Géniques-Univ. Pierre et Marie Curie<br />

Paris 6-CNRS-UMR7102.<br />

2 Lab. Neurosciences Cognitives-Univ. Bordeaux I-CNRS-UMR5106.<br />

MAP kinases of the ERK subtype (Extracellular signal-Regulated Kinase) play a critical role<br />

in long term neuronal adaptations, learning <strong>and</strong> memory. These long term events require gene<br />

regulations that depend on the presence of activated ERKs in the nucleus. ERKs are expressed<br />

<strong>and</strong> activated in neurites, at considerable distance from the nucleus, <strong>and</strong> shuttle towards the<br />

nucleus upon activation by a mechanism that is not clearly defined yet. Furthermore, we have<br />

previously shown that one characteristic of neuronal cells is that the transcription factor Elk-1,<br />

one of the major ERKs’ target, has the same expression pattern as ERK <strong>and</strong> is expressed in<br />

both cytoplasmic <strong>and</strong> nuclear compartments.<br />

In the present study, we analysed the sub-cellular fate of ERK <strong>and</strong> Elk-1 in a model of ERKdependent<br />

Immediate Early Gene (IEG) induction on striatal neurons. We show that ERK <strong>and</strong><br />

Elk-1 rapidly translocate to the nucleus upon stimulation with the same kinetics. Confocal<br />

analysis of ERK <strong>and</strong> Elk-1 staining reveal a “clustered” pattern with a high degree of colocalization<br />

with clathrin-coated vesicle (CCV) markers. Co-immunoprecipitation assays<br />

show an interaction of ERK <strong>and</strong> Elk-1 with markers of CCV, which is transiently increased<br />

upon stimulation. Despite the fact that glutamate stimulates endocytosis of both ionotropic<br />

<strong>and</strong> metabotropic glutamate receptors, we establish that ERK interact specifically with<br />

AMPA-containing vesicles. Interestingly, blockade of endocytosis inhibits glutamatedependent<br />

ERK <strong>and</strong> Elk-1 nuclear translocation without alteration of neither ERK activation<br />

nor activation of other MAP kinases. In <strong>our</strong> model, where inhibition of vesicular transport<br />

restricts ERK activation to the cytoplasm, we measured the consequences of the absence of<br />

nuclear translocation of activated ERKs. We underline the multiple roles of ERKs in this<br />

compartment <strong>and</strong> show their functions as key regulators of IEG expression as well as<br />

modulators of chromatin structure via the control of histone phosphorylation. This study<br />

underlies the key role of the vesicular transport in the synase-to-nucleus shuttling of ERK. We<br />

are currently analyzing the functional relevance of the vesicular transport-mediated ERK<br />

nuclear translocation in the establishment of plasticity in the striatum in vivo.<br />

- 575 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 73<br />

A novel human glucocorticoid receptor transcript splice variant: Identification <strong>and</strong><br />

tissue distribution of hGR[delta]313-338, an alternative exon 2 transactivation domain<br />

isoform.<br />

Jonathan D. Turner 1 , <strong>and</strong> Claude P. Muller 1,2<br />

1<br />

Institute of Immunology, Laboratoire National de Santé, 20A rue Auguste Lumière, L-<br />

1011, Luxemb<strong>our</strong>g<br />

2<br />

Department of Immunology, Graduate School of Psychobiology, University of Trier, D-<br />

54290, Germany<br />

Email: jonathan.turner@LNS.ETAT.LU <strong>and</strong> claude.muller@LNS.ETAT.LU<br />

All human glucocorticoid receptor isoforms (hGR) are encoded by the NR3C1 gene. NR3C1<br />

consists of 7 core exons (exons 2-8) common to all the known protein isoforms, has two<br />

major exon 8-9 splice variants, <strong>and</strong> a 5’ UTR consisting of 11 alternative splice variants.<br />

Here, we report the existence of a novel splice variant, hGR[delta]313-338, containing a 26<br />

residue (78 bp) deletion in the N-terminal region (encoded by exon 2) between amino acids<br />

313 <strong>and</strong> 338. This N-terminal region has previously been shown to include the 1<br />

transactivation domain, that is thought to make contact with proteins in the basal<br />

transcriptional apparatus, including the TATA box-binding protein (TBP). The<br />

hGR[delta]313-338 observed at the mRNA level represents a transcript variant encoding a<br />

protein isoform with a deletion between the tau 1 domain <strong>and</strong> the DNA binding domain<br />

(DBD) encoded by exons 3 <strong>and</strong> 4. Interestingly, the deleted residues show a relatively high<br />

abundance of potential phosphorylation sites (4 serines, 2 threonines, <strong>and</strong> 2 tyrosines, 28% of<br />

deleted residues) including serine 317 that has been shown to be phosphorylated. It is thought<br />

that phosphorylation plays an important role in transactivation action of hGR. It is also known<br />

from knockout mice that removal of the entire exon 2 covering both the tau 1 transactivation<br />

domain <strong>and</strong> <strong>our</strong> deleted region does not result in the expression of a transcriptionally inactive<br />

receptor; however, it produces an altered glucocorticoid- induced transcription pattern. Here<br />

we report the first observation of hGR[delta]313-338, <strong>and</strong> its sequence <strong>and</strong> its expression in a<br />

range of human tissues. We hypothesise that hGR[delta]313-338 represents an isoform of the<br />

hGR with an altered glucocorticoid induced transactivation profile.<br />

- 576 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 74<br />

Importance of the coordinate expression of PKC" <strong>and</strong> Ets1 for breast cancer cells<br />

Martina Vetter 1 , Dario Schunke 1 , Paul N. Span 2 , Fred Sweep 2 , Henrike Ronneburg 1 ,<br />

H.-J. Holzhausen 3 , Angela Dittmer 1 , Christoph Thomssen 1 <strong>and</strong> Jürgen Dittmer 1<br />

1 Clinic for Gynecology, University of Halle, Germany, 2 Department of Chemical<br />

Endocrinology, Radboud University Nijmegen Medical Centre, Nijmegen, Germany,<br />

3 Institute for Pathology, University of Halle, Germany<br />

Protein kinase C" (PKC") <strong>and</strong> the transcription factor Ets1 are often associated with<br />

advanced tumor progression. We have previously shown that PKC" is able to<br />

phosphorylate Ets1 <strong>and</strong> to increase its transcriptional activity. By using RNA interference<br />

we could now show that PKC" also positively influences Ets1 expression in a variety of<br />

cancer cell lines. One major way by which PKC" regulates Ets1 expression is by<br />

increasing Ets1 protein stability. The relationship between PKC" <strong>and</strong> Ets1 was confirmed<br />

by the finding that, in primary breast cancers, Ets1 expression correlates with that of<br />

PKC". In searching for similar effects of PKC" <strong>and</strong> Ets1 we found that the response of<br />

MDA-MB-231 cells to mithramycin or UV-light were similarly affected by PKC"- <strong>and</strong><br />

Ets1-specific siRNAs, whereas only PKC"-specific siRNA modulated cell morphology<br />

<strong>and</strong> anchorage-independent growth. In an effort to identify PKC" <strong>and</strong> Ets1-responsive<br />

genes by microarray analysis we found that the RNA levels of thirty-two genes were<br />

similarly affected by both the PKC"- <strong>and</strong> Ets1-specific siRNAs. Among those genes were<br />

MMP1 <strong>and</strong> MMP9 known to be regulated by Ets1 as well as the Ets1 co-activators SP100<br />

<strong>and</strong> AML-1. Another responder gene was Rho-GDI#, an inhibitor of Rho-GTPases some<br />

of which are important regulators of cellular migration. In MDA-MB-231 cells, PKC"- or<br />

Ets1-specific siRNA downregulated both the RNA- as well as the protein level of Rho-<br />

GDI#. In primary breast cancer, expression of Rho-GDI#, but not of Rho-GDI" <strong>and</strong> Rho-<br />

GDI$, correlated with the levels of PKC" <strong>and</strong> Ets1 suggesting that there is a general link<br />

between PKC"/Ets1 <strong>and</strong> Rho-GDI# in breast cancer cells. Interestingly, expression of<br />

Vav1, another Rho-regulating protein which cooperates with Rho-GDI# in T-cells to<br />

activate NFAT, was found to correlate with Rho-GDI#, PKC" <strong>and</strong> Ets1 expression. The<br />

importance of Rho-GDI# <strong>and</strong> Vav1 in breast cancer cells is currently under investigation.<br />

- 577 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 75<br />

c-Jun as a sensor of DNA damage<br />

Vinciguerra M.1-2, Esposito I.2, Cosentino C.2, Maggiolini M.1 <strong>and</strong> Musti A.M.1-2<br />

1. Dipart. Farmaco-Biologico Università della Calabria, Rende, Italy<br />

2. Dipart. Biol. Patol. Cell. Mol. I Università di Napoli “Federico II”, Italy<br />

DNA damage elicits a cellular response resulting in the onset of cell cycle delay, DNA repair<br />

or apoptosis. Central to this process is the DNA-damage dependent activation of three IP3<br />

kinase family members ATM, ATR <strong>and</strong> DNA-PK, which in turn initiate phosphorylation of<br />

proteins involved in the DNA damage pathway. The inability to respond properly to DNA<br />

damage leads to genetic instability, which in turn may enhance the rate of cancer<br />

development. Emerging evidence suggest that, in early stages of oncogenesis, the surviving<br />

transformed cells can be reprogrammed to cell death trough the stress signalling activated by<br />

DNA-damaging anticancer chemotherapies. However advance tumors are frequently resistant<br />

to these treatments, a response that in certain cancer cells seems to be mediated by DNA-PK<br />

over-activity, or by active c-Jun/AP1 transcription factor. c-Jun represents the intersection of<br />

multiple pathways eliciting quite opposite programs, as cellular survival or apoptosis. So far<br />

the regulatory signals leading to this divergence have not been identified. We have<br />

characterized a novel c-Jun consensus phosphorylation site for ATM/ATR kinases. Our<br />

results indicate that DNA-damage dependent phosphorylation of c-Jun at the ATM/ATR<br />

consensus site in turn switches on c-Jun pro-apoptotic phosphorylation at JNK-specific sites.<br />

Furthermore, <strong>our</strong> results suggest that differential regulation of c-Jun phosphorylation by<br />

apical DNA-sensor kinases may play an important role in the chemoresponsiveness of tumor<br />

cells exposed to DNA-damaging agents such as the topoisomerase<br />

II inhibitor etoposide.<br />

- 578 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 76<br />

Blocking NF-kB activation by engineering proteins targeted to the minimal<br />

oligomerization domain of NEMO<br />

Emanuel Wyler*, Monika Kaminska, Andreas Plückthun, Michel Veron, <strong>and</strong> Fabrice<br />

Agou<br />

The Unit of Enzymatic Regulation of Cell Activities, Pasteur Institut, Paris, France,<br />

*Present address: Institut of Biochemistry, ETH, Zurich, Switzerl<strong>and</strong><br />

wyler@bc.biol.ethz.ch,kaminska@pasteur.fr,plueckthun@bioc.unizh.ch<br />

mveron@pasteur.fr, fagou@pasteur.fr<br />

The link between NF-kB signal transduction pathway <strong>and</strong> cancer is now well established.<br />

Thus, inhibiting this pathway is a promising clinical approach through a pro-apoptotic effect<br />

in the treatment of certains cancers. It has already proved to be efficient in therapy, in<br />

combination with chemotherapy <strong>and</strong> radiation for a variety of cancers. The IKK complex is a<br />

priviledged target for designing inhibitors due to its central role of the pathway. We<br />

previously defined a minimal oligomerization domain of NEMO <strong>and</strong> showed that<br />

oligomerization was necessary for NEMO function. This allowed us to develop new<br />

inhibitory peptides of the NF-kB pathway targeting NEMO oligomerization (1).<br />

Ankyrins constitute an attractive class of stable <strong>and</strong> small repeat proteins that provide variable<br />

<strong>and</strong> modular binding surfaces to a target protein. We used the ribosom display method to<br />

select ankyrins binding to the NEMO minimal oligomerization domain from a native ankyrin<br />

library. After f<strong>our</strong> rounds of selection, several ankyrins with affinity in the low nanomolar<br />

range were isolated.When expressed in 293T cells, the selected ankyrins strongly inhibit<br />

TNF-a-mediated NF-kB activation while having no effect on the basal activity. Controls with<br />

native ankyrin or null plasmid were without effect. Furthermore, we could show that this NFkB<br />

inhibition occurs through a specific interaction between ankyrin binders <strong>and</strong> the<br />

endogenous NEMO, resulting in the IKK inhibition. Our findings indicate that high-affinity<br />

binders selected from peptide or chemical libraries against the minimal oligomerization<br />

domain of NEMO can be a promising strategy to search for specific NF-kB inhibitors. Our<br />

binders might also be used for structural studies for both NEMO <strong>and</strong> the minimal<br />

oligomerization domain.<br />

F. Agou et al. (2004) J.Biol.Chem. 279, 54248-54257<br />

- 579 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 77<br />

Diphenyleneiodonium inhibits cerulein-induced apoptosis in pancreatic acinar cells<br />

Ji Hoon Yu, Joo Weon Lim, Kyung Hwan Kim, <strong>and</strong> Hyeyoung Kim*<br />

Department of Pharmacology, Brain Korea 21 Project for Medical Science, Yonsei<br />

University, College of Medicine, Seoul 120-752, Korea. E-mail: jihoonyu@hotmail.com<br />

Apoptosis linked to oxidative stress has been implicated in pancreatitis. Recently we<br />

demonstrated that NADPH oxidase subunits Nox1, p27phox, p47phox <strong>and</strong> p67phox are<br />

constitutively expressed in pancreatic acinar cells. Diphenyleneiodonium (DPI) is widely used<br />

as an inhibitor of flavoenzymes, particularly NADPH oxidase. We investigated whether<br />

inhibition of NADPH oxidase with treatment of DPI suppresses ROS production <strong>and</strong> the<br />

activation of NADPH oxidase in pancreatic acinar AR42J cells stimulated with cerulein, <strong>and</strong><br />

whether cerulien induces the expression of apoptosis-inducing factor (AIF), caspase-3<br />

activation <strong>and</strong> apoptosis, which is inhibited by DPI. To determine the effect of DPI on<br />

cerulein-evoked Ca++ influx, intracellular Ca2++ level was monitored in the cells treated<br />

with or without DPI <strong>and</strong> cultured in the presence of cerulein. As a result, cerulein induced the<br />

activation of NADPH oxidase, determined by ROS production <strong>and</strong> translocation of cytosolic<br />

subunits p47phox <strong>and</strong> p67phox to the membrane, which is in parallel with increase in<br />

apoptotic indices, including the expression of AIF, caspase-3 activation, TUNEL staining,<br />

DNA fragmentation <strong>and</strong> decrease in cell viability. Treatment with DPI inhibited ceruleininduced<br />

activation of NADPH oxidase <strong>and</strong> apoptosis, but not Ca++ influx in pancreatic acinar<br />

cells. These results demonstrate that inhibition of NADPH oxidase by DPI may alleviate<br />

ROS-mediated apoptosis in pancreatic acinar cells.<br />

- 580 -


Session XIV : Transcriptional <strong>and</strong> translational control Poster XIV, 78<br />

NFAT <strong>and</strong> bone disease: a possible “decoy” therapeutic approach<br />

Margherita Zennaro, Elisabetta Lambertini, Roberto Gambari, Letizia Penolazzi <strong>and</strong><br />

Roberta Piva.<br />

Department of Biochemistry <strong>and</strong> Molecular Biology, University of Ferrara, Italy. Email:<br />

piv@unife.it<br />

Bone is a dynamic tissue, under constant remodelling all life long. This process results from<br />

the balance between the activity of osteoblasts (OBs) <strong>and</strong> osteoclasts (OCs), that deposit <strong>and</strong><br />

resorb bone respectively. Any modification of this delicated equilibrium can affect bone<br />

integrity, leading to patologies such as osteopenic disorders or osteopetrosis. A central role in<br />

the control of OCs functions <strong>and</strong> in some OBs activities too, was recently described for<br />

Nuclear Factor of Activated T-cells (NFAT) c1. In this study, we suggest a possible<br />

involvment of NFATc1 in the transcriptional regulation of human estrogen receptor alpha<br />

(ER) gene in both kind of cells. It is well known that estrogen influences OBs <strong>and</strong> OCs<br />

activities, playing a fundamental role in protection of bone mass, as indicated, for example, by<br />

association of osteoporosis <strong>and</strong> menopause.<br />

We previously demonstrated that a specific oligonucleotide (RA4-3’) used as decoy molecule<br />

can increase ER expression in bone cells, by interfering with the activity of an unidentified<br />

negative transcription factor. Interestingly, RA4-3’ sequence contains a putative binding site<br />

(TGAAAA) for NFAT that is the aim of these investigations. First of all we detected a high<br />

level of NFATc1 expression in OCs <strong>and</strong> in SaOS-2 osteoblastic cells, treated with PMA <strong>and</strong><br />

ionomycin. We then verified the specificities of the interaction between NFAT protein <strong>and</strong><br />

<strong>our</strong> sequence, by using cross competition in electrophoretic mobility shift assay. Preliminary<br />

results indicat that a specific interaction between NFAT <strong>and</strong> <strong>our</strong> sequence occurs, probably<br />

mediated by some NFAT co-transcription factors, such as AP-1. Next, we verified NFAT<br />

functionality in OCs <strong>and</strong> SaOS-2 through the transfection of pNFAT-TA luc cis-reporter<br />

vector, containing three t<strong>and</strong>em copies of the NFAT-consensus sequence upstream of the<br />

luciferase gene, in combination with RA4-3’. This caused a decrease of Luc activity, due to<br />

the recruitment of NFAT proteins by RA4-3’. In addition, we demonstrated that RA4-3’<br />

selectively induces apoptosis in OCs but not in OBs. In view of a possible therapeutic<br />

application of NFAT decoy, the role of NFAT in this issue was also investigated.<br />

- 581 -


Notes<br />

- 582 -


Notes<br />

Notes<br />

- 583 -


- 584 -


Session XV : Reactive oxygen species <strong>and</strong> cell <strong>signaling</strong><br />

- 585 -


Session XV : Reactive oxygen species <strong>and</strong> cell <strong>signaling</strong> Poster XV, 1<br />

Evidence for a defective lung NF-kB activation by oxidative stress in cystic fibrosis cell<br />

culture <strong>and</strong> mouse models<br />

Emilie Boncoeur, Olivier Tabary, Elise Bonvin, Annick Clément, Alex<strong>and</strong>ra Henrion-<br />

Caude, Jacky Jacquot.<br />

Inserm U 719, Hôpital Saint-Antoine, UPMC ParisVI, 75571, Paris, France<br />

Lung in patients with cystic fibrosis (CF) is characterized by structural damage <strong>and</strong> altered<br />

repair due to oxidative stress. To gain insights into the oxidative stress-related damage in CF<br />

lung, we studied the effects of hyperoxia (95 % O2) in CF <strong>and</strong> normal mouse lung <strong>and</strong> cell<br />

culture models. In normal lung epithelium, protection against hyperoxic injury has been<br />

shown to be related to an increased expression of the cell cycle inhibitor p21WAF1/CIP1 thus<br />

confering a survival advantage to damaged cells <strong>and</strong> to increased NF-kB activity by involving<br />

a cytoplasm-to-nucleus translocation after degradation of the inhibitor protein IkB alpha by<br />

the proteasome machinery. Here we demonstrate in CF in vivo <strong>and</strong> in vitro models that<br />

hyperoxia did not induce neither increased p21WAF1/CIP1 expression nor lung NF-kB<br />

activation IkB alpha degradation, nor induction of caspase 3 <strong>and</strong> subsequent apoptosis but,<br />

unexpectedly caused enhanced proteasome activity, in contrast to that observed in normal in<br />

vivo <strong>and</strong> in vitro controls. Interestingly, ectopic expression of p21WAF1/CIP1 in hyperoxiaexposed<br />

CF lung epithelial cells or treatment with the selective proteasome inhibitor MG132<br />

restored the NF-kB activation <strong>and</strong> IkB alpha degradation which was associated with an<br />

increased caspase-3 activity <strong>and</strong> apoptotic cell death. Our data suggest that the use of<br />

proteasome inhibitors or related substances modulating p21WAF1/CIP1 degradation,<br />

promoting the activation of NF-kB in hyperoxic conditions could be a valuable approach to<br />

protect lung epithelial cells from oxidative stress in CF patients.<br />

- 586 -


Session XV : Reactive oxygen species <strong>and</strong> cell <strong>signaling</strong> Poster XV, 2<br />

Production of DNA str<strong>and</strong> breaks by metal-induced oxygen radicals<br />

Ezzatollah Keyhani(1,2), Farnoosh Attar(1), Fatemeh Abdi-Oskoui(1), Jacqueline<br />

Keyhani(2)<br />

(1)Institute of Biochemistry <strong>and</strong> Biophysics, University of Tehran, 13145 Tehran, <strong>and</strong><br />

(2)Laboratory for Life Sciences, Tehran 19979, Iran. E-mail: keyhanie@ibb.ut.ac.ir<br />

Elevated levels of reactive oxygen species (ROS) have been implicated in the etiology of<br />

cancer, neurodegenerative <strong>and</strong> cardiovascular diseases, as well as in the aging process <strong>and</strong> as<br />

a condition promoting breast cancer metastases. H2O2, a product of the cell metabolism, is a<br />

potential s<strong>our</strong>ce of ROS. In small concentrations (10-6 M), H2O2 is a <strong>signaling</strong> molecule<br />

capable of inducing chemotactic activity, stimulating the synthesis of cytoskeleton elements,<br />

<strong>and</strong> causing changes in cytosolic calcium concentrations. H2O2 is normally disposed of by<br />

specialized enzymes such as catalases <strong>and</strong> peroxidases. However, various physiologic<br />

perturbations of cellular homeostasis may lead to a dramatic increase in the amount of H2O2<br />

within a cell. In the presence of transition metals, H2O2 is rapidly converted to the highly<br />

reactive <strong>and</strong> highly toxic hydroxyl radical. The latter is responsible for lipid peroxidation,<br />

oxidative damage to proteins <strong>and</strong> breakage of DNA str<strong>and</strong>s. In this study, the extent of DNA<br />

damage caused by H2O2 in the presence of various transition metals was evaluated in vitro.<br />

Purified Salmonella typhimurium DNA was exposed to 0.1 to 100 mM H2O2 <strong>and</strong> either<br />

Fe2+, Fe3+, Cu2+, Ni2+, or Cd2+ in increasing concentrations from 0.01 to 0.1 mM.<br />

Damage to DNA was assessed by electrophoresis of the DNA preparations in 1% agarose gel.<br />

Breakage of the DNA str<strong>and</strong>s would produce a series of DNA fragments of various sizes<br />

resulting in a smear in the gel after electrophoresis while intact DNA would produce a single<br />

b<strong>and</strong>. Results showed that all of the metals investigated triggered DNA breakage in the<br />

presence of H2O2. The extent of breakage depended on the metal ion, its concentration, <strong>and</strong><br />

H2O2 concentration. Addition of either EDTA or catalase to the reaction mixture completely<br />

inhibited the DNA degradation, confirming the involvement of both the metal ion <strong>and</strong> the<br />

H2O2 in the breakage of DNA str<strong>and</strong>s. Production of the hydroxyl radical when H2O2 <strong>and</strong> a<br />

metal ion were both present in the reaction mixture was verified by the thiobarbituric acid<br />

method. The extent of DNA breakage caused by the metal ions was as follows:<br />

Cu2+>Fe2+>Fe3+>Ni2+>Cd2+.<br />

- 587 -


Session XV : Reactive oxygen species <strong>and</strong> cell <strong>signaling</strong> Poster XV, 3<br />

Antioxidant enzymes during hypoxia-anoxia <strong>signaling</strong> events in Crocus sativus L. corm<br />

Ezzatollah Keyhani(1,2), Jacqueline Keyhani(2), Mahnaz Hadizadeh(1), Lila<br />

Ghamsari(1)<br />

(1)Institute of Biochemistry <strong>and</strong> Biophysics, Univ. of Tehran, 13145 Tehran, Iran, <strong>and</strong><br />

(2)Laboratory for Life Sciences, 19979 Tehran, Iran. E-mail: keyhanie@ibb.ut.ac.ir<br />

Saffron has been shown to have therapeutic <strong>and</strong> preventive effects for various cancers <strong>and</strong><br />

other ailments. The saffron plant (Crocus sativus L.) is propagated only via its corms. Thus a<br />

better knowledge of the corm biochemistry <strong>and</strong> of its response to stresses is m<strong>and</strong>atory.<br />

Reactive oxygen species (ROS) are involved in the response to hypoxia/anoxia (H/A), <strong>and</strong> it<br />

was also shown that anoxia stress led to hydrogen peroxide formation in plants. H/A was<br />

produced in Crocus sativus L. corms, either dormant or cultivated for 3 days, by flooding<br />

them. Flooded corms were withdrawn after 1, 2, 3, 4, 8, 10 <strong>and</strong> 14 days under water <strong>and</strong> used<br />

immediately, as well as control corms cultivated under normoxic conditions, for extract<br />

preparation. ROS scavenging enzymes activity was studied in the obtained extracts. Catalase<br />

activity was 2.5 times the control value in dormant corms flooded for 8 days, but exhibited a<br />

f<strong>our</strong>-fold increase compared to control in corms cultivated for 3 days <strong>and</strong> then flooded for just<br />

1 day. The activities of o-dianisidine <strong>and</strong> ascorbate peroxidases decreased in dormant corms<br />

maintained in H/A, while they consistently increased in corms cultivated for 3 days prior to<br />

H/A. Superoxide dismutase (SOD) activity exhibited a two-fold increase in dormant corms<br />

flooded for 2 to 4 days, then returned to the control value. In corms cultivated for 3 days<br />

prior to H/A, SOD activity remained at the control level except for a two-fold increase at day<br />

8. Glutathione peroxidase activity was consistently higher than the control, both in dormant<br />

corm <strong>and</strong> in corm cultivated for 3 days prior to H/A. Data showed that 1) H/A stimulated<br />

catalase, SOD <strong>and</strong> gluthatione peroxidase activities in dormant corms; 2) H/A stimulated all<br />

five enzymes studied in corms cultivated under normoxic conditions prior to flooding with a<br />

maximum stimulation for catalase, followed, in decreasing order, by o-dianisidine peroxidase,<br />

ascorbate peroxidase, glutathione peroxidase <strong>and</strong> SOD; 3) there is a fine regulation of ROS<br />

scavenging enzymes response depending on whether H/A occured in dormant corms, or in<br />

corms grown for 3 days under normoxic conditions prior to H/A.<br />

- 588 -


Session XV : Reactive oxygen species <strong>and</strong> cell <strong>signaling</strong> Poster XV, 4<br />

4-Hydroxyestradiol Promotes Neoplastic Transformation of Human Breast Epithelial<br />

Cells through Generation of Reactive Oxygen Species<br />

Sin-Aye Park, Eun-Hee Kim, Hye-Kyung Na, <strong>and</strong> Young-Joon Surh<br />

National Research Laboratory of Molecular Carcinogenesis <strong>and</strong> Chemoprevention,<br />

College of Pharmacy, Seoul National University, Seoul 151-742, South Korea<br />

Epidemiological studies <strong>and</strong> animal experiments have revealed that estrogens are<br />

carcinogenic. Catechol estrogens are considered to be critical intermediates in estrogeninduced<br />

carcinogenesis. It has been suggested that further oxidation of catechol estrogens to<br />

quinones through redox cycling produces reactive oxygen species (ROS) which can cause<br />

oxidative DNA damage in cells. In the present study, we have examined whether<br />

carcinogenic potential of catechol estrogens is associated with production of ROS <strong>and</strong><br />

subsequent activation of cellular <strong>signaling</strong> pathways. 4-Hydroxyestradiol (4-OHE2), a<br />

representative catechol estrogen, significantly enhanced colony formation in human<br />

mammary epithelial cells (MCF10A) initiated with 9,10-dimethyl-1,2-benz(a)anthracene<br />

(DMBA). 4-OHE2-treated MCF10A cells also accumulated the intracellular ROS, which was<br />

abolished by the antioxidant Trolox. N-acetyl-L-cysteine (NAC) significantly inhibited the 4-<br />

OHE2-promoted anchorage-independent colony formation initiated with DMBA. NF-!B, a<br />

representative redox sensitive transcription factor, has been known to be involved in<br />

carcinogenesis. 4-OHE2 induced NF-!B DNA binding <strong>and</strong> transcription activity, but did not<br />

affect AP-1 DNA binding. 4-OHE2 induced expression of cyclooxygenase-2 (COX-2) <strong>and</strong><br />

inducible nitric oxide synthase in MCF10A cells. 4-OHE2 also activated ERK1/2 <strong>and</strong> AKT<br />

which are up-stream target molecules of NF-!B. To determine whether 4-OHE2-enhanced<br />

ROS production is associated with phosphorylation of ERK <strong>and</strong> AKT, MCF10A cells were<br />

treated with NAC in the presence of 4-OHE2. NAC inhibited the phosphorylation of ERK1/2<br />

<strong>and</strong> AKT induced by 4OHE2. NAC <strong>and</strong> pharmacological inhibitors of ERK <strong>and</strong> AKT<br />

attenuated the 4-OHE2-induced NF-!B DNA binding activity <strong>and</strong> also COX-2 expression in<br />

MCF10A cells. Taken together, above findings suggest that 4-OHE2-induced ROS<br />

production contributes to neoplastic transformation of MCF10A cells via activation of<br />

AKT/ERK/NF-!B <strong>signaling</strong>.<br />

- 589 -


Session XV : Reactive oxygen species <strong>and</strong> cell <strong>signaling</strong> Poster XV, 5<br />

Functional Genetic Screens to Identify Genes in Adaptative Responses to Hypoxia<br />

Celine Rofel1, Iris Partouns1, Raymond Hessing1, Elena Bardina1, Marianne<br />

Koritzinsky2, Brad Wouters2, Jan Willem Voncken1<br />

1) Department of Molecular Genetics<br />

2) Department of Radiation Oncology; Research Institute Growth & Development,<br />

Maastricht University, Universiteitssingel 50 6229 ER; Maastricht, The Netherl<strong>and</strong>s<br />

Tumors need oxygen <strong>and</strong> nutrient to grow, but these two factors are rapidly depleted, due to a<br />

rapid growth, leading to hypoxic tissue in the tumor. Hypoxia remains a constant feature of<br />

these tumors <strong>and</strong> contributes to tumor progression by triggering adaptative mechanisms.<br />

HIF-1 is one of most important transcription factor up-regulated during hypoxia.<br />

HIF-1 alpha appear to be important for tumor vascularization <strong>and</strong> metabolic adaptation to<br />

hypoxia, which are essential for tumor progression.<br />

To further investigate genes involved in mechanism of adaptation to hypoxia we screened a<br />

placenta retroviral cDNA library for genes capable to bypass a growth arrest induced by<br />

hypoxic conditions.<br />

The HCT116 colon carcinoma cell line were selected to screen the library based on their<br />

capacity to form colonies, to be genetically stable <strong>and</strong> sensitive to hypoxia.<br />

The HCT116 were infected by the cDNA retroviral library 2 days before applying the hypoxic<br />

treatment of 0% during 5 days, <strong>and</strong> then placed under normoxia (21%) for 10 days. The DNA<br />

from the cells was isolated <strong>and</strong> a PCR was performed to amplify the integrated cDNA. Two<br />

intense b<strong>and</strong>s of 1kb <strong>and</strong> 4kb were observed. The 1kb b<strong>and</strong> was isolated, subcloned into a<br />

PCR4 topo plasmid <strong>and</strong> sequenced. 2 clones were identified as placental lactogen cDNA. The<br />

cDNA of placental lactogen was cloned into a retroviral plasmid to validate the gene function<br />

<strong>and</strong> elucidate the mechanism. To confirm that placental lactogen increase the survival of the<br />

cells submitted to hypoxia reoxygenation we will analyze HCT116 expressing placental<br />

lactogen survival by colony formation assay. To evaluate the capacity of placental lactogen to<br />

give a growth advantage under hypoxia we will submit the cells to 0% during 2 <strong>and</strong> 3 days to<br />

achieve a growth curve.<br />

Based on the literature this gene plays a role in suppression of apoptosis pathway, thus it<br />

could be interesting to investigate whether it also plays a role in hypoxia-induced apotosis.<br />

Therefore we will assess the number of apoptotic cells submitted to hypoxic condition<br />

expressing or not placental lactogen.<br />

- 590 -


Notes<br />

- 591 -


Session XVI : Chemopreventive agents<br />

- 592 -


Session XVI : Chemopreventive agents Poster XVI, 1<br />

Anti-neoplastic <strong>and</strong> anti-clastogenic properties of curcumin<br />

Tzvetan Alaikov1, Spiro M. Konstantinov2,4, Tzvetomira Tzanova2, Kyril Dinev2,<br />

Margarita Topashka-Ancheva3, Martin R. Berger4<br />

1University Hospital St. Anna, Medical University of Sofia, Department of Internal<br />

Medicine, 1 D. Mollov Str, 1000 Sofia, Bulgaria; e-mail: Alaikov@abv.bg<br />

2Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str, 1000 Sofia, Bulgaria; email:<br />

Pgen@TU-Sofia.bg<br />

3Inst. of Zoology, Bulgarian Academy of Sciences, 1 Tzar Osvoboditel Blvd., 1000 Sofia,<br />

Bulgaria; e-mail: Topashka.mn@lycos.com<br />

4Unit of Toxicology <strong>and</strong> Chemotherapy; German Cancer Research Center, Im<br />

Neuenheimer Feld 280, 69120 Heidelberg, Germany; e-mail: M.Berger@DKFZ.de<br />

Curcumin is natural pigment of turmeric <strong>and</strong> is reported as signal transduction modulator <strong>and</strong><br />

inhibitor of transcription factors, e.g. NF-kB. In <strong>our</strong> study we found a concentrationdependent<br />

cytotoxic activity of curcumin in a panel of leukemic cell lines: SKW-3, CEM, U-<br />

937, HL-60, HL-60/Dox, K-562, LAMA-84 <strong>and</strong> AR-230. Additive to synergistic interactions<br />

were recorded for combinations with bendamustine <strong>and</strong> idarubicine in SKW-3 <strong>and</strong> LAMA-84<br />

cells. Noteworthy, a strong potentiation of the efficacy of bendamustine by concomitant<br />

curcumin application was found in multiple myeloma (MM) cells (RPMI-8226 <strong>and</strong> U-266).<br />

Moreover, curcumin increased the bendamustine cytotoxicity in primary cultures, isolated<br />

from the bone marrow of patients with MM. This interaction could be explained by NF-kB<br />

inhibition, because NF-kB is activated in many human cancers, especially leukaemia <strong>and</strong><br />

MM. Curcumin is characterized by low toxicity in vivo <strong>and</strong> was described to have<br />

chemoprotective activity. Therefore, the intracellular level of reduced glutathione (GSH) was<br />

measured <strong>and</strong> a concentration dependent increase of GSH level was recorded in AR-230 <strong>and</strong><br />

SKW-3 cells for concentrations ranging from 5 to 25 mcM. Experiments with mice showed<br />

significant protection against cisplatin induced chromosomal aberrations (clastogenic effect)<br />

<strong>and</strong> inhibition of mitoses in bone marrow cells. Curcumin alone caused significant reduction<br />

of the mitotic index, but ameliorated this parameter when combined with cisplatin. Our<br />

experimental data indicate that curcumin has pleyotropic effects on signal transduction by<br />

inhibiting transcription cascades <strong>and</strong> thus exerting direct antitumor activity <strong>and</strong> by enhancing<br />

the free radical scavenging that can explain the protective <strong>and</strong> anti-clastogenic activity.<br />

- 593 -


Session XVI : Chemopreventive agents Poster XVI, 2<br />

In vivo effects of the chemopreventive agent curcumin in the 5T33MM myeloma model.<br />

Jo Caers, Els Van Valckenborgh, Benjamin Van Camp, Karin V<strong>and</strong>erkerken.<br />

Laboratory of hematology & immunology. Vrije Universiteit Brussel, Brussels, Belgium<br />

Multiple Myeloma (MM) is a plasma cell malignancy characterised by the accumulation of<br />

monoclonal plasma cells in the bone marrow. The disease is usually preceded by an agedependent<br />

pre-malignant condition called monoclonal gammopathy of undetermined<br />

significance (MGUS). Since these MGUS patients are observed off therapy, we believe that<br />

these patients might benefit from a preventive treatment. In this study, we wanted to<br />

investigate the effects of such a chemopreventive agent, namely curcumin, on MM disease<br />

progression by using the murine 5T33MM myeloma model. In vitro, cell proliferation was<br />

assessed by 3H thymidine uptake. The IC50 for curcumin was about 7,5 µM. Compared to<br />

dexamethasone, a classic drug in MM therapy, curcumin was 50 times more effective. After<br />

overnight, curcumin increased the apoptosis rate of myeloma cells about twofold. In addition,<br />

an inihibitory effect could be seen on neo-vascularisation using a rat aortic ring assay with a<br />

reduction in total number, length <strong>and</strong> branching of newly created vessels by curcumin<br />

treatment. In vivo analysis of tumor burden was analyzed by injecting two groups of 10 mice<br />

with 5T33MM cells, one group of 10 naive mice was included as negative control. Intraperitoneal<br />

treatment with curcumin (50 mg/kg) was started two days before tumor<br />

inoculation. At week 3, when the vehicle controls showed signs of morbidity, the mice were<br />

sacrificed <strong>and</strong> tumor load was analyzed by determining bone marrow invasion, liver <strong>and</strong><br />

spleen mass <strong>and</strong> serum paraprotein concentrations. In treated mice, a 47 % reduction in serum<br />

paraprotein concentrations <strong>and</strong> a 30% reduction in the percentage of 5T33MM idiotype<br />

positive cells in the BM were monitored. Treatment further reduced the splenomegaly by 42%<br />

<strong>and</strong> the hepatomegaly with 30 %. In conclusion, these are the first in vivo data on the effects<br />

of curcumin on myeloma development. In human myeloma disease, curcumin was earlier<br />

described as inhibitor of NF-kappa b <strong>and</strong> the JAK-STAT pathway. The inhibitory effects of<br />

curcumin on these pathways will be further assessed on murine 5T33MM cells.<br />

- 594 -


Session XVI : Chemopreventive agents Poster XVI, 3<br />

A fully dissociated compound of plant origin for inflammatory gene repression.<br />

Karolien De Bosscher*, Wim V<strong>and</strong>en Berghe*, Ilse M.E. Beck*, Wim Van Molle#,<br />

Nathalie Hennuyer§, Janet Hapgood‡, Claude Libert#, Bart Staels§, Ann Louw‡ <strong>and</strong><br />

Guy Haegeman*.<br />

* Laboratory of Eukaryotic Gene Expression <strong>and</strong> Signal Transduction (LEGEST),<br />

Department of Molecular Biology, Ghent University, K. L. Ledeganckstraat 35, B-9000<br />

Gent, Belgium. E-mail: Guy.Haegeman@Ugent.be. # Department for Molecular<br />

Biomedical Research, Fl<strong>and</strong>ers Interuniversity Institute for Biotechnology (V.I.B.) <strong>and</strong><br />

Ghent University (UGent), 'Fiers-Schell-Van Montagu' building,Technologiepark 927,<br />

B-9052 Gent (Zwijnaarde), Belgium. § Département d’Arthérosclérose – U.545, Institut<br />

National de la Santé et de la Recherche Médicale, Institut Pasteur de Lille, 1 Rue<br />

Calmette BP245, 59019 Lille Cedex, France .‡ Department of Biochemistry, University<br />

of Stellenbosch, Matiel<strong>and</strong> 7602, Stellenbosch, Rep. of South Africa.<br />

The identification of selective glucocorticoid receptor (GR) modifiers, which separate<br />

transactivation <strong>and</strong> transrepression properties, represents an important goal for steroid<br />

pharmacology. While the gene-activating properties of GR are mainly associated with<br />

undesirable side effects, its negative interference with the activity of transcription factors,<br />

such as NF-kB, greatly contributes to its anti-inflammatory <strong>and</strong> immune-suppressive<br />

capacities. We found that Compound A (CpdA), a plant-derived phenyl aziridine precursor,<br />

although not belonging to the steroidal class of GR-binding lig<strong>and</strong>s, does mediate geneinhibitory<br />

effects by activating the glucocorticoid receptor (GR). We demonstrated that CpdA<br />

exerts an anti-inflammatory potential by down-modulating TNF-induced pro-inflammatory<br />

gene expression, but interestingly, does not at all enhance GRE-driven genes or induces GR<br />

binding to GRE-dependent genes in vivo. Furthermore, we have shown that the specific<br />

gene-repressive effect of CpdA depends on the presence of functional GR, displaying a<br />

differential phosphorylation status with CpdA as compared to DEX treatment. The antiinflammatory<br />

mechanism involves both a reduction of the in vivo DNA-binding activity of<br />

p65 as an interference with the transactivation potential of NF-kB. Finally, we present<br />

evidence that CpdA is as effective as DEX in counteracting acute inflammation in vivo, <strong>and</strong><br />

does not cause a hyperglycemic side effect. Taken together, this compound may be a lead<br />

compound of a novel class of anti-inflammatory agents with fully dissociated properties <strong>and</strong><br />

might thus hold great potential for therapeutic use.<br />

- 595 -


Session XVI : Chemopreventive agents Poster XVI, 4<br />

Inhibition of TNF-alpha induced NF-kappa B activation by kava (Piper methysticum)<br />

derivatives<br />

Folmer Florence1, Marc Diederich2, Marcel Jaspars1, Romain Blasius2, Jioji<br />

Tabudravu1<br />

1Marine Natural Products Laboratory, Department of Chemistry, University of<br />

Aberdeen, Aberdeen AB24 3UE, Scotl<strong>and</strong>. E-mail: f.folmer@abdn.ac.uk<br />

2Laboratoire de biologie moleculaire et cellulaire du cancer (LBMCC), Hopital du<br />

Kirchberg, 9, rue Edward Steichen, L-2540 Luxemb<strong>our</strong>g, Luxemb<strong>our</strong>g. Email :<br />

marc.diederich@lbmcc.lu<br />

The inducible transcription factor NF-kappa B plays a central role in the regulation of<br />

immune, inflammatory, <strong>and</strong> carcinogenic responses. While normal activation of NF-kappa B<br />

is required for cell survival <strong>and</strong> immunity, its deregulated expression is characteristic for<br />

inflammatory <strong>and</strong> infectious diseases <strong>and</strong> for cancer. NF-kappa B has hence become a major<br />

target in anti-inflammatory <strong>and</strong> anti-cancer drug discovery. In the present study, we<br />

investigated molecular mechanisms induced by lactones <strong>and</strong> chalcones isolated from Fijian<br />

kava (Piper methysticum) used in traditional medicine against urinary tract infections <strong>and</strong><br />

asthma. In order to underst<strong>and</strong> underlying regulatory mechanisms, inhibition of both NFkappa<br />

B-driven reporter gene expression <strong>and</strong> TNF-alpha induced NF-kappa B binding to a<br />

consensus response element were assayed. Total inhibition was achieved at the concentrations<br />

of 320 microM (flavokavain A), 175 microM (flavokavain B), <strong>and</strong> 870 microM (kavain <strong>and</strong><br />

dihydrokavain). Moreover, treatment with either kavain, flavokavain A, or flavokavain B led<br />

to the inhibition of both the degradation of the inhibitor of kappa B (IkappaB) <strong>and</strong> the<br />

subsequent translocation of the p50 <strong>and</strong> p65 subunits of NF-kappa B from the cytoplasm to<br />

the nucleus, as shown by western blot analysis. Additionally, kinase specific screening <strong>and</strong><br />

co-transfections with IKK (kinase of IkappaB) <strong>and</strong> IKK dominant negative expression genes<br />

showed that flavokavain A, but not kavain, nor flavokavain B, inhibits IKK. Flavokavain A<br />

was also shown to inhibit PRAK (p38 regulated/activated kinase), MAPKAP-K3 (MAPKactivated<br />

protein kinase 3), DYRK1A (dual-specificity tyrosine-phosphorylated <strong>and</strong> regulated<br />

kinase 1A), <strong>and</strong> Aurora B. Altogether, these results give a first insight into anti-inflammatory<br />

mechanisms triggered by traditionally used chemopreventive kava derivatives.<br />

- 596 -


Session XVI : Chemopreventive agents Poster XVI, 5<br />

Resveratrol induces apoptosis in chemo-resistant cancer cells via modulation of AMPK<br />

<strong>signaling</strong> pathway<br />

Jin-Taek Hwang 1 , Dong Wook Kwak 2 , Sun Kyo Lin 2 , Hye Min Kim 2 , Young Min Kim 2<br />

<strong>and</strong> Ock Jin Park<br />

Department of Food <strong>and</strong> Nutrition, Hannam University, 133 Ojeong-dong Daedeok-gu,<br />

Daejeon 306-791, Korea, E-mail: ojpark@hannam.ac.kr<br />

1 Department of Biochemistry <strong>and</strong> Molecular Biology, Medical Research Center for<br />

Bioreaction to Reactive Oxygen Species, Kyung Hee University College of Medicine,<br />

Seoul 130-791, Korea<br />

2 Department of Biological Sciences, Hannam University, Daejeon 306-791, Korea<br />

Resveratrol has been reported to possess therapeutic effects for various cancers including<br />

colon cancers. In the present study, the molecular basis of resveratrol with emphasis on its<br />

ability to control intracellular <strong>signaling</strong> cascades of AMP-activated kinase (AMPK)<br />

responsible for inducing apoptosis in cancer drug-resistant cells was investigated. Recently,<br />

the evolutionarily conserved serine/threonine kinase, AMPK, emerges as a possible target<br />

molecule of cancer control. We have investigated the effects of resveratrol on apoptosis in<br />

relation to AMPK in HT-29 cells made chemo-resistant by the treating a cancer drug<br />

etoposide. Resveratrol exhibited a variety of molecular events in etoposide-based<br />

combination therapy in HT-29 colon cancer cells including the AMPK activation, inhibition<br />

of cell growth, induction of apoptosis <strong>and</strong> ROS generation. The involvement of AMPK<br />

<strong>signaling</strong> cascade in resveratrol based cancer therapy was clearly shown by comparing the<br />

conditions of AMPK activated states <strong>and</strong> inactivated states. We have identified ROS as an<br />

upstream regulator of AMPK.<br />

- 597 -


Session XVI : Chemopreventive agents Poster XVI, 6<br />

Combined effects of gallic acid <strong>and</strong> resveratrol on gap junction intercellular<br />

communication <strong>and</strong> matrix metalloproteinase<br />

Jong Hoon Kim, Ki Won Lee, Hyo Jin Kim, <strong>and</strong> Hyong Joo Lee*<br />

Department of Food Biotechnology, School of Agricultural Biotechnology, <strong>and</strong> Center<br />

for<br />

Agricultural Biomaterials, Seoul National University, Seoul 151-742, Republic of Korea<br />

Dietary or pharmaceutical augmentation of endogenous antioxidant defense capacity has been<br />

considered one of the plausible ways to prevent reactive oxygen species (ROS)-mediated<br />

human diseases <strong>and</strong> aging, but antioxidant therapy has been at best equivocal. The present<br />

study investigated the possible combined effect of gallic acid <strong>and</strong> resveratrol, which is major<br />

antioxidants in red wine, on gap junction intercellular communication (GJIC) <strong>and</strong> on matrix<br />

metalloproteinase-2 (MMP-2) closely related to carcinogenesis. Resveratrol, but not gallic<br />

acid, prevented inhibition of GJIC <strong>and</strong> hyperphosphorylation of connexin 43 (Cx43) induced<br />

by hydrogen peroxide (H2O2) in normal rat liver epithelial cells. The sustained production of<br />

