30.01.2013 Views

Abstract book (download .pdf file) - Redox and Inflammation ...

Abstract book (download .pdf file) - Redox and Inflammation ...

Abstract book (download .pdf file) - Redox and Inflammation ...

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.

Luxembourg,<br />

January 26th to 28th, 2011<br />

New Conference Center Kirchberg<br />

NCCK - Luxembourg<br />

Cell Signal-omics 2011<br />

Integrated cellular pathology<br />

Systems biology of human disease<br />

Proceedings<br />

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

Program<br />

Marc Diederich <strong>and</strong> Guido Kroemer<br />

Organized by<br />

Recherches Scientifiques Luxembourg asbl<br />

Under the auspices of<br />

the European Research Institute for Integrated Cellular Pathology


����������������<br />

�����������<br />

������������������<br />

������������������<br />

��������� �����<br />

�����������������������<br />

�����������<br />

����������������������� �������������� ����� � ��������������� �������������������<br />

������ �������� ������������ �������� ���������� � �������� ��� �� ������������� ������� ����� ��������������� � ������� ����� �� �


Luxembourg,<br />

January 26th to 28th, 2011<br />

New Conference Center Kirchberg<br />

NCCK - Luxembourg<br />

Cell Signal-omics 2011<br />

Integrated cellular pathology - Systems biology of human disease<br />

Proceedings <strong>and</strong> Program<br />

Editor<br />

Marc Diederich<br />

This meeting is organized <strong>and</strong> financed by<br />

Recherches Scientifiques Luxembourg asbl<br />

Under the auspices of<br />

the European Research Institute for Integrated Cellular Pathology<br />

This meeting is co-financed by<br />

the Fonds National de la Recherche, Luxembourg<br />

Printing of the Proceedings sponsored by<br />

the Fondation de Recherche Cancer et Sang (Luxembourg)


Table of content<br />

!"#$%&#'''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''()<br />

*&+,-./#012#,34''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''5)<br />

6#,#"%/)7,$-"2%38-,''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''9)<br />

:;


Preface<br />

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

expression in Luxembourg. This type of meeting was originally thought to teach doctoral<br />

students of the molecular <strong>and</strong> cellular biology master training program of the University<br />

of Nancy I (France).<br />

Since then, more than 6000 fundamental, clinical <strong>and</strong> industrial researchers were<br />

gathering in Luxembourg for eight different meetings in order to discuss therapeutic<br />

applications in the field of signal transduction, transcription <strong>and</strong> translation related to novel<br />

therapeutic applications. These meetings allow new insights into this rapidly moving field.<br />

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

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

therapeutic approaches for the next century.<br />

For the 2011 edition of meeting, with 350 participants, I am convinced that this meeting will<br />

be a great success.<br />

Welcome to Luxembourg!<br />

Marc Diederich<br />

1


Acknowledgments<br />

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

Recherches Scientifiques asbl<br />

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

The Fonds National de la Recherche, Luxembourg<br />

Under the auspices of :<br />

the European Research Institute for Integrated Cellular Pathology<br />

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

The Fonds National de la Recherche, Luxembourg<br />

Recherches Scientifiques asbl<br />

Kuwait Petroleum SA - Luxembourg<br />

The City of Luxembourg<br />

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

Computer Home, Luxembourg<br />

Canon Luxembourg<br />

Your onsite organization team:<br />

Claudia Cerella, Sebastien Chateauvieux, Marc Diederich, Cristina Florean, Elodie Frenger,<br />

Francois Gaascht, Anthoula Gaigneaux, Stefania Gonfloni, Cindy Gr<strong>and</strong>jenette, Christina<br />

Grigorakaki, Sheherazade Hajjouli, Estelle Henry, Liliane Hermes, Monika Jain, Tommy<br />

Karius, Mareike Kelkel, Noemie Legr<strong>and</strong>, Fabienne Mack, Franck Morceau, Florian Muller,<br />