H2O2 by phenazine methosulfate (PMS), a chemical generating intracellular ROS, induced<br />

activation of MMP-2 in HT1080 human fibrosarcoma cells. Resveratrol prevented activation<br />

of pro-MMP-2 by PMS, but gallic acid did not exert these effects. Gallic acid rather induced<br />

inhibition of GJIC <strong>and</strong> activation of MMP-2 without H2O2 in a dose-dependent manner. Both<br />

GA <strong>and</strong> resveratrol generated H2O2 in the media without cells, <strong>and</strong> GA generated almost 5<br />

times more H2O2 than resveratrol in a dose- <strong>and</strong> time-dependant manner. The inhibition of<br />

GJIC by gallic acid was mediated by hyperphosphorylation of connexin 43 (Cx43) <strong>and</strong><br />

activation of extracellular signal-regulated kinase 1/2 (ERK1/2) through generation of ROS,<br />

while catalase <strong>and</strong> resveratrol significantly reversed gallic acid-induced inhibition of GJIC.<br />

The activation of MMP-2 <strong>and</strong> increase of motility by gallic acid was also partly abolished by<br />

catalase <strong>and</strong> resveratol. These results indicate potential prooxidant activity of gallic acid <strong>and</strong><br />

the combined effect of red wine phenolics on carcingenesis. Taken together, above findings<br />

suggest that antioxidants may act differently in ROS-mediated carcinogenesis depending on<br />

the conditions <strong>and</strong> structure.<br />

- 598 -


Session XVI : Chemopreventive agents Poster XVI, 7<br />

Jaceosidin, a pharmacologically active flavone derived from Artemisia plants, induces<br />

apoptosis in ras-transformed human breast epithelial (MCF10A-ras) cells<br />

Min-Jung Kim1, Do-Hee Kim1, Ki Won Lee1, Do-Young Yoon2, <strong>and</strong> Young-Joon Surh1<br />

1National Research Laboratory of Molecular Carcinogenisis & Chemoprevention,<br />

College of Pharmacy, Seoul National University, Seoul 151-742 <strong>and</strong> 2Department of<br />

Molecular Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea<br />

Extracts of Artemisia plants possess anti-inflammatory <strong>and</strong> anti-oxidative activities. Eupatilin<br />

(5,7-dihydroxy-3,4,6-tri-methoxy-flavone), a pharmacologically active flavone derived from<br />

Artemisia asiatica, was shown to inhibit phorbol ester-induced cyclooxygenase-2 induction<br />

<strong>and</strong> NF-kB activation in mouse skin (H.J. Seo et al., Int. J. Cancer, 100: 456-62, 2002), <strong>and</strong> to<br />

induce cell cycle arrest in ras-transformed human mammary epithelial (MCF10A-ras) cells<br />

(D.H. Kim et al., Biochem Pharmacol., 68: 1081-7, 2004). In the present study, we examined<br />

the ability of jaceosidin (4 ,5,7-trihydroxy-3 ,6-dimethoxyflavone) isolated from Artemisia<br />

argyi to induce apoptosis in MCF10A-ras cells. Jaceosidin inhibited the growth of MCF10Aras<br />

cells to a greater extent than eupatilin. Jaceosidin-induced apoptosis was mediated by<br />

intracellular ROS accumulation in MCF10A-ras cells, which was blocked by the antioxidant<br />

N-acetylcysteine (NAC). Jaceosidin has an additional hydroxyl moiety at the 4 -position<br />

which was replaced by the methoxy group in eupatilin. To better assess the pro-apoptotic<br />

effects of jaceosidin, we analyzed the treated cells by the flow cytometry. MCF10A-ras cells<br />

treated with jaceosidin (100 µM) exhibited the increased proportion of hypodiploid or<br />

apoptotic cells (48.72% as composed to 7.78% in control cells). Jaceosidin treatment also<br />

decreased the ratio of pro-apoptotic Bax to the anti-apoptotic Bcl-2 <strong>and</strong> induced the cleavage<br />

of caspase-3 <strong>and</strong> poly(ADP-ribose)polymerase (PARP). Moreover, jaceosidin elevated<br />

expression of p53 <strong>and</strong> p21, while inhibited the activation of ERK1/2 which is an important<br />

component of cell survival pathways. In conclusion, the pro-apoptotic activity of jaceosidin<br />

is associated with ROS accumulation <strong>and</strong> inhibition of ERK1/2 activation in MCF10A-ras<br />

cells.<br />

- 599 -


Session XVI : Chemopreventive agents Poster XVI, 8<br />

Epigallocatechin gallate inhibits phorbol ester-induced expression of COX-2 <strong>and</strong><br />

activation of NF-kappaB <strong>and</strong> CREB in mouse skin in vivo<br />

Joydeb Kumar Kundu <strong>and</strong> Young-Joon Surh<br />

National Research Laboratory of Molecular Carcinogenesis <strong>and</strong> Chemoprevention,<br />

College of Pharmacy, Seoul National University, Seoul 151-742, South Korea<br />

Despite accumulating evidence supporting the cancer chemopreventive properties of green<br />

tea, the underlying molecular mechanisms are not fully clarified. The representative green tea<br />

polyphenol epigallocatechin gallate (EGCG) has been reported to inhibit mouse skin tumor<br />

promotion. Since an inappropriate expression of cyclooxygenase-2 (COX-2) has been<br />

frequently implicated in the pathogenesis of cancer, we attempted to determine the effects of<br />

EGCG on 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced COX-2 expression in mouse<br />

skin. EGCG administered by gavage inhibited COX-2 expression in TPA-treated mouse skin.<br />

To further elucidate the underlying mechanisms of COX-2 inhibition, we examined the<br />

effects of EGCG on the activation of different transcription factors that regulate COX-2<br />

expression. While EGCG inhibited TPA-induced DNA binding of NF-kappaB <strong>and</strong> cyclic<br />

AMP response element binding protein (CREB), the compound failed to block the activation<br />

of AP-1 <strong>and</strong> CCAAT/enhancer binding protein (C/EBP) in TPA treated-mouse skin. EGCG<br />

diminished TPA-induced phosphorylation <strong>and</strong> degradation of IkappaBalpha <strong>and</strong> nuclear<br />

translocation of p65. Moreover, TPA-induced activation of extracellular signal-regulated<br />

protein kinase <strong>and</strong> p38 mitogen-activated protein (MAP) kinase was suppressed by EGCG.<br />

Pretreatment of mouse skin with pharmacological inhibitors of MAP kinases revealed that<br />

SB203580, but not U0126, inhibited TPA-induced CREB DNA binding. Taken together, <strong>our</strong><br />

study indicates that EGCG inhibits TPA-induced COX-2 expression <strong>and</strong> activation of NF-!B<br />

<strong>and</strong> CREB in mouse skin by downregulation of p38 MAP kinase.<br />

- 600 -


Session XVI : Chemopreventive agents Poster XVI, 9<br />

Wogonin, a plant flavone, potentiates etoposide-induced apoptosis in tumor cells<br />

Eibai Lee1, Riyo Enomoto1, Chie Suzuki1, Masataka Ohno1, Toshinori Ohashi1, Azusa<br />

Miyauchi1, Eriko Tanimoto1, Kaori Maeda1, Hiroyuki Hirano2, Toshio Yokoi2 <strong>and</strong><br />

Chiyoko Sugahara1<br />

1Department of Pharmacology <strong>and</strong> 2Department of Pharmaceutical Chemistry, Faculty<br />

of Pharmaceutical Sciences, Kobe Gakuin University, Ikawadani-cho, Nishi-ku, Kobe<br />

651-2180 Japan. E-mail : elee@pharm.kobegakuin.ac.jp<br />

Etoposide, a podophylotoxin anticancer agent, induces apoptotic cell death in normal <strong>and</strong><br />

cancer cells. Etoposide-induced apoptosis plays a role in not only anticancer effect but also<br />

adverse reaction such as myelosuppression. Since we have found that wogonin, a flavone<br />

found in Scutellaria baicalensis, suppresses thymocyte apoptosis induced by various<br />

compounds including etoposide, we examined the effect of this flavone on etoposide-induced<br />

apoptosis in cancer cells. Although wogonin itself hardly affected biochemical <strong>and</strong><br />

morphological features of apoptosis in leukemia cells such as Jurkat <strong>and</strong> HL-60 cells, this<br />

flavone significantly potentiated etoposide-induced apoptosis in these cells. Similarly,<br />

wogonin accelerated etoposide-induced cell death in lung cancer cells. Since wogonin had no<br />

effect on the action of other anticancer agents such as 5-FU <strong>and</strong> cisplatin, this flavone seems<br />

to accelerate only apoptotic cell death induced by etoposide in cancer cells. These results<br />

suggest that the modification of etoposide-induced apoptosis by wogonin may be available to<br />

reduce the adverse reaction of this agent.<br />

- 601 -


Session XVI : Chemopreventive agents Poster XVI, 10<br />

Resveratrol inhibits the growth of PA-1 ovarian cancer cells by down-regulating<br />

eEF1A2 expression<br />

Mee-Hyun Lee1, Bu-Young Choi2 <strong>and</strong> Young-Joon Surh1<br />

1National Research Laboratory of Molecular Carcinogenesis <strong>and</strong> Chemoprevention,<br />

College of Pharmacy, Seoul National University, Seoul 151-742 <strong>and</strong> 2C&C Research<br />

Labs, Hwasung City, Gyeonggi-do 445-970, South Korea<br />

The eukaryotic elongation factor 1A2 (eEF1A2), a subtype of eEF1A that is a key factor in<br />

the translational process of protein synthesis, promotes the transfer of animoacylated tRNAs<br />

to the A site of the ribosome. In human, eEF1A2 expression is restricted to the heart, brain,<br />

<strong>and</strong> skeletal muscle. Recently, eEF1A2 has been shown to be a potential oncogene that is<br />

over-expressed in ovarian cancer. Therefore, the regulation of inappropriate expression of<br />

eEF1A2 is considered as a rational strategy for ovarian cancer therapy. Resveratrol (3,4’,5trihydroxy<br />

stilbene), a phytoalexin present in grapes, has been reported to possess antioxidant,<br />

anti-inflammatory, <strong>and</strong> anti-carcinogenic activities. In the present study, we examined the<br />

anti-carcinogenic effect of resveratrol in PA-1 ovarian cancer cells considering eEF1A2 as a<br />

potential target. Resveratrol inhibited the growth of PA-1 cells. Treatment of PA-1 cells with<br />

insulin (10 µg/ml) or FBS (1%) resulted in the induction of eEF1A2. Pretreatment of PA-1<br />

cells with resveratrol suppressed insulin- or FBS-induced expression of eEF1A2. Previous<br />

studies have demonstrated that eEF1A2 confers anti-apoptotic effect by down-regulating<br />

caspase-3 activity. Resveratrol was found to induce caspase-3 activity <strong>and</strong> cause apoptosis in<br />

eEF1A2 over-expressing cells, suggesting that the induction of apoptosis by resveratrol in<br />

PA-1 cells is mediated, at least in part, via down-regulation of eEF1A2 expression. Taken<br />

together, these results suggest that oncogenic eEF1A2 is a potential target for ovarian cancer<br />

chemoprevention with resveratrol.<br />

- 602 -


Session XVI : Chemopreventive agents Poster XVI, 11<br />

Resveratrol Inhibits IL-1#-Induced Stimulation of Caspase 3 <strong>and</strong> Apoptosis in Human<br />

Articular Chondrocytes in vitro<br />

Shakibaei M1, John T2, Seifarth C1, Mobasheri A3<br />

1Musculoskeletal Research Group, Institute of Anatomy, Ludwig-Maximilian-<br />

University Munich, Pettenkoferstrasse 11, 80336 Munich; 2Charité Medicine University<br />

Berlin, Campus Benjamin Franklin, Department for Trauma Surgery, Berlin,<br />

Germany; 3Connective Tissue <strong>and</strong> Molecular Pathogenesis Research Groups, Faculty of<br />

Veterinary Science, University of Liverpool, United Kingdom.<br />

Resveratrol (trans-3,4´-trihydroxystilbene) is a polyphenolic phytoalexin that is present in<br />

various fruits, in the skin of red grapes, peanuts <strong>and</strong> root extracts. Recent studies have shown<br />

that resveratrol exhibits potent antioxidant properties <strong>and</strong> is able to exert anti-inflammatory<br />

<strong>and</strong> anti-catabolic properties in several cell types. The pro-inflammatory cytokine interleukin<br />

1 # (IL-1#) plays a pivotal role in the pathogenesis of osteoarthritis (OA) in humans <strong>and</strong><br />

animals. In this study we investigated whether resveratrol is able to block the proinflammatory<br />

effects of IL-1#, specifically the activation of caspase 3 <strong>and</strong> subsequent<br />

induction of apoptosis in human articular chondrocytes.<br />

Cultures of human articular chondrocytes were pre-stimulated with 10 ng/ml IL-1# for 1, 12<br />

<strong>and</strong> 24 h before being co-treated with IL-1# <strong>and</strong> 100 µM/ml resveratrol or 50µM the caspase<br />

inhibitor Z-DEVD-FMK for 1, 12 <strong>and</strong> 24 h respectively in vitro.<br />

Resveratrol significantly increased the IL-1#-induced inhibition the attenuated expression of<br />

cartilage specific collagen type II <strong>and</strong> signal transduction receptor integrin #1 in a time<br />

dependent manner. Incubation of chondrocytes with IL-1# resulted in activation of the<br />

cysteine protease caspase 3, PARP cleavage <strong>and</strong> apoptotic cell death. In this report we present<br />

evidence from ultrastructural studies that mitochondria play a major role in IL-1#-induced<br />

apoptosis. The treatment of human chondrocytes with IL-1# induced mitochondrial swelling<br />

in a time-dependent manner. These changes were observed as early as 1 h<strong>our</strong> after treatment<br />

of cells with IL-1#. These effects (activation of caspase 3, cleavage of PARP <strong>and</strong><br />

mitochondrial changes) were abolished through the co-treatment with resveratrol.<br />

Furthermore, co-treatment of the IL-1#–stimulated cells with the caspase inhibitor Z-DEVD-<br />

FMK blocked apoptosis as shown by electron microscopy <strong>and</strong> Western blotting, suggesting<br />

that this process is a caspase-dependent pathway.<br />

In summary, <strong>our</strong> results confirm that resveratrol is an effective in vitro inhibitor of caspase 3<br />

<strong>and</strong> apoptosis in human chondrocytes. These findings suggest that this dietary polyphenolic<br />

compound may have future applications in the nutraceutical based therapy of human <strong>and</strong><br />

animal OA.<br />

- 603 -


Session XVI : Chemopreventive agents Poster XVI, 12<br />

Possible link between NO concentrations <strong>and</strong> COX-2 expression in systems treated with<br />

soy-isoflavones<br />

Jang-In Shin 1 , Yoon-Kyung Lee 1 , Young Min Kim 2 <strong>and</strong> Ock Jin Park 1<br />

1 Department of Food <strong>and</strong> Nutrition, Hannam University, 133 Ojeong-dong Daedeok-gu,<br />

Daejeon 306-791, Korea, E-mail: ojpark@hannam.ac.kr<br />

2 Department of Biological Sciences, Hannam University, 133 Ojeong-dong Daedeok-gu,<br />

Daejeon 306-791, Korea,<br />

The production of nitric oxide (NO) emerges as an essential determinant in auto- <strong>and</strong><br />

paracrine <strong>signaling</strong>. NO is known to be generated under inflammatory conditions,<br />

carcinogenesis <strong>and</strong> circulatory shock. The large amount of NO produced in response to<br />

cytokines plays an important role in inflammatory conditions. Cyclooxygenase (COX), the<br />

central enzyme in prostanoid biosynthesis, is involved in the first step of prostanoid synthesis<br />

from arachidonic acid. The reported studies to evaluate the relationship between NO <strong>and</strong><br />

COX-2 have revealed both inhibitory <strong>and</strong> stimulatory effects of NO on COX-2 expression.<br />

Genistein, one of soy-isoflavones, is a polyphenolic flavonoid <strong>and</strong> a potent antioxidant <strong>and</strong><br />

anti-inflammatory agent. In the present study, the effect of soy-isoflavones on NO production<br />

<strong>and</strong> COX-2 gene expression was examined. NO production by soy-isoflvaones was greatly<br />

increased even though eNOS <strong>and</strong> iNOS expression were not different from non-treated<br />

control. The increment of NO was accompanied with the elevated expression of COX-2 <strong>and</strong><br />

the concentrations of PGE 2. The COX-2 stimulatory effect of soy-isoflavones appeared to be<br />

modulated by ERK1/2 <strong>and</strong> p38.<br />

- 604 -


Session XVI : Chemopreventive agents Poster XVI, 13<br />

Antiinflammatory, Antioxidative, <strong>and</strong> Chemopreventive Effects of Zerumbone, a<br />

Sesquiterpene Derived from Tropical Ginger<br />

Jun-Wan Shin1, Yoshimasa Nakamura2, Akira Murakami3, Hajime Ohigashi3 <strong>and</strong><br />

Young-Joon Surh1<br />

1College of Pharmacy, Seoul National University, Seoul 151-742, South Korea,<br />

2Graduate School of Natural Science <strong>and</strong> Technology, Okayama University, Okayama<br />

700-8530 & 3Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan<br />

Zerumbone (ZER), a sesquiterpene derived from tropical ginger Zingiber zerumbet Smith<br />

(Zingiberaceae), was found to suppress chemically-induced colon <strong>and</strong> skin carcinogenesis.<br />

Dextran sulfate sodium-induced colitis as well as production of proinflammatory cytokines<br />

(TNF-alpha, interleukin-1 alpha <strong>and</strong> interleukin-1 beta) <strong>and</strong> prostagl<strong>and</strong>in E2 was attenuated<br />

in mice fed ZER. In another experiment, topical application of ZER inhibited phorbol esterinduced<br />

COX-2 expression <strong>and</strong> tumpor promption in mouse skin. Treatment of mouse<br />

epidermal JB-6 cells with zerumbone induced the expression of heme oxygenase-1 (HO-1), a<br />

respresentative antioxidant enxyme that degrades heme to produce iron, carbon monoxide <strong>and</strong><br />

biliverdin. Zerumbone-induced expression of HO-1 in JB-6 cells appears to be mediated<br />

through antioxidant response elements (ARE)-driven activation of the redox-sensitive<br />

transcription factor Nrf2. Likewise, zerumbone upregulated HO-1 <strong>and</strong> several other<br />

antioxidant enzymes via ARE-Nrf2 siganling in the rat hepatic epithelial cell line. These<br />

findings suggest that chemopreventive effects of zerumbone is attributable to its<br />

antiinflammatory <strong>and</strong> antioxidant properties.<br />

- 605 -


Notes<br />

- 606 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease<br />

- 607 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 1<br />

Mitochondrial “Movement” <strong>and</strong> Lens Optics Following Oxidative Stress from UV-B<br />

Radiation: Cultured Bovine Lens <strong>and</strong> Cultured Human Retinal Pigment Epithelial Cells<br />

(ARPE-19) as Examples<br />

Vladimir Bantseev <strong>and</strong> Hyun-Yi Youn<br />

School of Optometry/Department of Biology, University of Waterloo, Waterloo, ON<br />

N2L 3G1, Canada. E-mail: vbantsee@uwaterloo.ca<br />

Mitochondria provide energy generated by oxidative phosphorylation <strong>and</strong> at the same time<br />

play a central role in apoptosis <strong>and</strong> aging. As a by-product of respiration, the electron<br />

transport chain is known to be the major intracellular site for the generation of reactive<br />

oxygen species (ROS). <strong>Expo</strong>sure to solar <strong>and</strong> occupational ultraviolet (UV) radiation, <strong>and</strong><br />

thus production of ROS <strong>and</strong> subsequent cell death, has been implicated in a large spectrum of<br />

skin <strong>and</strong> ocular pathologies, including cataract. Retinal pigment epithelial cell apoptosis<br />

generates photoreceptor dysfunction <strong>and</strong> ultimately visual impairment.<br />

The purpose of this study was to characterize in vitro changes following oxidative stress with<br />

UV-B radiation in 1) ocular lens optics <strong>and</strong> cellular function in terms of mitochondrial<br />

dynamics of bovine lens epithelium <strong>and</strong> superficial cortical fiber cells <strong>and</strong> 2) human retinal<br />

pigment epithelial (ARPE-19) cells. Cultured bovine lenses <strong>and</strong> confluent cultures of ARPE-<br />

19 cells were irradiated with broadb<strong>and</strong> UV-B radiations at energy levels of 0.5 <strong>and</strong> 1.0<br />

J/cm2. Lens optical function (spherical aberration) was monitored daily up to 14 days using<br />

an automated laser scanning system that was developed at the University of Waterloo. This<br />

system consists of a single collimated scanning helium-neon laser s<strong>our</strong>ce that projects a thin<br />

(0.05mm) laser beam onto a plain mirror mounted at 450 on a carriage assembly. This mirror<br />

reflects the laser beam directly up through the scanner table surface <strong>and</strong> through the lens<br />

under examination. A digital camera captures the actual position <strong>and</strong> slope of the laser beam<br />

at each step. When all steps have been made, the captured data for each step position is used<br />

to calculate the back vertex distance for each position <strong>and</strong> the difference in that measurement<br />

between beams. To investigate mitochondrial movement, the mitochondria-specific<br />

fluorescent dye Rhodamine 123 was used. Time series were acquired with a Zeiss 510<br />

(configuration Meta 18) confocal laser scanning microscope (Carl Zeiss Inc., Toronto<br />

Canada) equipped with an inverted Axiovert 200M microscope <strong>and</strong> 40-x water-immersion C-<br />

Apochromat objective (NA 1.2).<br />

The optical analysis showed energy level-dependent increases in back vertex distance<br />

variability (loss of sharp focus) from 0.39±0.04mm (control, n=11) to 1.63±0.33mm (1.0<br />

J/cm2, n=10) <strong>and</strong> 0.63±0.13mm (0.5 J/cm2, n=9). Confocal laser scanning microscopy<br />

analysis of both bovine lenses <strong>and</strong> ARPE-19 cells showed that following treatment at 0.5<br />

J/cm2 the mitochondria stopped moving immediately whereas at 1.0 J/cm2 not only did the<br />

mitochondria stop moving, but fragmentation <strong>and</strong> swelling was seen. Untreated control tissue<br />

exhibited up to 15µm/min of movement of the mitochondria. This could represent normal<br />

morphological change, presumably allowing energy transmission across the cell from regions<br />

of low to regions of high ATP dem<strong>and</strong>. Lack of mitochondrial movement, fragmentation <strong>and</strong><br />

swelling of mitochondria may represent early morphological changes following oxidative<br />

stress that may lead to activation of caspase-mediated apoptotic pathways.<br />

- 608 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 2<br />

2-Methoxyestradiol inhibits superoxide anion generation while enhaces superoxide<br />

dismutase activity in swine granulosa cells<br />

Giuseppina Basini, Sujen E. Santini, Francesca Grasselli<br />

Dipartimento di Produzioni Animali, Biotecnologie Veterinarie, Qualità e Sicurezza<br />

degli Alimenti, Sezione di Fisiologia Veterinaria, Università di Parma, 43100 Parma,<br />

Italy. E-mail: basini@unipr.it<br />

Angiogenesis <strong>and</strong> antiangiogenesis physiologically take place during ovarian follicular<br />

growth <strong>and</strong> regression. Antiangiogenesis has been the subject of intense interest because of its<br />

implications for restricting tumor growth. 2-Methoxyestradiol (2-ME) is an estradiol<br />

metabolite with antiangiogenic <strong>and</strong> antitumor activity. It is formed by granulosa cell <strong>and</strong> is<br />

present in the normal follicle at high concentrations. In this unique microenvironment, it may<br />

regulate selected cell types via autocrine <strong>and</strong>/or paracrine action. Identification of reactive<br />

oxygen specie role in the molecular pharmacology of 2-ME is an active area of research. To<br />

this purpose we evaluated the effect of 2-ME on both superoxide anion (O2-) production<br />

(WST-1 test, Roche, Mannheim, Germany) <strong>and</strong> on superoxide dismutase activity (SOD Assay<br />

kit Dojndo Molecular Technologies, Japan) in swine granulosa cells. Cells were obtained<br />

from follicles > 5 mm, seeded in 200 ul M199 in 96well plates <strong>and</strong> treated for 48 h with 1 uM<br />

2-ME. O2- assay was performed on 104 cells/well <strong>and</strong> 20 ul WST-1 were added during the<br />

last 4 h of incubation. SOD assay was performed on 2'105 cells/well; after treatment, cells<br />

were lysed <strong>and</strong> assayed for enzyme activity. In both assays the absorbance was read at 450<br />

nm against 620 nm. 2-ME inhibited (p


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 3<br />

Hypoxic responses in acute myeloid leukaemia (AML)<br />

Siv Lise Bedringaas, Kimberley Hatfield, Gry Sjøholt, Lars Helgel<strong>and</strong>#, Jørn Skavl<strong>and</strong>,<br />

Olav Dahl§, Øystein Bruserud, Bjørn Tore Gjertsen<br />

Hematology section, Institute of Medicine, University of Bergen <strong>and</strong> Department of<br />

Internal Medicine, Haukel<strong>and</strong> University Hospital, N-5021 Bergen. #Department of<br />

Pathology, The Gade Institute, University of Bergen, Haukel<strong>and</strong> University Hospital.<br />

§Section of Oncology, Institute of Medicine <strong>and</strong> Department of Oncology, Haukel<strong>and</strong><br />

University Hospital, Bergen, Norway.<br />

Acute myeloid leukaemia (AML) is a haematological malignancy characterized with block in<br />

differentiation <strong>and</strong> numerous, but also recurrent, chromosomal translocations. The most<br />

common recognized mutation (approx. 30%) is in the class III receptor FMS-related tyrosine<br />

kinase 3 (Flt3/Stk1/Flk2). Even though AML blasts are in a normoxic environment in the<br />

peripheral blood, <strong>our</strong> hypotheses is that excessive growth of malignant blasts in the bone<br />

marrow create a hypoxic environment. Tum<strong>our</strong> hypoxia is known to promote chemo<br />

resistance <strong>and</strong> aggressive disease.<br />

In this study we have investigated the hypoxic effect in cryopreserved primary AML cells <strong>and</strong><br />

AML cell lines (HL-60, NB4) by Western blot, ELISA, flow cytometry, two-dimensional gel<br />

electrophoresis (2DE) <strong>and</strong> mass spectrometric identification of proteins, as well as the twodimensional<br />

differential gel electrophoresis (DIGE). In particular the protein levels of<br />

Hypoxia Inducible Factor-1" (HIF-1"), vascular epithelial growth factor (VEGF),<br />

osteopontin (OPN), cyclin-dependent kinase inhibitor 1A (CDKN1A/p21/CIP1), <strong>and</strong> Flt3<br />

were also investigated in the patient samples, <strong>and</strong> HDM2 in cell lines were examined.<br />

We investigated AML cells in hypoxia (1% O2) <strong>and</strong> Co2+ that mimics hypoxic conditions.<br />

Preliminary results show that 30% of patients (10/30) <strong>and</strong> both cell lines responded to<br />

hypoxia with increased protein level of HIF-1". More than 50% of the patients responded<br />

with VEGF production (17/30), but a significant fraction of the VEGF responders were not<br />

HIF-1" responders (7/17). The VEGF non-responders had non-detectable levels of VEGF<br />

before <strong>and</strong> during hypoxia. Approximately 50% of the patients responded with OPN increase.<br />

CDKN1A/p21 <strong>and</strong> Flt3 showed no response to hypoxia in any patients. The level of HDM2<br />

was reduced in both cell lines tested during hypoxia.<br />

Preliminary results suggest that DIGE is more sensitive than conventional 2DE in detection of<br />

protein modulation induced by hypoxia (1% O2) <strong>and</strong> Co2+. Ongoing experiments are aimed<br />

to map the protein networks that differ between hypoxic HIF-1" responsive <strong>and</strong> nonresponsive<br />

AML.<br />

- 610 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 4<br />

Strongly reduced density of parvalbumin-immunoreactive projection neurons in the<br />

mammillary bodies in schizophrenia: further evidence for limbic neuropathology<br />

Hans-Gert Bernstein1, Stephanie Krause2, Dieter Krell1, Henrik Dobrowolny1, Renate<br />

Stauch1, Karin Ranft1, Peter Danos2 <strong>and</strong> Bernhard Bogerts1<br />

1Department of Psychiatry, University of Magdeburg, Leipziger Str. 44, D-39120<br />

Magdeburg <strong>and</strong> 2Department of Psychiatry, 2University of Giessen, Germany; E-mail:<br />

Hans-Gert.Bernstein@medizin.uni-magdeburg.de<br />

The mammillary bodies are important relay nuclei within limbic <strong>and</strong> extralimbic connections.<br />

They receive information from the hippocampus <strong>and</strong> are reciprocally connected to the anterior<br />

thalamic nuclei, the septum <strong>and</strong> the midbrain. The mammillary bodies are known to play roles<br />

in memory formation <strong>and</strong> are affected in alcoholism <strong>and</strong> vitamin B1 deficiency. Their<br />

strategic position linking temporolimbic to frontothalamic brains structures make the<br />

mammillary bodies c<strong>and</strong>idates for alterations in schizophrenia. We studied 15 postmortem<br />

brains of schizoprenics <strong>and</strong> 15 matched control brains from the Magdeburg Brain Collection.<br />

A demographic <strong>and</strong> clinical history was available for all cases. Brains were fixed in formalin,<br />

embedded in paraffin <strong>and</strong> cut into 20 µm thick coronal sections. Brain sections were stained<br />

either for Heidenhain-Woelcke, calretinin or parvalbumin. We determined the volumes of the<br />

mammillary bodies <strong>and</strong> performed cell countings using stereological principles <strong>and</strong> a<br />

computerized image analysis system. Our volumetric calculations revealed that the volumes<br />

of mammillary bodies do not differ between schizophrenics <strong>and</strong> controls. However, in<br />

schizophrenia the neuronal densities were significantly reduced on both sides (on left side by<br />

35.9%, on right side by 22 %). No changes were seen in the density of calretinin-containing<br />

neurons, whereas the number of parvalbumin-immunoreactive mammillary neurons was<br />

profoundly reduced (by more than 50%). This cell loss (as a result of developmental<br />

malformation <strong>and</strong>/or neurodegeneration) points to a prominent involvement of the<br />

mammillary bodies in the pathomorphology of schizophrenia. Parvalbumin-immunoreactive,<br />

GABAergic interneurons have repeatedly been reported to be diminished in several brain<br />

regions in schizophrenia. However, in the mammillary bodies parvalbumin labels a<br />

subpopulation of glutamate/aspartate-containing neurons projecting to the anterior thalamus.<br />

Thus, <strong>our</strong> data provide new evidence for impaired limbic circuits in schizophrenia.<br />

Supported by NBL-3 <strong>and</strong> Stanley-Foundation.<br />

- 611 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 5<br />

Sustained gene transfer with photochemical internalisation in breast cancer cells.<br />

Sanae Bouali, Jihane Mirouah, Jean L. Merlin<br />

Laboratoire de recherche en oncologie, EA 3452, Centre Alexis Vautrin, Avenue de<br />

B<strong>our</strong>gogne, 54511 V<strong>and</strong>oeuvre Les Nancy cedex, France. E-MAIL :<br />

s.bouali@nancy.fnclcc.fr<br />

Although the vectors used to deliver therapeutic genes to targeted cells have traditionally been<br />

modified viruses, non viral synthetic vectors are also receiving much attention as an<br />

alternative method. Most non viral gene therapy vectors deliver transgene into cells through<br />

the endocytic pathway. Lack of escape from endocytic vesicles in many cases constitutes a<br />

barrier for delivery of the transgene. A new biotechnology method was developed named<br />

photochemical internalisation (PCI). PCI has been described as an innovative procedure for<br />

releasing macromolecules from endocytic vesicles to the cytosol of target cells. This<br />

technology is based on light induced delivery of macromolecules such us DNA, proteins, <strong>and</strong><br />

other therapeutic molecules in a cytosol.<br />

METHODS: Human breast cancer cells were preincubated with the photosensitiser mesotetraphenylporphine<br />

(TPPS2a) followed by treatment with plasmid encoding enhanced Green<br />

Fluorescent Protein complexed with Polyethylenimine or Eudragit RL or transfectin <strong>and</strong> light<br />

exposure. The expression of the GFP gene was scored by flow cytometry.<br />

RESULTS: The photochemical using light doses improve the efficiency of transfection<br />

mediated by polyethylenimine , but not by Eudragit RL <strong>and</strong> transfectin. This study was<br />

designed to optimise the PCI of Polyethylenimine, Eudragit RL <strong>and</strong> tranfectin-DNA<br />

complexes gene transfer in vitro.<br />

CONCLUSION: In the present study, results confirm the potency of combining the PCI with<br />

Polyethylenimine mediated gene transfer.<br />

- 612 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 6<br />

GENETIC AND EPIGENETIC CONTROL OF HEMATOPOIETIC STEM CELLS<br />

PROLIFERATION.<br />

Leonid V. Bystrykh, Alice Gerrits, Leonie M. Kamminga, Ronald van Os, Albertina<br />

Ausema, Ellen Weersing, Bert Dontje, Gerald de Haan.<br />

Department of Cell Biology, Section Stem Cell biology. University Medical Center<br />

Groningen, University of Groningen, 9713 AV Ant.Deusinglaan 1, The Netherl<strong>and</strong>s.<br />

Previously, we identified two major loci controlling HSC behavi<strong>our</strong>, namely Scp2 at ch11<br />

(55-85 Mb) affecting proliferation, <strong>and</strong> a locus at ch18 (12-32 Mb) affecting HSC pool size.<br />

Genome-wide microarray-based profiling of gene expressions using mRNA isolated from<br />

murine hematopoietic stem cells was deposited in the www.genenetwork.org database [1].<br />

Having used a set of 30 recombinant BXD inbred lines we were able to map controlling loci<br />

(QTLs) for thous<strong>and</strong>s of transcripts. From a list of 12422 probe sets on the Affymetrix Murine<br />

Genome U74Av2 GeneChip we identified 3615 transcripts with heritable variation in<br />

expression levels. Of these, 399 were highly significantly cis acting (cutoff -10logP 2.93)<br />

<strong>and</strong> 3216 trans-acting transcripts. Our studies with BXDs suggest that the HSC controlling<br />

locus on ch18 is likely to be regulated by chromatin remodeling genes.<br />

It is known that serial transplantation of mouse HSC causes gradual deterioration of stem cell<br />

function, reminiscent with senescence phenotype. Our microarray analysis of serially<br />

transplanted HSC showed 300 genes to be either up- or downregulated. Comparison of these<br />

data with recently published microarray data, allowed us to identify essential components of<br />

a genomic network operating in HSCs. These include components of PcG <strong>and</strong> TrxG<br />

epigenetic complexes, which appear to cross talk with Shh, Wnt ant Notch pathways.<br />

Collectively, these genes are largely responsible for maintenance <strong>and</strong> proliferation of HSC.<br />

Currently we are verifying <strong>our</strong> network by selective retroviral overexpression of some<br />

essential genes (Ezh2, Msi1h).<br />

Literature<br />

[1] Bystrykh L, Weersing E, Dontje B, Sutton S, Pletcher MT, Wiltshire T, Su AI, Vellenga<br />

E, Wang J, Manly KF, Lu L, Chesler EJ, Alberts R, Jansen RC, Williams RW, Cooke MP, de<br />

Haan G. Uncovering regulatory pathways that affect hematopoietic stem cell function using<br />

'genetical genomics'. Nat Genet. 2005;37(3):225-32.<br />

- 613 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 7<br />

Expression <strong>and</strong> role of phosphatidylcholine-specific phospholipase C (PC-PLC) in<br />

human NK cells <strong>and</strong> its relationship with CD16 receptor.<br />

Serena Cecchetti, Francesca Spadaro, Franca Podo <strong>and</strong> Carlo Ramoni.<br />

Department of Cell Biology <strong>and</strong> Neurosciences, Istituto Superiore di Sanità, Viale<br />

Regina Elena 299, 00161, Rome Italy.<br />

Phosphatidylcholine hydrolysis by a specific phospholipase C (PC-PLC) is a widespread<br />

response elicited by most growth factors, cytokines, neurotrasmitters, hormones <strong>and</strong> other<br />

extra-cellular signals. Our previous studies showed that PC-PLC is involved in the functional<br />

role of NK-mediated cell killing <strong>and</strong> in the lytic granules exocytosis process. PC-PLC was<br />

detected both on specific cytoplasmic compartments <strong>and</strong> on the outer membrane surface, in<br />

which the enzyme translocated upon cell activation <strong>and</strong> lytic ability maturation.<br />

Interestingly, PC-PLC expression on NK membrane surface was directly proportional to that<br />

of CD16, the low-affinity receptor for the Fc fragment of IgG, suggesting a possible<br />

correlation between the enzyme <strong>and</strong> NK cell maturation.<br />

In the present work we analyzed the trafficking from the plasma membrane to cytoplasmic<br />

regions of CD16 receptor <strong>and</strong> PC-PLC enzyme. Both proteins were internalized upon<br />

antibody engagement, degraded <strong>and</strong> newly synthetized, moreover, they needed an integral <strong>and</strong><br />

functional actin cytoskeleton during their trafficking. Pre-incubation of NK cells with the PC-<br />

PLC inhibitor tricyclodecan-9-yl-potassium xanthate (D609) determined a dramatic decrease<br />

of PC-PLC fraction expressed on NK plasma membrane. Surprisingly, D609 also downmodulated<br />

the membrane expression of CD16 receptor. Among phenotype markers of<br />

peripheral blood lymphocytes analyzed after D609 treatment, only CD16 was deeply downmodulated,<br />

thus suggesting a possible specific role of PC-PLC enzyme in regulating CD16<br />

receptor expression. Moreover, we demonstrated that PC-PLC was activated, within 5<br />

minutes, upon CD16 stimulation, thus suggesting an involvement of the enzyme in CD16<br />

signal transduction. We also evidenced that in antibody-dependent cellular cytotoxicity<br />

process PC-PLC played a double functional role both in regulating CD16 membrane<br />

expression <strong>and</strong> in transducing CD16 receptor signals, since D609 totally abolished this<br />

important lytic mechanism.<br />

- 614 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 8<br />

Different ways to arrest proliferation of cancer cells by dsRNA<br />

Elena L.Chernolovsksya, Tatyana O. Kabilova, Al`bina V. Vladimirova, Valentin V.<br />

Vlassov<br />

Institute of Chemical Biology <strong>and</strong> Fundamental Medicine SB RAS, Lavrentiav ave., 8,<br />

Novosibirsk 630090, Russia, E-mail: elena_ch@niboch.nsc.ru<br />

Deregulation of genes encoding components of <strong>signaling</strong> pathways are considered as a key<br />

factor in the development of different types of tumors in humans. We investigated inhibition<br />

of MYC genes by dsRNAs in KB-3-1, SC-N-MC <strong>and</strong> IMR-32 cells. Sequence-specific<br />

siRNA (si3ex) targeted to the third exon of c-myc gene was found to decrease the level of cmyc,<br />

but not N-myc mRNA. si3ex decreased the rate or even arrested the proliferation of cmyc<br />

overexpressing cell lines KB-3-1 <strong>and</strong> SC-N-MC, but did not affect the proliferation of<br />

IMR-32 (which overexpress N-myc). si2ex homological to the conservative region of the<br />

second exon of both c- <strong>and</strong> N-myc was able to downregulate both genes <strong>and</strong> to reduce<br />

proliferation of KB-3-1, SC-N-MC <strong>and</strong> IMR-32 cells. PKR or/<strong>and</strong> OAS1 mRNA levels were<br />

not effected. Long double str<strong>and</strong>ed RNAs <strong>and</strong> some short dsRNA can stimulate innate<br />

cytokine responses in mammals, which can induce proliferation blockage, differentiation or<br />

apoptosis in cancer cells. Long double str<strong>and</strong>ed RNAs: dsMyc homologous to the 3 exon of cmyc<br />

gene, dsGFP homologous to mRNA of EGFP gene <strong>and</strong> GU-rich siRNA (siI),<br />

homologous to the intron sequence of human MDR1 gene were found to inhibit proliferation<br />

<strong>and</strong> to decrease the mRNA level of interferon-sensitive genes: c-myc <strong>and</strong> beta-actin when<br />

delivered into cancer human cells. The level of downregulation was march higher than that of<br />

synthetic interferon inducer poly(I:C) <strong>and</strong> is likely depends on the properties of dsRNA: the<br />

thermodynamic stability, nuclease resistance <strong>and</strong> affinity to dsRNA-binding proteins.<br />

Antiproliferation activity of long dsRNA, displayed in the absence of transfection reagent,<br />

was cell line specific <strong>and</strong> correlated with the ability of dsRNA to inhibit the expression of cmyc<br />

<strong>and</strong> beta-actin genes. The elevation of PKR or/<strong>and</strong> OAS1 mRNA levels was detected in<br />

all cells affected by long dsRNA <strong>and</strong> does not correlates with proliferation blockage. The data<br />

suggest, that double str<strong>and</strong>ed RNAs can serve as antiproliferative agents, acting sequencespecifically<br />

or activating innate immunity response.<br />

This work was supported by RFBR (grant No. 06-04-49480- ), RAS programs “Molecular<br />

<strong>and</strong> Cellular Biology” <strong>and</strong> “Fundamental science for medicine”, Interdisciplinary grant from<br />

SB RAS, FCSTP RI-012/001/254.<br />

- 615 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 9<br />

Progressive Factors in Uterine Cervical Cancer associated with High-risk Human<br />

Papillomavirus<br />

Yoon Pyo Choi*, Suki Kang, Owen Ding*, Nam Hoon Cho*<br />

Dept of Pathology, Yonsei University College of Medicine<br />

Brain Korea 21 Project for Medical Science, Yonsei University*<br />

Human papillomavirus (HPV) infections play a crucial role in the progress of cervical cancer.<br />

In the present study, we aimed to establish the proteomic profiles <strong>and</strong> characterization of the<br />

tumor related proteins by using 2-DE <strong>and</strong> MALDI-TOF MS in 6 cervical swab samples,<br />

which were obtained from the patients with infection of HPV 16 <strong>and</strong> 18. We compared<br />

nuclear protein <strong>and</strong> cytoplasmic protein, separately, by using the subcellular fraction in the<br />

proteomic analysis. On comparison of differential protein spots between cervical cancer<br />

tissues with HPV 16 or HPV 18 <strong>and</strong> HaCaT cell lines as the control, we confirmed that<br />

proteins, such as PCNA, 14-3-3 sigma, CDC25A, Mdm2, Mdm4, minimal chromosomal<br />

maintenance (MCM) 3, MCM4 <strong>and</strong> MCM8 according to surrogate endpoint biomarkers<br />

(SEB), were upregulated in nuclear fractions of cervical cancer tissues <strong>and</strong> that also CDC25B<br />

was downregulated in nuclear fractions, whereas being upregulated in cytoplasmic fractions<br />

of the cervical cancer tissues. This subcellular shuttling protein, CDC25B, indicates that it is<br />

probably associated with G2 arrest in the carcinogenesis of cervical cancer by HPV infection.<br />

And then, for validation of proteomic analysis, on application of c<strong>and</strong>idates to have been<br />

screened in the proteomic analysis on 201 archival samples of cervical neoplasia associated<br />

with various HPV types infection (75 CIN 1, 28 CIN II, 82 CIN III <strong>and</strong> 16 cancer) by<br />

immunohistochemistry, PCNA, MDM2, CDC25A, MCM4 <strong>and</strong> MCM7, were found to be<br />

significantly correlated to the disease progression. MCM8 also revealed one of the most<br />

specific <strong>and</strong> correlative markers to the disease progression <strong>and</strong> even to the poor outcome of<br />

the cervical cancer. In conclusion, we have demonstrated that HPV can cause the unstable<br />

regulation of a lot of carcinogenesis-related proteins, such as proliferation factors, cell cycle<br />

regulatory factors, cell <strong>signaling</strong> factors, oncogenes etc. without the significant difference of<br />

any specific molecules being found according to the HPV genotypes, in the ongoing process<br />

of cervical cancer. MCM8 appears to be another novel marker in addition to well-known<br />

biomarkers to predict poor outcome in uterine cervical cancer. Until a marker of such<br />

versatility is found, knowing everything possible about the markers that we have is crucial,<br />

<strong>and</strong> this study has shown some new findings about these known markers.<br />

- 616 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 10<br />

Androgens induce while PPAR-lig<strong>and</strong>s inhibit prostate cancer cells migration.<br />

Joanna Duli"ska-Litewka, Dorota Gil <strong>and</strong> Piotr Laidler<br />

Department of Medical Biochemistry, Jagiellonian University Medical College, ul.<br />

Kopernika 7, 31-034 Kraków, POLAND; e-mail: mblitewk@kinga.cyf-kr.edu.pl,<br />

tel/fax:+48(12)4223272<br />

Purpose: Matrix metalloproteinases (MMPs) are a family of proteolytic enzymes which<br />

degrade the extracellular matrix or components of the basement membrane. MMP have been<br />

found to be associated with prostate cancer metastasis. Prostate growth, survival <strong>and</strong><br />

pathology is generally dependent on <strong>and</strong>rogen stimulation mediated by the <strong>and</strong>rogen receptor<br />

(AR). Several lig<strong>and</strong>s of peroxisome proliferators activated receptors (PPARs) were shown to<br />

decrease the proliferation <strong>and</strong> enhance apoptosis in various cancers, also in prostate cancer<br />

thus somehow opposing <strong>and</strong>rogen effects. Therefore we sought to determine if <strong>and</strong> how<br />

<strong>and</strong>rogens <strong>and</strong> PPAR-lig<strong>and</strong>s influence MMPs expression <strong>and</strong> prostate cancer cells migration.<br />

Methods: Studies were carried out on human prostate cancer cell lines: LNCaP, Du-145 <strong>and</strong><br />

PC-3. We studied the effect of testosterone <strong>and</strong> PPAR$ lig<strong>and</strong>s (ciglitazone, arachidonic acid)<br />

on cells’ migration (Boyden’s chamber), expression <strong>and</strong> activity of MMPs (zymography) <strong>and</strong><br />

selected proteins engaged in cell <strong>signaling</strong> (RT-PCR, Western blot). The antagonist of PPAR$<br />

– GW9662 <strong>and</strong> inhibitor of 5"-reductase – finasteride were also included.<br />

Results: Treatment of LNCaP cells with testosterone resulted in an increase of pro-MMP-2<br />

expression <strong>and</strong> MMP-2 activity <strong>and</strong> led to the increased proliferation <strong>and</strong> decreased<br />

expression of PPAR$. Finasteride clearly suppressed this effects. Selective PPAR$ lig<strong>and</strong>s<br />

significantly decreased proliferation, migration <strong>and</strong> the level of active MMPs. RT-PCR <strong>and</strong><br />

Western blot analyses basically indicated the opposite effects of testosterone <strong>and</strong> ciglitazone<br />

on expression of AR, PPAR$, c-myc <strong>and</strong> Akt. Inhibition of the expression of phosphorylated<br />

Akt by ciglitazone did not affect total Akt levels, indicating the functional effect of PPAR$<br />

lig<strong>and</strong> on Akt signal transduction.<br />

Conclusions: The results suggest that the expression <strong>and</strong> regulation of the activity of AR <strong>and</strong><br />

PPAR$ significantly <strong>and</strong> clearly in opposite manner affect prostate cancer cell proliferation,<br />

migration <strong>and</strong> MMP expression. It seems that important <strong>signaling</strong> step in <strong>and</strong>rogen-induced<br />

migration might be inhibited by PPAR$ lig<strong>and</strong>s. Moreover, Akt signal transduction seems to<br />

be linked to AR <strong>and</strong> PPAR lig<strong>and</strong>s modulation of these basic cell properties through as yet<br />

not fully recognized mechanism. This results may provide a novel target of PPAR$ activators<br />

<strong>and</strong> indicate their potential as useful therapeutic agents in the treatment of prostate cancer.<br />

This work was financially supported by the KBN: Jagiellonian University Medical College –<br />

grant W,/253/P/L <strong>and</strong> the State Committee for Scientific Research - grant No 6 PO5A 074 21,<br />

Pol<strong>and</strong><br />

- 617 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 11<br />

The orphan nuclear receptor RORalpha activity is regulated by post-translational<br />

modifications : the role of phorbol esters <strong>and</strong> protein kinase C.<br />

E. DUPLUS, V. SOUBEYRE, Y. LEMAIGRE-DUBREUIL, B. BRUGG<br />

Nuclear receptor superfamily includes lig<strong>and</strong>-dependant transcription factors that control<br />

numerous genes involved in differentiation, proliferation <strong>and</strong> cell homeostasis. Some of these<br />

receptors have also been described to be regulated by post-translational modifications like<br />

phosphorylations. For a long time, Retinoid-related orphan receptor alpha (RORalpha) has<br />

been considered as a constitutively active transcription factor with no known lig<strong>and</strong> nor<br />

effector. However, recent new data suggest that this nuclear receptor is controled by lig<strong>and</strong><br />

binding. It is expressed in a wide range of organs, particularly in the cerebellum where it<br />

seems to be implicated in neuronal survival <strong>and</strong> differentiation. No regulatory pathways of<br />

these biological functions have been described to control RORalpha activity so far. The aim<br />

of <strong>our</strong> work is to identify different <strong>signaling</strong> protein kinases, usually described to regulate<br />

nuclear receptor, to directly modulate RORalpha transcriptionnal activity. Among these<br />

proteins, some of protein kinase C isoforms (PKCs) are of great interest considering their<br />

involvement, as RORalpha, in neurone survival <strong>and</strong> differentiation. In this study, we show<br />

that PKC activator, Phorbol 12-Myristate 13-Acetate (PMA) inhibits RORalpha activity in<br />

COS-7 cell line. This effect is totally prevented by PKC inhibitor Bisindolylmaleimide 1<br />

suggesting that PKCs are involved in PMA effect. Western blot experiments in COS-7 cells<br />

demonstrated that PKC are also implicated in post-translational modifications of RORalpha<br />

leading to an electrophoretic migration shift. In order to identify theses changes, we carried<br />

out metabolic labeling experiments which suggested that 1) RORalpha is a phosphoprotein, 2)<br />

Its phosphorylation state is enhanced by PKC activation in COS-7 cells<br />

Thus, these data constitute the first evidence of PKC-dependent RORalpha phosphorylation<br />

<strong>and</strong> inhibition of its transcriptional activity. They open important therapeutic prospects,<br />

especially for neurodegenerative diseases.<br />

- 618 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 12<br />

Are key factors of signalling intracellular pathways involved in the regulation of<br />

neuromuscular changes after disuse conditions ?<br />

Erwan Dupont, Marie-Hélène Canu, Maurice Falempin, Yvonne Mounier <strong>and</strong> Laurence<br />

Stevens<br />

Laboratoire de Plasticité Neuromusculaire, EA 1032, IFR 118, Unité de Neurosciences et<br />

Physiologie Adaptatives, Bât SN4, Université des Sciences et Technologies de Lille,<br />

59655 Villeneuve d’Ascq cedex, France<br />

The performance of the neuromuscular system is highly dependent on activity. A situation of<br />

disuse induced by hypodynamia-hypokinesia (HH) in rats induces profound changes in<br />

skeletal muscles like phenotype transformations, <strong>and</strong> a marked atrophy resulting from a loss<br />

in mass <strong>and</strong> protein content. HH is also a model of hindpaw sensory deprivation since in these<br />

conditions, the cutaneous receptors located on the foot sole are deactivated. HH produces a<br />

cortical reorganisation of the somatosensory cortex (SmI), indicating the existence of a<br />

neuronal plasticity.<br />

The factors responsible for these impairments are still unknown <strong>and</strong> the cellular <strong>and</strong><br />

molecular processes sustaining this “plasticity” are not well described. The major aims here<br />

were to better underst<strong>and</strong> the mechanisms i) that characterized muscular plasticity,<br />

phenotypical transitions <strong>and</strong> atrophy, <strong>and</strong> ii) involved in cortical plasticity (sensory<br />

conduction <strong>and</strong>/or cortical excitability changes, regulation of neurotransmitter <strong>and</strong><br />

neurotrophin expression). Among multiple intracellular signalling pathways, one major is<br />

implied in controlling more specifically muscle <strong>and</strong>/or nervous plasticity : the MAPKinase<br />

(Mitogen-Activated Protein Kinase) cascade. We induced modulations of transformations<br />

using different periods of disuse (from 7 to 28 days of HH) in rats. Key markers of<br />

MAPKinase cascade were followed : ERK, JNK <strong>and</strong> p38. Their protein expressions were<br />

evaluated by western blots (using specific antibodies) performed on all the animal groups, as<br />

well at the muscular as at the cortical levels. For the muscles, the expression of all MAPK<br />

markers was increased in a time-dependent manner of disuse, excepted for 14 days of HH, the<br />

increase being lower than for 7 <strong>and</strong> 28 days. At the cortical level, the increase was less<br />

pronounced <strong>and</strong> not disuse-dependent.<br />

Taken together, these results may help to develop strategies that aim at attenuating the effects<br />

of pathologies, like long-term immobilizations, on the neuromuscular system.<br />

- 619 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 13<br />

#1 - INTEGRIN REGULATION OF MECHANOTRANSDUCTION IN<br />

OSTEOARTHRITIC HUMAN ARTICULAR CHONDROCYTES.<br />

Kerry J. Elliot*, Tina T. Chowdhury** <strong>and</strong> Donald M. Salter*.<br />

* The Queen’s Medical Research Institute, The University of Edinburgh, 47 Little<br />

France Crescent, Edinburgh, UK, EH16 4TJ. E-mail: kelliot@staffmail.ed.ac.uk<br />

** Queen Mary, University of London, Department of Engineering, Mile End Road,<br />

London, UK, E1 4NS.<br />

Introduction: Normal <strong>and</strong> osteoarthritic (OA) human articular chondrocytes (HAC) respond<br />

to 0.33 Hz cyclical mechanical stimulation (MS) via a "5#1 integrin mediated<br />

mechanotransduction pathway although downstream signalling pathways are different. In<br />

both normal <strong>and</strong> OA cells, the pathway involves tyrosine kinase activity <strong>and</strong> protein kinase C<br />

(PKC). However, while normal HAC respond to 0.33 Hz mechanical stimulation with<br />

increases in aggrecan gene expression, aggrecan mRNA levels of OA HAC are unchanged.<br />

Experiments were performed utilising a panel of function-modifying antibodies to investigate<br />

whether modulation of #1 integrin function influenced OA chondrocyte mechanotransduction.<br />

Methods: OA HAC were incubated with function modifying #1 integrin receptor antibodies<br />

JB1a, P4C10 <strong>and</strong> TS2/16 for 30 min prior to 0.33 Hz MS. RNA was extracted 24 h<strong>our</strong>s post-<br />

MS <strong>and</strong> relative levels of aggrecan mRNA were assessed by semi-quantitative RT-PCR. PKC<br />

translocation <strong>and</strong> FAK phosphorylation in response to MS were assessed by western blotting.<br />

Results: Treatment with anti #1 integrin antibodies for 30 mins did not alter aggrecan mRNA<br />

levels, however following 24 h<strong>our</strong>s antibody incubation significant decreases in aggrecan<br />

mRNA levels were seen with the #1 integrin antibody P4C10 but not JB1a. Following MS,<br />

there was no alteration in aggrecan mRNA levels in untreated OA cells <strong>and</strong> MS had no effects<br />

on P4C10 down-regulation of aggrecan mRNA levels. Cells treated with JB1a showed a<br />

significant increase in aggrecan mRNA levels following MS. 30 seconds MS of chondrocytes<br />

resulted in translocation of PKC- <strong>and</strong> PKC. to the cell membrane which is inhibited by<br />

P4C10 but not by JB1a or TS2/16. 1 minute MS induced FAK phosphorylation which was<br />

unaffected by P4C10 or TS2/16. In the presence of JB1a a second enhanced phosphorylation<br />

of FAK was seen following 5 minutes MS.<br />

Conclusions: The results suggest that HAC mechanotransduction can be manipulated by<br />

function modifying #1 integrin receptor antibodies. The results also suggest that JB1a, in<br />

conjunction with MS acts in a positive manner raising the possibility that anti #1 integrin<br />

therapy may be used to alter the balance between catabolic <strong>and</strong> anabolic activity in<br />

osteoarthritis.<br />

- 620 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 14<br />

Menin protracts Chk1 phosphorylation <strong>and</strong> increases the ratio of homology-directed<br />

DNA repair<br />

Esposito I.1, Gallo A.2, Terrazzano G.1 <strong>and</strong> Musti A.M.1,3<br />

1 Dip.Biol.Patol.Cell.Mol., University of Naples “FedericoII, Italy<br />

2 I.E.O.S. of CNR c/o University of Naples “FedericoII”, Italy<br />

3 Dip. Farmaco-biologico, University of Calabria, Rende (Cs), Italy<br />

Multiple Endocrine Neoplasia type 1 (MEN1) is an inherited tumor disease characterised by<br />

pancreatic, parathyroid <strong>and</strong> anterior pituitary adenomas Besides, many MEN-1 patients also<br />

develop tumors in a range of non-endocrine tissues, suggesting that MEN1 may control tumor<br />

development in different tissues, depending on genetic interactions with other cancerassociated<br />

genes. Menin, the nuclear protein encoded by the MEN1 gene, interacts with a<br />

large number of proteins, among which nuclear proteins involved in chromatin modification<br />

or DNA repair, as Tritorax, RPA <strong>and</strong> FANCD2. Besides, loss of menin expression leads to<br />

increased sensitivity to DNA damage, both in embryonic mouse fibroblasts <strong>and</strong> Drosophila<br />

larvae.<br />

We have investigated the effect of Menin over-expression on the cellular response to DNA<br />

damage. We found that menin protracts the ATM/ATR-dependent phosphorylation of the Sphase<br />

checkpoint kinase Chk1, in response to etoposide or UV radiation, without affecting the<br />

rate of DNA damage. However, menin-induced persistence of Chk1 activation had no effect<br />

on either etoposide-induced S-phase arrest or apoptosis, but increased the ratio of<br />

homologous-directed DNA repair. These results provide novel insights into the<br />

oncosuppressor activity of MEN-1 by linking menin to DNA-repair pathways.<br />

- 621 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 15<br />

Nogo-A promotes denervation in an amyotrophic lateral sclerosis model<br />

Anissa Fergani1, Natasa Jokic1, Jose-Luis Gonzalez De Aguilar, Leda Dimou2, Shuo<br />

Lin3, Markus A. Ruegg3, Martin E. Schwab2, Luc Dupuis1 & Jean-Philippe Loeffler1<br />

1Laboratoire de Signalisations Moléculaires et Neurodégénérescence, INSERM U-692,<br />

Université Louis Pasteur, Faculté de Médecine, 11 rue Humann, 67000 Strasb<strong>our</strong>g,<br />

France,<br />

e-mail: anissa.fergani@neurochem.u-strasbg.fr, jnataly@yahoo.com,<br />

loeffler@neurochem.u-strasbg.fr, 2Brain Research Institute, University of Zurich <strong>and</strong><br />

Department of Biology, ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich,<br />

Switzerl<strong>and</strong>, schwab@hifo.unizh.ch, 3Biozentrum, University of Basel,<br />

Klingelbergstrasse 70, CH-4056 Basel, Switzerl<strong>and</strong>,<br />

e-mail: shuo.lin@unibas.ch, markus-a.ruegg@unibas.ch.<br />

The pathogenesis of ALS still remains unclear. Growing evidence suggests that initial<br />

alterations in skeletal muscle, preceding the onset of disease symptoms, are related to a loss of<br />

neuromuscular junction integrity, axonal degeneration <strong>and</strong> muscle denervation, rather than<br />

motor neuron death. Our previous studies showed a characteristic expression pattern of the<br />

three major isoforms of the neurite outgrowth inhibitor Nogo (including Nogo-A, -B <strong>and</strong> -C)<br />

in skeletal muscles of ALS patients <strong>and</strong> SOD1(G86R) mice. We found that the increased<br />

levels of Nogo-A <strong>and</strong> Nogo-B, which had been barely detectable in muscles of control<br />

subjects, correlate significantly with the severity of motor impairment of ALS patients, as<br />

determined by the clinically validated ALS functional rating scale. We wished to determine<br />

the impact of knocking down Nogo-A on the progression of ALS pathology in SOD1(G86R)<br />

mice <strong>and</strong> gain insight into the role of Nogo up-regulation in skeletal muscle. We crossbred<br />

mice knockout for Nogo-A with mice overexpressing the ALS-related mutation G86R, <strong>and</strong><br />

followed the survival time. We also performed in vivo skeletal muscle transfection of a vector<br />

expressing Nogo-A in Thy-1/YFP mice, <strong>and</strong> looked at the morphology of the neuromuscular<br />

junction (NMJ). Double-transgenic G86R/Nogo-A(-/-) mice survived longer than<br />

G86R/Nogo-A(+/+) mice. Transient expression of Nogo-A in skeletal muscle fibers was<br />

sufficient to injury the NMJ by inducing dismantlement of the post-synaptic structures <strong>and</strong><br />

loss of pre-synaptic terminals. The ectopic expression of Nogo-A in skeletal muscle initiates a<br />

cascade of events leading to loss of NMJs <strong>and</strong> denervation. This deletereous effect may be<br />

relevant to ALS since mice suffering from an ALS-like pathology <strong>and</strong> lacking Nogo-A live<br />

longer. Supported by AFM <strong>and</strong> ARS.<br />

- 622 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 16<br />

Molecular regulation of the expression of human A#H-J-J Locus, encoding Aspartyl-#hydroxylase,<br />

Junctin <strong>and</strong> Junctate<br />

Giordana Feriotto1, Alessia Finotti1, Giulia Breveglieri1, Pompeo Volpe3, Susan<br />

Treves2, Francesco Zorzato2, <strong>and</strong> Roberto Gambari1<br />

Dept of Biochemistry <strong>and</strong> Molecular Biology1, Dept of Experimental <strong>and</strong> Diagnostic<br />

Medicine2, Ferrara University, Italy; Dept of Biomedical Experimental Sciences3,<br />

Padova University, Italy; E-mail: gam@unife.it<br />

We have recently cloned <strong>and</strong> characterised the A#H-J-J locus, a genomic sequence that<br />

generates three functionally distinct proteins, the enzyme aspartyl #-hydroxylase (A#H),<br />

junctin, a structural protein of sarcoplasmic reticulum membrane, <strong>and</strong> the membrane-bound<br />

protein junctate (1, 2). The expression of the A#H-J-J locus is regulated by (a) transcription<br />

directed by the P1 <strong>and</strong> the P2 promoters (located upstream of exon 1 <strong>and</strong> exon 2 respectively),<br />

<strong>and</strong> (b) alternative splicing. In order to functionally characterise the two promoter sequences,<br />

we first demonstrate that mRNAs from P1 promoter are actively transcribed in all the human<br />

tissues <strong>and</strong> cell lines analysed, while the expression directed from the P2 promoter is tissue<br />

specific, in particular restricted to skeletal muscle, heart <strong>and</strong> brain. The P1 region responsible<br />

for maximal transcription contains at least twelve GC-box homologous to Sp1 consensus<br />

binding sequence; by EMSA we identified three GC-rich elements which bind Sp family<br />

nuclear factors with high efficiency (3). On the contrary, the P2 promoter contains sequences<br />

which bind to different transcription factors, including MEF-2 <strong>and</strong> MEF-3 (4). The<br />

transcription directed by the P2 promoter is induced by high expression of MEF-2 <strong>and</strong><br />

inhibited by HDAC4. ChIP analysis showed that MEF-2 efficiently binds chromatin of the P2<br />

promoter in C2C12 myotubes (4). In addition, we found that A#H <strong>and</strong> junctate transcripts<br />

exhibit single or multiple exons skipping in the region coding for the Ca++ binding domain<br />

leading to a number of A#H <strong>and</strong> junctate mRNA variants.<br />

1. Treves S et al. (2000) J. Biol. Chem. 275, 39555; 2. Treves S et al. (2004) J. Cell. Biol.<br />

166, 537; 3. Mischiati C et al. (1999) J. Biol. Chem. 274, 33114; 4. Feriotto G et al. (2005)<br />

Mol. Cell. Biol. 25, 3261.<br />

- 623 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 17<br />

Combined supplementation of folic acid <strong>and</strong> vitamin E diminishes diabetes-induced<br />

dysmorphogenesis in rat embryos<br />

Mattias Gäreskog, Ulf J Eriksson <strong>and</strong> Parri Wentzel<br />

Department of Medical Cell Biology, Uppsala University, Sweden<br />

Husargatan 3, Box 571, 751 23 Uppsala<br />

Mattias.Gareskog@medcellbiol.uu.se<br />

Diabetic embryopathy <strong>and</strong> growth disturbances are 3-5 fold more common in infants of<br />

diabetic mothers than in offspring of non-diabetic pregnancy. The mechanisms causing these<br />

disturbances are likely to be multifactorial.<br />

It has been suggested that oxidative stress <strong>and</strong> apoptosis plays an important roles during<br />

organogenesis. This notion may also be involved in the induction of embryonic<br />

dysmorphogenesis in diabetic pregnancy. We aimed to investigate the effect of administration<br />

of folic acid alone or combined with vitamin E on embryonic malformations <strong>and</strong> different<br />

markers of apoptosis, such as NFkB activity, Bcl-2, Bax protein levels <strong>and</strong> p-53 protein <strong>and</strong><br />

mRNA expression.<br />

Diabetes was induced by a single injection of streptozotocin. Pregnant, non-diabetic <strong>and</strong><br />

diabetic, rats were treated with daily injections of folic acid or treated with both folic acid <strong>and</strong><br />

5% vitamin E in the diet. Untreated rats from both groups served as controls. Embryos were<br />

collected on gestational day 10 or 11 <strong>and</strong> were evaluated with regard to malformations <strong>and</strong><br />

growth retardation. Some embryos were used for NFkB assay, western blot <strong>and</strong> cDNA<br />

synthesis.<br />

We found increased malformations <strong>and</strong> growth retardation in a diabetic milieu compared to<br />

normal environment. Supplementation of folic acid alone <strong>and</strong> combined with vitamin E<br />

normalized these parameters<br />

In addition, we found decreased NFkB activity <strong>and</strong> Bcl-2 protein, increased expression of Bax<br />

<strong>and</strong> p53 protein <strong>and</strong> mRNA in embryos of diabetic rats compared to normal rats.<br />

Administration of folic acid to the diabetic rats increased NFkB activity <strong>and</strong> Bcl-2 protein.<br />

Combined administration of folic acid <strong>and</strong> vitamin E normalized Bcl-2 expression in the<br />

diabetic environment.<br />

Combined folic acid <strong>and</strong> vitamin E supplementation to pregnant diabetic rats diminished<br />

diabetes-induced dysmorphogenesis <strong>and</strong> had several beneficial effects on embryonic<br />

apoptotic rate.<br />

- 624 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 18<br />

Alterations in salivary antioxidants, nitric oxide, <strong>and</strong> transforming growth factor-# in<br />

relation to disease activity in Crohn’s disease patients<br />

1Fakhteh Ghorbani, 1Azadeh Eshghtork, 2Ali Rezaie, 1Mohammad J. Zamani,<br />

1Shekoufeh Nikfar, 1Gholamreza Dehghan, 1Bardia Taghavi, 3Nasser E. Daryani, <strong>and</strong><br />

1Mohammad Abdollahi<br />

2Department of Community Health Sciences, Faculty of Medicine, University of<br />

Calgary, Canada, 1Department of Toxicology <strong>and</strong> Pharmacology, Faculty of Pharmacy,<br />

<strong>and</strong> Pharmaceutical Sciences Research Center, <strong>and</strong> 3Department of Gastrointestinal<br />

Diseases, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran,<br />

Email: mohammad.abdollahi@utoronto.ca<br />

It has been postulated that oxidative stress, nitric oxide (NO) <strong>and</strong> transforming growth factor<br />

#1 (TGF- #1) have major roles in the pathophysiology of Crohn’s disease (CD). The aim of<br />

this study was to determine the salivary levels of total antioxidant capacity (TAC), specific<br />

antioxidants (i.e. uric acid, albumin, transferrin <strong>and</strong> thiol molecules), lipid peroxidation<br />

(LPO), NO <strong>and</strong> TGF- #1in CD patients <strong>and</strong> control subjects <strong>and</strong> also investigate their<br />

correlation with activity of the disease. Twenty-eight patients with confirmed CD diagnosis<br />

were enrolled <strong>and</strong> whole saliva samples were obtained. Smokers, diabetics, those who<br />

suffered from periodontitis, <strong>and</strong> those who were consuming antioxidant supplements were<br />

excluded from the study. Crohn’s Disease Activity Index (CDAI) was used to determine the<br />

severity of the disease. Twenty healthy subjects were also recruited. In CD patients significant<br />

reductions in salivary levels of TAC (0.248±0.145 vs. 0.342±0.110 mmol/L), albumin<br />

(1.79±0.42 vs. 2.3±0.2 µg/ml) <strong>and</strong> uric acid (3.1±1.4 vs. 4.1±2.0 mg/dl) were found. TGF-#1<br />

was significantly increased in CD patients compared to healthy subjects (3.02±1.54 vs.<br />

2.36±0.52 ng/ml). A f<strong>our</strong>-fold increase in NO levels (198.8±39.9 vs. 50.2±21.3 µmol/L) along<br />

with a five-fold increase in LPO concentration (0.146±0.064 vs. 0.027±0.019 µmol/L) was<br />

documented in CD patients in comparison to control group. CDAI significantly correlated<br />

with the TAC, LPO <strong>and</strong> the interaction between TAC <strong>and</strong> LPO (r2=0.625, r2=0.8, F test’s<br />

P


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 19<br />

TAT fusion proteins as a drug delivery system<br />

Mira Grdi)a, Ana-Matea Mikecin <strong>and</strong> Miroslav Pozni',<br />

Division of Molecular Medicine, Rudjer Boskovic Institute, Bijenicka 54, 10 000 Zagreb,<br />

Croatia Email: grdisa@irb.hr<br />

TAT protein, a basic domain of human immunodeficiency virus type 1 (HIV-1), possess the<br />

ability to traverse biological membranes efficiently in a process termed «protein<br />

transduction». Protein transduction can be described as the direct uptake by the cell of<br />

exogenous proteins/peptides, as a result of a specific property of the protein/peptide<br />

component. The mechanism is unknown. Transduction occurs in receptor-transporter-<br />

independent manner that appears to target the lipid bilayer directly. Thus, HIV-1 Tat proteins<br />

have tramendous potential to deliver large-sized compounds (drugs) into the cells.<br />

In the rpesent study, transduction of full-length Tat-p27, pt-mutated Tat-p27 <strong>and</strong> N'- Tat-p27<br />

(truncated p27 on the C-terminal end) fusion proteins into human tumor cell lines was<br />

examined <strong>and</strong> whether these transduced proteins induced apoptosis in the cells.<br />

The proteins (p27, p23, Mp27) were transdused into different cell lines (NALM, MOLT, Raji,<br />

SuDHL, K562, RKO <strong>and</strong> MiaPaCa2) <strong>and</strong> their effect on proliferation of the cells were<br />

measured. The fusion proteins influenced moderately on the proliferation of different cell<br />

lines <strong>and</strong> varied among the proteins <strong>and</strong> type of the cells. A transduced p27 did not<br />

remarkable influence on proliferation of examined cell lines. Mutated p27 inhibited the<br />

proliferation of examined cell lines up to 30 %. On the other h<strong>and</strong>, a transduction of p23<br />

protein, truncated form of p27, inhibited the proliferation all of examined cell lines 30-60 %.<br />

Also the effects on expression of host p27 protein were examined, as well as an influence on<br />

induction of apoptosis.<br />

The expression of cell cycle regulatory proteins (cycD2, D3, cycE) were not remarkable<br />

affected with transduced proteins. It seems that transduced proteins induced apoptosis in<br />

examined cell lines. According the results, different apoptotic pathways were induced,<br />

depending on the type of cells.<br />

- 626 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 20<br />

Apoptotic cell <strong>signaling</strong> in lymphocytes from HIV+ patients during successful therapy<br />

S<strong>and</strong>ro Grelli,1 Emanuela Balestrieri,2 Claudia Matteucci,1 Antonella Minutolo,1<br />

Gabriella d’Ettorre,3 Fiorella Di Sora,4 Francesco Montella,4 Vincenzo Vullo,3 Stefano<br />

Vella,5 Cartesio Favalli,1 Antonio Mastino,6 Beatrice Macchi,2,7<br />

1Dept. of Exp. Med. <strong>and</strong> Bioch. Sci. <strong>and</strong> 2Dept. of Neuroscience, Univ. of Rome ‘Tor<br />

Vergata’, Via Montpellier 1, 00133 Rome, Italy. E-mail: grelli@med.uniroma2.it 3Dept<br />

of Infect. <strong>and</strong> Trop. Dis., Univ. of Rome ‘La Sapienza’, Rome;4Clin. Immunol. Unit, S.<br />

Giovanni Hospital, Rome; 5Istituto Superiore di Sanità, Rome; 6Dept. of Microbiol.,<br />

Genet. <strong>and</strong> Mol. Sci., Univ. of Messina, Messina; 7IRCCS S. Lucia, Rome. Italy<br />

The simultaneous expression of 15 genes involved in apoptotic cell signalling was analyzed<br />

by RNA-protection assay in peripheral blood mononuclear cells of HIV-infected patients<br />

before <strong>and</strong> during highly active antiretroviral therapy. After 12 months of successful therapy,<br />

characterized by an increase in CD4+ T cells <strong>and</strong> a decrease in viral load <strong>and</strong> in spontaneous<br />

apoptosis of lymphocytes, the expression of the pro-apoptotic genes FAS, FASL, FAF1,<br />

FADD, CASPASE 8, DR3, TRAIL, TNFR1, TRADD <strong>and</strong> BAX was significantly downregulated<br />

with respect to time 0, while that of BCL-2, BCL-XL <strong>and</strong> MCL-1 was up-regulated.<br />

Moreover, non-parametric bivariate Spearman’s analysis of correlation showed that apoptosis<br />

was directly correlated significantly with mRNA levels for TRAIL, FASL <strong>and</strong> CASPASE 8.<br />

Conversely, apoptosis levels were significantly inversely correlated with those of mRNA for<br />

BCL-2, BCL-XL <strong>and</strong> MCL-1. These results contribute to better define the signalling<br />

pathways that control apoptosis during HIV infection, showing for the first time in vivo the<br />

possible involvement of some death signalling mediators, such as FAF1,FADD, DR3 <strong>and</strong><br />

TRADD, or regulators, such as MCL-1 or BAX. Overall <strong>our</strong> data suggest that inhibition of<br />

apoptosis in HIV+ patients under successful therapy is the results of a complex network of<br />

signalling, involving both death <strong>and</strong> survival of lymphocytes.<br />

- 627 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 21<br />

Selection of non-apoptotic human spermatozoa provides superior tyrosine<br />

phosphorylation<br />

during capacitation <strong>and</strong> improved acrosome reaction<br />

Sonja Grunewald, Thomas Baumann, Uwe Paasch, Hans-Juergen Gl<strong>and</strong>er<br />

EAA Center University of Leipzig, Philipp-Rosenthal-Str. 23-25, 04103 Leipzig,<br />

Germany<br />

sonja.grunewald@medizin.uni-leipzig.de<br />

Introduction: Capacitation <strong>and</strong> acrosome reaction (AR) of human spermatozoa are<br />

prerequisites for fertilization. Major changes during capacitation result from tyrosine<br />

phosphorylations, a process regulated by a cAMP-dependent pathway involving protein<br />

kinase A. Annexin-V-MACS is able to separate apoptotic from non-apoptotic sperm based on<br />

their externalization of phosphatidylserine (EPS). The non-apoptotic (EPS-) fraction is<br />

characterized by lowest amounts of caspase activation, disrupted mitochondrial potential <strong>and</strong><br />

DNA fragmentation. The aim of <strong>our</strong> study was to investigate the separation effect of<br />

Annexin-V-MACS on capacitation-related tyrosine phosphorylations <strong>and</strong> acrosome reaction.<br />

Methods: Semen specimens from 10 healthy donors were separated into 2 samples each, one<br />

was left untreated (control) <strong>and</strong> the second was subjected to Annexin-V-MACS. Two aliquots<br />

of both, the control as well as the EPS negative fraction after Annexin-V-MACS were<br />

incubated in BWW at 37°C, 5% CO2 for 3h either with 3% BSA (capacitation) or without<br />

additives. Capacitation (CAP) was monitored by tyrosine phosphorylation (TyrP, western<br />

blot). AR was determined by labeling with mab CD46-FITC before <strong>and</strong> after stimulation with<br />

calcium ionophore A23187, followed by flow cytometric evaluation of the percentage of<br />

CD46+ sperm.<br />

Results: Densitometric analyses of the 105kDa <strong>and</strong> 80 kDa b<strong>and</strong>s of the TyrP western blots<br />

demonstrated significantly higher TyrP in the capacitated aliquots compared to the noncapacitated<br />

aliquots. Furthermore, EPS- samples presented with significant more TyrP<br />

compared to the non-separated semen samples (TyrP: Control 100%, Control-CAP 136±30%,<br />

EPS- 115±13%. EPS--CAP 165±34%). There was no difference in spontaneous AR in all<br />

groups (CD46+ sperm: Control 3.5±0.7%, Control-CAP 4.8±1.2%, EPS- 3.4±1.6%, EPS--<br />

CAP 4.9±1.9%). In contrast, after induction of AR the capacitated as well as EPS- aliquots<br />

showed significantly increased amounts of CD46 positive sperm. AR was best inducible in<br />

EPS- sperm after capacitation (CD46+ sperm: control 25.9±11.4%, control-CAP 44.3±9.3%,<br />

EPS- 37.4±8.0%, EPS--CAP 55.7±15.1%).<br />

Conclusions: Non-apoptotic human spermatozoa are characterized by superior ability to<br />

capacitate <strong>and</strong> consequently by maximum potential to perform acrosome reaction after<br />

stimulation. Selection of EPS negative sperm by Annexin-V-MACS may be of advantage for<br />

assistant reproduction in order to prepare the sperm subpopulation with the highest fertilizing<br />

potential.<br />

- 628 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 22<br />

The Skeletal Muscle Transcriptome In Amyotrophic Lateral Sclerosis.<br />

Benoît HALTER1, Luc DUPUIS1, Marc DE TAPIA1, Franck DI SCALA1, Christa<br />

Niederhause-Wiederkehr2, Philippe Demougin2, Michael Primig2, Jean-Philippe<br />

LOEFFLER1 & Jose-Luis GONZALEZ de AGUILAR1<br />

1Laboratoire de Signalisation Moléculaire et Neurodégénérescence – INSERM U692 -<br />

Faculté de Médecine - 11, rue Humann - 67085 Strasb<strong>our</strong>g Cedex FRANCE. Tel.: (+33)<br />

390 24 30 8! Fax: (+33) 390 24 30 65. Mail: halter@neurochem.u-strasbg.fr;<br />

gonzalez@neurochem.u-strasbg.fr; loeffler@neurochem.u-strasbg.fr<br />

2Biozentrum <strong>and</strong> Swiss Institute of Bioinformatics, University of Basel Switzerl<strong>and</strong><br />

The mechanism underlying ALS pathogenesis is still unclear but growing evidence suggests<br />

that initial alterations in skeletal muscle, preceding the onset of disease symptoms, may<br />

contribute to the disease process. We compared the muscle transcriptome of ALS-related<br />

SOD1(G86R) mice with that of sciatic nerve-axotomized mice. We used a high-density<br />

oligonucleotide microarray <strong>and</strong> applied advanced bio-informatics protocols to provide a high<br />

throughput <strong>and</strong> accurate analysis of gene expression on a large scale. An unsupervised<br />

clustering algorithm identified genes that are highly regulated in SOD1(G86R) mice. While<br />

some of these genes represented the characteristic denervation process occurring in ALS,<br />

others were specifically regulated under the pathological condition but not after experimental<br />

denervation. Skeletal muscle provides an additional s<strong>our</strong>ce of information for the<br />

comprehension of the pathological mechanisms acting in ALS. The systematic approach at<br />

the gene level presented herein may serve to identify new targets for therapeutic intervention<br />

<strong>and</strong> diagnosis.<br />

Supported by AFM (Association Française contre les Myopathies) <strong>and</strong> ARS (Association<br />

p<strong>our</strong> la Recherche sur la Sclérose Latérale Amyotrophique).<br />

- 629 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 23<br />

THE ANALGESIC AND ULCEROGENIC EFFECTS OF TRIBULUS TERRESTRIS<br />

EXTRACTS IN ANIMAL MODELS<br />

Mahmoud Reza Heidari, Jalal, Vafazadeh, Sayed Ahmad Hoseini , Mohammad Ghazi<br />

Khansari <strong>and</strong> Masood Yakhchali<br />

Dept. of Toxicology <strong>and</strong> Pharmacology, Faculty of Pharmacy, Neuroscience <strong>and</strong><br />

Physiology Research Center, Kerman, Iran, Heidarimr@yahoo.com<br />

Tribulus terrestris has been used in traditional medicine as releiving reuhmatic pain <strong>and</strong><br />

analgesic plant from long ago. Extraction of the fruits of plant was done by two different<br />

methods(suxheletion <strong>and</strong> perculation) with methanol 80%. The perculated extract with<br />

different doses 50, 100, 200, 400 <strong>and</strong> 800 mg/Kg were injected intaperitoneally to mice. The<br />

analgesic effects of the methanolic extract of this plant were evaluated by formalin <strong>and</strong> tailflick<br />

test in male albino mice. Ulcerogenicity test was done by Azuumi method using J Score.<br />

The dose of 100 mg/kg perculated extract had the highest analgesic effect in Formalin <strong>and</strong><br />

Tail-flick test. The ulcerogenicity of plant extract is lower than the indomethacin. The extracts<br />

have had less pathologic effect in rat,s stomach.<br />

- 630 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 24<br />

Expression <strong>and</strong> protective role of heme oxygenase-1 in the delayed myocardial<br />

preconditioning<br />

Gabor Jancso, Barbara Cserepes, Balazs Borsiczky, Andrea Ferenc, Boglarka Racz,<br />

Janos Lantos, E Roth<br />

Department of Surgical Research <strong>and</strong> Techniques, Faculty of Medicine, University of<br />

Pécs, Pécs, Hungary ; e-mail: jancsogabor@hotmail.com<br />

Ischaemic preconditioning (PC) is an endogenous adaptation response of the myocardium to<br />

stress, whereby short ischaemic-reperfusion periods increase the myocardium’s tolerance to a<br />

longer ischaemic attack. Heme oxygenase (HO)-1, that degrades the pro-oxidant heme to<br />

carbon monoxide <strong>and</strong> to the antioxidant bilirubin, has been shown to protect cardiomyocytes<br />

against oxidative injury. We aimed to demonstrate the expression <strong>and</strong> protective effect of HO-<br />

1 in the delayed PC. Neonatal rat cardiac myocytes were exposed to ischaemic PC (ischaemic<br />

medium for 20 min) <strong>and</strong> pharmacological (adenosine, norepinephrine, opioid)<br />

preconditioning (group 1). 24 h<strong>our</strong>s later cells were subjected to a test ischaemia (TI) –<br />

culturing for 3 h<strong>our</strong>s in ischaemic medium, following with a 2 h reperfusion in normal<br />

medium – <strong>and</strong> then lactate dehydrogenase (LDH), live/death ratio, <strong>and</strong> apoptosis were<br />

measured. In group 2. HO-1 enzymatic activity was competitively inhibited by administration<br />

of zinc protoporphyrin (ZnPPIX) after PC. In group 3. HO-1 synthesis was blocked with HO-<br />

1 siRNA before PC. In group 4. HO-1 expression was induced by administration of cobalt<br />

protoporphyrin (CoPPIX). In the group 5. (control) we made only test ischaemia without<br />

preconditioning. Furthermore we demonstrated HO-1 expression in the various groups with<br />

immuno-staining. Our results showed a significant decrease of LDH release in PC groups vs.<br />

control group, that has been risen in ZnPPIX <strong>and</strong> HO-1 siRNA treated groups. Increased<br />

apoptosis <strong>and</strong> cell death were seen in PC groups which were treated with ZnPPIX <strong>and</strong> HO-1<br />

siRNA. CoPPIX pretreatment resulted in decreased LDH level, apoptosis <strong>and</strong> cell death,<br />

which was comparable to PC groups. HO-1 immuno-staining showed an appreciable HO-1<br />

expression in PC groups, which was abolished with HO-1 siRNA administration, but not in<br />

ZnPPIX group. Our results suggest that HO-1 expression increases in both ischaemic <strong>and</strong><br />

pharmacological PC, <strong>and</strong> HO-1 has cellular protective effect against cell death <strong>and</strong> apoptosis<br />

in ischaemia-reperfusion induced oxidative injury. Supported by OTKA T-048851; OTKA<br />

F046593; OTKA F046504; OTKA T 038035.<br />

- 631 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 25<br />

Array <strong>and</strong> conventional comparative genomic hybridization reveal genomic copy<br />

number changes are closely linked to outcome of salivary gl<strong>and</strong> carcinomas.<br />

Kowan J. Jee1 Kristiina Heikinheimo2, Silvana DiPalma3, Antti Mäkitie4, Sakari<br />

Knuutila1 <strong>and</strong> Ilmo Leivo*1<br />

1 Department of Pathology, Haartman Institute <strong>and</strong> Helsinki University Central<br />

Hospital, FIN-00014 University of Helsinki, Finl<strong>and</strong><br />

2Department of Oral <strong>and</strong> Maxillofacial Surgery, Institute of Dentistry, University of<br />

Turku, FIN-20520 Turku, Finl<strong>and</strong><br />

3Department of Histopathology, Royal Surrey County Hospital, Surrey, United<br />

Kingdom (SDP)<br />

4Department of Otorhinolaryngology, Helsinki University Central Hospital, FIN-00290<br />

HUS (AM), Finl<strong>and</strong><br />

Genetic abnormalities at the subchromosomal level including whole genome survey which is<br />

capable to providing a precise abnormal localization on chromosomes are not well<br />

documented in salivary gl<strong>and</strong> carcinomas. We employed both conventional comparative<br />

genomic hybridization (cCGH) <strong>and</strong> oligonucleotide based array CGH (aCGH) technologies to<br />

identify genes for amplifications <strong>and</strong> deletions in salivary gl<strong>and</strong>s. We used eleven cases of<br />

mucoepidermoid carcinoma (MEC), nine cases of adenoid cystic carcinoma (AdCC) <strong>and</strong> eight<br />

cases of salivary duct carcinoma (SDC) of paraffin embedded tissues.<br />

In cCGH, the DNA copy number changes were detected in 19 of 28 carcinomas (67.9%). The<br />

most common gains were found on 1q <strong>and</strong> chromosome X, <strong>and</strong> the most common losses were<br />

whole chromosome 18, on 18q only, <strong>and</strong> minimum common regions on 18q21-qter.<br />

Interestingly,<br />

low-grade MEC showed almost normal DNA copy number (in average 0.33 changes per<br />

tumor) <strong>and</strong> clear contrast of the number of aberrations showed in high-grade carcinomas.<br />

Analysis of aCGH showed a detail genomic aberration in a case of high-grade MEC in which<br />

we compared both results. The results provided more precise genomic localization <strong>and</strong> target<br />

genes. Additional gains of 8q21.3-qter, 19q13.12 <strong>and</strong> losses of 5q13.2-q22.2 were identified<br />

in this case. Even in a pooled low-grade MEC, <strong>and</strong> AdCC which were shown be normal<br />

cCGH pattern, we found an agreement results with cCGH in MEC, however, interestingly a<br />

restricted amplification of 6p22 in AdCC was identified.<br />

In conclusions, aCGH is a useful method to identify fine resolution of chromosomal<br />

aberrations by using paraffin materials <strong>and</strong> the results are indicating in both methods, the<br />

degree of DNA copy number change is directly associated with rapidly aggressive behavior of<br />

salivary gl<strong>and</strong> carcinomas.<br />

Furthermore, the target genes may useful for developments for new drugs.<br />

- 632 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 26<br />

Antioxidative effects of plant polyphenols: from protection of G-protein <strong>signaling</strong> to<br />

prevention of age-related diseases<br />

Viktor Jefremov1,2, Mihkel Zilmer1, Kersti Zilmer1, Nenad Bogdanovic3 <strong>and</strong> Ello<br />

Karelson1<br />

1Department of Biochemistry, Tartu University, 50411 Tartu, Estonia. E-mail:<br />

ello.karelson@ut.ee; mihkel.zilmer@ut.ee; 2Department of Neurology <strong>and</strong><br />

Neurosurgery, Tartu University Hospital, 50411 Tartu, Estonia. E-mail:<br />

viktor.jefremov@kliinikum.ee 3Geriatric Department, Neurotec, Karolinska Institute,<br />

Karolinska University Hospital, S-14186 Huddinge, Stockholm, Sweden. E-mail:<br />

nenad.bogdanovic@neurotec.ki.se<br />

The oxidative stress hypothesis of aging <strong>and</strong> age-related diseases suggests beneficial effects<br />

of antioxidative plant polyphenols in preventing <strong>and</strong> suppressing the neurodegeneration <strong>and</strong><br />

atherogenesis. Recent data reported that certain plant polyphenols, incl. phytoestrogens, can<br />

improve cognitive function in aging <strong>and</strong> in Alzheimer’s disease (AD) <strong>and</strong> slow<br />

atherosclerosis development (Joseph et al., 2005; Manach et al., 2005). However, the<br />

mechanisms behind these protective actions of polyphenols remain to be elucidated. This<br />

study was undertaken to elucidate the antioxidant potency of three polyphenols (resveratrol,<br />

curcumin, genistein) by using the two human models: 1) down-regulation of pro-oxidant<br />

stimulated G-proteins in the postmortem brain membranes of aging <strong>and</strong> AD subjects; 2)<br />

elevation of oxy-resistance (lag-phase) of low-density lipoproteins (LDL) in the human<br />

serum. We used [35S]-GTP$S binding to investigate the effects of polyphenols on the activity<br />

of the stimulated G-proteins in the human brain membranes (Mahlapuu et al., 2003). The<br />

effect of the agents on duration of lag-phase <strong>and</strong> maximal rate of the Cu2+-induced LDL<br />

oxidation was determined by formation of conjugated dienes at 234 nm. In aging <strong>and</strong> AD<br />

frontocortical (FC) membranes, 3-10 µM of investigated polyphenols dose-dependently<br />

depressed the G-protein stimulation induced by 10 µM hydrogen peroxide or 500 µM<br />

homocysteine (an approximate 25% stimulation by both pro-oxidants was observed). In aging<br />

FC, resveratrol revealed higher antioxidant effects towards the stimulated G-proteins than<br />

genistein whereas the effect of curcumin was lowest. In AD, the antioxidant potency for<br />

curcumin showed significant decline from the value in normal aging whereas the antioxidant<br />

effects for resveratrol <strong>and</strong> genistein did not reduce remarkably. In both, aging <strong>and</strong> AD brain<br />

FC, the antioxidant effect of 3-10 µM 17-#-estradiol on the stimulated G-proteins showed<br />

moderate values, similarly to genistein.<br />

In the plasma concentration (1 µM), resveratrol, curcumin <strong>and</strong> genistein significantly<br />

increased the resistance of LDL to oxidation, prolonging the oxidation lag-phase 4.5, 2.8 <strong>and</strong><br />

1.5 fold, respectively. The ability of curcumin <strong>and</strong> genistein to significantly reduce the<br />

maximal rate of LDL oxidation (compared with control) might represent as a specific<br />

mechanism enabling the moderate antioxidant (antiatherogenic) activity for these compounds.<br />

Our results suggest that plant polyphenols have structure-dependent antioxidant<br />

(antiatherogenic) potency which might realize via protection of G-proteins from oxidative<br />

damage in normal aging <strong>and</strong> neurodegeneration as well as via prevention of LDL from<br />

abnormal (excessive) oxidation in atherogenesis.<br />

- 633 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 27<br />

Dissecting the mTOR Pathway<br />

Manel Joaquin1, Takahiro Nobukini1, Marta Roccio2, Stephen G. Dann3, So Young<br />

Kim1, Pawan Gulati3, Maya P. Byfield4, Jonthan M. Backer4, François Natt5, Johannes<br />

L. Boss2, Fried J.T. Zwartkruis2 <strong>and</strong> George Thomas1<br />

1Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel<br />

Switzerl<strong>and</strong>; 2Department of Physiological Chemistry <strong>and</strong> Centre for Biomedical<br />

Genetics, University Medical Center Utrech, Universiteitsweg 100, 3584 CG, Utrech,<br />

The<br />

Netherl<strong>and</strong>s; 3Genome Research Institute, University of Cincinnati, 2180E Galbraith<br />

Road, Cincinnati, OH 45237; 4Department of Molecular Pharmacology, Albert Eisntein<br />

College of Medicine, NY 10461; 5Novartis Institue for Biomedical Research, Lichstrasse<br />

35, 4002 Basel, Switzerl<strong>and</strong>. manel.joaquin@upf.edu<br />

During the evolution of metazoans <strong>and</strong> the rise of systemic hormonal regulation, the<br />

insulincontrolled<br />

class 1 phosphatidyl-inositide-3OH-kinase (PI3K) pathway was merged with the<br />

primordial amino acid driven mammalian Target of Rapamycin (mTOR) pathway to control<br />

the growth <strong>and</strong> development of the organism. Insulin regulates mTOR function through a<br />

recently-described canonical <strong>signaling</strong> pathway, which is initiated by the sequential activation<br />

of class 1 PI3K <strong>and</strong> Protein Kinase B (PKB) <strong>and</strong> subsequent repression of the Tuberous<br />

Sclerosis tumor suppressor complex proteins (TSC1/2), culminating in the activation of the<br />

Ras homologue enriched in brain (Rheb). However, how the amino acid input is integrated<br />

with that of insulin <strong>signaling</strong> pathway is unclear. Here we employed a number of molecular,<br />

biochemical <strong>and</strong> pharmacological approaches to address this issue. Unexpectedly, we found<br />

that the major pathway by which amino acids control mTOR <strong>signaling</strong> is distinct from that of<br />

insulin <strong>and</strong> that instead of <strong>signaling</strong> through components of the insulin/class 1 PI3K pathway,<br />

amino acids mediate mTOR activation by <strong>signaling</strong> through the class 3 PI3K, hVps34.<br />

- 634 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 28<br />

C-MIP: A NOVEL REGULATORY PROTEIN IMPLICATED IN T CELL<br />

SIGNALING.<br />

Maud Kamal , Karine Dahan, Marina C<strong>and</strong>elier, Philippe Lang, Georges Guellaen,<br />

Djillali Sahali.<br />

Nephrology department <strong>and</strong> INSERM U 581-équipe Avenir, Hôpital Henri Mondor,<br />

Créteil 94010, France. e-mail : kamal@im3.inserm.fr.<br />

It is believed that the pathogenesis of idiopathic nephrotic syndrome involves T cell<br />

dysfunction. Using subtractive cloning <strong>and</strong> differential screening, we isolated a new<br />

transcript, c-mip (c-maf inducing protein), which is up-regulated during the active phase of<br />

the disease. The transcript encodes an 85 kDa protein characterized by a pleckstrin homology<br />

(PH) domain <strong>and</strong> a Leucine rich repeat (LRR) domain. The localization of c-mip is restricted<br />

to the thymus, fetal liver, T cells <strong>and</strong> kidneys. We have demonstrated that the stimulation of T<br />

cells with Phytohemagglutinin (PHA) <strong>and</strong> Concanavalin A (Con A) downregulates c-mip.<br />

The expression of c-mip in stimulated T cells is further downregulated by Calphostin C, a<br />

PKC inhibitor. On the other h<strong>and</strong>, the inhibition or the activation of the MAPKs ERK1/2, by<br />