Karoline Noworyta, Marie-Anne Olinger, Barbora Orlikova, Michael Schnekenburger,<br />

Christiane Schuld, Marc Schumacher, Carole Seidel, Cyril Sobolewski, Marie-Helene Teiten,<br />

Anne Trecul, Elodie Viry.<br />

3


European Research Institute for Integrated Cellular Pathology (ERI-ICP)<br />

About ERI-ICP<br />

ERI-ICP is a professional organization with some 100 Founding Members including many of<br />

the most prominent Europea researchers (<strong>and</strong> several Nobel laureates) working on integrated<br />

cellular pathology. The principal objective of ERI-ICP, a virtual research Institute, will be to<br />

promote cell biology research applied to major human diseases in Europe, including (but not<br />

limited to) neurodegenerative diseases, myopathies, mitochondriopathies, infectious diseases,<br />

cancer <strong>and</strong> pathological aging. ERI-ICP will promote the implementation <strong>and</strong> utilization of<br />

systems biology (or integrated biology) by establishing a continuum between fundamental,<br />

translational <strong>and</strong> clinical research. ERI-ICP will strive for excellence, independence,<br />

leadership, diversity <strong>and</strong> flexibility. Throughout its work, ERI-ICP will provide independent,<br />

authoritative <strong>and</strong> evidence-based advice to underpin policy for stimulating the implementation<br />

of systems biology applicable to the investigation, classification, diagnosis <strong>and</strong> therapeutic<br />

management of major human diseases.<br />

ERI-ICP will host a website <strong>and</strong> will disseminate “official” (but ameliorable) methods <strong>and</strong><br />

protocols applicable to integrated cellular pathology. ERI-ICP will also participate in efforts<br />

of data integration, provide a web forum for researchers working in the area of systems<br />

biology applied to human disease, <strong>and</strong> organize annual conferences. By the conjunction of<br />

these activities, ERI-ICP should be able to set up the st<strong>and</strong>ards for data quality, for data<br />

exchange <strong>and</strong> storage that are crucial for systems biology, as applied to human disease.<br />

Therefore, ERI-ICP will serve as an instrument to crystallize <strong>and</strong> streamline all efforts within<br />

this area in the European Research Area. The Founding Members of ERI-ICP have been<br />

selected among highly renowned scientists from all areas of disease-oriented fundamental <strong>and</strong><br />

clinical research. The Founding Group will be responsible for defining the overall objectives<br />

of the Institute, addressing issues such as credibility of the institution, membership <strong>and</strong><br />

governance. To steer the process, a ‘Governing Body’ consisting of a President, a Vice-<br />

President, a Secretary-General <strong>and</strong> five fellows representing all disciplines Europe-wide, will<br />

be established. To address issues as they arise, ad hoc taskforces will be organized amongst<br />

members of the Institute, specific to their area of expertise. As a Founding Member of<br />

ERIICP you can anticipate contributing to the preparation of position papers, interacting with<br />

EU <strong>and</strong> national institutions to facilitate proposals <strong>and</strong> the implementation of new policies<br />

that ensure that disease-oriented systems biology remain at the top of the European research<br />

agenda.<br />

The official launch of ERI-ICP will be one of the highlights at the opening of the 1st<br />

Conference on Integrated Cellular Pathology, at the Pasteur Institute, in Paris, on April 22-24,<br />

2010. The second annual conference is planned for January 26-29, 2011, in Luxembourg.<br />

http://kroemerlab.com/ERI-ICP_About.html<br />

5


General Information<br />

Meeting Venue<br />

All meeting sessions are held at the New Congress Center Kirchberg<br />

4, Place de l’Europe<br />

L-1499 Luxembourg-Kirchberg<br />

This new building is hidden located behind the Philharmonie Concert Hall (left side) <strong>and</strong> is in<br />

fact integrated into the basis of the white Tower building (see map with photos for details).<br />

Registration will take place in the at the registration desk open daily (9h00-19h00).<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 13h00 - 15h00. Additional<br />