PD 98059 <strong>and</strong> IFNalpha respectively, does not affect the level of expression of c-mip in these<br />

stimulated T cells. These results suggest that PKC is involved in c-mip regulation<br />

independently of RAS-GRP. Moreover, we have demonstrated that the overexpression of cmip,<br />

in Jurkat cells, activates the ERK1/2 pathway <strong>and</strong> increases the expression of the<br />

regulatory subunit of the PI3 kinase, p85alpha. Overexpression of c-mip inhibits NFkB<br />

activation without any direct interference with NFkB p65. Altogether, these results suggest<br />

that c-mip recruits p85alpha <strong>and</strong> plays a negative regulatory role on NFkB <strong>signaling</strong> via<br />

mechanisms that are still under investigation.<br />

- 635 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 29<br />

Over-expression of transforming growth factor <strong>and</strong> nitric oxide in saliva of ulcerative<br />

colitis patients with various disease activities<br />

1Sara Khalaj, 2Ali Rezaie, 1Maryam Shabihkhani, 1Mohammad J. Zamani,<br />

1Shekoufeh Nikfar, 3Naser E. Daryani, <strong>and</strong> 1Mohammad Abdollahi<br />

2Department of Community Health Sciences, Faculty of Medicine, University of<br />

Calgary, Canada, 1Department of Toxicology <strong>and</strong> Pharmacology, Faculty of Pharmacy,<br />

<strong>and</strong> Pharmaceutical Sciences Research Center, <strong>and</strong> 3Department of Gastrointestinal<br />

Diseases, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.<br />

Email: mohammad.abdollahi@utoronto.ca<br />

Growth factors <strong>and</strong> nitric oxide (NO) play a major role in dysregulated immune response in<br />

ulcerative colitis (UC). Recent evidence showed increased levels of transforming growth<br />

factor-#1 (TGF-#1) in UC <strong>and</strong> suggested an anti-inflammatory effect for this factor. Based on<br />

<strong>our</strong> recent study, dysfunctional immunoregulation is present in saliva of UC patients, we<br />

hypothesized that salivary level of NO <strong>and</strong> TGF-#1 may differ by severity of UC <strong>and</strong> be<br />

useful to determine the activity of the disease. Thirty-seven consecutive patients with<br />

confirmed UC were enrolled <strong>and</strong> saliva samples were obtained. Fifteen healthy control<br />

subjects were also recruited. Truelove-Witts severity index <strong>and</strong> modified Truelove-Witts<br />

severity index were used to determine the severity of the disease. NO <strong>and</strong> TGF-#1 levels were<br />

detected in saliva of all patients <strong>and</strong> control subjects using Enzyme-Linked Immunosorbent<br />

Assay. Twenty-one patients had mild disease while 8 had moderate <strong>and</strong> 8 had severe colitis.<br />

Adjusted for baseline characteristics, the levels of NO <strong>and</strong> TGF-#1 in different groups were<br />

compared. Salivary NO <strong>and</strong> TGF-#1 levels were higher in UC patients comparing to controls<br />

(P


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 30<br />

15-Deoxy-&12,14-prostagl<strong>and</strong>in J2 covalently modifies <strong>and</strong> functionally inactivates p53<br />

in human breast cancer (MCF-7) cells<br />

Do-Hee Kim, Hye-Kyung Na <strong>and</strong> Young-Joon Surh<br />

National Research Laboratory of Molecular Carcinogenesis <strong>and</strong> Chemoprevention,<br />

College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea<br />

Cyclopentenone prostagl<strong>and</strong>ins (cyPGs) exert diverse cellular functions, such as antiinflammatory<br />

<strong>and</strong> cytoprotective effects. 15-Deoxy- &12,14-prostagl<strong>and</strong>in J2 (15d-PGJ2), a<br />

representative cyPG of the J series, has been reported to exert biphasic effects on proliferation<br />

<strong>and</strong> apoptosis. In the present study, we found that MCF-7 human breast cancer cells treated<br />

with 15d-PGJ2 accumulated the tumor suppressor gene product p53 in both cytosolic <strong>and</strong><br />

nuclear fractions. Under the same experimental conditions, however, the mRNA level of p53<br />

remained unchanged. This prompted us to investigate the molecular mechanisms responsible<br />

for the 15d-PGJ2-mediated accumulation of p53 protein. Murine double-minute 2 (MDM2), a<br />

zinc finger protein, is known to negatively regulate p53. 15d-PGJ2 treatment caused failure of<br />

proper p53-MDM2 interaction in MCF-7 cells which could stabilize p53 protein by blocking<br />

MDM2-mediated degradation. We observed that 15d-PGJ2 covalently modify cysteine<br />

residues of p53 in MCF-7 cells. Covalent modification of p53 by 15d-PGJ2 is considered to<br />

interfere with DNA binding of p53. In support of this assumption, 15d-PGJ2 inhibited the<br />

DNA binding ability of both recombinant p53 <strong>and</strong> MCF-7 nuclear extracts in a concentrationrelated<br />

manner. Likewise, p53 conjugation with 15d-PGJ2 may hamper its interaction with<br />

MDM2, <strong>and</strong> subsequent degradation. The majority of accumulated p53 in 15d-PGJ2-treated<br />

MCF-7 cells represents a functionally inactive or mutant form as assessed by<br />

immunoprecipitation <strong>and</strong> immunofluorescence analysis. It is anticipated that the electrophilic<br />

carbon center located in the alpha,#-unsaturated carbonyl moiety of the cyclopentenone ring<br />

might be critical for the 15d-PGJ2-induced covalent modification of p53, cysteine thiol,<br />

which retarded its degradation. MCF-7 cells treated with 9,10-dihydro-15-deoxy-&12,14prostagl<strong>and</strong>in<br />

J2 that lacks the electrophilic alpha #-unsaturated functionality failed to<br />

accumulate p53 protein, lending further support to the above assumption. In summary, 15d-<br />

PGJ2 directly conjugates with cysteine thiol residues of p53, interfering its interaction with<br />

MDM2, thereby stabilizing p53. Covalent modification of p53 by 15d-PGJ2 also hampers p53<br />

DNA binding, leading to functional inactivation of this tumor suppressor.<br />

- 637 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 31<br />

EFFECT OF ENDOTHELIN ON THE SODIUM/HYDROGEN EXCHANGER (NHE)<br />

ACTIVITY OF HUMAN MONOCYTES AND ATHEROSCLEROSIS-RELATED<br />

FUNCTIONS<br />

Koliakos G1., Befani Chr. 2, Paletas K.3, Kaloyianni M.2<br />

1Laboratory of Biological Chemistry, Medical School, Aristotle University of<br />

Thessaloniki, 54124<br />

2Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle<br />

University of Thessaloniki, 54124<br />

3 B’ Medical Clinic, Medical School, Aristotle University of Thessaloniki, 54124<br />

Aims: The aim of the present study is to investigate the influence of endothelin on human<br />

monocyte sodium/hydrogen exchanger (NHE-1) activity <strong>and</strong> on the atherosclerosis-related<br />

monocyte functions. NHE-1 is an integral membrane protein that exchanges one intracellular<br />

H+ ion for an extracellular Na+ ion. It plays an essential role in all cell types regulating the<br />

internal pH (pHi), cell growth, differentiation <strong>and</strong> apoptosis. Endothelin is a potent<br />

vasoconstrictive <strong>and</strong> mitogenic 21-amino-acid peptide that has been implicated in the<br />

pathogenesis of atherosclerosis. Monocytes are key contributors to the atherosclerotic process.<br />

Methods: The effect of endothelin (10pg/ml) on monocyte NHE-1 activity was<br />

studied by means of pHi (using the fluorescent indicator BCECF-AM) <strong>and</strong> 22Na uptake.<br />

Specific inhibitors were used in order to avoid interference from other sodium-exchanging<br />

pumps. In an attempt to investigate the signal transduction pathway from endothelin to NHE 1<br />

we used PD 98059 MEK1 (50µ)) (MAPKp42/44 inhibitor), GF-109203X (10µ)) (inhibitor<br />

of all the isoforms of PKC) <strong>and</strong> Gö 6976 (500nM) (specific inhibitor of the isoform PKCs). In<br />

addition, the effect of endothelin (10pg/ml) on the ability of monocytes to bind on the<br />

basement membrane glycoprotein laminin <strong>and</strong> to migrate on trans-well culture inserts was<br />

estimated. We also measured the density of CD36 receptor using a fluorescein isothiocyanate<br />

(FITC)-linked anti CD36 monoclonal antibody.<br />

Results: Endothelin caused an increase in pHi (p


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 32<br />

Implication of TRIP6, a novel molecular partner of the MAGI-1b scaffolding molecule<br />

in invasiveness<br />

Larissa Kotelevets1, Erik Bruyneel3, Alexei Kruglov1,4, Marc Bracke3, Frans van Roy2<br />

<strong>and</strong> Eric Chastre1<br />

1 INSERM U683, Faculté de Médecine X. Bichat, Paris, France<br />

2 Department for Molecular Biomedical Research, VIB-Ghent University, B-9052 Ghent<br />

3 Laboratory of Experimental Cancerology, Ghent University Hospital, B-9000 Ghent,<br />

Belgium<br />

4 Institute of Theoretical <strong>and</strong> Experimental Biophysics, Russian Academy of Sciences,<br />

Pushchino, Russia.<br />

We recently established the critical role of the PTEN/MAGI-1b signalosome in the<br />

stabilization of cell-cell contacts <strong>and</strong> the suppression of invasiveness. The PTEN tumor<br />

suppressor is recruited to E-cadherin junctional complexes through the binding to the 2nd<br />

PDZ domain of the MAGI-1b scaffolding molecule, whereas #-catenin interacts with the 5th<br />

PDZ domain of MAGI-1b (Kotelevets et al, FASEB J 19,115,2005). To identify additional<br />

effector molecules that might regulate the activity <strong>and</strong> the composition of the PTEN / MAGI-<br />

1b complexes, we used yeast-two hybrid screening. Among the clones identified, we focused<br />

on TRIP6 (Thyroid Receptor Interacting Protein 6)/ZRP-1 (Zyxin-related Protein 1). TRIP6<br />

has been reported to associate with p130Cas in focal adhesion complexes. We demonstrated<br />

that TRIP6 interacted directly with MAGI-1b through binding to the 5th PDZ-binding motif.<br />

Ectopic expression of TRIP6 induced invasiveness in the epithelial kidney MDCK <strong>and</strong><br />

MDCKts-src cells in a PI3-kinase dependant manner, <strong>and</strong> reverted the E-cadherin-dependent<br />

cell-cell aggregation. In this connection, we observed a slight increase in AKT activity in<br />

MDCKts-src derivatives transfected with TRIP6 expression vectors, compared to parental<br />

cells. The TRIP6Stop473 mutant, lacking the PDZ binding motif was unable to promote cell<br />

invasiveness <strong>and</strong> did not interfere with cell-cell aggregation. The competing peptides<br />

corresponding to the C-terminus of TRIP6 or b-catenin promoted invasiveness in TRIP6<br />

Stop473-transfected MDCKts-src cells but not in the parental cell line. These results suggest<br />

that TRIP6-induced invasiveness in epithelial cells involves the induction of cell motility by<br />

the TRIP6 core molecule, whereas the C-terminus is required to destabilizes E-cadherin<br />

junctional complexes by competing with #-catenin for the interaction with MAGI-1b.<br />

- 639 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 33<br />

Microgravity signal ensnarls cell adhesion <strong>and</strong> cytoskeleton of rat osteoblasts: osteopontin,<br />

CD44, osteonectin, <strong>and</strong> "-tubulin<br />

Yasuhiro Kumei 1 , Sadao Morita 1 , Hitoyata Shimokawa 1 , Kei-ichi Ohya 1 , Hisako Katano 2 ,<br />

Hideo Akiyama 3 , Masahiko Hirano 3<br />

1 Graduate School of Tokyo Medical <strong>and</strong> Dental University, Tokyo, Japan<br />

2 University of Tokyo Medical Science Institute, Tokyo, Japan<br />

3 Toray Research Center, Kamakura, Japan<br />

We examined gene expression of osteopontin (OP), osteonectin (ON), CD44, <strong>and</strong> "-tubulin in<br />

rat osteoblasts that were cultured aboard Space Shuttle for 4 days. During the last 24 hrs, all<br />

the cultures were treated with 1",25 dihydroxyvitamin D3, <strong>and</strong> solubilized by guanidine<br />

solution. The relative mRNA levels were determined by quantitative RT-PCR, followed by<br />

normalization to glyceraldehyde 3-phosphate dehydrogenase. Microgravity decreased the OP<br />

mRNA levels by 40%, while increased the CD44 mRNA levels by 280%, as compared to the<br />

ground controls. OP is a major extracellular matrix protein in bone, <strong>and</strong> important for<br />

hydroxyapatite crystal formation <strong>and</strong> osteoclast attachment to the bone surface. The<br />

hyaluronic acid receptor CD44 interacts with OP <strong>and</strong> FN, however, fibronectin gene<br />

expression was not altered by microgravity. Microgravity also increased the ON mRNA<br />

levels by 40%, but decreased the "-tubulin mRNA levels by 40%, as compared to ground<br />

control levels. ON is another matrix protein associated with tubulin. Association/dissociation<br />

of these molecules may mediate cell adhesion, motility, <strong>and</strong> <strong>signaling</strong> via rearrangement of<br />

cytoskeletal structure under microgravity conditions.<br />

- 640 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 34<br />

Induction of accumulation of $-globin mRNA in human erythroid cells treated with<br />

psoralens<br />

Ilaria Lampronti1, Nicoletta Bianchi1, Cristina Zuccato1, Francesco Dall’Acqua2,<br />

Margherita Zennaro1, Daniela Vedaldi2, Giampietro Viola2 <strong>and</strong> Roberto Gambari1,3.<br />

1Department of Biochemistry <strong>and</strong> Molecular Biology, University of Ferrara,<br />

2Department of Pharmaceutical Sciences, University of Padova, 3GenTech-for-Thal,<br />

Laboratory for the Development of Pharmacological <strong>and</strong> Pharmacogenomic Therapy of<br />

Thalassemia, Ferrara<br />

The identification of potential therapeutic agents for treatment of hematological diseases,<br />

including #-thalassemia <strong>and</strong> sickle cell anemia (SCA), can be based on pharmacologicallymediated<br />

regulation of the expression of human $-globin genes, leading to an increase of the<br />

production of fetal hemoglobin (HbF). DNA-binding drugs appear to be of great interest <strong>and</strong><br />

represent a promising approach to control gene expression. Among molecules able to interact<br />

with DNA, psoralens <strong>and</strong> related compounds could be relevant. In this study, we analysed<br />

several linear <strong>and</strong> angular psoralens. To verify the activity of these furano-cumarins, we<br />

employed two experimental cell systems, the human leukemic K562 cell line for preliminary<br />

studies <strong>and</strong> the two-phase liquid culture of human erythroid progenitors isolated from normal<br />

donors for further analysis on the most active compounds. Erythroid differentiation was<br />

analysed by benzidine-staining; expression of $-globin genes by quantitative RT-PCR assays<br />

using the Perkin-Elmer 7700 Sequence Detector. The results of <strong>our</strong> investigation suggest that<br />

angelicin, bergapten <strong>and</strong> several structurally-related compounds (trimethylangelicin, psoralen,<br />

8-methoxy-psoralen, 4-methyl-psoralen <strong>and</strong> 5’-methyl-psoralen) are powerful inducers of<br />

erythroid differentiation <strong>and</strong> $-globin mRNA accumulation both in K562 cells <strong>and</strong> in human<br />

erythroid precursors. The activity of trimethylangelicin <strong>and</strong> 8-methoxy-psoralen were found<br />

higher than that displayed by hydroxyurea, commonly used as HbF inducers in $-thalassemia<br />

<strong>and</strong> SCA patients. Our results could have practical impact, as it is well know that an increase<br />

in $-globin mRNA <strong>and</strong> fetal hemoglobin production could ameliorate the clinical status of<br />

patients with $-thalassemia <strong>and</strong> sickle cell anemia.<br />

Work supported by CIB, AVLT (Associazione Veneta per la Lotta alla Talassemia), EU<br />

(Fondi Strutturali Obiettivo 2), Fondazione CARIPARO (Cassa di Risparmio Padova-<br />

Rovigo), ER-SPINNER <strong>and</strong> PRRIITT.<br />

- 641 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 35<br />

Regulation of fibroblasts adhesion <strong>and</strong> migration by protease nexin-1<br />

Xiaobiao Li<br />

Friedrich Miescher Institute, Basel<br />

Protease Nexin-1 (PN-1), a 43 KDa glycoprotein inhibits extracellular serine proteases<br />

activity. It also triggers signal transduction by interacting with members of distinct surface<br />

receptor families, such as low-density lipoprotein protein receptors (e.g. LRP1), <strong>and</strong> heparan<br />

sulfate proteoglycans (e.g. syndecan-1). These interactions lead to activation of either PKA or<br />

ERK <strong>signaling</strong>, <strong>and</strong> the final outcome depends on the cross talk between these two pathways.<br />

In wild type mouse embryonic fibroblasts, PN-1 activates PKA via interaction with LRP1,<br />

leading to inhibition of Ras-ERK <strong>signaling</strong>. In cells deficient in LRP1, interaction between<br />

PN-1 <strong>and</strong> syndecan-1 activates ERK <strong>signaling</strong> (Li et al., submitted).<br />

In this study, we observed that PN-1 activated ERK <strong>signaling</strong> <strong>and</strong> its down stream effector<br />

Rac1, causing lamellipodia formation <strong>and</strong> enhanced cell migration in LRP1-/- MEF cells.<br />

This effect was mediated by the interaction between PN-1 <strong>and</strong> syndecan-1. Overexpression of<br />

PN-1 activated ERK <strong>signaling</strong> constitutively <strong>and</strong> increased cell adhesion <strong>and</strong> migration on<br />

vitronectin. The effect was even more significant when LRP1-/- MEF cells were<br />

overexpressing syndecan-1. We also observed that stimulated LRP1-/- MEF cell migration<br />

was inhibited by preincubation with antibodies against syndecan-1, <strong>and</strong> integrin #3, which is<br />

functionally coupled to this proteoglycan. Furthermore PN-1 was specifically coimmunoprecipitated<br />

with the integrin #3 subunit. Consequently <strong>our</strong> data indicate that PN-1<br />

affects ERK <strong>signaling</strong> <strong>and</strong> cell migration through its interactions with integrin #3 <strong>and</strong><br />

syndecan-1.<br />

- 642 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 36<br />

Differential activation of Akt <strong>and</strong> VEGF in tumor-derived microvascular endothelial<br />

cells: a novel mechanism for acquired chemoresistance in liver cancers<br />

Fanyin Meng, Roger Henson, Hania Wehbe, Molly Lang, <strong>and</strong> Tushar Patel<br />

Scott <strong>and</strong> White Clinic, Texas A&M University System Health Science Center College of<br />

Medicine, 2401 South 31st Street, Temple, TX 76508, U.S.A. E-mail:<br />

fmeng@swmail.sw.org<br />

Targeting endothelial cells that line tumor blood vessels is a promising new strategy for the<br />

treatment of cancers such as liver cancer. The transcription factor Nuclear Factor Kappa B<br />

(NF-kB) can regulate the expression of key mediators of angiogenesis <strong>and</strong> cell survival such<br />

as protein kinase B (PKB/Akt) <strong>and</strong> vascular endothelial growth factor (VEGF). However, the<br />

involvement <strong>and</strong> role of these mechanisms in tumor derived endothelial cells is unknown.<br />

Methods: Hepatocellular carcinoma was induced in BALB/c mice using Ndiethylnitrosamine.<br />

Microvascular endothelial cells (MVEC) were isolated from liver tumors<br />

(T-MVECs) as well as normal liver tissues (N-MVECs) using a magnetic bead<br />

immunoaffinity technique. In vitro angiogenesis was quantitated using a commercial assay kit<br />

(Chemicon). Results: In both types of MVEC, transfection with NF-kB increased Akt<br />

phosphorylation <strong>and</strong> VEGF expression. However, the increased VEGF expression was<br />

blocked by dominant negative Akt in T-MVECs, but not N-MVECs. Moreover, overexpression<br />

of Akt directly increased VEGF expression <strong>and</strong> NF-kB dependent angiogenesis in<br />

T-MVECs. Incubation with gemcitabine to induce chemotherapeutic stress dramatically<br />

increased NF-kB activation in both T-MVECs <strong>and</strong> N-MVECs, but increased Akt <strong>and</strong> VEGF<br />

expression only in T-MVECs. In addition, Akt activation <strong>and</strong> VEGF expression was<br />

dependent on NF-kB in T-MVECs. Over-expression of Akt decreased gemcitabine-induced<br />

apoptosis in T-MVECs, but not in N-MVECs. In vitro angiogenesis was increased by<br />

chemotherapeutic stress in T-MVECs, not in N-MVECs, <strong>and</strong> was abolished by inhibition of<br />

either NF-kB or Akt. Conclusions: Akt is selectively activated in tumor derived endothelial<br />

cells <strong>and</strong> may contribute to NF-kB dependent VEGF expression <strong>and</strong> angiogenesis <strong>and</strong><br />

survival during chemotherapeutic stress. Targeting NF-kB dependent up-regulation of Akt<br />

<strong>and</strong> VEGF may be useful to decrease chemoresistance <strong>and</strong> tumor progression in liver cancers.<br />

- 643 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 37<br />

$-Glutamyltransferase is upregulated after oxidative stress through the Ras signal<br />

transduction pathway in colon carcinoma cell lines<br />

Seila Møller, Serhyi Pankiv, Ugo Moens, Nils-Erik Huseby<br />

Department of Medical Biochemistry, Faculty of Medicine, University of Tromsø,<br />

Norway. email: nilseh@fagmed.uit.no<br />

Gamma-glutamyltransferase (GGT) has a central role in glutathione (GSH) homeostasis.<br />

After oxidative stress <strong>and</strong> depletion of GSH, upregulation of GGT has been reported. As<br />

oxidative stress may activate various Ras signal transduction pathways, we examined whether<br />

the regulation of GGT is mediated through these pathways. Acute exposure of rat colon<br />

carcinoma cells to menadione resulted in elevated GGT activity <strong>and</strong> protein levels detectable<br />

after 24 h. Quantitative PCR showed increased total GGT mRNA levels 6 h after the<br />

exposure. Actinomycin D <strong>and</strong> cycloheximide reduced the elevation. When the oxidative stress<br />

exposures were performed in the presence of protein kinase inhibitors that block downstream<br />

targets p38, PI-3K <strong>and</strong> MEK1/2 of Ras, the upregulation of GGT was attenuated. GGT is<br />

transcribed from five promoters into multiple mRNAs. Using semiquantitative RT-PCR,<br />

promoter II was found increased after acute oxidative stress. Transient cotransfection studies<br />

using this promoter II region in a luciferase plasmid together with an active Ras plasmid,<br />

resulted in increased luciferase activity, whereas no activity was measured with dominant<br />

negative Ras. This promoter contains putative binding sites for AP1, AP2, NFkB, SRE <strong>and</strong><br />

SP1. Using plasmids with binding sites for the mentioned trancription factors we found that<br />

SRE <strong>and</strong> AP1 were active in transcribing the enzyme.<br />

The present study shows an upregulation of GGT after exposure to oxidative stress through a<br />

de novo transcription of the GGT gene being mediated through several Ras signal<br />

transduction pathways. The cells will benefit from the enzyme activity in maintaining the<br />

intracellular level of GSH. Thus, the enzyme will add to the protective measures of the tumor<br />

cells during oxidative stress.<br />

- 644 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 38<br />

HYPOXIA IS RESPONSIBLE OF sVEGF-R1 INDUCTION IN VILLOUS<br />

TROPHOBLAST EXPLANTS CULTURE<br />

Carine Munaut , Sarah Berndt , Sophie Lorquet, Christel Pequeux, Francis Frankenne,<br />

Van den Brûle Frédérique <strong>and</strong> Jean-Michel Foidart.<br />

Laboratory of Tumor <strong>and</strong> Development Biology, CRCE, CHU, GIGA, EMBIC partners.<br />

University of Liège, T<strong>our</strong> de Pathologie (B23), Sart Tilman, B-4000 Liège, Belgium.<br />

Preeclampsia is a disorder unique to human pregnancy characterized by a generalized<br />

systemic maternal inflammatory response, associated with diffuse endothelial cell<br />

dysfunction. It is one of the leading causes of maternal <strong>and</strong> perinatal morbidity <strong>and</strong> mortality,<br />

affecting 5% to 7% of all pregnancies, yet the etiology <strong>and</strong> pathogenesis have not been fully<br />

defined.<br />

Oxygen plays a central role in this pathology. Recently, preeclampsia has been described as a<br />

state of imbalance between pro-angiogenic <strong>and</strong> anti-angiogenic factors. Main pro-angiogenic<br />

factors that promote angiogenesis in the placenta belong to VEGF family.<br />

We have previously shown that preeclampsia was associated with low levels of circulating<br />

PlGF <strong>and</strong> increased levels of total VEGF-A <strong>and</strong> soluble VEGF-R1 (Tsatsaris et al, 2003,<br />

J.Clin.Endocrinol.Metab). Here, <strong>our</strong> study was undertaken to test the hypothesis that high<br />

levels of those angiogenic factors could be related to hypoxic status of placenta in<br />

preeclampsia.<br />

Small fragments of placental villi from first trimester (11-14 weeks) voluntary interrupted<br />

pregnancies were used for explant culture under normoxia (20% O2, <strong>and</strong> 5% CO2) or hypoxia<br />

(1% O2, 5% CO2 <strong>and</strong> 94% N2). Under hypoxia, villous trophoblast explants expressed higher<br />

levels of VEGF-A, VEGF-R1, sVEGF-R1 <strong>and</strong> VEGF-R2 mRNAs than under normoxia<br />

culture. By contrast, PlGF mRNA was decreased in hypoxia. Protein secretion of VEGF-A<br />

<strong>and</strong> sVEGF-R1, determined by ELISA, were also found elevated in hypoxic explant cultures.<br />

No histological or morphological modifications were observed under hypoxia as compared to<br />

normoxia.<br />

Our data show that villous trophoblast is the likely s<strong>our</strong>ce of increased plasma levels of<br />

VEGF-A <strong>and</strong> VEGF-R1 in pregnant woman. Our results also support the hypothesis that<br />

overproduction of sVEGF-R1 by villous trophoblast submitted to extended hypoxia in<br />

preeclampsia could account for the depletion of maternal blood free VEGF.<br />

- 645 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 39<br />

Mechanisms of selenium cytotoxicity in a malignant mesothelioma model – targeting<br />

Thioredoxin Reductase-1<br />

Gustav Nilsonne, Branka Kocic, Aristi Potamitou Fern<strong>and</strong>es, Agnes Stein, Anna-Klara<br />

Rundlöf, Mikael Björnstedt, Anders Hjerpe, <strong>and</strong> Katalin Dobra.<br />

All authors: Karolinska Institutet, Institution for Laboratory Medicine, Dept. of<br />

Pathology. Karolinska Univ Hosp Huddinge F-46, S-141 86 Huddinge, Sweden.<br />

Malignant mesothelioma (MM) is an aggressive tum<strong>our</strong> arising from the serous cavities. MM<br />

cells may differentiate into an epithelioid or a sarcomatoid phenotype, <strong>and</strong> the latter is more<br />

chemoresistant <strong>and</strong> associated with a worse prognosis. The thioredoxin (Trx) system is<br />

upregulated in many tumors, <strong>and</strong> the highest reported levels have been found in MM. The<br />

system comprises Trx, Thioredoxin Reductase (TrxR1) <strong>and</strong> NADPH. It functions mainly to<br />

maintain intracellular redox balance, but also counteracts apoptosis. Sodium selenite causes<br />

oxidative stress.<br />

The objectives of this study were to investigate whether selenite could induce apoptosis in a<br />

sarcomatoid <strong>and</strong> an epithelioid MM cell line, <strong>and</strong> what mechanisms would mediate the<br />

cytotoxic effects.<br />

Apoptosis was measured by three independent methods <strong>and</strong> was induced in the sarcomatoid<br />

cell line at low concentrations. The IC 50 was 7,5 µM. IC 50 of the epithelioid cell line, with<br />

greater expression of the Trx system, was 21 µM. The DCF probe revealed ROS formation<br />

following selenite treatment, analysed with confocal microscopy. Selenite concentrations of<br />

10 µM or more inhibited TrxR1 effectively.<br />

ICC revealed p53 activation in both cell lines. Immunostaining with FACS analysis revealed<br />

that Bax was activated by selenite. The DioC6 marker analysed by FACS showed loss of<br />

mitochondrial membrane potential. Inhibition of JNK using SP600125 had no effect in either<br />

cell line. Inhibition of p38 decreased the activation of Bax. No caspase activity was detected<br />

after selenite treatment. The pan-caspase inhibitor z-VAD-fmk did not attenuate apoptosis.<br />

In conclusion, selenite generated ROS, inhibited TrxR1 <strong>and</strong> caused apoptosis in two MM cell<br />

lines. The therapy resistant sarcomatoid cells were more sensitive. The effects were mediated<br />

through p53 <strong>and</strong> Bax, but appear to be independent of both caspases <strong>and</strong> JNK. This is a<br />

promising new treatment option for an aggressive <strong>and</strong> chemoresistant tumor.<br />

- 646 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 40<br />

Signaling pathways regulating production of hyaluronic acid in pig oocyte-cumulus cell-<br />

complexes<br />

Radek Procházka <strong>and</strong> Eva Nagyová<br />

Institute of Animal Physiology <strong>and</strong> Genetics, Academy of Sciences of the Czech<br />

Republic, Lib2chov, 277 21 Czech Republic. E-mail: prochazka@iapg.cas.cz<br />

Cumulus cells undergo an extensive expansion following the preovulatory surge of<br />

gonadotrophins due to increased production of hyaluronic acid (HA) <strong>and</strong> its deposition in<br />

extracellular spaces. In vitro, the expansion can be stimulated with FSH in all mammalian<br />

species. The insulin like-growth factor-I (IGF-I) can modify the action of FSH on cumulus<br />

cells. The objective of this study was to characterize mechanisms by which the IGF-I affects<br />

the FSH-stimulated production <strong>and</strong> deposition of HA in pig oocyte-cumulus complexes<br />

(OCCs). For this purpose, <strong>signaling</strong> pathways activated in cumulus cells by IGF-I were<br />

studied <strong>and</strong> their importance for production of HA was assessed. OCCs isolated from 3-5 mm<br />

follicles were cultured in M-199 medium supplemented with 3 mg/ml polyvinyl pyrrolidone<br />

under an atmosphere of 5% CO2 in air for 24 h. To quantify production of HA, OCCs were<br />

cultured in the presence of 2.5 µCi of tritiated glucosamine hydrochloride <strong>and</strong> the<br />

incorporated label was measured by a liquid scintillation counter in medium containing OCCs<br />

(total HA) or within the complexes alone (retained HA). We found that production <strong>and</strong><br />

retention of HA was significantly higher in OCCs cultured in medium with FSH (10 ng/ml)<br />

than in control group cultured without FSH (P


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 41<br />

Inactivation of human melanoma cells induced by alkylating agents <strong>and</strong>/or highly<br />

ionizing radiation<br />

Aleks<strong>and</strong>ra M. Risti'-Fira1, Ivan M. Petrovi'1, Lela B. Kori'anac1, Danijela V.<br />

Todorovi'1, Lucia M. Valastro2, Giacomo Cuttone2, Giuseppe Privitera3<br />

1 Vin(a Institute of Nuclear Sciences, Belgrade, Serbia <strong>and</strong> Montenegro<br />

2 Istituto Nazionale di Fisica Nucleare, LNS, Catania, Italy<br />

3 Institute of Radiology <strong>and</strong> Radiation Oncology, University of Catania, Italy<br />

E-mail: aristic@vin.bg.ac.yu<br />

The response of human HTB140 melanoma cells to chemotherapeutic drugs fotemustine<br />

(FM) <strong>and</strong> dacarbazine (DTIC) as well as of proton irradiation were studied. Viability of cells<br />

treated with 100 <strong>and</strong> 250 micromolar drugs was assessed after incubation of 6, 24, 48, 72 <strong>and</strong><br />

96 h. Proton irradiations of exponentially growing cells were performed at the CATANA<br />

(Centro di Adro Terapia e Applicazzioni Nucleari Avanzati) facility for treatment of eye<br />

melanoma at INFN, LNS – Catania, delivering to the cell monolayer single doses of 2, 4, 8,<br />

12 <strong>and</strong> 16 Gy. Cell viability was evaluated 7 days after irradiation. For all treatments,<br />

inactivation level was estimated using microtetrasolium (MTT) <strong>and</strong> sulforhodamine B (SRB)<br />

assays. The combined effects of each drug <strong>and</strong> protons, were carried out using the same drug<br />

concentrations, while proton doses applyed were those used in therapy, i.e. 12 <strong>and</strong> 16 Gy. In<br />

this case viability was estimated using only SRB staining since it was shown to be more<br />

reliable than MTT assay for the incubation of 48 h. With the increase of the drug<br />

concentration or irradiation dose, the level of cell inactivation was more pronounced, reaching<br />

approximately 60 %, 48 h after drug treatment or 7 days after irradiation at 16 Gy.<br />

Considering the rate of drug concentrations used, as well as the level of doses applied, it<br />

appears that HTB140 cells are more resistant to proton irradiation than to alkylating agents<br />

tested. The combined treatment with FM or DTIC <strong>and</strong> protons did not show significant<br />

changes of cell viability as compared to the effects of single agents. Slightly better cell<br />

inactivation was obtained for 250 micromolar DTIC combined with 16 Gy proton irradiation.<br />

Taking into account the fact that the chosen time point for measuring cumulative effects of<br />

drug <strong>and</strong> irradiation was 48 h post-irradiation, it seams that the obtained level of viability<br />

could be attributed almost only to the effects of drugs.<br />

- 648 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 42<br />

GSK-3 mediates differentiation <strong>and</strong> activation of pro-inflammatory dendritic cells<br />

Elena Rodionova, Eugene Maraskovsky, Michael Hess, Michael Kirsch, Anthony D. Ho,<br />

<strong>and</strong> Thomas Luft<br />

Glycogen synthase kinase (GSK)-3 is a multifunctional enzyme critical for cellular<br />

differentiation, apoptosis, self renewal <strong>and</strong> motility, <strong>and</strong> dysregulation of GSK-3 is linked to<br />

immune deviations associated with diabetes, cancer, schizophrenia <strong>and</strong> Alzheimer’s disease.<br />

We demonstrate that GSK-3 is constitutively active in human monocytes. During<br />

differentiation of monocyte-derived dendritic cells (MoDC), GSK-3 inhibits macrophage<br />

development <strong>and</strong> allows DC to accumulate in an immature stage by inhibiting spontaneous<br />

maturation.<br />

However, in the context of DC activation GSK-3 acquires a pro-inflammatory function<br />

mediating high levels of IL-12p70, IL-6 <strong>and</strong> TNF-alpha without influencing migration or<br />

secretion of IL-10.<br />

These paradoxical inhibitory <strong>and</strong> pro-inflammatory effects of GSK-3 highlight changes in<br />

intracellular “primed” GSK-3 targets due to the activity of other kinases. The proinflammatory<br />

effect of GSK-3 in the context of DC activation is immediately counterbalanced<br />

by Akt-1. Akt-1 is phosphorylated during DC activation <strong>and</strong> partially inhibits GSK-3 activity<br />

by Ser21/9 phosphorylation. Thus, IL-12p70 secretion results from the competition for the<br />

phosphate-binding pocket of GSK-3 between GSK-3 target proteins (primed by other kinases)<br />

<strong>and</strong> the inhibitory Ser9/21 moiety (phosphorylated by Akt-1).<br />

Our results demonstrate that GSK-3 is a crucial enzyme mediating the differentiation <strong>and</strong><br />

activation of pro-inflammatory DC.<br />

- 649 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 43<br />

Adaptive functional differentiation of dendritic cells – integrating the network of extra-<br />

<strong>and</strong> intracellular signals<br />

Elena Rodionova1, Eugene Maraskovsky3,4, Michael Kirsch2, Michael Hess2 <strong>and</strong><br />

Thomas Luft1,2.<br />

Phenotypic maturation, cytokine secretion <strong>and</strong> migration are distinct functional characteristics<br />

of dendritic cells (DCs). These functions are independently regulated by a number of<br />

extracellular variables, such as type, strength <strong>and</strong> persistence of an array of soluble <strong>and</strong><br />

membrane-bound mediators. Since the exact composition of these variables in response to<br />

infection may differ between individuals, the intracellular signalling pathways activated by<br />

these extracellular networks may more closely correlate with DC function <strong>and</strong> predict the<br />

c<strong>our</strong>se of adaptive immunity. We found that activation of p38K, ERK1/2 <strong>and</strong> PC-PLC<br />

enhanced cytokine secretion, whereas p38K, cAMP <strong>and</strong> PC-PLC enhanced migration. In<br />

contrast, PI3K/Akt-1 <strong>and</strong> cAMP inhibited cytokine secretion whilst ERK1/2 inhibited<br />

migration. Migration <strong>and</strong> cytokine secretion further differed in their sensitivity to inhibition<br />

over time. However, although DCs could be manipulated to express migration, cytokine<br />

secretion or both, the level of activation or persistence of intracellular pathway signalling was<br />

not predictive. Our results suggest a modular organization of function. We hypothesize that<br />

the expression of specific DC functions integrates a large variety of activating <strong>and</strong> inhibitory<br />

variables, <strong>and</strong> is represented by the formation of a functional unit of molecular networks - the<br />

signal response module (SRM). The combined activities of these modules define the<br />

functional outcome of DC activation.<br />

- 650 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 44<br />

Alterations in cell <strong>signaling</strong> in the model organism Saccharomyces cerevisiae caused by<br />

heterologous expression of enterobacterial virulence factors<br />

Isabel Rodríguez-Escudero,1 Ainel Alemán,1 Philip R. Hardwidge,2 B. Brett Finlay, 2<br />

Rafael Rotger, 1 Víctor J. Cid1 <strong>and</strong> María Molina1<br />

1Dpto. de Microbiología II, Fac. de Farmacia, Universidad Complutense, Pza. de<br />

Ramón y Cajal s/n, 28040 Madrid, Spain. E-mail: isabelre@farm.ucm.es<br />

2Biotechnology Laboratory, Room 237, Wesbrook Building, 6174 University Boulevard,<br />

University of British Columbia, Vancouver, BC, V6T 1Z3. Canada.<br />

Certain proteins from pathogenic bacteria, injected into the cell via a specialized type III<br />

secretion system (TTSS), account for the induction of local actin polymerization by<br />

interfering with host cell <strong>signaling</strong>. Enteropathogenic E. coli (EPEC) strains cause severe<br />

diarrhea by inducing characteristic “effacing” lesions in enterocytes through the development<br />

of actin-supported pedestals at the site of bacterial adhesion. Pathogenesis requires a<br />

functional TTSS, which injects into the host cell the intimin receptor, Tir, as well as other<br />

effectors. We have made use of the model organism Saccharomyces cerevisiae to probe the<br />

functions of several EPEC effector proteins, studying their effects on cell growth, cytoskeletal<br />

function <strong>and</strong> <strong>signaling</strong> pathways. We found that some EPEC effectors were able to interfere<br />

with cytoskeletal function <strong>and</strong> to activate MAPK pathways when expressed in yeast, offering<br />

a feasible host cell model for molecular studies on these proteins. On the same trend, we have<br />

used this model to study the function of the SigD/SopB TTSS effector from Salmonella, a<br />

bacteria that invades epithelial cells by modifying epithelial cell <strong>signaling</strong> to induce its own<br />

internalization. SigD/SopB shares homology with mammalian phosphatidylinositol 4phosphatases.<br />

By expressing this protein in S. cerevisiae we have been able to dissect two<br />

functional regions into SigD, the catalytic C-terminal region <strong>and</strong> an N-terminal extension that<br />

is able to disrupt actin. We have isolated yeast Cdc42 as a suppressor by overexpression of<br />

the actin-depolarization phenotype caused by heterologous SigDR468A, a novel catalytically<br />

inactive version of this protein. This provides evidence that small GTPases within the host<br />

cell are putative targets for this bacterial virulence factor.<br />

- 651 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 45<br />

Delta-opioid receptor function is regulated by phosducin-like protein long (PhLPL) in<br />

cultured astroglyal cells<br />

Pilar Sánchez-Blázquez, Carlos Montero <strong>and</strong> Javier Garzón.<br />

Laboratory of Neuropharmacology, Instituto Cajal, Avenida Doctor Arce 37, Madrid<br />

28002. Spain. Email: psb@cajal.csic.es<br />

Affinity-purified IgGs raised against different PhLPL sequences recognised two major b<strong>and</strong>s<br />

of about 38 (non-glycosylated) <strong>and</strong> 55 kDa (N-glycosylated) in Western blots of proteins<br />

from resting astroglial cells. While in resting astrocytes little PhLPL was found in the nucleus,<br />

stimulation of delta-opioid receptors with the selective agonist DPDPE, induces a<br />

redistribution of PhLPL, <strong>and</strong> a large fraction of the cell membrane protein pool translocated to<br />

the nucleus. Although the mechanism by which PhLPL reaches the nucleus remains unknown,<br />

nuclear phosphorylation of PhLPL might facilitate its binding to 14-3-3, allowing the Cterminal<br />

domain of PhLPL to act as an activator of transcription. In <strong>our</strong> work, Gbeta coimmunoprecipitated<br />

with the PhLPL, <strong>and</strong> the association increased in presence of the delta<br />

opioid agonist DPDPE. At longer intervals (6 <strong>and</strong> 24 h) the association diminished while that<br />

of PhLPL with the P-Ser-binding protein 14-3-3 augmented. This observation suggests that<br />

the stimulation of delta opioid-receptors markedly increase phosphorylation of PhLPL over<br />

the very modest observed in resting astrocytes. We next examine the kinases involved in<br />

PhLPL phosphorylation analyzing the effects of different cell-permeable inhibitors (0.1 to 100<br />

nM): the PKA inhibitor peptide-fragment 6-22 amide; the CaMKII inhibitors, KN-93 <strong>and</strong><br />

autoclamide 2-related inhibitory peptide; the selective inhibitor of PKC, chelerythrine; <strong>and</strong> the<br />

CK2 inhibitor, DRB. At the concentrations <strong>and</strong> exposure times used, none of these kinase<br />

inhibitors interfered with the shape or viability of the astroglial cells. In the whole astrocyte<br />

we found that PKC, CaMKII <strong>and</strong> to a lesser extent CK2, were all important kinases in PhLPL<br />

phosphorylation. However, differences were observed between the N-glycosylated <strong>and</strong> nonglycosylated<br />

forms of PhLPL. Thus, G protein signalling in astroglial cells induces the<br />

cellular redistribution of PhLPL <strong>and</strong> its compartment-dependent phosphorylation by different<br />

kinases.<br />

(This work was supported by grants MCYT SAF2003-0112 <strong>and</strong> BMC2002-03228.)<br />

- 652 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 46<br />

Modification of GSTP1-1 by cyclopentenone prostagl<strong>and</strong>ins<br />

Fancisco J. Sánchez-Gómez, Javier Gayarre, Konstantinos Stamatakis <strong>and</strong> Dolores<br />

Pérez-Sala<br />

Departamento de Estructura y Función de Proteínas. Centro de Investigaciones<br />

Biológicas, C.S.I.C., 28040 Madrid, Spain. Email: dperezsala@cib.csic.es<br />

Glutathione-S-transferases (GST) constitute a family of enzymes which play a key role in the<br />

detoxification of electrophilic compounds by catalyzing their conjugation with glutathione.<br />

Therefore, these enzymes play an important role in cell defense mechanisms, including the<br />

development of resistance of tumor cells towards anticancer drugs. Cyclopentenone<br />

prostagl<strong>and</strong>ins are electrophilic compounds that have shown antiproliferative activity against<br />

a variety of cancer cell lines, which is related to their ability to modify cellular proteins by<br />

Michael addition. It is known that cyclopentenone PG can inhibit the activity of several GST<br />

isoforms, both in vitro <strong>and</strong> in intact cells. The molecular basis for this effect appears to be<br />

complex, but to date, covalent interactions between GST <strong>and</strong> cyclopentenone PG have not<br />

been documented. Here we show that the cyclopentenone PG 15-deoxy-Delta-12,14-PGJ2<br />

covalently binds to GSTP1-1 in vitro. This binding can be demonstrated by MALDI-TOF<br />

mass spectrometry, as well as by the use of a biotinylated 15d-PGJ2 analog. Using this analog<br />

we have evidenced the binding of 15d-PGJ2 to GSTP1-1 in intact human T-cell leukemia<br />

Jurkat cells. In addition, treatment of Jurkat cells with cyclopentenone PG induces the<br />

oligomerization of GSTP1-1 <strong>and</strong> a concomitant increase in the phosphorylation of JNK <strong>and</strong> of<br />

c-Jun. This effect is associated with the induction of apoptosis. Monomeric, but not<br />

oligomeric GST has been shown to act as an endogenous inhibitor of JNK. In the light of this<br />

evidence <strong>our</strong> results provide a basis to explore the involvement of the GST-JNK pathway in<br />

the apoptotic process <strong>and</strong> put forward a potential mechanism for the activation of JNK by<br />

cyclopentenone PG in Jurkat cells.<br />

- 653 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 47<br />

M-CSF-induced proliferation <strong>and</strong> LPS-dependent activation of macrophages requires<br />

Raf-1 phosphorylation to induce MKP-1 (DUSP1) expression<br />

Ester Sánchez-Tilló, Monica Comalada, Consol Farrera, Annabel F. Valledor, Jorge<br />

Lloberas <strong>and</strong> Antonio Celada<br />

Macrophage Biology Group, Institute of Biomedical Research, Barcelona Science Park,<br />

University of Barcelona, Barcelona, Spain. E-mail: acelada@ub.edu<br />

Macrophages are key regulators of immune responses. Bone marrow-derived macrophages<br />

undergo proliferation in response to their specific growth factor, M-CSF. The addition of<br />

activating agents, such as lipopolysaccharide (LPS), results in macrophage growth arrest <strong>and</strong><br />

engagement in a pro-inflammatory response, which produces nitric oxide (NO) <strong>and</strong> cytokines<br />

including tumor necrosis alpha (TNF-"), IL-1 <strong>and</strong> IL-6. Although activation of ERK is<br />

required for both macrophage proliferation <strong>and</strong> activation, the kinetics of these two processes<br />

differs. In <strong>our</strong> cellular model, early peak of ERK activation (5 min) correlates with cellular<br />

proliferation whereas a later peak (15 min) is associated with the activation program. M-CSF<br />

<strong>and</strong> LPS also induce with different time-c<strong>our</strong>se the dual specificity MAP kinase phosphatase<br />

MKP1 (DUSP1), which correlates with the dephosphorylation of ERK. Therefore, MKP1 is a<br />

key regulator of the time-c<strong>our</strong>se of ERK activity. Using RNA interference <strong>and</strong><br />

pharmacological inhibitors, here we provide evidence that macrophagic MKP1 expression<br />

induced by M-CSF <strong>and</strong> LPS is dependent on Raf-1 activation. In addition, Raf-1 interacts<br />

with PKC', which is also involved in MKP-1 induction. Concordantly, the distinct timec<strong>our</strong>se<br />

of Raf-1 <strong>and</strong> MEK-1/2 activation correlates with that of ERK-1/2 induced by M-CSF<br />

or LPS. ERK activation in response to M-CSF is Raf-1-dependent, but an alternative pathway<br />

induces ERK activation in response to LPS. Blockage of Raf-1 activity results in increased<br />

expression of cyclin dependent kinase inhibitors p21Waf-1 <strong>and</strong> p27Kip-1 <strong>and</strong> macrophage<br />

growth arrest even in the presence of M-CSF. In contrast, LPS stimulation has no effect on<br />

the expression of pro-inflammatory cytokines <strong>and</strong> inducible nitric oxide synthase.<br />

- 654 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 48<br />

Role of ATP in trauma associated cytokine release by P2X7 ion channel stimulation<br />

E. Marion Schneider1, Katrin Vorlaender1, Xueling Ma1, Weidong Du1, Manfred<br />

Weiss2,<br />

1Sektion Experimentelle Anaesthesiologie, Klinische Anaesthesiologie2,<br />

Universitaetsklinikum Ulm, Ulm, Germany;<br />

Marion.schneider@uni-ulm.de<br />

Objective: Trauma <strong>and</strong> massive tissue damage contribute to immediate cytokine release <strong>and</strong> systemic<br />

inflammatory response syndromes (SIRS) which may eventually lead to life threatening sepsis <strong>and</strong><br />

septic shock. Knowledge about mechanisms explaining hyperinflammation in the absence of<br />

infectious complications would improve preventive therapeutic options. We asked whether P2X7 ion<br />

channel activation may be involved.<br />

Background: The P2X7 receptor is ubiquitiously expressed <strong>and</strong> belongs to a family of lig<strong>and</strong>-gated<br />

channels activated by extracellular ATP. Binding of extracellular ATP activates multimerization of<br />

P2X7 subunits into trimers <strong>and</strong> hexamers, allowing transmembrane passage of cations <strong>and</strong> small<br />

molecules. Short stimulation with extracellular ATP upregulates cytokine release. Prolonged or<br />

repeated exposure to ATP induces the formation of a cytolytic pore <strong>and</strong> triggers cell death. An A to C<br />

single nucleotide polymorphism (SNP) at position 1513 (1513A_C) of the Glu-496 to Ala residue in<br />

the intracellular C-terminal tail leads to nonfunctional P2X7, whereas heterozygous individuals have a<br />

lower response.<br />

Methods: Cell isolates were plated with 2x105 cell/ml <strong>and</strong> ATP stimulation was performed with 1 <strong>and</strong><br />

5 mM ATP for 6h <strong>and</strong> 24h. Cytokines released were determined by a chemiluminescence assay<br />

(Immulite, DPC-Biermann, Bad Nauheim, Germany). P2X7 SNPs were determined by<br />

pyrosequencing (Biotage, Uppsala, Sweden). Ca++ signalling was studied by Fluo-4 (Ca++ dye)<br />

assisted confocal laser scanning microscopy using a Leica-TCS NT microscope <strong>and</strong> argon/krypton<br />

laser using semi-confluent HeLa, microvascular endothelium <strong>and</strong> macrophages.<br />

Results: Effects of P2X7 stimulation on the release of inflammatory cytokines in spontaneous B cell<br />

lines as well as in whole blood <strong>and</strong> in Ficoll isolated blood mononuclear cells (PBMC) were as<br />

follows: ATP induced solely IL-8 but neither IL-1ß, nor IL-6 or IL-10 in B cells. By contrast, PBMC<br />

of healthy donors responded with positive IL-1ß in addition to IL-8, no TNF-a was induced. Thus in<br />

addition to the effect on caspase 1 leading to IL-1ß secretion, ATP activates the release of IL-8 as an<br />

universal oxidative stress chemokine. Responses were stronger in wildtype P2X7 as compared to the<br />

1513A_C heterozygous cells. P2X7 signalling involves a Ca++ release response generated by<br />

phosphatidylinositol-3-phosphate (IP3) from intracellular stores. A number of inhibitors applied<br />

(Ryanodine, Paxilline: 1µM, Xestospongin C: 10µM, oxATP: 1µM) clearly demonstrated that the<br />

calcium signal involves both, intracellular Ca++ stores <strong>and</strong> extracellular Ca++ influx as well. In<br />

addition to the involvement of phospholipase C (PLC) to generate IP3-induced intracellular Ca++<br />

release, the blockade by paxilline further suggests a role by large conductance Ca++ -activated K+<br />

channels (BK). Upon decline of the Ca++ response, strong fluorescent labelling was transiently<br />

detected in swollen mitochondria. Apoptosis was associated with impressive blebbing of the plasma<br />

membrane.<br />

Conclusions: ATP stimulation of the P2X7 receptor induces the release of IL-1ß <strong>and</strong> IL-8 in<br />

monocytic cells <strong>and</strong> IL-8 as a universal stress marker in B cells <strong>and</strong> in other, also non hematopoietic<br />

cells. In vivo, P2X7 stimulation may well occur in severe trauma, may lead to neutrophil recruitment<br />

via IL-8-induced chemotaxis as well as cell damage in endothelial <strong>and</strong> epithelial cells. Via P2X7,<br />

ATP-induced apoptosis is dependent on PLC activation, intracellular Ca++ release <strong>and</strong> the activation<br />

of large conductance potassium channels as well.<br />

- 655 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 49<br />

Control of Human Herpesvirus Type 8-associated diseases by NK cells<br />

1Maria C. Sirianni, 1Massimo Campagna, 1Donato Scaramuzzi, 1Maurizio Carbonari,<br />

2Elena Toschi, 2Ilaria Bacigalupo, 2Paolo Monini <strong>and</strong> 2Barbara Ensoli<br />

1Clinical Immunology, Department of Clinical Medicine, University of Rome “La<br />

Sapienza”, 00161 Rome <strong>and</strong> 2National AIDS Center, Istituto Superiore di Sanita’,<br />

00161 Rome, Italy. E-mail: mariacaterina.sirianni@uniroma1.it<br />

The ‘natural killer’ (NK) cells, a first line defense mechanism against tumors <strong>and</strong> viruses,<br />

preferentially kill target cells lacking surface Major Histocompatibility Complex class I<br />

(MHC-I) molecule expression. NK cells recognize these targets through membrane receptors,<br />

which can trigger either activating or inhibitory signals for killing. Several tumor or virus<br />

infected cells down-regulate MHC-I expression as a mechanism to evade recognition <strong>and</strong><br />

killing by cytotoxic T lymphocytes (CTL). They, however, become targets for NK cells<br />

cytotoxic activity.<br />

NK cell activity is reduced during disease progression in Human Immunodeficiency Virus<br />

(HIV) infection, <strong>and</strong> in associated tumors latently infected by the oncogenic Human<br />

Herpesvirus Type 8 (HHV8) such as Kaposi’s sarcoma (KS) <strong>and</strong> primary effusion lymphomas<br />

(PEL). We have demonstrated that HIV-related KS (AIDS-KS) is characterized by an<br />

increased expression on T lymphocytes of inhibitory receptors recognizing HLA-C molecules<br />

(1), whereas non-HIV infected patients with KS (classic KS, C-KS) have a substantial number<br />

of peripheral blood NK cells bearing these receptors (2). NK cells from patients with C-KS,<br />

are equipped with cytolytic molecules including granzyme A <strong>and</strong> perforin. However, the<br />

cytotoxic activity of NK cells is reduced in patients with C-KS, AIDS-KS or PEL patients,<br />

which are all infected by the HHV8, <strong>and</strong> this correlates with disease severity (3-5). Moreover,<br />

we have found that HHV8 infected cell lines established from PELs have a reduced surface<br />

expression of MHC-I molecules <strong>and</strong> are sensitive to the lysis mediated by NK cells (4, 5).<br />

Since PEL cells express the same HHV8 latency program as KS cells, these data point to<br />

MHC-I downregulation by HHV8 as a primary immune evasion mechanism against CTL<br />

responses, further reinforced by up-regulation of inhibitory receptors on T <strong>and</strong> NK cells in the<br />

setting of HIV <strong>and</strong> HHV8 infection. Thus, studies on killing receptor regulation <strong>and</strong> <strong>signaling</strong><br />

in T <strong>and</strong> NK cells may shed light on HHV8-associated tumors both in HIV-infected or non<br />

infected patients.<br />

- 656 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 50<br />

Proteomic identification of cellular targets for posttranslational modification by antiinflammatory<br />

cyclopentenone prostagl<strong>and</strong>ins<br />

Konstantinos Stamatakis, Javier Gayarre, Francisco J. Sánchez-Gómez <strong>and</strong> Dolores<br />

Pérez-Sala<br />

Departamento de Estructura y Función de Proteínas. Centro de Investigaciones<br />

Biológicas, C.S.I.C., 28040 Madrid, Spain. Email: kostas@cib.csic.es<br />

Prostagl<strong>and</strong>ins with cyclopentenone structure have been shown to display protective effects in<br />

numerous cellular <strong>and</strong> animal models of inflammation <strong>and</strong> injury. These effects have been<br />

related to their ability to block the inflammatory response <strong>and</strong> to elicit a cellular heat shock<br />

response. An important determinant in the beneficial effects of cyclopentenone PG is their<br />

ability to form covalent adducts with thiol groups in proteins by Michael addition. Here we<br />

have explored the ability of biotinylated analogs of the cyclopentenone PG, 15d-PGJ2 <strong>and</strong><br />

PGA1, to mimic the effect of their parent PG. Biotinylated 15d-PGJ2 inhibited the response<br />

of renal mesangial cells to proinflammatory stimuli <strong>and</strong> elicited a cellular stress response. In<br />

addition, both biotinylated analogs induced the activation of MAPK in murine fibroblasts.<br />

Therefore, we have used these biotinylated PG to identify, through a proteomic approach,<br />

some of the protein targets the modification of which may be involved in the biological<br />

effects of cyclopentenone PG. Extracts from cells incubated in the presence of biotinylated<br />

PG were analyzed by 2D electrophoresis <strong>and</strong> the spots showing positive biotin staining were<br />

analyzed by tryptic digestion <strong>and</strong> MALDI-TOF mass spectrometry. In biotinylated 15d-PGJ2treated<br />

cells we have identified several proteins involved in the regulation of cellular<br />

architecture <strong>and</strong> dynamics, such as actin, tubulin, vimentin <strong>and</strong> tropomyosin. Modification of<br />

vimentin was associated with a collapse of the intermediate filament network in mesangial<br />

cells, which was reminiscent of a heat shock response. In addition, several proteins known to<br />

be regulated by oxidative stress are also targets for the addition of 15d-PGJ2, including<br />

Hsp90, nucleoside diphosphate kinase <strong>and</strong> methyl thioadenosine phosphorylase. The<br />

structural <strong>and</strong> functional implications of the modification of these various targets <strong>and</strong> their<br />

potential involvement in the biological effects of cyclopentenone PG will contribute to a<br />

better underst<strong>and</strong>ing of the selectivity <strong>and</strong> therapeutic potential of these prostanoids.<br />

- 657 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 51<br />

Primer Design <strong>and</strong> Polymerase Chain Reaction (PCR) Amplification of the Mutation<br />

Cluster Regon (MCR) of the Adenomatous Polyposis Coli (APC) Gene<br />

Mary Eloisa Torres-Rowshangah <strong>and</strong> Filipinas F. Natividad<br />

Department of Biology, College of Science, University of the Philippines Baguio,<br />

Governor Pack Road, Baguio City 2600, Philippines E-mail :metorres@eudoramail.com<br />

Research <strong>and</strong> Biotechnology Division, St. Luke’s Medical Center, 279 E. Rodriguez<br />

Senior Boulevard, Quezon City 1102 Phlippines E-mail :ffnatividad@st luke.com.ph<br />

Colorectal cancer is a major health concern worldwide. The Adenomatous Polyposis Coli<br />

(APC) gene, a tumor suppressor gene, controls colonocyte epithelial proliferation.. Mutation<br />

in the APC gene is also implicated in cell migration, cell-cell adhesion, chromosomal<br />

stability, cell cycle control <strong>and</strong> apoptosis. The APC gene is a suitable marker for colorectal<br />

cancer detection because APC gene mutation is an initiating event in colorectal tumorigenesis<br />

for both sporadic <strong>and</strong> hereditary colorectal cancers.Germline mutations are scattered<br />

throughout the 5’half of the APC gene while somatic mutations are found in a Mutation<br />

Cluster Region (MCR) in exon 15 that spans codons 1250 to 1500 with hotspots at codons<br />

1309 <strong>and</strong> 1450. A pair of primers encompassing the MCR of the APC gene was designed<br />

using OLIGO software <strong>and</strong> the Polymerase Chain Reaction (PCR) mix <strong>and</strong> conditions were<br />

determined using DNAsis software. After obtaining informed consent from 109 patients<br />

clinically diagnosed with sporadic colorectal cancer with no prior treatment at the Institute of<br />

Digestive Diseases of the St. Luke’s Medical Center (SLMC) included in the study,<br />

Deoxyribonucleic acid (DNA was extracted from a portion of the surgical tissue samples <strong>and</strong><br />

the MCR of the APC gene was amplified using U3612 <strong>and</strong> L4500 <strong>and</strong> the PCR mix <strong>and</strong><br />

conditions determined earlier. The obtained PCR product was run in 1.5% agarose gel<br />

yielding positive results.This new protocol offers an effective way of amplifying the MCR of<br />

the APC gene. The advantages of this procedure includes lower cost, reduced h<strong>and</strong>s-on time,<br />

greater simplicity <strong>and</strong> reliability that is suitable for research <strong>and</strong> clinical management.<br />

- 658 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 52<br />

N-(4-hydroxyphenyl)retinamide interferes with functionally active IGF-1-mediated<br />

survival pathways in human retinoblastoma cells.<br />

Roberta Venè1, Giuseppe Arena1, Douglas M. Noonan2, Adriana Albini,1 <strong>and</strong><br />

Francesca Tosetti1<br />

1Experimental Oncology A Laboratory, National Cancer Research Institute (IST), L.go<br />

R. Benzi 10, 16132 Genova, Italy. 2Università degli Studi dell'Insubria, Varese, Italy. Email:<br />

francesca.tosetti@istge.it , roberta.vene@istge.it<br />

The cancer chemopreventive retinoid N-(4-hydroxyphenyl)retinamide (4HPR) kills Y79<br />

retinoblastoma cells through a complex cell death pathway involving ROS elevation,<br />

lysosomal damage <strong>and</strong> irreversible mitochondrial dysfunction. The Insulin-like growth factor<br />

I (IGF-I) sustains the survival of tumor cells by different mechanisms including protection<br />

from cell death <strong>and</strong> resistance to chemotherapeutic agents. Since IGF-I is also a survival<br />

factor in the human retina, we asked whether IGF-I was able to protect Y79 cells against<br />

4HPR cytotoxicity. IGF-I increased Y79 cell proliferation, in contrast, IGF-I could not<br />

prevent 4HPR-induced DNA fragmentation, loss of MTT reduction, LDH release, <strong>and</strong> late<br />

ATP depletion. IGF-I induced a rapid phosphorylation of AKT at Ser473 <strong>and</strong> mTOR at<br />

Ser2448 that were sustained over time. Phosphorylation of AKT, but not of mTOR , was<br />

almost completely suppressed by 4HPR at 24h. To confirm AKT inactivation by 4HPR, we<br />

considered GSK3beta, a downstream substrate of AKT that is inactivated by phosphorylation<br />

at Ser9 <strong>and</strong> is involved in the survival response induced by a number of growth factors <strong>and</strong><br />

chemical inhibitors of mitochondrial membrane depolarization. Surprisingly, we found that<br />

not only 4HPR sustains IGF-I-induced GSK3beta phosphorylation at Ser9, but that 4HPR<br />

itself is able to induce GSK3beta phosphorylation. The antioxidant NAC reversed this effect,<br />

suggesting the involvement of a ROS-mediated mechanism in stimulation of GSK3 beta<br />

phosphorylation by 4HPR.<br />

- 659 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 53<br />

HSP90 INHIBITION BY GELDANAMYCIN UPREGULATES EXPRESSION OF<br />

HEAT SHOCK PROTEINS IN PC-12 CELLS.<br />

Christina B. Wieg<strong>and</strong> 1, Madhavi J. Rane3, Leroy R. Sachleben Jr 2, Rachel Wu 3 <strong>and</strong><br />

Evelyne Gozal 1,2<br />

Depts of Pharm. & Tox.1, Pediatrics2, Medicine3, University of Louisville, Louisville,<br />

KY<br />

Heat shock proteins (Hsps) play a role in cellular injury, proteins repair <strong>and</strong> ubiquitination, or<br />

induction of apoptosis when cells are beyond repair. Hsp90 regulates the function of its client<br />

proteins <strong>and</strong> can bind Akt kinase or heat shock factor (HSF). Upon cellular stress HSF is<br />

released to the nucleus to induce Hsps transcription. Geldanamycin (GA) binds Hsp90 ATPbinding<br />

site, inhibiting its ATPase function <strong>and</strong> disrupting protein associations. We<br />

previously showed that 6h sustained hypoxia (SH; 0.1% O2) increased Akt-Hsp90 binding<br />

<strong>and</strong> Akt phosphorylation in PC-12 cells, without altering cell survival. However, 24h SH<br />

decreased Akt phosphorylation, coinciding with the onset of cell death. GA induced cell death<br />

<strong>and</strong> decreased Akt expression in a dose dependent manner, with higher toxicity in normoxia<br />

(RA; 21% O2) than in SH, <strong>and</strong> disrupted Hsp90-Akt association without affecting SHinduced<br />

Akt phosphorylation. Because of Hsps dual function in cellular repair <strong>and</strong> apoptosis,<br />

we hypothesized that GA may affect cell survival by disrupting Hsp90 binding with<br />

additional Hsps or with HSF. To determine whether GA modulates Hsps expression, GAtreated<br />

cells were exposed to 6h, 12h, <strong>and</strong> 24h RA or SH. GA increased Hsp25, Hsp70 <strong>and</strong><br />

Hsp90 expression independently from FiO2, while Akt/Hsp90 binding was differentially<br />

altered in SH <strong>and</strong> RA. Thus, increased Hsps expression observed in GA-treated PC-12 cells<br />

may result from the dissociation of HSF from Hsp90 <strong>and</strong> its subsequent translocation<br />

independently from the hypoxic stress. Increased Hsps expression may initially protect cells<br />

from cytotoxic effects of low GA concentrations, as suggested by GA dose-dependent effect<br />

on cell survival. However, longer GA exposures may increase cell death resulting from<br />

accumulating damaged proteins or SH-related energy depletion. We are currently<br />

investigating changes in HSF association with the Hsp90 complex in SH <strong>and</strong> RA GA-treated<br />

cells. These findings suggest that disruption of Hsp90 chaperone functions by GA leads to<br />

inactivation of survival pathways, alterations in the protein associations of Hsp90 complexes,<br />

<strong>and</strong> induction of stress proteins targeting cells to apoptosis. NIH F30-NS051998-01, HL-<br />

074296, 2 P20 RR15576-06, <strong>and</strong> AHA 0335278N.<br />

- 660 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 54<br />

The effect of UV-irradiation on telomerase activity <strong>and</strong> other stress-related proteins in<br />

lens epithelial cells<br />

WU Zhi-hong,(!"!) ZHANG Jin-shong#"#$%<br />

Department of Ophthalmology, the Forth Affiliated Hospital, China Medical University.<br />

Shenyang 110001, China<br />

Corresponding author: WU Zhi-hong<br />

Email: fswuzhihong@sina.com&bjwuzhihong@yahoo.com<br />

Background To investigate the change <strong>and</strong> role of telomerase activity <strong>and</strong> other stressrelated<br />

proteins UV-induced DNA damage <strong>and</strong> repair in lens epithelial cells. Methods Human<br />

lens epithelial cells were irradiated at UV-doses 0.0(control group) <strong>and</strong><br />

0.5'1.5'2.5'3.5'5.0'7.5'10.0 mJ/cm2(treated 1~7group). telomerase activity was<br />

determined by Telomerase Repeat Amplification Protocol-Enzyme Linked Immunosorbent<br />

Assay#TRAP-ELISA%(P53'P21'growth arrest <strong>and</strong> DNA damage inducible<br />

(GADD45)'proliferating cell nuclear antigen(PCNA) <strong>and</strong> P16 protein levels were analyzed<br />

by Western blotting.<br />

Results Telomerase activity in control group <strong>and</strong> treated 1~7group showed increased<br />

tendency, compairing in 8 groups, there are significant difference (F=45.65,P


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 55<br />

PI3 kinase inhibition provides a novel mechanism for the action of valproic acid<br />

Xuehua Xu1, Helmut Kae2, Annette Müller-Taubenberger3, Kathryn Adley4, Nadine<br />

Pawolleck5, Vivian Lee6, Talvinder Sihra6, Claudia Wiedemann7, Gerry Weeks2, Jan<br />

Faix8, Markus Maniak5, Tian Jin1 <strong>and</strong> Robin S.B. Williams4<br />

1. Chemotaxis Signal Sect, NIAID, Rockville, MD20852, USA. TJIN@niaid.nih.gov.<br />

2. Microbiol. Immunol. UBC, Vancouver, V6T1Z3, Canada. weeks@ciml.univ-mrs.fr.<br />

3. M-Plank Biochem, LRZ, Munich, 80336, Germany. amueller@lrz.uni-muenchen.de.<br />

4. Dept Biol <strong>and</strong> WIBR, UCL, London, WC1E 6BT, UK. robin.williams@ucl.ac.uk.<br />

5. Inst Cell Bio, University of Kasse, Kassel, 34132, Germany. maniak@uni-kassel.de.<br />

6. Dept Pharmacol., UCL, London, WC1E6BT, UK. t.sihra@ucl.ac.uk.<br />

7. Mol Cell Bio, UCL Med. School, NW32PF, UK. c.wiedemann@medsch.ucl.ac.uk.<br />

8. Hanover Med School, Hanover, D-30623, Germany. hans@imap.bpc.mh-hannover.de.<br />

Valproic acid (VPA, 2 propyl pentanoic acid) is a short chain fatty acid that was accidentally<br />

found to be an effective treatment for epilepsy in 1962, <strong>and</strong> is now also used to treat bipolar<br />

disorder <strong>and</strong> migraine, <strong>and</strong> is undergoing trials for cancer therapy. Despite its wide use, the<br />

therapeutic targets of VPA are not clear. We have identified an effect of VPA in blocking<br />

chemotaxis, a process controlled by phosphoinositide 3-kinase (PI3K) signalling, using the<br />

social amoeba Dictyostelium discoideum. In this model, we show that VPA attenuates PI3K<br />

activity by inhibiting signal-induced PIP3 production, that both VPA <strong>and</strong> a PI3K inhibitor<br />

reverse the phenotype of activated Rac1A, reduce Ras activation <strong>and</strong> VPA also inhibits<br />

vesicle trafficking. These results suggest that VPA functions through the reduction of PIP3<br />

production to modulate downstream effectors including small GTPases <strong>and</strong> vesicle release.<br />

To examine the therapeutic potential of this effect, we show that the human PI3Kgamma<br />

enzyme is weakly but directly inhibited by VPA <strong>and</strong> that both VPA <strong>and</strong> a PI3K inhibitor<br />

suppress depolarization-dependent neurotransmitter release in purified rat nerve terminals.<br />

These results thus suggest that VPA may function to reduce PI3K activity as a therapeutic<br />

mechanism for the treatment of epilepsy, bipolar disorder, migraine, <strong>and</strong> cancer.<br />

- 662 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 56<br />

Determination of oxidative stress status <strong>and</strong> concentration of TGF-B1 in the blood <strong>and</strong><br />

saliva of osteoporotic patients<br />

1Gholamreza Yousefzadeh, 1Bagher Larijani, 2Azadeh Mohammadirad, 1Ramin<br />

Heshmat, 2Roja Rahimi, <strong>and</strong> 2Mohammad Abdollahi<br />

1Endocrinology <strong>and</strong> Metabolism Research Centre, <strong>and</strong> Pharmaceutical Sciences<br />

Research Center, Tehran University of Medical Sciences, Tehran, Iran<br />

Email: mohammad.abdollahi@utoronto.ca<br />

Introduction: The maintenance of bone mass is influenced by genetic, race, hormonal,<br />

mechanical <strong>and</strong> nutritional factors which modulate the local <strong>and</strong> systemic mechanisms<br />

regulating bone turnover. It seems that oxidative stress <strong>and</strong> interleukins particularly TGF-#1<br />

also influences bone mass.<br />

Objectives: to determine if oxidative stress <strong>and</strong> TGF-#1 have any role in the bone mass<br />

density.<br />

Methods: Blood <strong>and</strong> saliva samples of twenty two osteoporotic patients were collected.<br />

TBARS <strong>and</strong> FRAP assay respectively were used to determine levels of lipid peroxidation <strong>and</strong><br />

antioxidant status in these samples. The concentration of TGF-#1 antigen was measured by<br />

using the capture s<strong>and</strong>wich ELISA.<br />

Results: Total antioxidant power in blood <strong>and</strong> saliva of osteoporotic patients was lower<br />

(p


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 57<br />

Erufosine: a membrane targeting antineoplastic agent with signal transduction<br />

modulating effects<br />

Maya M. Zaharieva1,2, Spiro M. Konstantinov1,2, Martin R. Berger2<br />

1Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str, 1000 Sofia, Bulgaria<br />

2German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg,<br />

Germany<br />

The ether lipid analogue erufosine (erucylphospho-N,N,N,-trimethyl-propylammonium,<br />

ErPC3) has high activity against leukaemic cells without affecting the normal hematopoiesis.<br />

It belongs to the group of alkylphosphocholines that are inhibitors of protein kinase C <strong>and</strong> of<br />

phospholipase C. However, the mechanism of action of erufosine remains rather unclear. We<br />

focused on combination effects with the tyrosine kinase inhibitor imatinib mesylate (gleevec,<br />

former STI-571 or CGP-57148) against chronic myeloid leukemia (CML) derived cell lines.<br />

The influence of erufosine on proteins involved in the phosphatidylinositol-3-phosphate<br />

pathway <strong>and</strong> on the expression of the retinoblastoma protein Rb as key component for the cell<br />

cycle entry <strong>and</strong> progression in mammalian cells was studied. A panel of three cell lines:<br />

SKW-3 (chronic T-cell leukemia), BV-173 <strong>and</strong> K-562 (CML) were treated with erufosine (at<br />

concentrations ranging from 2 to 50 mcM). Whole cell lysates were prepared for Western<br />

Blot analysis. The combination effect of erufosine with imatinib on BV-173 <strong>and</strong> K-562 was<br />

estimated after the treatment using MTT-dye reduction assay. The consecutive treatment of<br />

K-562 <strong>and</strong> BV-173 cells with erufosine (2.5, 5, 15, 30 mcM) <strong>and</strong> imatinib (0.05, 0.1 mcM)<br />

led to synergism <strong>and</strong> therefore such combinations could be beneficial for relapsed patients<br />

with drug resistant disease. Erufosine caused decrease of pAkt <strong>and</strong> BCR-ABL protein<br />

expression, while it induced the Rb in K-562 cells. These alterations in the signal<br />

transduction could be an explanation for the drug interaction found. Furthermore, Rb is a<br />

substrate of caspases <strong>and</strong> it is cleaved during apoptosis. Such Rb degradation was observed in<br />

SKW-3 <strong>and</strong> BV-173 cells after incubation with erufosine for 14 h. Our experimental findings<br />

suggest that erufosine acts through induction of changes in protein <strong>signaling</strong> <strong>and</strong> especially<br />

through Rb induction. This unique mode of action makes it an attractive partner for<br />

combination therapies, e.g. with imatinib-based therapy for CML.<br />

- 664 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 58<br />

Impact of Rb knock down on the modulation erufosine signal transduction in human<br />

leukemia cells<br />

Maya M. Zaharieva1,2, Milen Kirilov2, Minqiang Chai2, Stefan Berger3, Spiro M.<br />

Konstantinov1,2, Martin R. Berger2<br />

1Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str, 1000 Sofia, Bulgaria<br />

2German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg,<br />

Germany<br />

3Central Institute for Mental Help, Department of Molecular Biology, J5, 68 159<br />

Mannheim<br />

The retinoblastoma gene Rb1 is a prototypical tum<strong>our</strong> supressor. Consistent with its tom<strong>our</strong>supressor<br />

function Rb is known to inhibit proliferation. Recent studies indicate its role in<br />

suppressing apoptosis, too. Loss of Rb sensitizes cells to apoptosis, but loss of Rb can only<br />

contribute to tumor development under conditions in which apoptosis response is already<br />

compromised. The new alkylphosphocholine erufosine is an inductor of Rb in leukemic cells.<br />

Therefore, we decided to suppress the expression of RB using conventional phosphorotioate<br />

antisense oligonucleotides <strong>and</strong> siRNAs. Unfortunately, both types of oligonucleotides used<br />

didn’t cause stable <strong>and</strong> reproducible knock-down <strong>and</strong> we initiated viral transfection<br />

experiments for stable Rb knock-down in SKW-3, BV-173 <strong>and</strong> K-562 cells. The antisense<br />

sequence was firstly cloned in the pSUPER vector <strong>and</strong> tested on HEK 293T cells. For<br />

normalizing the transfection efficiency an EGFP-expressing vector was used in addition. To<br />

realize the transfer of the H1-shRNA cassette directed to the Rb-mRNA, the cells were<br />

infected with LentiLox virus which co-express an EGFP-protein. The resulting levels of target<br />

mRNA were measured by real-time RT-PCR after excluding of non-fluorescent cells by flow<br />

cytometry <strong>and</strong> cell sorting. The highest decrease of the Rb-mRNA (about 86%) was found in<br />

SKW-3 cells. A concomitent knock-down at the protein level was evidenced by Western<br />

blotting. The proliferation activity of the infected cells was estimated by colorimetric MTTdye<br />

reduction assay before <strong>and</strong> after erufosine treatment. Comparison of Rb knock-down <strong>and</strong><br />

wild type leukemic cells revealed the presence of significant differences in cell cycle duration<br />

<strong>and</strong> sensitivity to erufosine, thus confirming the impact of Rb modulation for the mode of<br />

action of alkylphosphocholines which are known as promising antineoplastic signal<br />

transduction modulators.<br />

- 665 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 59<br />

Labile zinc <strong>and</strong> zinc transporter-4 (ZnT4) expression in mast cells <strong>and</strong> airway<br />

epithelium<br />

1Zalewski, P., 1Grosser, D., 2Murgia, C., 1Lang, C., 1Ho, L., 1Truong-Tran, A.,<br />

3Danscher, G., 3Stoltenberg, M <strong>and</strong> 1Ruffin, R.<br />

1Department of Medicine, University of Adelaide, The Queen Elizabeth Hospital,<br />

Woodville, South Australia 5011. 2Istituto Nazionale Ricerca per gli Alimenti e la<br />

Nutrizione, 00178, Rome, Italy. 3Department of Anatomy, University of Aarhus,<br />

Denmark.<br />

In addition to tightly-bound Zn(II) in metalloenzymes, zinc fingers <strong>and</strong> other metalloproteins,<br />

cells contain more dynamic, exchangeable pools of labile Zn(II). Labile Zn(II) is involved in<br />

processes as diverse as mast cell secretion, caspase-regulation <strong>and</strong> neurotransmission. Our<br />

previous studies, using the Zn(II)-specific fluorophore Zinquin, have shown localization of<br />

abundant pools of labile Zn(II) in secretory granules of mast cells <strong>and</strong> islet cells <strong>and</strong> in apical<br />

vesicles of human <strong>and</strong> animal airway epithelium (AE) [1-3]. Using autometallography to<br />

detect labile Zn(II) by electron microscopy, we have demonstrated the presence of large,<br />

vesicular-like aggregates of Zn(II) in the apical cytoplasm of rat AE. How Zn is incorporated<br />

into these organelles is unclear, but a role for specific intracellular Zn(II) transporters appears<br />

likely. Here we show abundant expression (mRNA <strong>and</strong> protein) for the vesicular Zn<br />

transporter ZnT4 in human <strong>and</strong> rodent mast cells <strong>and</strong> AE. Immunofluorescence staining for<br />

ZnT4 in AE was strikingly similar to that of Zinquin fluorescence of Zn(II), both showing<br />

vesicular <strong>and</strong> apical cytoplasmic distributions. In addition, some of the AE cells had a rim of<br />

fluorescence at the basolateral membrane <strong>and</strong> occasionally around the entire plasma<br />

membrane, suggesting a role for this Zn(II) transporter in membrane uptake of Zn(II), as well<br />

as redistribution to the apical vesicles. Since apical AE Zn(II) is markedly decreased in a<br />

murine model of allergic airway inflammation, we studied the effects of airway inflammation<br />

on expression of ZnT4. Balb/c mice were sensitized to ovalbumin (OVA) <strong>and</strong> challenged with<br />

aerosolized OVA. There was a significant decrease in whole lung ZnT4 mRNA expression<br />

(by Real-time PCR) as well as ZnT4 immunofluorescence in the bronchial epithelium of the<br />

OVA-treated mice when compared to saline-treated controls. We propose that ZnT4 plays an<br />

important role in vesicular/granular uptake of Zn(II) in mast cells <strong>and</strong> AE.<br />

1. Carter JE, Truong-Tran AQ, Grosser D, Ho L, Ruffin RE <strong>and</strong> Zalewski PD. 2002<br />

Involvement of <strong>Redox</strong> events in caspase activation in Zn-depleted airway epithelial cells.<br />

Biochem Biophys Res Commun 279, 1062-70.<br />

2. Zalewski, P. D., Millard, S. H., Forbes, I. J., Kapaniris, O., Slavotinek, A. <strong>and</strong> Betts, W. H.<br />

(1994) Video image analysis of labile zinc in viable pancreatic islet cells using a specific<br />

fluorescent probe for zinc. J. Histochem. Cytochem. 42, 877-884.<br />

3. Ho LH, Ruffin RE, Murgia C, Li L, Krilis SA, Zalewski PD. 2004. Labile zinc <strong>and</strong> zinc<br />

transporter ZnT4 in mast cell granules: Role in regulation of caspase activation <strong>and</strong> NF-kB<br />

translocation J Immunol 172, 7750-60.<br />

- 666 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 60<br />

Animal model of tumor progression: a study of combined therapy to overcome the<br />

multiple drug resistance syndromes of malignant cells<br />

Marina Zenkova, Nadezda Mironova, Olga Shklyaeva, Ekaterina Andreeva, <strong>and</strong><br />

Valentin Vlassov<br />

Institute of Chemical Biology <strong>and</strong> Fundamental Medicine SB RAS<br />

8, Lavrentiev ave., 630090, Novisibirsk, Russian Federation E-mail:<br />

marzen@niboch.nsc.ru<br />

In this work we developed experimental animal model of tumor progression on the base of<br />

mice lymphosarcomas LS <strong>and</strong> RLS. LS tumor displays high sensitivity to cyclophosphamide,<br />

which is widely used in anticancer therapy to initiate apoptosis. RLS was derived from LS by<br />

passaging at low concentration of cyclophosphamide in mice (10-20 mg/kg) <strong>and</strong> characterized<br />

by resistance to high dose of cyclophosphamide (50-150 mg/kg).<br />

The primary cultures (LS <strong>and</strong> RLS)) of these tumors were obtained <strong>and</strong> characterized by<br />

differences in expression of the genes involved in formation of multiple drug resistant<br />

phenotype. We show that RLS tumors are characterized by high level of mdr1b <strong>and</strong> bcl-2<br />

genes expression as compared with LS <strong>and</strong> low level of p53 gene expression. The study of<br />

RLS sensitivity to cytostatics revealed that about 10% of cells display MDR phenotype <strong>and</strong><br />

survive even at high dose of cytostatics. By cultivation of RLS on medium with increasing<br />

vinblastine concentrations cell line RLS40 was obtained. RLS40 exhibited high levels of<br />

mdr1a/1b genes expression as compared to RLS <strong>and</strong> 20-folds increase of resistance to<br />

cytostatics. Drug resistant RLS40 cells were transplanted into CBA mice <strong>and</strong> sensitivity of<br />

formed tumors (solid <strong>and</strong> ascetic) to anticancer drugs was tested. We show that RLS40<br />

tumors were resistant to a number of cytostatics usually used in st<strong>and</strong>ard protocols of<br />

antitumor therapy (cyclophosphamide, cysplatine, adriamicine, rubomicine <strong>and</strong> TNF): these<br />

therapeutics have no effect on tumor growth. Thus, RLS40 tumor can be used as an animal<br />

model of tumors which display high drug resistance <strong>and</strong> poor prognosis of treatment. This<br />

model corresponds to tumor status observed in patients after one or several c<strong>our</strong>ses of<br />

chemotherapy <strong>and</strong> can be used for testing combined conventional therapy <strong>and</strong>/or developing<br />

gene-targeted therapeutics (siRNAs, antisense oligonucleotides, ribozymes) affecting<br />

expression of mdr1a/1b <strong>and</strong> bcl-2 genes.<br />

This work was supported by RAS programs Molecular <strong>and</strong> cellular biology <strong>and</strong> Science to<br />

medicine, FCNTP grant RI-012/001/254.<br />

- 667 -


Session XVII : Cell <strong>signaling</strong> in health <strong>and</strong> disease Poster XVII, 61<br />

Intracrine angiotensin II induces intracellular <strong>signaling</strong> in rabbit kidney convoluted<br />

proximal tubule cells<br />

Jia L. Zhuo, Oscar A. Carretero <strong>and</strong> Xiao C. Li.<br />

Division of Hypertension <strong>and</strong> Vascular Research, Henry Ford Hospital, Detroit, MI<br />

48202, USA. E-mail: jzhuo1@hfhs.org<br />

Although the role of extracellular Ang II <strong>and</strong> the underlying mechanisms mediated by cell<br />

surface AT1 receptors have been well studied in the kidney, whether intracellular Ang II<br />

plays a physiological role in renal proximal tubule cells remains poorly understood. We tested<br />

the hypothesis that extracellular Ang II is taken up by proximal tubule cells through AT1<br />

receptor-mediated endocytosis, <strong>and</strong> that internalized Ang II induces intracellular calcium<br />

mobilization, activates NF!B, <strong>and</strong> increases RNA transcription by stimulating cytoplasmic<br />

<strong>and</strong> nuclear AT1 receptors. When proximal tubule cells were exposed to extracellular Ang II<br />

(1 nM), intracellular Ang II levels were increased by 60% (161 ± 6 vs. 98 ± 5 pg/mg protein;<br />

p < 0.001). The increases in intracellular Ang II were blocked by the endocytotic inhibitors<br />

colchicine (10-6 M) (p < 0.01), phenylarsine oxide (PAO) (10-6 M), which inhibits tyrosine<br />

phosphatase (p < 0.01), <strong>and</strong> the AT1 receptor blocker losartan (10-5 M) (p < 0.01), but not by<br />

the AT2 receptor antagonist PD 123319 (10-5 M). To determine whether intracellular Ang II<br />

has a physiological role, Ang II was microinjected into single proximal tubule cells (1 nM),<br />

while cell surface AT1 receptors were blocked with extracellular losartan (10-5 M).<br />

Intracellular Ang II induced a robust increase in intracellular calcium, which peaked at 15 to<br />

30 s. When Ang II was microinjected together with losartan, the calcium responses were<br />

blocked while concurrent microinjection of Ang II <strong>and</strong> PD 123319 had no effect. Moreover,<br />

blockade of AT1 receptor-mediated Ang II internalization by colchicine or losartan inhibited<br />

Ang II-induced activation of NF!B (p < 0.01). Ang II (1 nM) also significantly increased<br />

RNA transcription in freshly isolated proximal tubule cell nuclei (p < 0.01) <strong>and</strong> the effect was<br />

inhibited by losartan. We conclude that extracellular Ang II is internalized in proximal tubule<br />

cells through AT1 receptor-mediated endocytosis <strong>and</strong> intracellular Ang II may exert<br />

important intracrine effects by activating cytoplasmic <strong>and</strong> nuclear AT1 receptors. This work<br />

is supported by grants from National Institutes of Health (RO1DK067299), American Heart<br />

Association Greater Miwest Affiliate (0355551Z), <strong>and</strong> National Kidney Foundation of<br />