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

Exhibits are open daily form January 26 th to 28 th , 2011.<br />

The Exhibit opens on Wednesday January 26 th (Coffee break 10h30) <strong>and</strong> ends on Friday<br />

January 28 st , 2011 after the afternoon coffee break.<br />

Posters<br />

All posters will be up from Wednesday January 26 th until Friday January 28 th :<br />

Wednesday January 26 th 14h00 – 16h00: Posters with even numbers<br />

Thursday January 27 th 14h00 – 16h00: Posters with odd numbers<br />

Friday January 28 th 14h00 – 16h00: All posters<br />

On Friday January 28 th 16h00, all poster presenters are requested to recover their<br />

posters (the boards will be removed).<br />

Welcome reception offered by the City of Luxembourg (expo area)<br />

On Wednesday January 26, 2011 from 19h30-20h30 at the expo surface.<br />

Gala Dinner<br />

The Gala Dinner will take place at the Hôtel Le Royal 12, boulevard Royal L-2449<br />

Luxembourg Tel: + 352 24 16 16-1, Fax: + 352 22 59 48 on Thursday, January 27th starting<br />

at 20h30. Additional tickets are NOT available at the registration desk.<br />

Transportation<br />

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

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

from your hotel depending on the distance to the meeting center (Check timetable for details).<br />

The busses are red, orange <strong>and</strong> yellow <strong>and</strong> are from the bus company "Demy Cars".<br />

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

7


How to reach the congress center?<br />

- By taxi:<br />

from the airport (Luxembourg - 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<br />

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

Luxembourg-Centre"<br />

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

Plateau de Kirchberg Aéroport, choose exit number "8", orient towards "Quartier Europeen<br />

Sud Luxembourg Centre"<br />

From Germany: Highway A6 (E44) from Trier, direction Luxembourg, choose exit<br />

number "8", orient towards "Quartier Europeen Sud Luxembourg Centre".<br />

8


9<br />

Exhibition


Many thanks to all exhibitors<br />

Please visit our Expo !<br />

10


11<br />

Scientific Program


Wednesday January 26th, 2011<br />

8h00 - 19h00: Registration<br />

Keynote session:<br />

Chair: Marc Diederich (LBMCC, Luxembourg)<br />

9h30 - 10h30 Mario Capecchi (University of Utah School of Medicine, Salt Lake City, USA):<br />

Gene Targeting into the 21st Century: Mouse Models of Human Disease from Cancer to<br />

Neuropsychiatric Disorders<br />

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

Session 1: Cell death<br />

Chair: Marc Diederich (LBMCC, Luxembourg)<br />

11h00 - 11h30<br />

11h30 - 12h00<br />

12h00 - 12h30<br />

12h30 - 12h45<br />

Eileen White (Rutgers University, New Brunswick, NJ, USA):<br />

Therapeutic targeting of death pathways in cancer: mechanisms for activating cell death in<br />

cancer cells.<br />

Gerry Melino (University of Rome Tor Vergata, Italy):<br />

Why we need the p53 family? The Guardian of maternal reproduction<br />

Stefania Gonfloni (University of Rome Tor Vergata, Dept of Biology, Italy):<br />

DNA-damage stress response in female germ cells: is Abl a fine tuner or a dangerous<br />

amplifier?<br />

Cinzia Di Pietro (Università degli Studi di Catania, Italy):<br />

Altered expression of Apoptotic Machinery genes explains the decrease of oocytes<br />

competence with aging<br />

12h45 – 13h00<br />

Inna N. Lavrik (DKFZ, Heidelberg, Germany):<br />

Towards underst<strong>and</strong>ing of life <strong>and</strong> death regulation at CD95/Fas<br />

13h00 - 14h00 Lunch <strong>and</strong> visit of the expo<br />

Workshops 1 <strong>and</strong> Poster Session 1<br />

14h00 - 15h00<br />

15h00 - 16h00<br />

Géraldine Guérin-Peyrou (Polyplus-transfection, France)<br />

Winfried van Eyndhoven (Agilent Technologies, The Netherl<strong>and</strong>s)<br />