Michigan.<br />

- 668 -


Notes<br />

- 669 -


Notes<br />

Notes<br />

- 670 -


- 671 -


List of participants<br />

(by alphabetical order)<br />

- 672 -


- 673 -


Pr. Mohammad Abdollahi<br />

14155-6451 Tehran IRN<br />

Email: mohammad@tums.ac.ir<br />

Mr. Indra Aerts<br />

Cellular Biochemistry<br />

2610 Antwerp BEL<br />

Email: indra.aerts@ua.ac.be<br />

Mrs. Pia Agren<br />

6228 CV Maastricht Maastricht<br />

NLD<br />

Email:<br />

pia.agren@farmaco.unimaas.nl<br />

Dr. Maria Cristina Albertini<br />

61029 Urbino ITA<br />

Email: cemetbio@uniurb.it<br />

Pr. Dario Alessi<br />

MRC Protein Phosphorylation<br />

Unit<br />

DD1 5EH Dundee GBR<br />

Email: d.r.alessi@dundee.ac.uk<br />

Dr. Alex<strong>and</strong>ra Andreeva<br />

Department of Pharmacology<br />

60612 Chicago, IL USA<br />

Email: a<strong>and</strong>reev@uic.edu<br />

Mrs. Adrienn Angyal<br />

Dept. of Immunology<br />

1117 Budapest HUN<br />

Email: sarmayg@cerberus.elte.hu<br />

Dr. Josef Abel<br />

Toxicology<br />

40225 Düsseldorf DEU<br />

Email: josef.abel@uniduesseldorf.de<br />

Pr. Bharat Aggarwal<br />

Cytokine Research<br />

TX 77030 Houston USA<br />

Email: aggarwal@md<strong>and</strong>erson.org<br />

Pr. Hee Jung Ahn<br />

Pathology<br />

463-712 Sungnam KOR<br />

Email: hjahn@cha.ac.kr<br />

Dr. Veronica Albertini<br />

Laboratory of Experimental<br />

Oncology<br />

6500 Bellinzona CHE<br />

Email:<br />

veronica.albertini@irb.unisi.ch<br />

Mrs. Asude Alpman<br />

Ege University Medical Faculty<br />

Medical Genetics<br />

35100 Izmir TUR<br />

Email: asudealpman@gmail.com<br />

Dr. Allison-Lynn Andrews<br />

Brroke Laboratory<br />

SO16 6YD Southampton GBR<br />

Email: ala@soton.ac.uk<br />

Mr. Sébastien ANTHERIEU<br />

Equipe de Toxicologie Cellulaire<br />

et Moléculaire et Génomique<br />

6903 Sophia-Antipolis FRA<br />

Email: antherieu@antibes.inra.fr<br />

Mr. ERIC ADRIAENSSENS<br />

INSERM ERI-8<br />

59655 Villeneuve d'Ascq FRA<br />

Email: eric.adriaenssens@univlille1.fr<br />

Dr. Ioanna-Katerina Aggeli<br />

Animal & Human Physiology<br />

157 84 Athens GRC<br />

Email: iageli@hotmail.com<br />

Mrs. Cano Ainara<br />

Biochemistry<br />

48940 Leioa ESP<br />

Email: ainaroco@hotmail.com<br />

Dr. Alise Alesenko<br />

Molecular mechanisms of cell<br />

proliferation<br />

154 Moscow RUS<br />

Email: ales@sky.chph.ras.ru<br />

Mr. Joana Amaral<br />

1600-083 Lisbon PRT<br />

Email: jamaral@ff.ul.pt<br />

Dr. Peter Angel<br />

Division Signal Transduction <strong>and</strong><br />

Growth Control<br />

69120 Heidelberg DEU<br />

Email: p.angel@dkfz.de<br />

Dr. Frank Antonicelli<br />

Biochemistry-Dermatology<br />

51095 Reims FRA<br />

Email: frank.antonicelli@univreims.fr


Dr. Nassera Aouali<br />

L-1526 Luxemb<strong>our</strong>g LUX<br />

Email: nassera.aouali@crpsante.lu<br />

Mr. Thierry Arnould<br />

URBC<br />

5000 Namur BEL<br />

Email:<br />

thierry.arnould@fundp.ac.be<br />

Dr. Beatrice Bachmeier<br />

Inst. of Pathology<br />

D-81925 Munich DEU<br />

Email: bachmeier@med.unimuenchen.de<br />

Dr. Marek Baltaziak<br />

Department of Pathology<br />

15-269 Bialystok POL<br />

Email: sulek@zeus.amb.edu.pl<br />

Dr. Joana Barbosa Melo<br />

Institute Medical Biology <strong>and</strong><br />

Center for Neurosciences, Faculty<br />

of Medicine, University of<br />

Coimbra<br />

3004-504 Coimbra PRT<br />

Email: jbmelo@cnc.cj.uc.pt<br />

Mrs. Montserrat Barragan<br />

Unitat de senyalitzacio cel!lular<br />

E-08003 Barcelona ESP<br />

Email: montse.barragan@upf.edu<br />

Dr. Christa Baumstark-Khan<br />

Cellular Biodiagnostics,<br />

Department of Radiobiology,<br />

Institute of Aerospace Medicine<br />

51170 Köln DEU<br />

Email: christa.baumstarkkhan@dlr.de<br />

Dr. Jose Aramburu<br />

Department of Experimental <strong>and</strong><br />

Health Sciences<br />

8003 Barcelona ESP<br />

Email: jaramburu@imim.es<br />

Pr. Ami Aronheim<br />

31096 Haifa ISR<br />

Email: aronheim@tx.technion.ac.il<br />

Mrs. Hye Yeon BAEK<br />

Department of Pharmacology<br />

120-752 Seoul KOR<br />

Email: ehrehrwh@hanmail.net<br />

Pr. Vladimir Bantseev<br />

School of Optometry/Biology<br />

N2L 3G1 Waterloo CAN<br />

Email: vbantsee@uwaterloo.ca<br />

Mr. Fawzia Bardag-gorce<br />

90502 Torrance USA<br />

Email: fgorce@labiomed.org<br />

Dr. Giuseppina Basini<br />

Dipartimento di Produzioni<br />

Animali-Sezione di Fisiologia<br />

43100 Parma ITA<br />

Email: basini@unipr.it<br />

Mrs. Ilse Beck<br />

LEGEST<br />

B-9000 Gent BEL<br />

Email: IlseME.Beck@Ugent.be<br />

Dr. Vivienne Armbruester<br />

Department of Medicine<br />

10029 New York City USA<br />

Email:<br />

vivienne.armbruester@mssm.edu<br />

Dr. Sigridur Asgeirsdottir<br />

Dept Pathology & Laboratory<br />

Medicine, Medical Biology<br />

Section<br />

9713 GZ Groningen NLD<br />

Email:<br />

s.asgeirsdottir@med.umcg.nl<br />

Mrs. Denyse BAGREL<br />

Laboratoire d'Ingénierie<br />

Moléculaire et Biochimie<br />

Pharmacologique<br />

57070 Metz FRA<br />

Email: bagrel@univ-metz.fr<br />

Dr. Federica Barbieri<br />

pharmacology-department of<br />

oncology biology <strong>and</strong> genetics<br />

16132 Genova ITA<br />

Email: federica.barbieri@unige.it<br />

Mrs. Elena Bardina<br />

Molecular Genetics<br />

6229 ER Maastricht NLD<br />

Email: e.bardina@gen.unimaas.nl<br />

Mr. Eric BATTAGLIA<br />

LIMBP<br />

57070 Metz FRA<br />

Email: battaglia@univ-metz.fr<br />

Mrs. Siv Lise Bedringaas<br />

Hematology section<br />

5021 BERGEN NOR<br />

Email: siv.bedringaas@med.uib.no


Dr. Jeffrey Beekman<br />

Dept. of Immunology<br />

3584 EA Utrecht NLD<br />

Email: j.beekman@azu.nl<br />

Dr. Isidoros Beis<br />

Animal & Human Physiology<br />

157 84 Athens GRC<br />

Email: ibeis@biol.uoa.gr<br />

Dr. Antonino Belfiore<br />

Clinical <strong>and</strong> Experimental<br />

Department<br />

88100 Catanzaro ITA<br />

Email: belfiore@unicz.it<br />

Dr. Yinon Ben-Neriah<br />

Lautenberg Center for<br />

Immunology<br />

91120 Jerusalem ISR<br />

Email: yinonbn@yahoo.com<br />

Pr. Hans-Gert Bernstein<br />

Experimental Psychiatry<br />

D-39120 Magdeburg DEU<br />

Email: Hans-<br />

Gert.Bernstein@medizin.unimagdeburg.de<br />

Mr. Alex<strong>and</strong>re Berthaut<br />

75006 Paris FRA<br />

Email: alexberthaut@aol.com<br />

Mrs. Françoise Besançon<br />

U685 INSERM<br />

75010 Paris FRA<br />

Email:<br />

Francoise.Besancon@stlouis.inser<br />

m.fr<br />

Pr. Iris Behrmann<br />

Laboratoire de Biologie et<br />

Physiologie Intégrée<br />

1511 Luxemb<strong>our</strong>g LUX<br />

Email: iris.behrmann@uni.lu<br />

Mr. Lijs Beke<br />

department of biochemistry<br />

3000 Leuven BEL<br />

Email:<br />

Lijs.Beke@med.kuleuven.be<br />

Pr. Jean Louis Beneytout<br />

Biochimie<br />

87025 Limoges FRA<br />

Email: beneytout@unilim.fr<br />

Mr. Samir Benosman<br />

INSERM U692<br />

67000 Strasb<strong>our</strong>g FRA<br />

Email:<br />

samirbenosman@gmail.com<br />

Mrs. Saoussen BERRAHMOUNE<br />

Equipe Projet "Photobiologie en<br />

Cancérologie"<br />

54511 V<strong>and</strong>oeuvre les Nancy FRA<br />

Email:<br />

s.berrahmoune@nancy.fnclcc.fr<br />

Dr. Carlo Bertoni-Freddari<br />

Neurobiology of Aging Laboratory<br />

60121 Ancona ITA<br />

Email: c.bertoni@inrca.it<br />

Dr. CHARLES BESTWICK<br />

MOLECULAR NUTRITION<br />

AB21 9SB ABERDEEN GBR<br />

Email: V.Smith@rowett.ac.uk<br />

Mrs. Adeline BEILLEROT<br />

LIMBP<br />

57070 Metz FRA<br />

Email: beillerotadeline@yahoo.fr<br />

Mrs. Blanco Belen<br />

Janssen Cilag<br />

28042 Madrid ESP<br />

Email: amex.c.viajes@aexp.com<br />

Dr. Eyal Bengal<br />

Department of Biochemistry<br />

31096 Haifa ISR<br />

Email: bengal@tx.technion.ac.il<br />

Mrs. Sarah Berndt<br />

Laboratoire de Biologie des<br />

Tumeurs et du Développement<br />

4000 Liége BEL<br />

Email: sarah.berndt@ulg.ac.be<br />

Dr. Holger Bertelsmann<br />

14109 Berlin DEU<br />

Email: bertelsmann@hmi.de<br />

Mrs. Iris Bertram<br />

Exp. Psychiatry Lab.<br />

39120 Magdeburg DEU<br />

Email: iris.bertram@gmx.de<br />

Mr. Arnaud Bianchi<br />

UMR 7561 LPPA<br />

54505 V<strong>and</strong>oeuvre FRA<br />

Email:<br />

arnaud.bianchi@medecine.uhpnancy.fr


Dr. Giordano Bianchi<br />

Clinic of internal medicine I<br />

16132 Genoa ITA<br />

Email:<br />

giordano.bianchi@katamail.com<br />

Pr. Jan C. Biro<br />

Informatics<br />

94105 San Francisco USA<br />

Email: jan.biro@sbcglobal.net<br />

Dr. Romain Blasius<br />

LBMCC<br />

L-2540 Luxemb<strong>our</strong>g LUX<br />

Email: romain.blasius@lbmcc.lu<br />

Pr. Mathieu Bollen<br />

Cell <strong>signaling</strong> laboratory, Division<br />

of Biochemistry, Department of<br />

Molecular Cell Biology<br />

3000 Leuven BEL<br />

Email:<br />

Mathieu.Bollen@med.kuleuven.be<br />

Dr. Emilie Boncoeur<br />

Insem U719<br />

75012 Paris FRA<br />

Email: emilie.boncoeur@stantoine.inserm.fr<br />

Mrs. Meike Boosen<br />

Institute of Genetics<br />

53117 Bonn DEU<br />

Email: meike.boosen@unibonn.de<br />

Mr. Pedro Borralho<br />

1600-083 Lisbon PRT<br />

Email: borralho@ff.ul.pt<br />

Dr. Hendrik Bielau<br />

Department of Psychiatry<br />

39120 Magdeburg DEU<br />

Email:<br />

Hendrik.Bielau@Medizin.Uni-<br />

Magdeburg.de<br />

Dr. Konstantin Birukov<br />

IL 60637 Chicago USA<br />

Email:<br />

kbirukov@medicine.bsd.uchicago.<br />

edu<br />

Mrs. Rikke Bogebo<br />

2600 Glostrup DNK<br />

Email: rb@kvl.dk<br />

Dr. Lina BOLOTINE<br />

Equipe Projet "Photobiologie en<br />

Cancérologie"<br />

54511 V<strong>and</strong>oeuvre les Nancy FRA<br />

Email: l.bolotine@nancy.fnclcc.fr<br />

Mrs. SARA BONDIONI<br />

ISTITUTO DI SCIENZE<br />

ENDOCRINE<br />

<strong>2012</strong>2 MILANO ITA<br />

Email: sara.bondioni@unimi.it<br />

Dr. Jean-Paul Borg<br />

Molecular Pharmacology<br />

13009 Marseille FRA<br />

Email: borg@marseille.inserm.fr<br />

Mrs. Veronika Borutinskaite<br />

Division of Medical Microbiology<br />

SE-581 85 Linkoping SWE<br />

Email: verbu@imk.liu.se<br />

Dr. Walter Birchmeier<br />

13125 Berlin DEU<br />

Email: wbirch@mdc-berlin.de<br />

Dr. Anna Birukova<br />

IL 60637 Chicago USA<br />

Email:<br />

abirukov@medicine.bsd.uchicago.<br />

edu<br />

Dr. Maria Bokarewa<br />

Dep of Rheumatology <strong>and</strong><br />

<strong>Inflammation</strong> Research<br />

S-41346 Goteborg SWE<br />

Email:<br />

maria.bokarewa@rheuma.gu.se<br />

Dr. Sophia Bonanou<br />

Biochemistry, School of Medicine<br />

41222 Larissa GRC<br />

Email: sbonanou@med.uth.gr<br />

Mr. Guillaume Bonnot<br />

HP2<br />

38700 La Tronche FRA<br />

Email: Guillaume.Bonnot@e.ujfgrenoble.fr<br />

Dr. Jürgen Borlak<br />

Pharmaforschung und<br />

medizinische Biotechnologie<br />

30625 Hannover DEU<br />

Email: borlak@item.fraunhofer.de<br />

Pr. Johannes L. Bos<br />

Physiological Chemistry<br />

3584 CG Utrecht NLD<br />

Email: m.c.arpesella@med.uu.nl


Dr. Daniela Boselli<br />

Tumor Immunology Laboratory<br />

10126 Turin ITA<br />

Email: daniela.boselli@unito.it<br />

Mrs. Joelle BOTTI<br />

U 504<br />

94807 VILLEJUIF FRA<br />

Email: botti@vjf.inserm.fr<br />

Mrs. Sylvina Bouleau<br />

Laboratoire de génétique et<br />

biologie cellulaire<br />

78035 Versailles FRA<br />

Email: bouleau@genetique.uvsq.fr<br />

Pr. Elisabeth BRAMBILLA<br />

Lung Research Group<br />

38706 LA TRONCHE FRA<br />

Email: EBrambilla@chugrenoble.fr<br />

Mr. Michael Bright<br />

Cancer Cell Biology<br />

W1W 7BS London GBR<br />

Email: michael@ludwig.ucl.ac.uk<br />

Mr. Bernard BRUGG<br />

UMR7102 Neurobiologie des des<br />

Processus Adaptatifs<br />

75005 Paris FRA<br />

Email:<br />

bernard.brugg@snv.jussieu.fr<br />

Mrs. Isabelle Buck<br />

LBMCC<br />

L-2540 Luxemb<strong>our</strong>g LUX<br />

Email: isabelle.buck@lbmcc.lu<br />

Dr. Herbert Bosshart<br />

Innate Immunity Research<br />

Laboratory<br />

CH-8091 Zurich CHE<br />

Email:<br />

herbert.bosshart@puk.zh.ch<br />

Mrs. Sanae BOUALI<br />

Laboratoire de Recherche<br />

54511 V<strong>and</strong>oeuvre les Nancy FRA<br />

Email: s.bouali@nancy.fnclcc.fr<br />

Mrs. Lana Bozulic<br />

Brian Hemmings lab/Friedrich<br />

Miescher Institute<br />

4058 Basel CHE<br />

Email: lana.bozulic@fmi.ch<br />

Mrs. Malene Br<strong>and</strong>t<br />

2600 Glostrup DNK<br />

Email: mabr@kvl.dk<br />

Mr. Christophe Broca<br />

Equipe Avenir INSERM (U661)<br />

34094 Montpellier FRA<br />

Email:<br />

christophe.broca@igf.cnrs.fr<br />

Mr. Justin Brumbaugh<br />

Schultz<br />

69117 Heidelberg DEU<br />

Email: brumbaug@embl.de<br />

Dr. Mir<strong>and</strong>a Buitenhuis<br />

Dept. of Immunology<br />

3584 EA Utrecht NLD<br />

Email: m.buitenhuis@azu.nl<br />

Pr. Serge Bottari<br />

HP2<br />

38700 La Tronche FRA<br />

Email: Serge.Bottari@ujfgrenoble.fr<br />

Mrs. Nadia Bougarne<br />

LEGEST<br />

9000 Ghent BEL<br />

Email: nadia.bougarne@UGent.be<br />

Pr. Marcantonio Bragadin<br />

Dept. Environmental Sciences<br />

30123 Venice ITA<br />

Email: bragadin@unive.it<br />

Mr. Laurent Brault<br />

LIMBP<br />

57070 Metz FRA<br />

Email: brault@univ-metz.fr<br />

Mr. Elisabeth Brouwer<br />

9713 GZ Groningen NLD<br />

Email: E.Brouwer@int.umcg.nl<br />

Pr. Pedro Buc Calderon<br />

Pharmacokinetic, Metabolism,<br />

Nutrition <strong>and</strong> Toxicology<br />

1200 Bruxelles BEL<br />

Email: calderon@pmnt.ucl.ac.be<br />

Mrs. Katarzyna Bukalis<br />

14109 Berlin DEU<br />

Email: k.bukalis@hmi.de


Mrs. Isabel Burghardt<br />

Laboratory of Molecular Neuro-<br />

Oncology, Department of General<br />

Neurology<br />

72076 Tuebingen DEU<br />

Email: isabel.burghardt@gmx.net<br />

Mr. Leonid Bystrykh<br />

Stem Cell<br />

9713 AV Groningen NLD<br />

Email: bystrykh@med.umcg.nl<br />

Mrs. Ying Cai<br />

Diabetes Research Center<br />

1090 Brussels BEL<br />

Email: ying.cai@vub.ac.be<br />

Dr. Serena Cecchetti<br />

Dept. Cell Biology <strong>and</strong><br />

Neurosciences<br />

162 Rome ITA<br />

Email: ramoni@iss.it<br />

Dr. Eva M Cernuda-Morollon<br />

Cancer Cell Biology<br />

W1W 7BS London GBR<br />

Email: eva@ludwig.ucl.ac.uk<br />

Pr. Benjamin Chain<br />

W1T4JF London GBR<br />

Email: b.chain@ucl.ac.uk<br />

Dr. Wen-Hsiung Chan<br />

Department of Bioscience<br />

Technology<br />

32023 Chung-Li TWN<br />

Email: whchan@cycu.edu.tw<br />

Dr. Hauke Busch<br />

Theoretical Bioinformatics<br />

69120 Heidelberg DEU<br />

Email: h.busch@dkfz.de<br />

Dr. Jocelyne Caboche<br />

Signalisation neuronale et<br />

Régulations géniques<br />

75005 Paris FRA<br />

Email:<br />

jocelyne.caboche@svn.jussieu.fr<br />

Mrs. Isabelle CARLAVAN<br />

Research<br />

06902 cedex Sophia-<br />

Antipolis/Valbonne FRA<br />

Email:<br />

isabelle.carlavan@galderma.com<br />

Dr. Claudia Cerella<br />

133 Rome ITA<br />

Email: cerella@uniroma2.it<br />

Mrs. BORAM CHA<br />

department of Pharmacology<br />

120-752 Seoul KOR<br />

Email:<br />

chaboram@yumc.yonsei.ac.kr<br />

Mrs. Florence Chainiaux<br />

URBC<br />

5000 Namur BEL<br />

Email:<br />

florence.chainiaux@fundp.ac.be<br />

Mr. Alain Chariot<br />

Medical Chemistry, CTCM<br />

4000 Liége BEL<br />

Email: alain.chariot@ulg.ac.be<br />

Mrs. Helen Bye<br />

Biochemistry<br />

CB 2 1QW Cambridge GBR<br />

Email: hlc24@mole.bio.cam.ac.uk<br />

Dr. Jo Caers<br />

Laboratory of Hematology &<br />

Immunology<br />

1090 Brussels BEL<br />

Email: jcaers@vub.ac.be<br />

Dr. Anje CAUWELS<br />

Dept. of Molecular Biomedical<br />

Research<br />

B-9052 Zwijnaarde BEL<br />

Email:<br />

Anje.Cauwels@dmbr.UGent.be<br />

Mrs. Maria I. Cerezo-Guisado<br />

Departamento de Bioquimica y<br />

Biologia Molecular y Genética<br />

10071 Caceres ESP<br />

Email: mbcerezo@unex.es<br />

Mrs. Georgia Chachami<br />

Lab. of Biochemistry, School of<br />

Medicine<br />

41222 Larissa GRC<br />

Email: ghah@med.uth.gr<br />

Dr. Rachel Chambers<br />

Centre for Respiratory Research<br />

WC1E 6JJ London GBR<br />

Email: r.chambers@ucl.ac.uk<br />

Mr. Jean-François Charlot<br />

Laboratoire de Biologie Cellulaire<br />

et Moléculaire<br />

25000 BESANCON FRA<br />

Email: jfcharlot@hotmail.com


Mr. Eric CHASTRE<br />

75018 Paris FRA<br />

Email: chastre@bichat.inserm.fr<br />

Mr. Jun-Jie Chen<br />

Deparmemt<br />

10018 Taipei TWN<br />

Email: d91443001@ntu.edu.tw<br />

Dr. Elena Chernolovskaya<br />

630090 Novosibirsk RUS<br />

Email: elena_ch@niboch.nsc.ru<br />

Mrs. Soon Ok CHO<br />

Department of Pharmacology<br />

120-752 Seoul KOR<br />

Email: ehrehrwh@hanmail.net<br />

Pr. Kang-Yell Choi<br />

Molecular Complex Control<br />

120-749 Seoul KOR<br />

Email: kychoi@yonsei.ac.kr<br />

Dr. Tina Chowdhury<br />

Cell <strong>and</strong> Tissue Engineering, Dept<br />

of Engineering<br />

E1 4NS London GBR<br />

Email: t.t.chowdhury@qmul.ac.uk<br />

Dr. Victor Cid<br />

Dpto. Microbiologia II<br />

28040 Madrid ESP<br />

Email: vicjcid@farm.ucm.es<br />

Dr. Ioulia Chatzistamou<br />

Biochemistry<br />

115 27 Athens GRC<br />

Email: ihatzist@med.uoa.gr<br />

Mrs. Miao-Der Chen<br />

Animal Science<br />

912-01 Ping Tung County TWN<br />

Email:<br />

miaoder124@yahoo.com.tw<br />

Dr. Susanna Chiocca<br />

Dept. of Experimental Oncology<br />

20141 Milan ITA<br />

Email: susanna.chiocca@ifom-ieocampus.it<br />

Dr. Claudia Choi<br />

30659 Hannover DEU<br />

Email: Choi.Claudia@MH-<br />

Hannover.de<br />

Mr. Yoon Pyo Choi<br />

Pathology<br />

120-752 Seoul KOR<br />

Email:<br />

yoonpyo@yumc.yonsei.ac.kr<br />

Mrs. Gitte Christensen<br />

2600 Glostrup DNK<br />

Email: glc@dcb-glostrup.dk<br />

Dr. Iwona Ciechomska<br />

Department of Biochemistry<br />

CB2 1QW Cambridge GBR<br />

Email: iac23@mole.bio.cam.ac.uk<br />

Pr. Ching-Chow Chen<br />

10018 Taipei TWN<br />

Email: ccchen@ha.mc.ntu.edu.tw<br />

Dr. Steven Cheng<br />

21746 Ijamsville, MD USA<br />

Email: scheng@cellogenetics.com<br />

Pr. Nam Hoon Cho<br />

Pathology<br />

120-752 Seoul KOR<br />

Email:<br />

cho1988@yumc.yonsei.ac.kr<br />

Mr. Jang Hyun Choi<br />

Signaling Network Lab.<br />

790-784 Pohang KOR<br />

Email: janghyun@postech.ac.kr<br />

Mr. Rasheduzzaman Chowdhury<br />

C. J. Schofield<br />

Ox1 3TA Oxford GBR<br />

Email:<br />

rasheduzzaman.chowdhury@chem<br />

.ox.ac.uk<br />

Dr. Hae-Yun Chung<br />

Department of Pharmacology<br />

120-752 Seoul KOR<br />

Email:<br />

hchung02@yumc.yonsei.ac.kr<br />

Dr. Aleks<strong>and</strong>ra Cierniewska-<br />

Cieslak<br />

92-215 Lodz POL<br />

Email: ola_cier@poczta.onet.pl


Pr. Czeslaw Cierniewski<br />

92-215 Lodz POL<br />

Email: cciern@zdn.am.lodz.pl<br />

Mr. Nicolas CLERE<br />

Laboratoire de Biologie Cellulaire<br />

et Moléculaire<br />

25040 BESANCON FRA<br />

Email: clere.nicolas@wanadoo.fr<br />

Pr. Paul Coffer<br />

Dept. of Immunology<br />

3584 EA Utrecht NLD<br />

Email: p.j.coffer@umcutrecht.nl<br />

Mrs. Anne Conradi<br />

Institute of Genetics<br />

53117 Bonn DEU<br />

Email: anne.conradi@uni-bonn.de<br />

Dr. Eva Corey<br />

GU Cancer Research Laboratory<br />

98008 Seattle USA<br />

Email: ecorey@u.washington.edu<br />

Dr. Evelyne Crapez<br />

Research Cancer Center<br />

34298 Montpellier FRA<br />

Email: ecrapez@valdorel.fnclcc.fr<br />

Mrs. Tania Cruz<br />

Division of Clinical Immunology<br />

<strong>and</strong> Rheumatology<br />

1100 DE Amsterdam NLD<br />

Email: k.a.reedquist@amc.uva.nl<br />

Mrs. Saskia Cillessen<br />

Pathology<br />

1081 HV Amsterdam NLD<br />

Email: sagm.cillessen@vumc.nl<br />

Dr. Daniel Closa<br />

Experimental Pathology<br />

8036 Barcelona ESP<br />

Email: dcabam@iibb.csic.es<br />

Dr. Eleanor Coffey<br />

Neuronal Signalling lab<br />

Fin-20521 Turku FIN<br />

Email: ecoffey@btk.fi<br />

Dr. Laura Conti<br />

Tumor Immunology Laboratory<br />

10126 Turin ITA<br />

Email: laura.conti@unito.it<br />

Dr. Sigrid CORNELIS<br />

Dept. of Molecular Biomedical<br />

Research<br />

B-9052 Zwijnaarde BEL<br />

Email: Sigrid@dmbr.UGent.be<br />

Dr. Pascale Crépieux<br />

Physiologie de la Reproduction et<br />

des Comportements<br />

37380 Nouzilly FRA<br />

Email: crepieux@t<strong>our</strong>s.inra.fr<br />

Dr. Barbara Cserepes<br />

Department of Surgical Research<br />

<strong>and</strong> Techniques<br />

7624 Pécs HUN<br />

Email: barbicska@yahoo.co.uk<br />

Mr. Thomas Clahsen<br />

Institut für Biochemie<br />

52074 Aachen DEU<br />

Email: thomas.clahsen@post.rwthaachen.de<br />

Dr. Laura Cobeno<br />

Dept. Pharmacology<br />

28040 Madrid ESP<br />

Email: acogolludo@ift.csic.es<br />

Dr. Angel Cogolludo<br />

Dept. Pharmacology<br />

28040 Madrid ESP<br />

Email: acogolludo@ift.csic.es<br />

Mrs. Elisabeth-Anne CORCELLE<br />

UMR 65 48<br />

6107 Nice FRA<br />

Email: ecorcell@unice.fr<br />

Dr. Michael C<strong>our</strong>tney<br />

Molecular Signalling Lab<br />

FIN 70211 KUOPIO FIN<br />

Email: c<strong>our</strong>tney@messi.uku.fi<br />

Mrs. Silvia Cristofanon<br />

LBMCC<br />

L-2540 Luxemb<strong>our</strong>g LUX<br />

Email:<br />

silvia.cristofanon@lbmcc.lu<br />

Mr. Alfredo Csibi<br />

HP2<br />

38700 La Tronche FRA<br />

Email: AlfredoCsibi@e.ujfgrenoble.fr


Mrs. Claire Cullmann<br />

69115 Heidelberg DEU<br />

Email: c.cullmann@dkfz.de<br />

Mrs. Lisa Dalla Puppa<br />

14109 Berlin DEU<br />

Email: dallapuppa@hmi.de<br />

Dr. Ben Davidson<br />

Department of Pathology<br />

N-0310 Oslo NOR<br />

Email:<br />

ben.davidson@radiumhospitalet.n<br />

o<br />

Mr. Maite De los Frailes<br />

Screening <strong>and</strong> Compound Profile<br />

28760 Tres Cantos- Madrid ESP<br />

Email:<br />

maite_de_los_frailes@gsk.com<br />

Dr. Maarten de Smit<br />

Biological Chemistry, Leiden<br />

University<br />

2333 CC Leiden NLD<br />

Email:<br />

m.smit@chem.leidenuniv.nl<br />

Mrs. Nicole DEFER<br />

INSERM U.581<br />

94010 Créteil FRA<br />

Email: defer@im3.inserm.fr<br />

Mr. Pim Dekker<br />

Corporate Research<br />

MK44 1LQ Sharnbrook GBR<br />

Email: pim.dekker@unilever.com<br />

Pr. Edgar F. da Cruz e Silva<br />

LaboratÛrio de TransduÁ„o de<br />

Sinais<br />

3810-193 Aveiro Aveiro PRT<br />

Email: dacruze@bio.ua.pt<br />

Mr. Avais DAULAT<br />

75014 Paris FRA<br />

Email: daulat@cochin.inserm.fr<br />

Pr. Roger Davis<br />

1605 Worcester USA<br />

Email:<br />

Roger.Davis@Umassmed.edu<br />

Dr. Milena De Nicola<br />

133 Rome ITA<br />

Email:<br />

milena.de.nicola@uniroma2.it<br />

Mrs. Fabienne DE TONI<br />

INSERM Unit 563<br />

31024 TOULOUSE FRA<br />

Email:<br />

fadetoni@toulouse.inserm.fr<br />

Mrs. Sylvie Defrére<br />

Gynecology<br />

1200 Bruxelles BEL<br />

Email:<br />

sylvie.defrere@gyne.ucl.ac.be<br />

Mrs. Anne-Isabelle Delattre<br />

URBC<br />

5000 Namur BEL<br />

Email: anneisabelle.delattre@fundp.ac.be<br />

Pr. Odete A. B. da Cruz e Silva<br />

LaboratÛrio de NeurociÍncias<br />

3810-193 Aveiro Aveiro PRT<br />

Email: odetecs@bio.ua.pt<br />

Dr. Frank Dautzenberg<br />

CNS Research<br />

2340 Beerse BEL<br />

Email: fdautzen@prdbe.jnj.com<br />

Dr. X<strong>and</strong>er de Haan<br />

Virology<br />

3584 CL Utrecht NLD<br />

Email: x.haan@vet.uu.nl<br />

Mrs. Aurélia De Pauw<br />

URBC<br />

5000 Namur BEL<br />

Email:<br />

aurelia.depauw@fundp.ac.be<br />

Dr. Wim DECLERCQ<br />

Dept. of Molecular Biomedical<br />

Research<br />

B-9052 Zwijnaarde BEL<br />

Email:<br />

Wim.Declercq@dmbr.UGent.be<br />

Mr. Julien Deheuninck<br />

CNRS UMR 8117<br />

59270 LILLE FRA<br />

Email: julien.deheuninck@ibl.fr<br />

Mrs. Daphne deLaunay<br />

Division of Clinical Immunology<br />

<strong>and</strong> Rheumatology<br />

1100 DE Amsterdam NLD<br />

Email: k.a.reedquist@amc.uva.nl


Mr. Igotz Delgado<br />

Biochemistry<br />

48940 Leioa ESP<br />

Email: ofbdebai@lg.ehu.es<br />

Mrs. Xiaoling Deng<br />

Centre for Respiratory Research<br />

WC1E 6JJ London GBR<br />

Email: caseydxl@yahoo.com<br />

Dr. Valentina Di Felice<br />

Molecular Anatomy<br />

90127 Palermo ITA<br />

Email: valentina.difelice@unipa.it<br />

Dr. Elitsa Dimova<br />

Biochemistry<br />

67663 Kaiserslautern DEU<br />

Email: dimova@chemie.uni-kl.de<br />

Dr. Katalin Dobra<br />

Department of Laboratory<br />

Medicine<br />

SE-14186 Stockholm SWE<br />

Email:<br />

katalin.dobra@labmed.ki.se<br />

Mrs. Sofia Dos Santos<br />

URBC<br />

5000 Namur BEL<br />

Email:<br />

sofia.dossantos@fundp.ac.be<br />

Mrs. Joe-Lin du Toit<br />

Dept of Physiological Sciences<br />

7600 Stellenbosch ZAF<br />

Email: jl@sun.ac.za<br />

Mrs. Raffaella Dell'Eva<br />

Experimental Oncology A<br />

16132 Genoa ITA<br />

Email: raffaelladelleva@yahoo.it<br />

Dr. Anne Devin<br />

IBGC<br />

33077 BORDEAUX FRA<br />

Email: anne.devin@ibgc.ubordeaux2.fr<br />

Dr. Marc Diederich<br />

LBMCC<br />

L-2540 Luxemb<strong>our</strong>g LUX<br />

Email: marc.diederich@lbmcc.lu<br />

Pr. Caroline Dive<br />

Clinical <strong>and</strong> Experimental<br />

Pharmacology Group<br />

M20 4BX Manchester GBR<br />

Email: cdive@picr.man.ac.uk<br />

Mrs. Sara C. T. S. Domingues<br />

LaboratÛrio de NeurociÍncias<br />

3810-193 Aveiro Aveiro PRT<br />

Email: a15922@alunos.bio.ua.pt<br />

Dr. Alex<strong>and</strong>ra Dreuw<br />

Institut für Biochemie<br />

52074 Aachen DEU<br />

Email: serge.haan@rwthaachen.de<br />

Dr. Lenka Dubska<br />

Department of Laboratory<br />

Medicine<br />

Msaryk Memorial Cancer Institute<br />

Brno 656 53 Czech Republic<br />

Email: dubska@mou.cz<br />

Dr. Maurice Dematteis<br />

40202 Louisville, KY USA<br />

Email:<br />

maurice.dematteis@louisville.edu<br />

Dr. Luciano Di Croce<br />

Epigenetic events in cancer<br />

8003 Barcelona ESP<br />

Email: luciano.dicroce@crg.es<br />

Mrs. Nathalie Dijsselbloem<br />

LEGEST<br />

9000 Gent BEL<br />

Email:<br />

nathalie.dijsselbloem@ugent.be<br />

Dr. Mojgan Djavaheri-Mergny<br />

Glycobiologie et signalisation<br />

cellulaire<br />

94807 Villejuif FRA<br />

Email: mergny@vjf.inserm.fr<br />

Dr. Rosanna DONO<br />

CNRS<br />

13288 Marseille FRA<br />

Email: dono@ibdm.univ-mrs.fr<br />

Dr. Sophia V. Drouva<br />

Interactions Cellulaires<br />

Neuroendocriniennes<br />

13916 Marseille FRA<br />

Email: drouva.sv@jeanroche.univ-mrs.fr<br />

Dr. Joanna Dulinska<br />

Department of Medical<br />

Biochemistry<br />

31-034 Krakow POL<br />

Email: mblitewk@cyf-kr.edu.pl


Mr. JACQUES DUMONT<br />

IRIBHM<br />

1070 BRUSSELS BEL<br />

Email: jedumont@ulb.ac.be<br />

Dr. Zdenek Dvorak<br />

Institute of Medical Chemistry <strong>and</strong><br />

Biochemistry<br />

77515 Olomouc CZE<br />

Email: moulin@email.cz<br />

Dr. Kerry Elliot<br />

E1 4NS London GBR<br />

Email: kelliot@staffmail.ed.ac.uk<br />

Dr. Anna-Mart Engelbrecht<br />

Dept of Physiological Sciences<br />

7600 Stellenbosch ZAF<br />

Email: ame@sun.ac.za<br />

Dr. Ilaria Esposito<br />

Prof. Anna Maria Musti<br />

80131 Napoli ITA<br />

Email: ila_esposito@libero.it<br />

Dr. Susanna Fagerholm<br />

Molecular studies on leukocyte<br />

adhesion, Carl G. Gahmberg<br />

14 Helsinki FIN<br />

Email:<br />

susanna.fagerholm@helsinki.fi<br />

Mrs. Consol Farrera-Sinfreu<br />

Group of Macrophage Biology<br />

8028 Barcelona ESP<br />

Email: cfarresi7@qui.ub.edu<br />

Mr. Eric DUPLUS<br />

UMR7102 Neurobiologie des des<br />

Processus Adaptatifs<br />

75005 Paris FRA<br />

Email: eric.duplus@snv.jussieu.fr<br />

Mrs. Souhayla El Maadidi<br />

LIMBP<br />

57070 Metz FRA<br />

Email: elnaadid@univ-metz.fr<br />

Mr. YALIN EMRE<br />

CNRS UPR 9078<br />

75730 CEDEX 15 PARIS FRA<br />

Email: emre@necker.fr<br />

Dr. Riyo Enomoto<br />

Department of Pharmacology<br />

651-2180 Kobe JPN<br />

Email:<br />

enomoto@pharm.kobegakuin.ac.jp<br />

Pr. Alfred Esser<br />

Cell Biology & Biophysics<br />

64110 Kansas City, Miss<strong>our</strong>i USA<br />

Email: essera@umkc.edu<br />

Dr. Alex<strong>and</strong>er Faltusz<br />

65824 Schwalbach/Ts DEU<br />

Email:<br />

alex<strong>and</strong>er.faltusz@merckbioscienc<br />

es.de<br />

Dr. Patrizia Fattoretti<br />

Neurobiology of Aging Laboratory<br />

60121 Ancona ITA<br />

Email: p.fattoretti@inrca.it<br />

Dr. Erwan Dupont<br />

Lab Neuromuscular Plasticity<br />

59655 Villeneuve d'Ascq FRA<br />

Email: Erwan.Dupont@univlille1.fr<br />

Mrs. Aleks<strong>and</strong>ra Ellert-<br />

Miklaszewska<br />

Dept. of Biochemistry <strong>and</strong> Clinical<br />

Chemistry<br />

02-097 Warsaw POL<br />

Email: aellert@nencki.gov.pl<br />

Dr. Takayuki Endoh<br />

Department of Physiology<br />

2618502 Chiba JPN<br />

Email: tendoh@tdc.ac.jp<br />

Mr. Morten Eskildsen<br />

2600 Glostrup DNK<br />

Email: me@dcb-glostrup.dk<br />

Dr. Beatrice EYMIN<br />

Lung Research Group<br />

38706 LA TRONCHE FRA<br />

Email: Beatrice.Eymin@ujfgrenoble.fr<br />

Dr. Margarida Fardilha<br />

LaboratÛrio de TransduÁ„o de<br />

Sinais<br />

3810-193 Aveiro Aveiro PRT<br />

Email: mfardilha@bio.ua.pt<br />

Mrs. Sylvie Fauconnet<br />

Laboratoire de Biologie Cellulaire<br />

et Moléculaire<br />

25000 BESANCON FRA<br />

Email: s.fauconnet@libertysurf.fr


Mrs. Anissa Fergani<br />

INSERM U-692<br />

67000 Strasb<strong>our</strong>g FRA<br />

Email: anissafergani@yahoo.fr<br />

Mrs. Nele FESTJENS<br />

Dept. of Molecular Biomedical<br />

Research<br />

B-9052 Zwijnaarde BEL<br />

Email: Nele@dmbr.UGent.be<br />

Mrs. Katherine G Finegan<br />

Faculty of Life Sciences<br />

M13 9PT Manchester GBR<br />

Email:<br />

K.G.Finegan@postgrad.mancheste<br />

r.ac.uk<br />

Dr. Daniela Flügel<br />

Biochemistry<br />

67663 Kaiserslautern DEU<br />

Email: dfluege@gwdg.de<br />

Mrs. Bénédicte Foveau<br />

CNRS UMR8117<br />

59021 Lille FRA<br />

Email: benedicte.foveau@ibl.fr<br />

Mrs. Giovanna Frazziano<br />

Dept. Pharmacology<br />

28040 Madrid ESP<br />

Email: acogolludo@ift.csic.es<br />

Mr. Michael Fricker<br />

Department of Biochemistry<br />

CB2 1QW Cambridge GBR<br />

Email: mf309@cam.ac.uk<br />

Mrs. Anne FERNANDEZ VIDAL<br />

INSERM Unit 563<br />

31024 TOULOUSE FRA<br />

Email:<br />

fervidal@toulouse.inserm.fr<br />

Mrs. Dragana Filipovic<br />

Laboratory for molecular biology<br />

<strong>and</strong> endocrinology 090<br />

11000 Belgrade YUG<br />

Email: dragana@vin.bg.ac.yu<br />

Dr. Alessia Finotti<br />

Dept of Biochemistry <strong>and</strong><br />

Molecular Biology<br />

44100 Ferrara ITA<br />

Email: gam@unife.it<br />

Dr. Emma Folch-Puy<br />

Experimental Pathology<br />

8036 Barcelona ESP<br />

Email: efpbam@iibb.csic.es<br />

Dr. Thierry Franck<br />

Centre de l'Oxygéne, Recherche et<br />

Développement<br />

4000 Liége BEL<br />

Email: T.franck@ulg.ac.be<br />

Dr. Samuel W. French<br />

90502 Torrance USA<br />

Email: sfrench@labiomed.org<br />

Mr. Hans Friedrichsen<br />

Department Biochemistry<br />

CB2 1QW Cambridge GBR<br />

Email: hfriedrichsen@gmail.com<br />

Mrs. Laura Ferrero-Miliani<br />

Lab. of Gastroenterology C 5403,<br />

Herlev Hospital<br />

2730 Herlev DNK<br />

Email: lali@stud.ku.dk<br />

Dr. Carmela Fimognari<br />

Pharmacology<br />

40126 Bologna ITA<br />

Email:<br />

carmela.fimognari@unibo.it<br />

Mr. KONSTANTINOS FLOROS<br />

BIOCHEMISTRY AND<br />

MOLECULAR BIOLOGY<br />

15701 ATHENS GRC<br />

Email: florksgr@yahoo.gr<br />

Mrs. Florence Folmer<br />

AB25 2WN Aberdeen GBR<br />

Email:<br />

florence_folmer@yahoo.com<br />

Mrs. Aurélie FRANCOIS<br />

Equipe Projet "Photobiologie en<br />

Cancérologie"<br />

54511 V<strong>and</strong>oeuvre les Nancy FRA<br />

Email: a.francois@nancy.fnclcc.fr<br />

Dr. Véronique FREUND-<br />

MICHEL<br />

EA3771<br />

67401 ILLKIRCH FRA<br />

Email: vfreund@pharma.ustrasbg.fr<br />

Dr. Ellen Fritsche<br />

Toxicology<br />

40225 Düsseldorf DEU<br />

Email: ellen.fritsche@uniduesseldorf.de


Mr. Thomas Frogne<br />

DK-2100 Copenhagen DNK<br />

Email: tfr@cancer.dk<br />

Mr. Gérald GAIBELET<br />

IPBS<br />

31077 TOULOUSE FRA<br />

Email: gerald.gaibelet@ipbs.fr<br />

Mr. Mattias Gäreskog<br />

Inst. Medical Cell Biology<br />

751 23 Uppsala SWE<br />

Email:<br />

Mattias.Gareskog@medcellbiol.uu<br />

.se<br />

Mr. Javier Gayarre<br />

28040 Madrid ESP<br />

Email: jgayarre@cib.csic.es<br />

Pr. Penka Genova<br />

CLMP<br />

1113 Sofia BGR<br />

Email: Pgen@TU-Sofia.bg<br />

Mrs. Lieke Gerris<br />

Xtrack<br />

3572GA Utrecht NLD<br />

Email: L.M.Gerris@students.uu.nl<br />

Dr. Lina Ghibelli<br />

133 Roma ITA<br />

Email: ghibelli@uniroma2.it<br />

Dr. François Fuks<br />

1070 Brussels BEL<br />

Email: ffuks@ulb.ac.be<br />

Dr. Catherine Gaitanaki<br />

Animal & Human Physiology<br />

157 84 Athens GRC<br />

Email: cgaitan@biol.uoa.gr<br />

Dr. Clare Garvey<br />

N1 9XW London GBR<br />

Email: c.garvey@nature.com<br />

Dr. Christoffer Gebhardt<br />

Division of Signal Transduction<br />

<strong>and</strong> Growth Control<br />

69120 Heidelberg DEU<br />

Email: c.gebhardt@dkfz.de<br />

Mr. Thomas Gerken<br />

C. J. Schofield<br />

Ox1 3TA Oxford GBR<br />

Email:<br />

thomas.gerken@bioch.ox.ac.uk<br />

Dr. Arieh Gertler<br />

The Hebrew University of<br />

Jerusalem<br />

76100 Rehovot ISR<br />

Email: gertler@agri.huji.ac.il<br />

Pr. Sankar Ghosh<br />

Section of Immunobiology<br />

6519 New Haven USA<br />

Email: sankar.ghosh@yale.edu<br />

Dr. Jozefa Gadek-Wesierski<br />

Dept. of Medicine I, Div.: Inst. of<br />

Cancer Research<br />

A-1090 Vienna AUT<br />

Email: jozefa.gadekwesierski@meduniwien.ac.at<br />

Pr. Roberto Gambari<br />

Dept of Biochemistry <strong>and</strong><br />

Molecular Biology<br />

44100 Ferrara ITA<br />

Email: gam@unife.it<br />

Dr. Jean-Stéphane Gatot<br />

Chimie médicale<br />

4000 Liége BEL<br />

Email: jsgatot@ulg.ac.be<br />

Mr. Christian Geest<br />

Dept. of Immunology<br />

3584 EA Utrecht NLD<br />

Email: C.Geest@azu.nl<br />

Dr. Sarah Gerlo<br />

LEGEST<br />

9000 Gent BEL<br />

Email: sarah.gerlo@ugent.be<br />

Dr. Mahmoud Ghazi-Khansari<br />

Tehran University of Medical<br />

Sciences<br />

13145 - 784 Tehran PCN<br />

Email: ghazikha@sina.tums.ac.ir<br />

Dr. Dorota Gil<br />

Department of Medical<br />

Biochemistry<br />

31-034 Krakow POL<br />

Email: dgil@poczta.fm


Dr. Mark Ginsberg<br />

Mail code 0726<br />

92093-0726 La Jolla USA<br />

Email: mhginsberg@ucsd.edu<br />

Mrs. Nadine Goebel<br />

40225 Duesseldorf DEU<br />

Email: nadine@wcv-mail.de<br />

Mr. Arnaud Goolaerts<br />

Physiopathology<br />

1070 Brussels BEL<br />

Email: agoolaer@ulb.ac.be<br />

Mrs. Delphine Goubau<br />

Dr J. Hiscott<br />

H3T 1E2 Montreal CAN<br />

Email:<br />

delphine.goubau@mcgill.ca<br />

Dr. Evelyne Gozal<br />

40202 Louisville, KY USA<br />

Email:<br />

evelyne.gozal@louisville.edu<br />

Dr. S<strong>and</strong>ro Grelli<br />

Dept. Exp.Med.Bioch.Sci.<br />

133 Rome ITA<br />

Email: grelli@med.uniroma2.it<br />

Dr. André Groyer<br />

Inserm U.683<br />

75870 Paris Cédex 18 FRA<br />

Email: groyer@bichat.inserm.fr<br />

Pr. Hans-Juergen Gl<strong>and</strong>er<br />

EAA Training Center<br />

4103 Leipzig DEU<br />

Email: glah@medizin.unileipzig.de<br />

Dr. Christoph Goemans<br />

Department of Biochemistry<br />

CB2 1QW Cambridge GBR<br />

Email: cgg20@mole.bio.cam.ac.uk<br />

Mrs. Morgane GORRIA<br />

INSERM U620<br />

35043 Rennes FRA<br />

Email: morgane.gorria@univrennes1.fr<br />

Mrs. Eleni G<strong>our</strong>gou<br />

Animal <strong>and</strong> Human Physiology<br />

15784 Athens GRC<br />

Email: elg<strong>our</strong>@biol.uoa.gr<br />

Mr. Igor Grbavac<br />

14109 Berlin DEU<br />

Email: grbavac@hmi.de<br />

Mrs. Femke Groeneweg<br />

Xtrack<br />

3572 GA Utrecht NLD<br />

Email:<br />

F.L.Groeneweg@students.uu.nl<br />

Dr. Sonja Grunewald<br />

EAA Training Center<br />

4103 Leipzig DEU<br />

Email:<br />

sonja.grunewald@medizin.unileipzig.de<br />

Mrs. Sophie Goblet<br />

URBC<br />

5000 Namur BEL<br />

Email: sophie.goblet@fundp.ac.be<br />

Dr. Reinaldo Gonzalez Ramos<br />

Gynecology<br />

1200 Bruxelles BEL<br />

Email:<br />

reinaldogonzalezr@yahoo.com<br />

Mrs. Valerie Gossye<br />

LEGEST<br />

9000 Gent BEL<br />

Email: valerie_gossye@ugent.be<br />

Mr. Ganjam Goutham Kumar<br />

Biochemistry<br />

67663 Kaiserslautern DEU<br />

Email: ganjam@chemie.uni-kl.de<br />

Dr. Mira Grdisa<br />

of molecular oncology<br />

10000 Zagreb HRV<br />

Email: grdisa@irb.hr<br />

Pr. Bernd Groner<br />

Institute for Biomedical Research<br />

D-60596 Frankfurt am Main DEU<br />

Email: groner@em.unifrankfurt.de<br />

Mrs. Barbara Guerra<br />

Institute of Biochemistry <strong>and</strong><br />

Molecular Biology<br />

5230 Odense DNK<br />

Email: bag@bmb.sdu.dk


Mr. Thierry GUILLAUDEUX<br />

UPRES-EA 3889<br />

35043 Rennes FRA<br />

Email: thierry.guillaudeux@univrennes1.fr<br />

Dr. Deepak Gupta<br />

All India Institue of Medical<br />

Sciences, New Delhi, India<br />

110029 Delhi IDN<br />

Email:<br />

drdeepakkumargupta@yahoo.com<br />

Dr. Claude Haan<br />

LBPI<br />

L-1511 Luxemburg LUX<br />

Email: claude.haan@uni.lu<br />

Dr. Yvette HABRAKEN<br />

Virology <strong>and</strong> Immunology<br />

4000 LIEGE BEL<br />

Email: Yvette.habraken@ulg.ac.be<br />

Mr. Eric Hajduch<br />

Unité 671<br />

75006 Paris FRA<br />

Email:<br />

eric.hajduch@bhdc.jussieu.fr<br />

Mr. Adam Hardy<br />

Schofield<br />

OX1 3TA Oxford GBR<br />

Email:<br />

adam.hardy@chem.ox.ac.uk<br />

Dr. Bettina Hartenstein<br />

Division of Signal Transduction<br />

<strong>and</strong> Growth Control<br />

69120 Heidelberg DEU<br />

Email: b.hartenstein@dkfz.de<br />

Dr. Nursel Gul<br />

Ankara Universitesi, Fen Fakultesi<br />

Biyoloji Bolumu<br />

Degol<br />

6100 TCA<br />

Email: ngul@science.ankara.edu.tr<br />

Mr. Eric Gutknecht<br />

CNS Research<br />

2340 Beerse BEL<br />

Email: egutknec@prdbe.jnj.com<br />

Dr. Serge Haan<br />

Institut für Biochemie<br />

52074 Aachen DEU<br />

Email: serge.haan@rwthaachen.de<br />

Pr. Guy HAEGEMAN<br />

Laboratory for Eukaryotic Gene<br />

Expression <strong>and</strong> Signal<br />

Transduction (LEGEST)<br />

9000 GENT BEL<br />

Email: guy.haegeman@ugent.be<br />

Dr. Reza Haji Hosseini<br />

Biology Group<br />

19569 Tehran IRN<br />

Email: hosseini@pnu.ac.ir<br />

Dr. Britta HARDY<br />

Felsenstein Medical Research<br />

Center<br />

49100 Petah-Tikva ISR<br />

Email: bhardy@post.tau.ac.il<br />

Mrs. Michal Hayun<br />

Prof. Benjamin Sredni<br />

52900 Ramat-Gan ISR<br />

Email: michalbgnm@netscape.net<br />

Mrs. Laura Gumy<br />

Department Biochemistry<br />

CB2 1QW Cambridge GBR<br />

Email: lfg23@cam.ac.uk<br />

Dr. Joohun Ha<br />

Biochemistry & Molecular<br />

Biology<br />

130-701 Seoul KOR<br />

Email: hajh@khu.ac.kr<br />

Mr. Thomas Haarmann-Stemmann<br />

Toxicology<br />

40225 Düsseldorf DEU<br />

Email: haarmann@uniduesseldorf.de<br />

Dr. Jody HAIGH<br />

Dept. of Molecular Biomedical<br />

Research<br />

B-9052 Zwijnaarde BEL<br />

Email:<br />

Jody.Haigh@dmbr.UGent.be<br />

Mr. Benoit Halter<br />

INSERM U692<br />

67000 Strasb<strong>our</strong>g FRA<br />

Email: ben_halter@yahoo.fr<br />

Dr. Shannon Harper<br />

Johannes L. Bos<br />

3584 CG Utrecht NLD<br />

Email: s.harper@med.uu.nl<br />

Mr. Rami Hayun<br />

Prof. Benjamin Sredni<br />

52900 Ramat-Gan ISR<br />

Email: rami_hayun@walla.co.il


Dr. Jochen Hefl<br />

Division of Signal Transduction<br />

<strong>and</strong> Growth Control<br />

69120 Heidelberg DEU<br />

Email: j.hess@dkfz.de<br />

Mr. Mahmoud Reza Heidari<br />

Toxicology <strong>and</strong> Pharmacology<br />

98 Kerman IRN<br />

Email: Heidarimr@yahoo.com<br />

Dr. Arnd Hengstermann<br />

Molecular Biology<br />

50939 Cologne DEU<br />

Email:<br />

arnd.hengstermann@pmintl.com<br />

Mr. Estelle Henry<br />

LBMCC<br />

L-2540 Luxemb<strong>our</strong>g LUX<br />

Email: estelle.henry@lbmcc.lu<br />

Dr. Andreas Herrmann<br />

52074 Aachen DEU<br />

Email: aherrmann@ukaachen.de<br />

Mrs. Majbrit Hjerrild<br />

2600 Glostrup DNK<br />

Email: mh@dcb-glostrup.dk<br />

Mr. CAROLE HONIGMANN<br />

inserm U-692<br />

67000 Strasb<strong>our</strong>g FRA<br />

Email: honigmann_c@yahoo.fr<br />

Mrs. Csilla Hegedüs<br />

Signal Transduction<br />

H-1113 Budapest HUN<br />

Email: hecsillu@yahoo.com<br />

Dr. Christine Hellweg<br />

Radiobiology<br />

51170 Koeln DEU<br />

Email: christine.hellweg@dlr.de<br />

Mrs. Rachel Henkens<br />

Anatomie et Cytologie<br />

Pathologiques<br />

4000 Liége BEL<br />

Email:<br />

R.Henkens@student.ulg.ac.be<br />

Dr. Thomas Herdegen<br />

24105 Kiel DEU<br />

Email:<br />

herdegen.office@pharmakologie.u<br />

ni-kiel.de<br />

Dr. Stefan Hippenstiel<br />

Dept. of Infectious Diseases<br />

13353 Berlin DEU<br />

Email:<br />

Stefan.Hippenstiel@charite.de<br />

Dr. Tjadine M. Holling<br />

Division of Molecular Biology,<br />

Department of<br />

Immunohematology <strong>and</strong> Blood<br />

Transfusion<br />

2333 ZA Leiden NLD<br />

Email: T.M.Holling@lumc.nl<br />

Mrs. Diana Hoogeboom<br />

Physiological Chemistry<br />

3584 CG Utrecht NLD<br />

Email: d.hoogeboom@med.uu.nl<br />

Dr. Jacqueline Heger<br />

Institute of Physiology<br />

35392 Gieflen DEU<br />

Email:<br />

Jacqueline.Heger@physiologie.me<br />

d.uni-giessen.de<br />

Dr. Brian Hemmings<br />

CH 4058 Basel CHE<br />

Email: brian.hemmings@fmi.ch<br />

Dr. Hanjo Hennemann<br />

53175 Bonn DEU<br />

Email: hennemann@caesar.de<br />

Mrs. Maria José Herrero<br />

Lab. Terapia Génica, Dpt.<br />

Farmacologia<br />

46010 Valencia ESP<br />

Email: mariajoseherrero@ono.com<br />

Pr. John Hiscott<br />

Molecular Oncology<br />

H3T 1E2 Montreal Quebec CAN<br />

Email: john.hiscott@mcgill.ca<br />

Dr. Olaf Holtkoetter<br />

Skin / Hair Physiology - Molecular<br />

Biology<br />

D-40191 Duesseldorf DEU<br />

Email:<br />

olaf.holtkoetter@henkel.com<br />

Mrs. Zuzana Horejsi<br />

Molecular Virology<br />

166 37 Prague CZE<br />

Email: zhora@img.cas.cz


Mr. Ruben Hoya-Arias<br />

LEGEST - Department Molecular<br />

Biology -<br />

9000 Gent BEL<br />

Email:<br />

Ruben.HoyaArias@UGent.be<br />

Mr. Haeng-Jeon Hur<br />

Department of food biotechnology<br />

151-742 Seoul KOR<br />

Email: snufst_hj@hotmail.com<br />

Dr. Britta Husse<br />

Julius Bernstein Institute of<br />

Physiology<br />

D-06097 Halle/Saale DEU<br />

Email: britta.husse@medizin.unihalle.de<br />

Mr. Aman Iqbal<br />

Schofield<br />

OX1 3TA Oxford GBR<br />

Email: aman.iqbal@chem.ox.ac.uk<br />

Mrs. Andrea Ivascu<br />

Cell Biology<br />

82377 Penzberg DEU<br />

Email: <strong>and</strong>rea.ivascu@roche.com<br />

Dr. Kowan Ja Jee<br />

Hartman Instutute, Department of<br />

Pathology<br />

FIN-00014, PO Box 21 Helsinki<br />

FIN<br />

Email: kowan.jee@helsinki.fi<br />

Mr. Namkyeong Jeon<br />

Department of oral pathlogy<br />

120-752 Seoul KOR<br />

Email: op1@yumc.yonsei.ac.kr<br />

Pr. Yan-Der Hsuuw<br />

Animal Science<br />

912-01 Ping Tung County TWN<br />

Email: hsuuw@yahoo.com.tw<br />

Dr. Am<strong>and</strong>ine Hurbin<br />

Lung Research Group<br />

38706 LA TRONCHE FRA<br />

Email: Am<strong>and</strong>ine.Hurbin@ujfgrenoble.fr<br />

Mr. Jin-Taek Hwang<br />

Biochemistry & Molecular<br />

Biology<br />

130-701 Seoul KOR<br />

Email: hjt153@hanmail.net<br />

Dr. Tarik Issad<br />

Institut Cochin<br />

75014 Paris FRA<br />

Email: issad@cochin.inserm.fr<br />

Dr. Gabor Jancso<br />

Department of Surgical Research<br />

<strong>and</strong> Techniques<br />

7624 Pécs HUN<br />

Email: jancsogabor@hotmail.com<br />

Mr. Viktor Jefremov<br />

Department of Biochemistry<br />

50411 Tartu EST<br />

Email:<br />

viktor.jefremov@kliinikum.ee<br />

Mr. Yong-Tark Jeon<br />

Laboratory of Gynecological<br />

Oncology<br />

110-799 Seoul KOR<br />

Email: rbssh9@snu.ac.kr<br />

Pr. Nam-ho Huh<br />

Cell Biology<br />

700-8558 Okayama JPN<br />

Email: namu@md.okayamau.ac.jp<br />

Pr. Nils-Erik Huseby<br />

Medical Biochemistry<br />

9037 Tromso Tromso UMI<br />

Email: nilseh@fagmed.uit.no<br />

Mr. Jin-Taek Hwang<br />

Dept of Food <strong>and</strong> Nutrition<br />

306-791 Daejeon KOR<br />

Email: ojpark@hannam.ac.kr<br />

Dr. Tarik Issad<br />

Institut Cochin<br />

75014 Paris FRA<br />

Email: issad@cochin.inserm.fr<br />

Dr. PATCHAREE<br />

JEARANAIKOON<br />

CLINICAL<br />

CHEMISTRY,FACULTY OF<br />

ASSOCIATED MED SCIENCES<br />

40002 KHON KAEN THA<br />

Email: patjea@kku.ac.th<br />

Mrs. Nam K Jeon<br />

depatrment of oral pathlogy<br />

120-752 seoul KOR<br />

Email: aroma990@hanmail.net<br />

Mrs. Min-A Jeong<br />

Department of food biotechnology<br />

151-742 Seoul KOR<br />

Email: accamy@hanmail.net


Mr. Jan Jepsen<br />

2100 Copenhagen DNK<br />

Email: jsj@cancer.dk<br />

Mr. Claus Johansen<br />

Department of Dermatology<br />

8000 Aarhus DNK<br />

Email: claus.johansen@ki.au.dk<br />

Mr. Kyung Don JU<br />

Department of Pharmacology<br />

120-752 Seoul KOR<br />

Email: clock1375@paran.com<br />

Dr. Magdalena Kacprzak<br />

Molecular Signalling Lab<br />

FIN 70211 KUOPIO FIN<br />

Email:<br />

Magdalena.Kacprzak@uku.fi<br />

Dr. Martha Kaloyianni<br />

Animal Physiology<br />

54124 Thessaloniki GRC<br />

Email: kaloyian@bio.auth.gr<br />

Pr. GAOUSSOU KANOUTE<br />

LABORATOIRE NATIONAL<br />

DU MALI<br />

BP 232 BAMAKO MLI<br />

Email:<br />

kanoutelaboratoire@yahoo.fr<br />

Dr. Lena Karlsson<br />

Clinical <strong>and</strong> Experimental<br />

Research Laboratory<br />

S-41346 Goteborg SWE<br />

Email: Lena.Karlsson@hjl.gu.se<br />

Mrs. Hanna Jerczynska<br />

Department of Mol. & Med.<br />

Biophysics<br />

92-215 Lodz POL<br />

Email: hanuka@zdn.am.lodz.pl<br />

Mrs. Anne-Laure Joly<br />

LBMCC<br />

2540 Luxemb<strong>our</strong>g LUX<br />

Email: anne-laure.joly@edu.univfcomte.fr<br />

Pr. Yong-Sung Juhnn<br />

Department of Biochemistry <strong>and</strong><br />

Molecular Biology<br />

110-799 Seoul KOR<br />

Email: juhnn@snu.ac.kr<br />

Mrs. PANAGIOTA KAKATSOU<br />

BIOCHEMISTRY AND<br />

MOLECULAR BIOLOGY<br />

15701 ATHENS GRC<br />

Email: florksgr@yahoo.gr<br />

Mrs. MAUD KAMAL<br />

INSERM U581 Equipe Avenir<br />

94010 CRETEIL FRA<br />

Email:<br />

maud.kamal@creteil.inserm.fr<br />

Dr. Elodie Kara<br />

Physiologie de la Reproduction et<br />

des Comportements<br />

37380 Nouzilly FRA<br />

Email: kara@t<strong>our</strong>s.inra.fr<br />

Mr. Atsufumi Kawabata<br />

Div. Physiol. Pathophysiol., Sch.<br />

Pharm. Sci.<br />

577-8502 Higashi-Osaka JPN<br />

Email:<br />

kawabata@phar.kindai.ac.jp<br />

Mr. Manel Joaquin<br />

Cell Signaling Unit<br />

8002 Barcelona ESP<br />

Email: manel.joaquin@upf.edu<br />

Dr. RHIANNON JONES<br />

ANTIONCOGENE<br />

SW3 6JB LONDON GBR<br />

Email: rhiannon.jones@icr.ac.uk<br />

Dr. Anna Jurewicz<br />

Neurology<br />

90-153 Lodz POL<br />

Email: ajur@afazja.am.lodz.pl<br />

Dr. Tuula Kallunki<br />

Apoptosis Department<br />

2100 Copenhagen DNK<br />

Email: tk@cancer.dk<br />

Mrs. Monika Kaminska<br />

Enzymatic Regulation of Cell<br />

Activities<br />

75724 Paris FRA<br />

Email: kaminska@pasteur.fr<br />

Mr. Emin Karaca<br />

Ege University Medical Faculty<br />

Medical Genetics<br />

35100 Izmir TUR<br />

Email: asudealpman@gmail.com<br />

Dr. Shirakawa Kazuo<br />

Saitama medical center<br />

350-8550 Kawagoe JPN<br />

Email: shirak@saitama-med.ac.jp


Mrs. Eirini Kefaloyianni<br />

Animal <strong>and</strong> Human Physiology<br />

15784 Athens GRC<br />

Email: ikefalog@biol.uoa.gr<br />

Pr. Ezzatollah Keyhani<br />

University of Tehran<br />

13145 Tehran IRN<br />

Email: keyhanie@ibb.ut.ac.ir<br />

Dr. Maria Kharlap<br />

Clinical<br />

Electrophysiology/Genetic<br />

Engineering<br />

121552 Moscow RUS<br />

Email: shadow1@mail.cnt.ru<br />

Mrs. Do-Hee Kim<br />

National Research Laboratory of<br />

Molecular Carcinogenesis <strong>and</strong><br />

Chemoprevention<br />

151-742 Seoul KOR<br />

Email: calladohee@yahoo.co.kr<br />

Pr. Hyeyoung Kim<br />

Department of Pharmacology<br />

120-752 Seoul KOR<br />

Email:<br />

kim626@yumc.yonsei.ac.kr<br />

Mrs. Mi Jeong Kim<br />

Oral Biology<br />

120-752 Seoul KOR<br />

Email:<br />

kimmi780507@hanmail.net<br />

Mrs. Seo-Young Kim<br />

Department of food biotechnology<br />

151-742 Seoul KOR<br />

Email: ssoy5200@hanmail.net<br />

Mrs. Lilian Kessels<br />

6200 MD Maastricht NLD<br />

Email:<br />

l.vanwylick@farmaco.unimaas.nl<br />

Dr. Jacqueline Keyhani<br />

University of Tehran<br />

13145 Tehran IRN<br />

Email: keyhanie@ibb.ut.ac.ir<br />

Dr. Hippokratis Kiaris<br />

Biochemistry<br />

115 27 Athens GRC<br />

Email: hkiaris@med.uoa.gr<br />

Mrs. Eun-Hee Kim<br />

National Research Laboratory of<br />

Molecular Carcinogenesis <strong>and</strong><br />

Chemoprevention<br />

151-742 Seoul KOR<br />

Email: idi@dreamwiz.com<br />

Mrs. Hyo-jin Kim<br />

Department of food biotechnology<br />

151-742 Seoul KOR<br />

Email: ipkhj@hanmail.net<br />

Mrs. Min-Jung Kim<br />

National Research Laboratory of<br />

Molecular Carcinogenesis <strong>and</strong><br />

Chemoprevention<br />

151-742 Seoul KOR<br />

Email: bluebeer99@hanmail.net<br />

Mrs. Su-Hyeong Kim<br />

110-799 Seoul KOR<br />

Email: rbssh9@snu.ac.kr<br />

Mr. HUR KEUN<br />

Cancer Research Institute<br />

110-744 Seoul PRK<br />

Email: blue2001@snu.ac.kr<br />

Dr. Khalid Khabar<br />

Program in BioMolecular<br />

Research<br />

11211 Riyadh SAU<br />

Email: khabar@kfshrc.edu.sa<br />

Dr. Simone Kiermayer<br />

Internal Medicine II<br />

66421 Homburg/Saar DEU<br />

Email: inskie@unikliniksaarl<strong>and</strong>.de<br />

Mrs. Hye Sun Kim<br />

Dept. of Anatomy, Embryology<br />

Lab.<br />

120-752 Seoul KOR<br />

Email: goldfish79@hanmail.net<br />

Mrs. Jin Eun Kim<br />

Department of Oral Biology<br />

120-752 Seoul KOR<br />

Email: judyjean@hanmail.net<br />

Pr. Myoung Hee Kim<br />

Dept. of Anatomy, Embryology<br />

Lab.<br />

170-752 Seoul KOR<br />

Email:<br />

mhkim1@yumc.yonsei.ac.kr<br />

Pr. SUNG KIM<br />

ANGIOGENESIS AND<br />

CHEMOPREVENTION<br />

449701 YONGIN KOR<br />

Email: sungkim7@khu.ac.kr


Pr. Young Min Kim<br />

Division of Biological Sciences<br />

306-791 Daejeon KOR<br />

Email: kym@hannam.ac.kr<br />

Mrs. Mélanie KIRCHMEYER<br />

Physiopathologie et<br />

Pharmacologie Articulaires<br />

54500 V<strong>and</strong>oeuvre lËs Nancy<br />

FRA<br />

Email: melkirch2002@yahoo.fr<br />

Mrs. Rasmus Boye Kjellerup<br />

Department of Dermatology<br />

8000 Aarhus DNK<br />

Email: rbk@studmed.au.dk<br />

Mrs. Anna Klemm<br />

Biophysic Group<br />

91052 Erlangen DEU<br />

Email: aklemm@biomed.unierlangen.de<br />

Mr. Holger Knobelspies<br />

Institute of Pharmacology <strong>and</strong><br />

Toxicology<br />

52074 Aachen DEU<br />

Email: hknobelspies@ukaachen.de<br />

Dr. George Koliakos<br />

Biological Chemistry<br />

54124 Thessaloniki GRC<br />

Email: koliakos@med.auth.gr<br />

Pr. Liliana Konarska<br />

Dept. of Biochemistry <strong>and</strong> Clinical<br />

Chemistry<br />

02-091 Warsaw POL<br />

Email: konarska@nencki.gov.pl<br />

Dr. Young Min Kim<br />

Dept of Food <strong>and</strong> Nutrition<br />

306-791 Daejeon KOR<br />

Email: ojpark@hannam.ac.kr<br />

Mr. Milen Ivanov Kirilov<br />

Experimental Chemotherapy<br />

1000 Sofia BGR<br />

Email: m.kirilov@dkfz.de<br />

Dr. Jean-Paul Klein<br />

Inserm UMR-S 392/E.A. 3950<br />

ULP<br />

67000 Strasb<strong>our</strong>g FRA<br />

Email: jeanpaul.klein@medecine.u-strasbg.fr<br />

Dr. Stefan Knapp<br />

Structural Genomics Consortium<br />

OX3 7LD Oxford GBR<br />

Email: stefan.knapp@sgc.ox.ac.uk<br />

Dr. Mariusz Koda<br />

Department of Pathology<br />

15-269 Bialystok POL<br />

Email: sulek@zeus.amb.edu.pl<br />

Dr. Natalia Koltovaya<br />

Laboratory of Radiation Biology<br />

141980 Dubna RUS<br />

Email: koltovaya@jinr.ru<br />

Dr. Paul Kong Thoo Lin<br />

School of Life Sciences<br />

AB25 1HG Aberdeen GBR<br />

Email: p.kong@rgu.ac.uk<br />

Mr. Matthew King<br />

Department of Biochemistry<br />

CB2 1QW Cambridge GBR<br />

Email:<br />

mak45@mole.bio.cam.ac.uk<br />

Dr. Paul Kirkham<br />

RH12 5AB Horsham GBR<br />

Email:<br />

paul.kirkham@novartis.com<br />

Mrs. Lilach Kleinberg<br />

Department of Pathology<br />

N-0310 Oslo NOR<br />

Email: lilachkl@odont.uio.no<br />

Mrs. Jelena Knezevic<br />

Laboratory for molecular oncology<br />

Institute<br />

10000 Zagreb HRV<br />

Email: jknezev@irb.hr<br />

Dr. Kersti Kokk<br />

50411 Tartu EST<br />

Email: kersti.kokk@ut.ee<br />

Dr. Radko Komers<br />

140 21 Prague CZE<br />

Email: rako@medicon.cz<br />

Pr. Spiro Konstantinov<br />

Experimental Chemotherapy<br />

1000 Sofia BGR<br />

Email: Pgen@TU-Sofia.bg


Mr. Daniel Korr<br />

Functional Genomics<br />

13342 Berlin DEU<br />

Email: daniel.korr@schering.de<br />

Pr. Peter H Krammer<br />

Tumorimmunology Program<br />

D-69120 Heidelberg DEU<br />

Email: p.krammer@dkfz.de<br />

Dr. Anna Krichevsky<br />

2115 Boston USA<br />

Email:<br />

akrichevsky@rics.bwh.harvard.ed<br />

u<br />

Mr. Aleks<strong>and</strong>ar Krstic<br />

Laboratory for human molecular<br />

genetics<br />

11010 Belgrade YUG<br />

Email: hmgbox@eunet.yu<br />

Mrs. Joanna Kuldo<br />

9713 GZ Groningen NLD<br />

Email: j.m.kuldo@med.umcg.nl<br />

Mrs. Young E. Kwak<br />

department of oral pathlogy<br />

120-752 Seoul KOR<br />

Email: op1@yumc.yonsei.ac.kr<br />

Dr. joerg Labahn<br />

IBI-2<br />

52435 Juelich DEU<br />

Email: j.labahn@fz-juelich.de<br />

Mr. Larissa Kotelevets<br />

inserm u683<br />

18 Paris FRA<br />

Email: koteleve@bichat.inserm.fr<br />

Dr. Bertolt Kreft<br />

Functional Genomics<br />

13342 Berlin DEU<br />

Email: bertolt.kreft@schering.de<br />

Mr. Fabian Kriebel<br />

Sektion Experimentelle<br />

Anaesthesiologie<br />

89075 Ulm DEU<br />

Email: fabiankriebel@web.de<br />

Dr. Jan K Kubicek<br />

IBI-2<br />

52425 Juelich DEU<br />

Email: j.kubicek@fz-juelich.de<br />

Mr. Joydeb Kumar Kundu<br />

National Research Laboratory of<br />

Molecular Carcinogenesis <strong>and</strong><br />

Chemoprevention<br />

151-742 Seoul KOR<br />

Email: kundujk@yahoo.com<br />

Mr. Jung-Yeon Kwon<br />

Department of food biotechnology<br />

151-742 Seoul KOR<br />

Email: nararav@hanmail.net<br />

Mrs. Marianne LABUSSIERE<br />

Histopathologie Expérimentale et<br />

Moléculaire<br />

54505 V<strong>and</strong>oeuvre les Nancy FRA<br />

Email:<br />

marianne.labussiere@voila.fr<br />

Dr. Jarmila Kralova<br />

Molecular Virology<br />

166 37 Prague CZE<br />

Email: kralova@img.cas.cz<br />

Dr. Stephanie Kreis<br />

LBPI<br />

L-1511 Luxemb<strong>our</strong>g LUX<br />

Email: stephanie.kreis@uni.lu<br />

Mr. Guido Kroemer<br />

CNRS-UMR8125<br />

94805 Villejuif FRA<br />

Email: kroemer@igr.fr<br />

Mrs. Satoko Kubo<br />

Div. Physiol. Pathophysiol., Sch.<br />

Pharm. Sci.<br />

577-8502 Higashi-Osaka JPN<br />

Email: 0544410001t@kindai.ac.jp<br />

Dr. Mikhail Kutuzov<br />

Department of Pharmacology<br />

60607 Chicago, IL USA<br />

Email: kutuzov@uic.edu<br />

Pr. Antonios Kyriakopoulos<br />

Molecular Trace Elements<br />

Research in the Life Science<br />

14109 Berlin DEU<br />

Email: Kyriakopoulos@hmi.de<br />

Mrs. Piret Laht<br />

molecular biology<br />

12618 Tallinn EST<br />

Email: piret.laht@ttu.ee


Mr. Christoph Lahtz<br />

AG Tumorgenetik<br />

6097 Halle (Saale) DEU<br />

Email: Christoph_Lahtz@gmx.de<br />

Mrs. Elise Langenkamp<br />

Medical Biology<br />

9713 GZ Groningen NLD<br />

Email:<br />

elise.langenkamp@gmail.com<br />

Mrs. Isabelle Lascombe<br />

Laboratoire de Biologie Cellulaire<br />

et Moléculaire<br />

25000 BESANCON FRA<br />

Email: isabelle.lascombe@voila.fr<br />

Mrs. Xuefen Le B<strong>our</strong>his<br />

INSERM ERI-8<br />

59655 Villeneuve d'Ascq FRA<br />

Email: xuefen.leb<strong>our</strong>his@univlille1.fr<br />

Dr. Eibai Lee<br />

Department of Pharmacology<br />

651-2180 Kobe JPN<br />

Email:<br />

elee@pharm.kobegakuin.ac.jp<br />

Mrs. Eun Young Lee<br />

Dept. of Anatomy, Embryology<br />

Lab.<br />

120-752 Seoul KOR<br />

Email: nannaya1210@paran.com<br />

Mr. Jeong-Sang Lee<br />

National Research Laboratory of<br />

Molecular Carcinogenesis <strong>and</strong><br />

Chemoprevention<br />

151-742 Seoul KOR<br />

Email: jslee11@yahoo.com<br />

Mrs. Jennifer Laing<br />

Virology <strong>and</strong> Immunology<br />

21201 Baltimore USA<br />

Email: jlain001@umaryl<strong>and</strong>.edu<br />

Dr. Sylvie LANTUEJOUL<br />

Lung Research Group<br />

38706 LA TRONCHE FRA<br />

Email: SLantuejoul@chugrenoble.fr<br />

Dr. Yves Laumonnier<br />

Department of Pharmacology of<br />

Natural Products & Clinical<br />

Pharmacology<br />

89081 Ulm DEU<br />

Email: yves.laumonnier@uniulm.de<br />

Mr. Chantal LEBRUN<br />

IPBS<br />

31077 TOULOUSE FRA<br />

Email: chantal.lebrun@ipbs.fr<br />

Mrs. EUN LEE<br />

ANGIOGENESIS AND<br />

CHEMOPREVENTION<br />

449701 YONGIN KOR<br />

Email: sungkim7@khu.ac.kr<br />

Pr. EunJu Lee<br />

department of oral pathlogy<br />

120-752 Seoul KOR<br />

Email: e16lee@yumc.yonsei.ac.kr<br />

Mr. Jong Hwa Lee<br />

Department of Pharmacology<br />

120-752 Seoul KOR<br />

Email: pico798@naver.com<br />

Dr. Elisabetta Lambertini<br />

Dept of Biochemistry <strong>and</strong><br />

Molecular Biology<br />

44100 Ferrara ITA<br />

Email: elambertini@yahoo.it<br />

Mrs. Maria Lapshina<br />

Molecular biology laboratory<br />

142432 Chernogolovka RUS<br />

Email: lapshina@icp.ac.ru<br />

Dr. Antigone Lazou<br />

Animal Physiology<br />

54124 Thessaloniki GRC<br />

Email: lazou@bio.auth.gr<br />

Mrs. Marielle Lebrun<br />

Virologie et Immunologie<br />

4000 Liege BEL<br />

Email: mlebrun@student.ulg.ac.be<br />

Pr. Eun Ju Lee<br />

Oral Cancer Research Institute,<br />

Department of Oral pathology<br />

120752 Seoul KOR<br />

Email: e16lee@yumc.yonsei.ac.kr<br />

Mr. Jangwon Lee<br />

Department of Pharmacology<br />

120-752 Seoul KOR<br />

Email: honorjw@hotmail.com<br />

Dr. Ki-won Lee<br />

Department of food biotechnology<br />

151-742 Seoul KOR<br />

Email: opening2@snu.ac.kr


Mrs. Mi-Hyun Lee<br />

National Research Laboratory of<br />

Molecular Carcinogenesis <strong>and</strong><br />

Chemoprevention<br />

151-742 Seoul KOR<br />

Email: mhyun_lee@yahoo.co.kr<br />

Mrs. Eija Lehtomaki<br />

Molecular Signalling Lab<br />

FIN 70211 KUOPIO FIN<br />

Email: elehtoma@messi.uku.fi<br />

Dr. Nathalie Leleu<br />

Laboratoire de génétique et<br />

biologie cellulaire<br />

78035 Versailles FRA<br />

Email:<br />

nathalie.leleu@genetique.uvsq.fr<br />

Mrs. Min Li<br />

1748 Hopkinton USA<br />

Email: minl@bios<strong>our</strong>ce.com<br />

Dr. Bertr<strong>and</strong> Liagre<br />

Biochimie<br />

87025 Limoges FRA<br />

Email: bertr<strong>and</strong>.liagre@unilim.fr<br />

Mrs. Yi-Chu Lin<br />

10018 Taipei TWN<br />

Email: f92443016@ntu.edu.tw<br />

Dr. Metoda Lipnik-ätangelj<br />

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

Experimental Toxicology<br />

SI-1000 Ljubljana SVN<br />

Email: metoda.lipnikstangelj@mf.uni-lj.si<br />

Mrs. Sun Kyoung Lee<br />

Department of Oral Biology<br />

120-752 Seoul KOR<br />

Email: l-pluto@hanmail.net<br />

Mr. Peter Leister<br />

Institute of Genetics<br />

53117 Bonn DEU<br />

Email: peterleister@uni-bonn.de<br />

Dr. Dina Lepik<br />

Institute of Molecular <strong>and</strong> Cell<br />

Biology<br />

51010 Tartu EST<br />

Email: dlepik@ebc.ee<br />

Dr. Xiao Li<br />

Receptor <strong>and</strong> Signal Transduction<br />

48202 Detroit USA<br />

Email: xcli1@hfhs.org<br />

Dr. S<strong>and</strong>ra Liekens<br />

virology <strong>and</strong> experimental<br />

chemotherapy<br />

3000 Leuven BEL<br />

Email:<br />

s<strong>and</strong>ra.liekens@rega.kuleuven.be<br />

Mrs. Asa Lindgren<br />

medical microbiology <strong>and</strong><br />

immunology<br />

41390 Goteborg SWE<br />

Email:<br />

asa.lindgren@microbio.gu.se<br />

Dr. David Litchfield<br />

Department of Biochemistry<br />

N6A 5C1 London CAN<br />

Email: litchfi@uwo.ca<br />

Mr. Taek-Sang Lee<br />

Laboratory of Gynecological<br />

Oncology<br />

110-799 Seoul KOR<br />

Email: rbssh9@snu.ac.kr<br />

Dr. Marilena E Lekka<br />

Biochemistry<br />

451 10 Ioannina GRC<br />

Email: mlekka@cc.uoi.gr<br />

Mrs. Mei-Hua Li<br />

National Research Laboratory of<br />

Molecular Carcinogenesis <strong>and</strong><br />

Chemoprevention<br />

151-742 Seoul KOR<br />

Email: maylee3737@yahoo.com<br />

Mrs. Xiaobiao Li<br />

Prof. Denis Monard<br />

4058 Basel CHE<br />

Email: xiaobiao@fmi.ch<br />

Dr. Temduang Limpaiboon<br />

40002 Khon Kaen THA<br />

Email: temduang@kku.ac.th<br />

Dr. Maria Maddalena Lino<br />

Prof. Monard<br />

4058 Basel CHE<br />

Email: linom@fmi.ch<br />

Mr. Yan-Zhi Liu<br />

Animal Science<br />

912-01 Ping Tung County TWN<br />

Email: f58725128@yahoo.com.tw


Pr. Etta Livneh<br />

84105 Beer Sheva ISR<br />

Email: etta@bgu.ac.il<br />

Dr. Maria J. Lorenzo<br />

Departamento de Bioquimica y<br />

Biologia Molecular y Genetica<br />

10071 Caceres ESP<br />

Email: mjlorenzo@unex.es<br />

Dr. Karen Lyle<br />

Johannes L. Bos<br />

3584 CG Utrecht NLD<br />

Email: k.s.lyle@med.uu.nl<br />

Dr. Shyamala Maheswaran<br />

Surgical Oncology<br />

2114 Boston USA<br />

Email:<br />

maheswaran@helix.mgh.harvard.e<br />

du<br />

Dr. Flavio Maina<br />

CNRS<br />

13288 Marseille FRA<br />

Email: maina@ibdm.univ-mrs.fr<br />

Pr. Livio Mallucci<br />

Pharmaceutical Sciences Research<br />

Division<br />

SE1 9NH London GBR<br />

Email: livio.mallucci@kcl.ac.uk<br />

Mrs. Sophie MARCHAL<br />

Equipe Projet "Photobiologie en<br />

Cancérologie"<br />

54511 V<strong>and</strong>oeuvre les Nancy FRA<br />

Email: s.marchal@nancy.fnclcc.fr<br />

Mrs. Elisabeth LONGAVENNE<br />

Laboratoire de Recherche<br />

54511 V<strong>and</strong>oeuvre les Nancy FRA<br />

Email: s.bouali@nancy.fnclcc.fr<br />

Dr. Sophie Lorquet<br />

Tum<strong>our</strong> <strong>and</strong> Development Biology<br />

Laboratory, CRCE<br />

4000 Liége, Sart-Tilman BEL<br />

Email:<br />

S.Lorquet@student.ulg.ac.be<br />

Mrs. Delphine Magard<br />

LBMCC<br />

L-2540 Luxemb<strong>our</strong>g LUX<br />

Email:<br />

delphine.magard@laposte.net<br />

Dr. Csaba Mahotka<br />

40225 Duesseldorf DEU<br />

Email: mahotka@med.uniduesseldorf.de<br />

Mrs. Adva Maissel<br />

prof Etta Livneh<br />

84105 Beer sheva ISR<br />

Email: advak@bgumail.bgu.ac.il<br />

Mr. Steve M<strong>and</strong>erscheid<br />

Institute of Genetics<br />

53117 Bonn DEU<br />

Email: stevem<strong>and</strong>er@web.de<br />

Dr. Stefania Mardente<br />

Dep Experimental Medicine<br />

162 Rome ITA<br />

Email:<br />

stefania.mardente@uniroma1.it<br />

Mr. Francisco Lopez Hern<strong>and</strong>ez<br />

Toxicologia<br />

37007 Salamanca ESP<br />

Email: flopezher@usal.es<br />

Mr. Thomas Luft<br />

Molecular Oncology <strong>and</strong><br />

Hematology<br />

69120 Heidelberg DEU<br />

Email: t.luft@dkfz.de<br />

Dr. Maria Magocsi<br />

Signal Transduction<br />

H-1113 Budapest HUN<br />

Email:<br />

m.magocsi@biomembrane.hu<br />

Mr. Allan Maillis<br />

Laboratoire de recherche<br />

CardioVasculaire<br />

L-1150 Luxemb<strong>our</strong>g LUX<br />

Email: allan.maillis@crpsante.healthnet.lu<br />

Mrs. Anu Maki-Hokkonen<br />

Molecular Signalling Lab<br />

FIN 70211 KUOPIO FIN<br />

Email: makihokk@hytti.uku.fi<br />

Mrs. Barbara Mänz<br />

Pharmakologie und Toxikologie<br />

52075 Aachen DEU<br />

Email: Barbara.Maenz@gmx.de<br />

Dr. Christiane Margue<br />

LBPI<br />

1511 Luxemb<strong>our</strong>g LUX<br />

Email: christiane.margue@uni.lu


Mrs. Nathalie Mari<br />

Cellular <strong>and</strong> Molecular Biology<br />

Unit<br />

5030 Gembloux BEL<br />

Email: mari.n@fsagx.ac.be<br />

Pr. Alberto Maria Martelli<br />

Cell Signalling<br />

40126 Bologna ITA<br />

Email: alberto.martelli@unibo.it<br />

Dr. Carlos Martinez-Salgado<br />

Unidad de Investigacion<br />

37007 Salamanca ESP<br />

Email: carlosms@usal.es<br />

Dr. Antonio Mastino<br />

Dept. Microbiol., Gen., Mol. Sci.<br />

98166 Messina ITA<br />

Email: antonio.mastino@unime.it<br />

Dr. Ray Mattingly<br />

Department of Pharmacology<br />

MI 48201 Detroit USA<br />

Email: r.mattingly@wayne.edu<br />

Mr. Laurent MEIJER<br />

Station Biologique de Roscoff<br />

29680 ROSCOFF FRA<br />

Email: meijer@sb-roscoff.fr<br />

Dr. Paul Mercer<br />

Centre for Respiratory Research<br />

WC1E 6JJ London GBR<br />

Email: paul.mercer@ucl.ac.uk<br />

Mr. Jean-Claude Marie<br />

75018 Paris FRA<br />

Email: ugh@bichat.inserm.fr<br />

Mrs. Maud Martin<br />

Cellular <strong>and</strong> Molecular Biology<br />

Unit<br />

5030 Gembloux BEL<br />

Email: martin.m@fsagx.ac.be<br />

Dr. John Marwick<br />

RH12 5AB Horsham GBR<br />

Email:<br />

john.marwick@novartis.com<br />

Mrs. Julie Mathieu<br />

INSERM U685<br />

75010 Paris FRA<br />

Email:<br />

Julie.Mathieu@stlouis.inserm.fr<br />

Dr. Mariola Matysiak<br />

Neurology<br />

90-153 Lodz POL<br />

Email: ajur@afazja.am.lodz.pl<br />

Dr. Fanyin Meng<br />

76504 Temple USA<br />

Email: fmeng@swmail.sw.org<br />

Pr. Ofer Merimsky<br />

Dept. of Cell <strong>and</strong> Developmental<br />

Biology<br />

69978 Tel-Aviv ISR<br />

Email: histol3@post.tau.ac.il<br />

Mrs. Giulia Marsili<br />

Department of Infectious, Parasitic<br />

<strong>and</strong> Immunomediated Diseases<br />

161 roma ITA<br />

Email: gmarsili@iss.it<br />

Mrs. Natalia Martinez<br />

Institute of Immunology<br />

L-1950 Luxemb<strong>our</strong>g LUX<br />

Email:<br />

natalia.martinez@LNS.ETAT.LU<br />

Dr. Aless<strong>and</strong>ro Massa<br />

pharmacology-department of<br />

oncology biology <strong>and</strong> genetics<br />

16132 Genova ITA<br />

Email: alemassa74@hotmail.com<br />

Dr. Janos Matko<br />

Immunology<br />

1117 Budapest HUN<br />

Email: matko@cerberus.elte.hu<br />

Dr. Ilya Mazo<br />

20850 Rockville USA<br />

Email:<br />

mazo@ariadnegenomics.com<br />

Mrs. Christina Mensger<br />

69115 Heidelberg DEU<br />

Email: c.mensger@dkfz.de<br />

Dr. Marie-Paule MERVILLE<br />

Medical Chemistry<br />

4000 Liége BEL<br />

Email: mpmerville@ulg.ac.be


Mrs. Rasa Merzvinskyte<br />

Department of Developmental<br />

Biology<br />

LT-08662 Vilnius LTU<br />

Email: sypssena@one.lt<br />

Mr. Olivier Meurette<br />

INSERM U620 UPRES EA 38-89<br />

35043 Rennes FRA<br />

Email: olivier.meurette@univrennes1.fr<br />

Pr. Jelena Milasin<br />

Human Genetics<br />

11000 Belgrade YUG<br />

Email: jelena_milasin@yahoo.com<br />

Dr. Martin Modriansky<br />

Institute of Medical Chemistry <strong>and</strong><br />

Biochemistry<br />

77515 Olomouc CZE<br />

Email: oregon23@email.cz<br />

Dr. Fabrizio Montecucco<br />

Clinic of internal medicine I<br />

16132 Genoa ITA<br />

Email:<br />

fabriziomontecucco@tiscali.it<br />

Dr. Franck Morceau<br />

LBMCC<br />

L-2540 Luxemb<strong>our</strong>g LUX<br />

Email: franck.morceau@lbmcc.lu<br />

Dr. Sophie Mouillet-Richard<br />

Laboratoire de différenciation<br />

cellulaire et prions<br />

94801 VILLEJUIF FRA<br />

Email: mouillet@vjf.cnrs.fr<br />

Dr. Pedro Mestres<br />

Anatomy <strong>and</strong> Cell Biology<br />

D-66421 Homburg/Saar DEU<br />

Email: anpmes@uniklinikumsaarl<strong>and</strong>.de<br />

Dr. Uwe Michelsen<br />

LSA R&D<br />

64293 Darmstadt DEU<br />

Email: uwe.michelsen@merck.de<br />

Dr. Gyesik Min<br />

Department of Microbiological<br />

Engineering<br />

660-758 Jinju KOR<br />

Email: g-min@jinju.ac.kr<br />

Mrs. Michele Moes<br />

Laboratoire de Biologie et de<br />

Physiologe intégrée<br />

L-1511 Luxemb<strong>our</strong>g LUX<br />

Email: michele.moes@uni.lu<br />

Mrs. Michaela Moors<br />

Toxicology<br />

40225 Düsseldorf DEU<br />

Email: moors@uni-duesseldorf.de<br />

Mrs. Laura Moreno<br />

Dept. Pharmacology<br />

28040 Madrid ESP<br />

Email: acogolludo@ift.csic.es<br />

Dr. Ange Mouithys-Mickalad<br />

Centre de l'Oxygéne, Recherche et<br />

Développement<br />

4000 Liége BEL<br />

Email: amouithys@ulg.ac.be<br />

Dr. Emmanuelle Meuillet<br />

85721 Tucson USA<br />

Email:<br />

emeuillet@azcc.arizona.edu<br />

Mrs. Frederique Mies<br />

physiopathologie<br />

1070 Bruxelles BEL<br />

Email: fmies@ulb.ac.be<br />

Dr. Marie-Christine Miquel<br />

UMR 7102 Neurobiologie des<br />

Processus Adaptatifs<br />

75005 Paris FRA<br />

Email: mariechristine.miquel@snv.jussieu.fr<br />

Mrs. Antonella Montalbano<br />

Dipartimento Medicina<br />

Sperimentale-Sezione Anatomia<br />

Umana<br />

90127 Palermo ITA<br />

Email: a.montalbano@unipa.it<br />

Dr. Ana Isabel Morales Martin<br />

Toxicologia<br />

37007 Salamanca ESP<br />

Email: amorales@usal.es<br />

Mrs. Andrea Morguet<br />

Anatomy <strong>and</strong> Cell Biology<br />

D-66421 Homburg/Saar DEU<br />

Email: anpmes@uniklinikumsaarl<strong>and</strong>.de<br />

Mr. Anice MOUMEN<br />

CNRS<br />

13288 Marseille FRA<br />

Email: moumen@ibdm.univmrs.fr


Dr. Marc Mousli<br />

Inserm UMR-S 392/E.A. 3950<br />

ULP<br />

67000 Strasb<strong>our</strong>g FRA<br />

Email: marc.mousli@medecine.ustrasbg.fr<br />

Dr. Thomas Mueller<br />

51149 Cologne DEU<br />

Email:<br />

thomas.mueller@pmintl.com<br />

Dr. Gerhard Müller-Newen<br />

Institut für Biochemie<br />

52074 Aachen DEU<br />

Email: mueller-newen@rwthaachen.de<br />

Dr. Anna Maria Musti<br />

Dipartimento Farmaco-Biologico<br />

87036 Rende ITA<br />

Email: ammusti@yahoo.it<br />

Dr. Elena Navolotskaya<br />

Laboratory of Peptide<br />

Bioregulators<br />

142290 Pushchino, Moscow<br />

Region RUS<br />

Email:<br />

navolots@fibkh.serpukhov.su<br />

Dr. Frank Neumann<br />

WR 13 6JW Upper Pendock GBR<br />

Email: fneumann@bioaxxess.com<br />

Mrs. Christina Nielsen<br />

Apoptosis Department<br />

2100 Copenhagen DNK<br />

Email: cn@cancer.dk<br />

Dr. Marc Mousli<br />

Inserm UMR-S 392/E.A. 3950<br />

ULP<br />

67000 Strasb<strong>our</strong>g FRA<br />

Email: marc.mousli@medecine.ustrasbg.fr<br />

Dr. Claude P. Muller<br />

Institute of Immunology<br />

L-1950 Luxemb<strong>our</strong>g LUX<br />

Email:<br />

claude.muller@LNS.ETAT.LU<br />

Dr. Carine Munaut<br />

Laboratoire de Biologie des<br />

Tumeurs et du Développement<br />

4000 Sart Tilman (Liége) BEL<br />

Email: c.munaut@ulg.ac.be<br />

Dr. ILIAS MYLONIS<br />

LABORATORY OF<br />

BIOCHEMISTRY, SCHOOL OF<br />

MEDICINE<br />

41222 LARISSA GRC<br />

Email: mylchem3@in.gr<br />

Mrs. Bartolomé Nerea<br />

Biochemistry<br />

48940 Leioa ESP<br />

Email: ofbbaesn@lg.ehu.es<br />

Dr. Dao Nguyen<br />

Section of Thoracic Oncology,<br />

Surgery Branch<br />

20892 Bethesda. Maryl<strong>and</strong> USA<br />

Email: dao_nguyen@nih.gov<br />

Mrs. Lotte B Nielsen<br />

2600 Glostrup DNK<br />

Email: lbn@dcb-glostrup.dk<br />

Mrs. Jihanne MRIOUAH<br />

Laboratoire de Recherche<br />

54511 V<strong>and</strong>oeuvre les Nancy FRA<br />

Email: d.leprince@nancy.fnclcc.fr<br />

Dr. Susanne Muller-Knapp<br />

Structural Genomics Consortium<br />

OX3 7LD Oxford GBR<br />

Email: susanne.mullerknapp@sgc.ox.ac.uk<br />

Dr. Georg Munz<br />

Institut für Biochemie<br />

52074 Aachen DEU<br />

Email: georg_munz@yahoo.com<br />

Mr. GEORGE NAKOS<br />

10563 ATHENS GRC<br />

Email:<br />

k.despotopoulou@heronia.com<br />

Pr. Andreas Nerlich<br />

Inst. of Pathology<br />

D-81925 Munich DEU<br />

Email: <strong>and</strong>reas.nerlich@extern.lrzmuenchen.de<br />

Mrs. Magali Nicolier<br />

Laboratoire de Biologie Cellulaire<br />

et Moléculaire<br />

25000 BESANCON FRA<br />

Email: magali_nicolier@yahoo.fr<br />

Dr. Shekoufeh Nikfar<br />

Drug Evaluation Commitee<br />

14155-6451 Tehran IRN<br />

Email: s_nikfar@yahoo.com


Mr. Gustav Nilsonne<br />

Department of Laboratory<br />

Medicine<br />

SE-14186 Stockholm SWE<br />

Email: gustav.nilsonne@ki.se<br />

Dr. Jill Norman<br />

Renal Cell Biology<br />

NW3 2PF London GBR<br />

Email:<br />

j.norman@medsch.ucl.ac.uk<br />

Dr. Silvia Nuccitelli<br />

133 Rome ITA<br />

Email: silvia.nuccitelli@libero.it<br />

Mrs. Ellen O'Dea<br />

Signaling Systems Laboratory<br />

92093 La Jolla, California USA<br />

Email: eodea@ucsd.edu<br />

Mrs. S<strong>and</strong>ra Olsson<br />

Macrophage Signaling<br />

22184 Lund SWE<br />

Email:<br />

s<strong>and</strong>ra.olsson@medkem.lu.se<br />

Pr. Moshe Oren<br />

Molecular Cell Biology<br />

76100 Rehovot ISR<br />

Email:<br />

moshe.oren@weizmann.ac.il<br />

Dr. Joost Oudejans<br />

Pathology<br />

1117HV Amsterdam NLD<br />

Email: jj.oudejans@vumc.nl<br />

Dr. Mario Nizzari<br />

Pharmacology-Department of<br />

oncology biologi <strong>and</strong> genetics<br />

16132 Genova ITA<br />

Email: mario.nizzari@unige.it<br />

Mr. Jiri Novotny<br />

Laboratory of Biochemistry of<br />

Membrane Receptors<br />

142 20 Prague CZE<br />

Email: novjiri@biomed.cas.cz<br />

Mrs. Susanna Nurmi<br />

Molecular studies on leukocyte<br />

adhesion<br />

14 (Helsinki) University of<br />

Helsinki FIN<br />

Email: Susanna.Nurmi@helsinki.fi<br />

Mrs. Andrea Oeckinghaus<br />

AG Scheidereit<br />

13125 Berlin DEU<br />

Email: an.oecki@gmx.de<br />

Mrs. Tove Olsson<br />

Department of Odontology<br />

SE-141 04 Huddinge SWE<br />

Email: tove.olsson@ki.se<br />

Dr. MarÌa Clara Ortiz Ruiz<br />

30100 Murcia ESP<br />

Email: clara@um.es<br />

Mrs. Ferda Ozkinay<br />

Ege University Medical Faculty<br />

Medical Genetics<br />

35100 Izmir TUR<br />

Email: ferdafo@yahoo.com<br />

Dr. Sylvia Nome Kvam<br />

7006 Trondheim NOR<br />

Email:<br />

sylvia.nome.kvam@stolav.no<br />

Mr. TOBIAS NUBEL<br />

CNRS UPR 9078<br />

75730 PARIS FRA<br />

Email: NUEBEL@NECKER.FR<br />

Mrs. Mieke Nuytten<br />

Department of Biochemistry<br />

3000 Leuven BEL<br />

Email:<br />

mieke.nuytten@med.kuleuven.be<br />

Mrs. Jung min Oh<br />

signal transduction<br />

110-799 Seoul KOR<br />

Email: snow99@snu.ac.kr<br />

Mr. Huseyin Onay<br />

Ege University Medical Faculty<br />

Medical Genetics<br />

35100 Izmir TUR<br />

Email: ferdafo@yahoo.com<br />

Mr. Pavel Ostasov<br />

Laboratory of Biochemistry of<br />

Membrane Receptors<br />

142 20 Prague CZE<br />

Email: ostasov@biomed.cas.cz<br />

Dr. Nesrin Ozsoy<br />

6100 T<strong>and</strong>ogan / ANKARA TUR<br />

Email:<br />

ozsoy@science.ankara.edu.tr


Dr. Elisabeth Paczoska-<br />

Eliasiewicz<br />

The Hebrew University of<br />

Jerusalem<br />

76100 Rehovot ISR<br />

Email: kozlicka103@yahoo.pl<br />

Dr. Valérie Palissot<br />

laboratoire d'hémato-cancérologie<br />

expérimentale<br />

L-1526 Luxemb<strong>our</strong>g LUX<br />

Email: lhce@crp-sante.lu<br />

Mrs. In-Ja Park<br />

Dept of Food <strong>and</strong> Nutrition<br />

306-791 Daejeon KOR<br />

Email: ojpark@hannam.ac.kr<br />

Pr. Ock Jin Park<br />

Dept of Food <strong>and</strong> Nutrition<br />

306-791 Daejeon KOR<br />

Email: ojpark@hannam.ac.kr<br />

Mrs. Ioana Parvu-Ferecatu<br />

Laboratoire de génétique et<br />

biologie cellulaire<br />

78035 Versailles FRA<br />

Email: Ioana-<br />

Costina.Parvu@genetique.uvsq.fr<br />

Pr. Jasminka Pavelic<br />

Rudjer Boökovic<br />

10000 Zagreb HRV<br />

Email: jpavelic@irb.hr<br />

Dr. Zofia Pawlowska<br />

Department of Mol. & Med.<br />

Biophysics<br />

92-215 Lodz POL<br />

Email: pawlow@zdn.am.lodz.pl<br />

Mr. Artur Padzik<br />

Neuronal Signalling Laboratory<br />

20521 Turku FIN<br />

Email: apadzik@btk.fi<br />

Mrs. Cornelieke Pals<br />

Dept. of Immunology<br />

3584 EA Utrecht NLD<br />

Email: c.e.g.m.pals@azu.nl<br />

Pr. Kwang-Kyun Park<br />

120-749 Seoul KOR<br />

Email: kkp304@hanmail.net<br />

Mrs. Sin-Aye Park<br />

National Research Laboratory of<br />

Molecular Carcinogenesis <strong>and</strong><br />

Chemoprevention<br />

151-742 Seoul KOR<br />

Email: qorwh7@empal.com<br />

Dr. Tushar Patel<br />

TX 76508 Temple USA<br />

Email: tpatel@swmail.sw.org<br />

Mrs. Catherine PAVOINE<br />

INSERM U.581<br />

94010 CRETEIL FRA<br />

Email: pavoine@im3.inserm.fr<br />

Mrs. Leticia Yamila Peche<br />

National Laboratory<br />

34012 Trieste ITA<br />

Email: cib@lncib.it<br />

Mrs. Beata Pajak<br />

02-776 Warsaw POL<br />

Email: bepaj@wp.pl<br />

Dr. Efrosyni Paraskeva<br />

Lab. of Biochemistry, School of<br />

Medicine<br />

41222 Larissa GRC<br />

Email: fparaskeva@med.uth.gr<br />

Pr. Ock Jin Park<br />

Division of Biological Sciences<br />

306-791 Daejeon KOR<br />

Email: ojpark@hannam.ac.kr<br />

Mrs. Lotta Parviainen<br />

Molecular Signalling Lab<br />

FIN 70211 KUOPIO FIN<br />

Email: leparvia@hytti.uku.fi<br />

Dr. Réjane Paumelle<br />

INSERM/U545<br />

59019 Lille FRA<br />

Email: rejane.paumelle@pasteurlille.fr<br />

Mrs. Patrycja Pawlikowska<br />

02-776 Warsaw POL<br />

Email: pmuras@wp.pl<br />

Mrs. Anastasia Pechtelidou<br />

Animal <strong>and</strong> Human Physiology<br />

15784 Athens GRC<br />

Email: apechtel@biol.uoa.gr


Mrs. Christel Péqueux<br />

Tum<strong>our</strong> <strong>and</strong> Development Biology<br />

Laboratory, CRCE<br />

4000 Liége, Sart-Tilman BEL<br />

Email: C.Pequeux@ulg.ac.be<br />

Dr. Richard Pestell<br />

Lombardi Comprehensive Cancer<br />

Center<br />

20057-1468 Washington, D.C.<br />

USA<br />

Email: r<strong>and</strong>p@georgetown.edu,<br />

pestell@georgetown.edu<br />

Mrs. Marlene Pickl<br />

Cell Biology<br />

82372 Penzberg DEU<br />

Email: marlene.pickl@roche.com<br />

Mrs. Kaie Pill<br />

molecular biology<br />

12618 Tallinn EST<br />

Email: kaie.pill@ttu.ee<br />

Dr. Sébastien PLANCON<br />

LBPI<br />

L-1511 Luxemb<strong>our</strong>g LUX<br />

Email: sebastien.plancon@uni.lu<br />

Mrs. Branka Popovic<br />

Human Genetics<br />

11000 Belgrade YUG<br />

Email: brankapo@verat.net<br />

Dr. Jacques Pouyssegur<br />

Institute of Signaling,<br />

Developmental Biology & Cancer<br />

Research<br />

6189 Nice FRA<br />

Email: pouysseg@unice.fr<br />

Dr. Dolores Pérez-Sala<br />

28040 Madrid ESP<br />

Email: dperezsala@cib.csic.es<br />

Mrs. Isidora Petrovic<br />

Laboratory for human molecular<br />

genetics<br />

11010 Belgrade YUG<br />

Email: hmgbox@eunet.yu<br />

Pr. Jacques Piette<br />

Virology & Immunology<br />

B-4000 Liége BEL<br />

Email: jpiette@ulg.ac.be<br />

Mrs. Sophie PINEL<br />

Histopathologie Expérimentale et<br />

Moléculaire<br />

54505 V<strong>and</strong>oeuvre les Nancy FRA<br />

Email: pinel.sophie@tiscali.fr<br />

Dr. Krzysztof Pluta<br />

Molecular Signalling Lab<br />

FIN 70211 KUOPIO FIN<br />

Email: Krzysztof.Pluta@uku.fi<br />

Mr. Ignacio Portero-Robles<br />

Zim II Hämatologie/Oncologie<br />

60590 Frankfurt am Main DEU<br />

Email: robles@em.uni-frankfurt.de<br />

Dr. Radek Prochazka<br />

Laboratory of Developmental<br />

Biology<br />

277 21 Libechov CZE<br />

Email: prochazka@iapg.cas.cz<br />

Dr. Francisco Pérez-Vizcaino<br />

Dept. Pharmacology<br />

28040 Madrid ESP<br />

Email: acogolludo@ift.csic.es<br />

Mrs. ERIKA PEVERELLI<br />

ISTITUTO DI SCIENZE<br />

ENDOCRINE<br />

<strong>2012</strong>2 MILANO ITA<br />

Email: erika.peverelli@unimi.it<br />

Dr. Albrecht Piiper<br />

Department of Internal Medicine II<br />

D-66421 Homburg/Saar DEU<br />

Email: inapii@uniklinikumsaarl<strong>and</strong>.de<br />

Dr. Michael PINKOSKI<br />

Email: mp191@leicester.ac.uk<br />

Pr. Alfonso Pompella<br />

Dept. of Experimental PAthology<br />

56126 Pisa ITA<br />

Email: apompella@biomed.unipi.it<br />

Mr. Benoit POURCET<br />

UR 545<br />

59000 LILLE FRA<br />

Email: benoit.p<strong>our</strong>cet@pasteurlille.fr<br />

Dr. Florian Puehler<br />

Experimental Oncology<br />

13342 Berlin DEU<br />

Email:<br />

Florian.Puehler@Schering.de


Pr. Alex<strong>and</strong>er Pukhalsky<br />

Laboratory of Immunogenetics<br />

115478 Moscow RUS<br />

Email: osugariver@medgen.ru<br />

Mr. Thibaut QUILLARD<br />

UMR 643<br />

44093 Nantes FRA<br />

Email: thibaut.quillard@univnantes.fr<br />

Pr. Parvaneh Rafiee<br />

53226 Milwaukee USA<br />

Email: prafiee@mcw.edu<br />

Dr. Annat Raiter<br />

Felsenstein Medical Research<br />

Center<br />

49100 Petah Tikva ISR<br />

Email: araiter@post.tau.ac.il<br />

Dr. Madhavi Rane<br />

Dept of Medicine<br />

40202 Louisville, KY USA<br />

Email: mrane@louisville.edu<br />

Mrs. Jane Rasaiyaah<br />

Dept Immunology <strong>and</strong> Molecular<br />

Pathology<br />

W1Y 4JF London GBR<br />

Email: rekajra@ucl.ac.uk<br />

Mr. Laurent REBER<br />

EA3771<br />

67401 ILLKIRCH FRA<br />

Email: lreber@pharma.u-strasbg.fr<br />

Mrs. Emma Quigley<br />

EC2A 4LQ London GBR<br />

Email: p.garner@ashley-pub.com<br />

Mrs. Claire Quiney<br />

75006 Paris FRA<br />

Email:<br />

claire.quiney@bhdc.jussieu.fr<br />

Dr. Irfan Rahman<br />

14620 Rochester, NY USA<br />

Email:<br />

Irfan_Rahman@urmc.rochester.ed<br />

u<br />

Mrs. Carole RAMACCI<br />

Laboratoire de Recherche<br />

54511 V<strong>and</strong>oeuvre les Nancy FRA<br />

Email: c.ramacci@nancy.fnclcc.fr<br />

Mrs. Carmen Ranftler<br />

Dept. of Medicine 1,Div.: Inst. of<br />

Cancer Research<br />

A-1090 VIENNA AUT<br />

Email: cfr1978@yahoo.de<br />

Dr. SID D. RAY<br />

MOLECULAR TOXICOLOGY,<br />

PHARM. SCI. DIV.<br />

11201 BROOKLYN, NY USA<br />

Email: sray@liu.edu<br />

Mr. Amélie REBILLARD<br />

INSERM U620<br />

35043 Rennes FRA<br />

Email: amelie.rebillard@univrennes1.fr<br />

Dr. Frédéric Quignon<br />

LIMBP<br />

F-57070 Metz FRA<br />

Email: frederic.quignon@univmetz.fr<br />

Mrs. Flavia Radogna<br />

133 Rome ITA<br />

Email: flavia.radogna@libero.it<br />

Dr. Souad Rahmouni<br />

Pathology<br />

4000 Liege BEL<br />

Email: srahmouni@ulg.ac.be<br />

Dr. Carlo Ramoni<br />

Dept. Cell Biology <strong>and</strong><br />

Neurosciences<br />

162 Rome ITA<br />

Email: ramoni@iss.it<br />

Pr. Ulf R. Rapp<br />

Institut für Medizinische<br />

Strahlenkunde und Zellforschung<br />

97078 Würzburg DEU<br />

Email: rappur@mail.uniwuerzburg.de<br />

Dr. Massimo Realacci<br />

Molecular biology<br />

161 Rome ITA<br />

Email:<br />

massimo.realacci@uniroma1.it<br />

Dr. Kris Reedquist<br />

Division of Clinical Immunology<br />

<strong>and</strong> Rheumatology<br />

1100 DE Amsterdam NLD<br />

Email: k.a.reedquist@amc.uva.nl


Dr. Gabriella Regis<br />

Tumor Immunology Laboratory<br />

10126 Turin ITA<br />

Email: gabriella.regis@unito.it<br />

Mrs. Patsy Renard<br />

URBC<br />

5000 Namur BEL<br />

Email: patsy.renard@fundp.ac.be<br />

Mr. Simone Reuter<br />

LBMCC<br />

L-2540 Luxemb<strong>our</strong>g LUX<br />

Email: simone.reuter@lbmcc.lu<br />

Mrs. Elena Rodionova<br />

Molecular Oncology <strong>and</strong><br />

Hematology<br />

69120 Heidelberg DEU<br />

Email: e.rodionova@dkfz.de<br />

Mrs. Lorena Rodriguez<br />

Biochemistry<br />

48940 Leioa ESP<br />

Email: ofbronol@lg.ehu.es<br />

Dr. Maria Rodriguez-Munoz<br />

Neuropharmacology<br />

28002 Madrid ESP<br />

Email: mrodriguez@cajal.csic.es<br />

Mr. David Romano<br />

Interactions Cellulaires<br />

Neuroendocriniennes (ICNE)<br />

13916 cedex 20 Marseille FRA<br />

Email: romano.d@nord.univmrs.fr<br />

Dr. Daniel U. Reimer<br />

6020 Innsbruck AUT<br />

Email: daniel.reimer@uibk.ac.at<br />

Mrs. Flore Renaud<br />

Laboratoire de Génétique et<br />

Biologie cellulaire<br />

78035 Versailles FRA<br />

Email: frenaud@genetique.uvsq.fr<br />

Pr. Michel Rigoulet<br />

IBGC<br />

33077 BORDEAUX FRA<br />

Email: michel.rigoulet@ibgc.ubordeaux2.fr<br />

Dr. Sophie Rodius<br />

LBPI<br />

L-1511 Luxemb<strong>our</strong>g LUX<br />

Email: sophie.rodius@uni.lu<br />

Dr. Alicia Rodriguez-Barbero<br />

Deparrtamento de Fisiologia y<br />

Farmacologia<br />

37007 Salamanca ESP<br />

Email: barberoa@usal.es<br />

Mrs. Celine Rofel<br />

Molecular Genetics<br />

6229 ER Maastricht NLD<br />

Email: c.rofel@gen.unimaas.nl<br />

Mr. Salvatore Romeo<br />

Pathology<br />

2333 ZA Leiden NLD<br />

Email: S.Romeo@lumc.nl<br />

Dr. Kerstin Reimers<br />

30659 Hannover DEU<br />

Email: Reimers.Kerstin@MH-<br />

Hannover.de<br />

Pr. Saverio Francesco Retta<br />

Department of Genetics, Biology<br />

<strong>and</strong> Biochemistry<br />

10126 Torino ITA<br />

Email: francesco.retta@unito.it<br />

Dr. Aleks<strong>and</strong>ra Ristic-Fira<br />

Laboratory for Molecular Biology<br />

<strong>and</strong> Endocrinology<br />

11001 Belgrade YUG<br />

Email: aristic@vin.bg.ac.yu<br />

Mr. Leonardo Rodrigues<br />

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

Molecular Biology<br />

05508-900 Sao Paulo BRA<br />

Email: leonardo@cecm.usp.br<br />

Dr. Isabel Rodriguez-Escudero<br />

Dpto. Microbiologia II<br />

28040 Madrid ESP<br />

Email: isabelre@farm.ucm.es<br />

Mrs. Catherine Rolvering<br />

LBPI<br />

L-1511 Luxemb<strong>our</strong>g LUX<br />

Email: catherine.rolvering@uni.lu<br />

Mr. Christian Rommel<br />

1228 Plan-les-Ouates, Geneva<br />

CHE<br />

Email:<br />

christian.rommel@serono.com


Mr. Guillaume Rommelaere<br />

URBC<br />

5000 Namur BEL<br />

Email:<br />

guillaume.rommelaere@student.fu<br />

ndp.ac.be<br />

Mr. Vladimir Rudajev<br />

Laboratory of Biochemistry of<br />

Membrane Receptors<br />

142 20 Prague CZE<br />

Email: rudajev@biomed.cas.cz<br />

Mrs. Iwona Sadzak<br />

Department of Microbiology <strong>and</strong><br />

Immunbiology<br />

A-1030 Vienna AUT<br />

Email: iwona.sadzak@univie.ac.at<br />

Mr. Carlos Salazar<br />

Theoretical Biophysics / Institute<br />

of Biology<br />

10115 Berlin DEU<br />

Email: carlos.salazar@rz.huberlin.de<br />

Dr. Pilar Sanchez-Blazquez<br />

Neuropharmacology<br />

28002 Madrid ESP<br />

Email: psb@cajal.csic.es<br />

Dr. Nijole Savickiene<br />

Department of Pharmaceutical<br />

Chemistry <strong>and</strong> Pharmacognosy<br />

3000 Kaunas LTU<br />

Email: savickienenijole@takas.lt<br />

Dr. Elisabeth Schaffner-Reckinger<br />

Laboratoire de Biologie et<br />

Physiologie Intégrée<br />

L-1511 Luxemb<strong>our</strong>g LUX<br />

Email: elisabeth.schaffner@uni.lu<br />

Mrs. Caroline Rouaux<br />

INSERM U-692<br />

67000 Strasb<strong>our</strong>g FRA<br />

Email: carolinerouaux@yahoo.fr<br />

Dr. M. Begona Ruiz-Larrea<br />

Dep. Physiology, Medicine School<br />

48080 Leioa ESP<br />

Email: mbego.ruizlarrea@ehu.es<br />

Dr. Xavier SAELENS<br />

Dept. of Molecular Biomedical<br />

Research<br />

B-9052 Zwijnaarde BEL<br />

Email:<br />

Xavier.Saelens@dmbr.UGent.be<br />

Mr. ALEX SALSMANN<br />

LBPI<br />

1134 LUXEMBOURG LUX<br />

Email: salsmann@cu.lu<br />

Dr. Sujen Eleonora Santini<br />

Dipartimento di Produzioni<br />

Animali-Sezione di Fisiologia<br />

43100 Parma ITA<br />

Email: basini@unipr.it<br />

Mrs. Claudia Schäfer<br />

Toxicology<br />

40225 Düsseldorf DEU<br />

Email: claudia.schaefer@uniduesseldorf.de<br />

Mrs. Barbara Schaljo<br />

Department of Microbiology <strong>and</strong><br />

Immunbiology<br />

A-1030 Vienna AUT<br />

Email:<br />

barbara.schaljo@univie.ac.at<br />

Dr. Molay Roy<br />

305-8642 Tsukuba JPN<br />

Email: mkroy@affrc.go.jp<br />

Dr. José Ignacio Ruiz-Sanz<br />

Dep. Physiology, Medicine School<br />

48080 Leioa ESP<br />

Email: joseignacio.ruizs@ehu.es<br />

Pr. Ronit Sagi-Eisenberg<br />

Chair, Dept. of Cell <strong>and</strong><br />

Developmental Biology<br />

69978 Tel-Aviv ISR<br />

Email: histol3@post.tau.ac.il<br />

Mrs. Ester Sanchez Tillo<br />

Group of Macrophage Biology<br />

8028 Barcelona ESP<br />

Email: esanchez@ub.edu<br />

Mr. Gabriella Sarmay<br />

Dept. of Immunology<br />

1117 Budapest HUN<br />

Email: sarmayg@cerberus.elte.hu<br />

Pr. Reinhold Schäfer<br />

Molecular Tumor Pathology<br />

D-10117 Berlin DEU<br />

Email:<br />

reinhold.schaefer@charite.de<br />

Pr. Gert Jan Scheffer<br />

Dept Anesthesiology<br />

6500 HB Nijmegen NLD<br />

Email: g.scheffer@anes.umcn.nl


Dr. Carsten Scheller<br />

97078 Wuerzburg DEU<br />

Email: scheller@vim.uniwuerzburg.de<br />

Mrs. Martine Schmitz<br />

LBPI<br />

1511 luxemb<strong>our</strong>g LUX<br />

Email: martine.schmitz@uni.lu<br />

Mr. Günter Schneider<br />

Technical University Munich, II.<br />

Department of Internal Medicine<br />

D-81675 Munich DEU<br />

Email:<br />

guenter.schneider@lrz.tum.de<br />

Mrs. Yvonne Schrage<br />

2333 ZA Leiden NLD<br />

Email: y.m.schrage@lumc.nl<br />

Mr. Marc Schumacher<br />

Laboratoire de chimie organique<br />

synthétique<br />

L-2162 Luxemb<strong>our</strong>g LUX<br />

Email: reenert@hotmail.com<br />

Dr. Anna Ivana Scovassi<br />

Istituto di Genetica Molecolare<br />

27100 Pavia ITA<br />

Email: scovassi@igm.cnr.it<br />

Dr. RITA SELVATICI<br />

44100 FERRARA ITA<br />

Email: svr@unife.it<br />

Dr. Bernd Schmeck<br />

Dept. of Infectious Diseases<br />

13353 Berlin DEU<br />

Email: Bernd.Schmeck@charite.de<br />

Dr. Benoït Schneider<br />

Laboratoire de différenciation<br />

cellulaire et prions<br />

94801 VILLEJUIF FRA<br />

Email: bschneid@vjf.cnrs.fr<br />

Dr. Marina Schorpp-Kistner<br />

Division of Signal Transduction<br />

<strong>and</strong> Growth Control<br />

69120 Heidelberg DEU<br />

Email: marina.schorpp@dkfz.de<br />

Dr. Jutta Schueller<br />

MO<br />

51149 Koeln DEU<br />

Email: jutta.schueller@pmintl.com<br />

Dr. Chantal Schwartz<br />

Hémato-Cancérologie<br />

expérimentale<br />

1526 Luxemburg LUX<br />

Email: chantal.schwartz@crpsante.lu<br />

Mrs. Claudia Seidel<br />

AG Tumorgenetik<br />

6097 Halle DEU<br />

Email:<br />

Claudia.Seidel@medizin.unihalle.de<br />

Dr. Gregg Semenza<br />

Institute for Cell Engineering<br />

21205 Baltimore, MD USA<br />

Email: gsemenza@jhmi.edu<br />

Mrs. Debora Schmitz<br />

Brian Hemmings lab/Friedrich<br />

Miescher Institute<br />

4058 Basel CHE<br />

Email: debora.schmitz@fmi.ch<br />

Pr. E. Marion Schneider<br />

Sektion Experimentelle<br />

Anaesthesiologie<br />

89075 Ulm DEU<br />

Email: marion.schneider@uniulm.de<br />

Mrs. Andrea Schote<br />

Institute of Immunology<br />

L-1950 Luxemb<strong>our</strong>g LUX<br />

Email:<br />

<strong>and</strong>rea.schote@LNS.ETAT.LU<br />

Dr. Wolfgang Schulz<br />

Urology<br />

40225 Duesseldorf DEU<br />

Email: wolfgang.schulz@uniduesseldorf.de<br />

Mr. Massimiliano Scolz<br />

National Laboratory<br />

34012 Trieste ITA<br />

Email: cib@lncib.it<br />

Mrs. Chantal Sellier<br />

Laboratoire de Régulation des<br />

signaux de division<br />

59655 Villeneuve d'Ascq FRA<br />

Email: chantal_sellier@yahoo.fr<br />

Mrs. Ji Hye Seo<br />

department of Pharmacology<br />

120-752 Seoul KOR<br />

Email: jhseo@yumc.yonsei.ac.kr


Mr. Christine Sers<br />

Laboratory of Molecular Tumor<br />

Pathology<br />

10117 Berlin DEU<br />

Email: christine.sers@charite.de<br />

Pr. Mehdi Shakibaei<br />

Institute of Anatomy<br />

80336 Munich DEU<br />

Email: mehdi.shakibaei@med.unimuenchen.de<br />

Dr. Galina Shmarina<br />

Laboratory of Immunogenetics<br />

115478 Moscow RUS<br />

Email: osugariver@medgen.ru<br />

Pr. Anna Siniscalchi<br />

44100 FERRARA ITA<br />

Email: snn@unife.it<br />

Dr. Asa Sjoling<br />

medical microbiology <strong>and</strong><br />

immunology<br />

413 90 Goteborg SWE<br />

Email: asa.sjoling@microbio.gu.se<br />

Mr. Gary Robert Smith<br />

Chemistry Department<br />

MK7 6AA Milton Keynes GBR<br />

Email:<br />

gary.smith@persescomms.com<br />

Dr. Ulrike Sommer<br />

Institut für Biochemie<br />

52074 Aachen DEU<br />

Email: ulrike.sommer@rwthaachen.de<br />

Dr. Jan Seyfried<br />

Faculty of Life Sciences<br />

M13 9PT Manchester GBR<br />

Email:<br />

jan.seyfried@manchester.ac.uk<br />

Dr. Yu Shaoqing<br />

Department of of<br />

Otorhinolaryngology<br />

250031 Jinan City, Sh<strong>and</strong>ong<br />

province CHN<br />

Email: yu_shaoqing@hotmail.com<br />

Dr. Georgios Simos<br />

Lab. of Biochemistry, School of<br />

Medicine<br />

41222 Larissa GRC<br />

Email: simos@med.uth.gr<br />

Pr. ANNA SINISCALCHI<br />

DEPT. CLINICAL EXPTL.<br />

MEDICINE, SECT.<br />

PHARMACOLOGY<br />

44100 FERRARA ITA<br />

Email: snn@unife.it<br />

Mrs. Iben Holst Eriksen Skjoth<br />

Biomedical Research <strong>and</strong><br />

Molecular Cell Biology Group<br />

5230 Odense M DNK<br />

Email: ibens@bmb.sdu.dk<br />

Dr. Dik Snijdelaar<br />

Department of Anesthesiology<br />

(550)<br />

6500 HB Nijmegen NLD<br />

Email: d.snijdelaar@anes.umcn.nl<br />

Mrs. Seung Wha Son<br />

120-749 Seoul KOR<br />

Email: spritlove@nate.com<br />

Mr. Marco Sgarbanti<br />

Department of Infectious, Parasitic<br />

<strong>and</strong> Immunomediated Diseases<br />

161 Roma ITA<br />

Email: marco.sgarbanti@iss.it<br />

Mr. Jun-Wan Shin<br />

National Research Laboratory of<br />

Molecular Carcinogenesis <strong>and</strong><br />

Chemoprevention<br />

151-742 Seoul KOR<br />

Email: senjon@hotmail.com<br />

Dr. Helle-Evi Simovart<br />

Institute of Anatomy<br />

50411 Tartu EST<br />

Email: helle-evi.simovart@ut.ee<br />

Pr. Maria Caterina Sirianni<br />

Clinical Immunology, Dept. of<br />

Clin. Medicine<br />

185 Rome ITA<br />

Email:<br />

mariacaterina.sirianni@uniroma1.i<br />

t<br />

Dr. Alina Smalinskiene<br />

Environmental <strong>and</strong> Health<br />

LT- 5009 Kaunas LTU<br />

Email: smalina@vector.kmu.lt<br />

Mrs. Mayra Solis<br />

Dr J. Hiscott<br />

H3T 1E2 Montreal CAN<br />

Email: somayr@hotmail.com<br />

Pr. Yong-Sang Song<br />

Laboratory of Gynecological<br />

Oncology<br />

110-799 Seoul KOR<br />

Email: rbssh9@snu.ac.kr


Mr. Yoo-Cheol Song<br />

Laboratory of Gynecological<br />

Oncology<br />

110-799 Seoul KOR<br />

Email: rbssh9@snu.ac.kr<br />

Dr. Christina Spiropoulou<br />

Special Pathogens Branch<br />

30333 Atlanta USA<br />

Email: ccs8@cdc.gov<br />

Pr. Dominique Stéhelin<br />

Email: Dominique.Stehelin@ibl.fr<br />

Dr. Simon Stockwell<br />

Anti-oncogene<br />

SW3 6JB London GBR<br />

Email: simons@icr.ac.uk<br />

Pr. Pann-Ghill Suh<br />

Signaling Network Lab.<br />

790-784 Pohang KOR<br />

Email: pgs@postech.ac.kr<br />

Dr. Rosanna Supino<br />

Unit of Chemotherapy <strong>and</strong><br />

Antitumor Preclinical<br />

Pharmacology<br />

20133 Milano ITA<br />

Email:<br />

rosanna.supino@istitutotumori.mi.<br />

it<br />

Mr. Anna Szypowska<br />

Physiological Chemistry<br />

3584 CG Utrecht NLD<br />

Email: a.a.szypowska@med.uu.nl<br />

Mr. Tavera Soraya<br />

Janssen Cilag<br />

28042 Madrid ESP<br />

Email: amex.c.viajes@aexp.com<br />

Mr. Konstantinos Stamatakis<br />

28040 Madrid ESP<br />

Email: kostas@cib.csic.es<br />

Pr. Laurence Stevens<br />

Lab Neuromuscular Plasticity<br />

59655 Villeneuve d'Ascq FRA<br />

Email: Laurence.Stevens@univlille1.fr<br />

Mr. Jiri Stohr<br />

Laboratory of Biochemistry of<br />

Membrane Receptors<br />

142 20 Prague CZE<br />

Email: stohr@biomed.cas.cz<br />

Dr. Stanislaw Sulkowski<br />

Department of Pathology<br />

15-269 Bialystok POL<br />

Email: sulek@zeus.amb.edu.pl<br />

Pr. Young-Joon Surh<br />

College of Pharmacy<br />

151-742 Seoul KOR<br />

Email: surh@plaza.snu.ac.kr<br />

Mr. Olivier Tabary<br />

Insem U719<br />

75012 Paris FRA<br />

Email: olivier.tabary@stantoine.inserm.fr<br />

Mrs. Tia Sorsa<br />

2600 Glostrup DNK<br />

Email: ts@dcb-glostrup.dk<br />

Mrs. Konstantina Stathopoulou<br />

157 84 Athens GRC<br />

Email: kstath@biol.uoa.gr<br />

Mr. Michael Stilmann<br />

AG Scheidereit<br />

13125 Berlin DEU<br />

Email: Stilmann@gmx.de<br />

Mr. Markus Streiff<br />

CH-4002 Basel CHE<br />

Email:<br />

markus.streiff@novartis.com<br />

Mr. Yoshiaki Sunami<br />

13092 Berlin DEU<br />

Email: y.sunami@mdc-berlin.de<br />

Mrs. Chie Suzuki<br />

Department of Pharmacology<br />

651-2180 Kobe JPN<br />

Email:<br />

suzuki@pharm.kobegakuin.ac.jp<br />

Mrs. Fatiha Tabet<br />

Ottawa Health Research Institute<br />

K1H 8M5 Ottawa CAN<br />

Email: ftabet@uottawa.ca


Mrs. Aurélie Tacheny<br />

URBC<br />

5000 Namur BEL<br />

Email:<br />

aurelie.tacheny@fundp.ac.be<br />

Mr. Marie-Helene Teiten<br />

LBMCC<br />

L-2540 Luxemb<strong>our</strong>g LUX<br />

Email:<br />

marie_helene.teiten@lbmcc.lu<br />

Dr. Stefano Thellung<br />

Pharmacology-Departmento of<br />

biology oncology <strong>and</strong> genetics<br />

16132 Genova ITA<br />

Email: thellung@yahoo.com<br />

Mr. Lionel Tintignac<br />

Brian Hemmings lab/Friedrich<br />

Miescher Institute<br />

4058 Basel CHE<br />

Email: lionel.tintignac@fmi.ch<br />

Dr. Alex<strong>and</strong>ra Tomova<br />

Laboratory of Genetic Engineering<br />

of Pathogenic Microorganisms<br />

123098 Moscow RUS<br />

Email: altomova@abv.bg<br />

Mr. Felix Tritschler<br />

Laboratory of Molecular Neuro-<br />

Oncology, Department of General<br />

Neurology<br />

72076 Tuebingen DEU<br />

Email: f.tritschler@gmx.de<br />

Dr. SOPHIE TURBAN-<br />

RAJAONAH<br />

Molecular Physiology<br />

DD1 4HN Dundee GBR<br />

Email: s.turban@dundee.ac.uk<br />

Dr. Tetsuya Tanaka<br />

Department of Applied Genetics,<br />

Institut de Biologie et de Médecine<br />

Moléculaires<br />

B-6041 Gosselies BEL<br />

Email: tetsuyat2003@hotmail.com<br />

Mrs. Silvia Tejerina<br />

URBC<br />

5000 Namur BEL<br />

Email: silvia.tejerina@fundp.ac.be<br />

Dr. Marily Theodoropoulou<br />

D80804 Munich DEU<br />

Email: marily@mpipsykl.mpg.de<br />

Dr. Alex<strong>and</strong>er Tokmakov<br />

Genomic Sciences Center<br />

230-0045 Yokohama JPN<br />

Email: tokmak@gsc.riken.go.jp<br />

Dr. Grazina Treigyte<br />

Department of Developmental<br />

Biology<br />

LT-08662 Vilnius LTU<br />

Email: grazina.treigyte@bchi.lt<br />

Dr. Andreas Tsakalof<br />

Lab. of Biochemistry, School of<br />

Medicine<br />

41222 Larissa GRC<br />

Email: atsakal@med.uth.gr<br />

Dr. Jonathan Turner<br />

Institute of Immunology<br />

L-1950 Luxemb<strong>our</strong>g LUX<br />

Email:<br />

jonathan.turner@LNS.ETAT.LU<br />

Pr. Mary Taub<br />

Taub Laboratory, Biochemistry<br />

Dept. 140 Farber Hall<br />

14214 Buffalo, NY USA<br />

Email: biochtau@buffalo.edu<br />

Mr. Bjorn Textor<br />

Division of Signal Transduction<br />

<strong>and</strong> Growth Control<br />

Heidelberg Heidelberg DEU<br />

Email: b.textor@dkfz.de<br />

Mrs. Angelica Timofeeva<br />

Genetic Engineering<br />

121552 Moscow RUS<br />

Email: Angelica_T@cardio.ru<br />

Dr. Aviva Tolkovsky<br />

Biochemistry<br />

CB 2 1QW Cambridge GBR<br />

Email: amt@mole.bio.cam.ac.uk<br />

Dr. Cristina Trejo-Solis<br />

Neuroimmunology<br />

14269 Mexico City MEX<br />

Email: trejosolis@yahoo.com.mx<br />

Dr. Jurg Tschopp<br />

Dept. Biochemistry<br />

1066 Epalinges CHE<br />

Email: jurg.tschopp@unil.ch<br />

Mrs. Tzvetomira Tzanova<br />

LIMBP<br />

57070 Metz FRA<br />

Email: tzvete_tzanova@abv.bg


Dr. Lucia Maria Valatro<br />

Laboratori Nazionali del Sud<br />

95100 Catania ITA<br />

Email: valastro@lns.infn.it<br />

Dr. Michiel van den Brekel<br />

1066 CX Amsterdam NLD<br />

Email: m.vd.brekel@nki.nl<br />

Mr. Edwin van der Linden<br />

Dept. of Immunology<br />

3584 EA Utrecht NLD<br />

Email: E.v<strong>and</strong>erlinden-6@azu.nl<br />

Pr. Aleyde Van Eynde<br />

Afdeling Biochemie<br />

3000 Leuven BEL<br />

Email:<br />

Aleyde.VanEynde@med.kuleuven<br />

.be<br />

Dr. Kenneth van Golen<br />

48109 Ann Arbor, Michigan USA<br />

Email: kgolen@umich.edu<br />

Dr. Anne Van Langendonckt<br />

Gynaecology<br />

1200 Brussels BEL<br />

Email:<br />

anne.vanlangendonckt@gyne.ucl.a<br />

c.be<br />

Dr. Tom VANDEN Berghe<br />

Dept. of Molecular Biomedical<br />

Research<br />

B-9052 Zwijnaarde BEL<br />

Email:<br />

Tom.V<strong>and</strong>enberghe@dmbr.UGent<br />

.be<br />

Mrs. Marjan Van Cleemput<br />

LEGEST<br />

9000 Ghent BEL<br />

Email:<br />

marjan.vancleemput@ugent.be<br />

Pr. Peter J. Van den Elsen<br />

Division of Molecular Biology,<br />

Department of<br />

Immunohematology <strong>and</strong> Blood<br />

Transfusion<br />

2333 ZA Leiden NLD<br />

Email: pjvdelsen@lumc.nl<br />

Mrs. Kristan van der Vos<br />

Dept. of Immunology<br />

3584 EA Utrecht NLD<br />

Email: K.v<strong>and</strong>ervos-<br />

3@umcutrecht.nl<br />

Mr. Joost van Galen<br />

Dept. Pathology<br />

1081 HV Amsterdam NLD<br />

Email: j.vangalen@vumc.nl<br />

Mrs. Tanja van Harn<br />

Xtrack<br />

3572GA Utrecht NLD<br />

Email: T.vanHarn@students.uu.nl<br />

Mr. Jorg van Loosdregt<br />

Dept. of Immunology<br />

3584 EA Utrecht NLD<br />

Email: J.vanloosdregt@azu.nl<br />

Dr. Wim V<strong>and</strong>en Berghe<br />

LEGEST<br />

9000 Gent BEL<br />

Email: w.v<strong>and</strong>enberghe@ugent.be<br />

Dr. Mark Van de Casteele<br />

Diabetes Research Center<br />

1090 Brussels BEL<br />

Email: mvdcaste@vub.ac.be<br />

Dr. Patrick van der Holst<br />

LBPI<br />

l-1511 Luxemb<strong>our</strong>g LUX<br />

Email: patrick.v<strong>and</strong>erholst@ist.lu<br />

Mr. Dave van Ditmarsch<br />

Xtrack<br />

3572GA Utrecht NLD<br />

Email:<br />

D.vanDitmarsch@students.uu.nl<br />

Dr. Cynthia van Golen<br />

48109 Ann Arbor, Michigan USA<br />

Email: kgolen@umich.edu<br />

Mrs. Marijn van Jaarsveld<br />

Xtrack<br />

3572GA Utrecht NLD<br />

Email:<br />

M.T.M.vanJaarsveld@students.uu.<br />

nl<br />

Dr. André van Puijenbroek<br />

Target Discovery Unit<br />

5340 BH Oss NLD<br />

Email:<br />

<strong>and</strong>re.vanpuijenbroek@organon.co<br />

m<br />

Pr. Peter VANDENABEELE<br />

Dept. of Molecular Biomedical<br />

Research<br />

B-9052 Zwijnaarde BEL<br />

Email:<br />

Peter.V<strong>and</strong>enabeele@dmbr.UGent<br />

.be


Dr. Peter Vanhoutte<br />

Signalisation neuronale et<br />

Régulations géniques<br />

75005 Paris FRA<br />

Email:<br />

peter.vanhoutte@svn.jussieu.fr<br />

Mrs. Roberta Vené<br />

Experimental Oncology A<br />

16132 Genoa ITA<br />

Email: robertavene@yahoo.it<br />

Mrs. Elisabeth VERCAMMEN<br />

Dept. of Molecular Biomedical<br />

Research<br />

B-9052 Zwijnaarde BEL<br />

Email: Elisav@dmbr.UGent.be<br />

Mr. Julien Verrax<br />

Pharmacokinetic, Metabolism,<br />

Nutrition <strong>and</strong> Toxicology<br />

1200 Bruxelles BEL<br />

Email:<br />

julien.verrax@pmnt.ucl.ac.be<br />

Mr. Eduardo Villamor<br />

6202 AZ Maastricht NLD<br />

Email: eiv@paed.azm.nl<br />

Pr. Athanase Visvikis<br />

MAEM UMR 7567<br />

54500 Nancy FRA<br />

Email: visvikis@maem.uhpnancy.fr<br />

Mr. J Willem Voncken<br />

Div. Molecular Genetics<br />

6229 ER Maastricht NLD<br />

Email:<br />

w.voncken@gen.unimaas.nl<br />

Mrs. Nele VANLANGENAKKER<br />

Dept. of Molecular Biomedical<br />

Research<br />

B-9052 Zwijnaarde BEL<br />

Email: Nelev@dmbr.UGent.be<br />

Dr. Valentina Venezia<br />

Pharmacology-Department of<br />

oncology biologi <strong>and</strong> genetics<br />

16132 Genova ITA<br />

Email: venezia@unige.it<br />

Mrs. Liesbeth Verhagen<br />

Dept. of Immunology<br />

3584 EA Utrecht NLD<br />

Email: L.verhagen@azu.nl<br />

Mrs. Lynn VERSTREPEN<br />

Dept. of Molecular Biomedical<br />

Research<br />

B-9052 Zwijnaarde BEL<br />

Email: Lynnv@dmbr.UGent.be<br />

Dr. Maria Vinciguerra<br />

Prof. Anna Maria Musti<br />

80131 Napoli ITA<br />

Email: kachera@libero.it<br />

Mr. Michael Vogt<br />

52074 Aachen DEU<br />

Email: michaelvogt1@gmx.de<br />

Dr. James Wachira<br />

department of biology<br />

21251 baltimore USA<br />

Email: jwachir@jewel.morgan.edu<br />

Mrs. Emilie VELOT<br />

Laboratoire de Recherche Cardio-<br />

Vasculaire<br />

1150 LUXEMBOURG LUX<br />

Email: emilie.velot@crp-sante.lu<br />

Dr. Donatella Verbanac<br />

Program Office<br />

10 000 Zagreb HRV<br />

Email: dverbanac@pliva.hr<br />

Dr. Helene Verheije<br />

Virology<br />

3584 CL Utrecht NLD<br />

Email: h.verheije@vet.uu.nl<br />

Dr. Valentina Villa<br />

pharmacology-department of<br />

oncology biology <strong>and</strong> genetics<br />

16132 Genova ITA<br />

Email: valentina.villa@unige.it<br />

Mrs. Elodie VIRY<br />

LIMBP<br />

57070 Metz FRA<br />

Email: elodieviry@hotmail.fr<br />

Mr. Joerg von Hagen<br />

LSA R&D<br />

64271 Darmstadt DEU<br />

Email: joerg.von.hagen@merck.de<br />

Dr. Vicki Waetzig<br />

24105 Kiel DEU<br />

Email:<br />

vicki.waetzig@pharmakologie.uni<br />

-kiel.de


Mrs. Jessica Wagener<br />

40225 Duesseldorf DEU<br />

Email: jessica.wagener@gmx.de<br />

Mr. Piotr Wardega<br />

751 24 Uppsala SWE<br />

Email:<br />

Piotr.Wardega@LICR.uu.se<br />

Dr. Thomas Welss<br />

Skin / Hair Physiology -<br />

Development Screening Systems<br />

D-40191 Duesseldorf DEU<br />

Email: thomas.welss@henkel.com<br />

Dr. Johanna Westra<br />

dept Rheumatology<br />

9713 GZ Groningen NLD<br />

Email: j.westra@med.umcg.nl<br />

Dr. Robin SB Williams<br />

Department of Biology<br />

WC1 E6BT London GBR<br />

Email: robin.williams@ucl.ac.uk<br />

Mrs. Ellen WIRAWAN<br />

Dept. of Molecular Biomedical<br />

Research<br />

B-9052 Zwijnaarde BEL<br />

Email: Ellenw@dmbr.UGent.be<br />

Dr. Verena Wünsche<br />

65824 Schwalbach/Ts DEU<br />

Email:<br />

verena.wuensche@merckbioscienc<br />

es.de<br />

Mrs. Samantha Wales<br />

Virology <strong>and</strong> Immunology<br />

21201 Baltimore USA<br />

Email: sqwales@yahoo.com<br />

Pr. Manfred Weiss<br />

Abteilung Klinische<br />

Anaesthesiologie<br />

89075 Ulm DEU<br />

Email:<br />

manfred.weiss.ulm@online.de<br />

Dr. Sibylle Wenzel<br />

Institute of Physiology<br />

35392 Giessen DEU<br />

Email: sibylle.wenzel@gmx.de<br />

Pr. Linda R. White<br />

Department of Neurology<br />

N-7006 Trondheim NOR<br />

Email: linda.white@ntnu.no<br />

Pr. Pat Wilson<br />

Renal Cell Biology<br />

NW3 2PF London GBR<br />

Email: pat.wilson@mssm.edu<br />

Mr. Jiong Wu<br />

1915 Beverly, Massachusetts USA<br />

Email: mdolan@cellsignal.com<br />

Mrs. Georgia Xenaki<br />

Molecular Pharmacology, Dr. C.<br />

Demonacos<br />

M13 9PL Manchester GBR<br />

Email:<br />

georgiaxenaki@hotmail.com<br />

Dr. Yihua Wang<br />

Laboratory of Cell <strong>and</strong> Molecular<br />

Biology<br />

100021 Beijing CHN<br />

Email: yihua_wang@hotmail.com<br />

Dr. Valerie Wells<br />

Cell Signalling laboratory,<br />

Pharmaceutical Sciences Research<br />

Division<br />

SE1 9NH London GBR<br />

Email: valerie.wells@kcl.ac.uk<br />

Mrs. Shannon Werner<br />

Signaling Systems Laboratory<br />

92093-0375 La Jolla, CA USA<br />

Email: slwerner@ucsd.edu<br />

Mrs. Christina Wieg<strong>and</strong><br />

40202 Louisville, KY USA<br />

Email:<br />

cbwieg01@gwise.louisville.edu<br />

Dr. Andrzej Wincewicz<br />

Department of Pathology<br />

15-269 Bialystok POL<br />

Email: sulek@zeus.amb.edu.pl<br />

Mrs. Kun Wu<br />

Nutrition <strong>and</strong> Food Hygiene<br />

Department<br />

150086 Harbin CHN<br />

Email: wukun@public.hr.hl.cn<br />

Dr. Ying Xia<br />

Department of Environmental<br />

Health<br />

45267 Cincinnati USA<br />

Email: xiay@email.uc.edu


Pr. Ningzhi Xu<br />

Laboratory of Cell <strong>and</strong> Molecular<br />

Biology<br />

100021 Beijing CHN<br />

Email: xningzhi@public.bta.net.cn<br />

Dr. Katsuji Yoshioka<br />

Cancer Research Institute<br />

920-0934 Kanazawa JPN<br />

Email: ysok-f01@siren.ocn.ne.jp<br />

Mrs. Maya Margaritova Zaharieva<br />

Experimental Chemotherapy<br />

1000 Sofia BGR<br />

Email: zaharieva26@yahoo.com<br />

Dr. Jana Zdychova<br />

VÌde&#328<br />

14021 Prague CZE<br />

Email:<br />

jana.zdychova@medicon.cz<br />

Dr. Margherita Zennaro<br />

Dept of Biochemistry <strong>and</strong><br />

Molecular Biology<br />

44100 Ferrara ITA<br />

Email:<br />

margheritazennaro@yahoo.it<br />

Mrs. Jun Zhao<br />

Physiological Chemistry<br />

3584 CG Utrecht NLD<br />

Email: j.zhao@med.uu.nl<br />

Pr. Aless<strong>and</strong>ra Zicari<br />

IMMUNO_BIOLOGY<br />

161 Rome ITA<br />

Email:<br />

aless<strong>and</strong>ra.zicari@uniroma1.it<br />

Dr. Michael B. Yaffe<br />

Center for Cancer Research<br />

2139 Cambridge, MA USA<br />

Email: myaffe@mit.edu<br />

Mr. Ji Hoon YU<br />

Department of Pharmacology<br />

120-752 Seoul KOR<br />

Email: jihoonyu@hotmail.com<br />

Dr. Peter Zalewski<br />

Department of Medicine<br />

5011 Woodville AUS<br />

Email:<br />

peter.zalewski@adelaide.edu.au<br />

Dr. Alain G. Zeimet<br />

6020 Innsbruck AUT<br />

Email: alain.zeimet@uibk.ac.at<br />

Dr. Ying Zhang<br />

Cellular And Molecular Biology<br />

20892-4256 Bethesda USA<br />

Email: yingz@helix.nih.gov<br />

Dr. Yuan Zhu<br />

Center of Reproductive Medicine,<br />

430030 Wuhan city, Hubei<br />

province CHN<br />

Email:<br />

huyuan0528@yahoo.com.cn<br />

Mrs. Rhyenne Zimmerman<br />

Biological Chemistry, Leiden<br />

University<br />

2333 CC Leiden NLD<br />

Email:<br />

r.zimmerman@chem.leidenuniv.nl<br />

Mrs. Christina Westmose Yde<br />

Biomedical Research <strong>and</strong><br />

Molecular Cell Biology Group<br />

5230 Odense M DNK<br />

Email: chriswy@bmb.sdu.dk<br />

Dr. Tülay Yucel-Lindberg<br />

141 04 Huddinge SWE<br />

Email: tulay.lindberg@ki.se<br />

Mrs. Darina Zaskodova<br />

Department of Medical<br />

Biochemistry<br />

500 38 Hradec Kralove CZE<br />

Email: zaskodovad@lfhk.cuni.cz<br />

Dr. Marina Zenkova<br />

630090 Novosibirsk RUS<br />

Email: marzen@niboch.nsc.ru<br />

Mr. Zhongchun Zhang<br />

Physiological Chemistry<br />

3584 CG Utrecht NLD<br />

Email: Z.Zhang@med.uu.nl<br />

Dr. Jia Zhuo<br />

Receptor <strong>and</strong> Signal Transduction<br />

48202 Detroit USA<br />

Email: jzhuo1@hfhs.org<br />

Mrs. Bea Zoer<br />

6200 MD Maastricht NLD<br />

Email:<br />

bea.zoer@farmaco.unimaas.nl


Pr. Moncef ZOUALI<br />

Centre Viggo Petersen<br />

75475 Paris Cedex 10 Paris FRA<br />

Email: moncef.zouali@wanadoo.fr

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

Saved successfully!

Ooh no, something went wrong!