Karoly Szuhai (Leiden University Medical Center Dept, The Netherl<strong>and</strong>s)<br />

Ian Majewski (Netherl<strong>and</strong>s Cancer Institute, The Netherl<strong>and</strong>s)<br />

14h00 - 15h30 Poster session (Even numbers) <strong>and</strong> visit of the expo<br />

15h30 – 16h00 Coffee break <strong>and</strong> visit of the expo<br />

13


Session 2: Cell signaling<br />

Chair: Ilya Shmulevich (Institute for Systems Biology, Seattle, USA)<br />

16h00 - 16h30<br />

Ilya Shmulevich (Institute for Systems Biology, Seattle, USA):<br />

Computational Systems Biology in Cancer Research<br />

Varadharajan Sundaramurthy (Max Planck Institute of Molecular Cell Biology <strong>and</strong><br />

16h30 - 17h00<br />

Genetics, Dresden, Germany):<br />

Systems Analysis of Endocytosis by Quantitative Image Analysis: Insights into Pathogenetic<br />

Mechanisms<br />

17h00 - 17h30<br />

Anne Grosse-Wilde (Institute for Systems Biology, Seattle, USA):<br />

Using single cell analysis tools to search for human breast cancer stem cells<br />

17h30 - 18h00<br />

Josef Penninger (Institute of Molecular Biotechnology, Vienna, Austria):<br />

Whole genome scans in drosophila to model human disease<br />

18h00 - 18h15<br />

Sébastien Chateauvieux (LBMCC, Luxembourg):<br />

Valproic acid perturbs hematopoietic differentiation networks<br />

18h15 - 18h30<br />

Ivana Scovassi (Istituto di Genetica Molecolare CNR, Pavia, Italy):<br />

Search for the effects of anticancer drugs: A look to apoptosis but also to autophagy<br />

18h30 - 18h45<br />

Vladimir L. Katanaev (University of Konstanz, Germany):<br />

Signaling by the Frizzled family of receptors: G protein-coupled mechanisms <strong>and</strong> targets<br />

19h00 - 19h30 Official talks<br />

19h30 - 20h30 Reception offered by the City of Luxembourg <strong>and</strong> visit of the expo<br />

20h30 - 21h00 Shuttles leave to the hotels from the meeting center<br />

Thursday January 27th, 2011<br />

Shuttle bus from the hotels to the meeting center (see map for details)<br />

8h00 - 19h00: Registration<br />

Session 3: Transcriptional control<br />

Chair: Young-Joon Surh (Seoul National University, South Korea)<br />

9h00 - 9h30<br />

Young-Joon Surh (Seoul National University, South Korea):<br />

<strong>Redox</strong> regulation of transcription factors by electrophilic lipid mediators<br />

9h30 - 10h00<br />

Luciano Di Croce (Center for Genomic Regulation, Barcelona, Spain):<br />

Novel mechanisms of transcriptional control in differentiation <strong>and</strong> cancer<br />

10h00 - 10h30<br />

Thomas Luft (University of Heidelberg, Germany):<br />

A dual role of JAK1: regulation of IFN-! dependent <strong>and</strong> independent IL-12p70 production<br />

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

Session 4: Immunology<br />

Chair: Eileen White (Rutgers University, New Brunswick, NJ, USA)<br />

11h00 - 11h30<br />

Bali Pulendran (Emory University, Atlanta, USA):<br />

Learning immunology from successful vaccines: innate immunity to systems vaccinology<br />

11h30 - 12h00<br />

Marie-Lise Gougeon (Institut Pasteur, Paris, France):<br />

Immunity <strong>and</strong> death in chronic viral infections<br />

Kasper Hoebe (University of Cincinnati College of Medicine, USA):<br />

12h00 - 12h30 Reduced T cell survival leads to loss of T cell quiescence <strong>and</strong> subsequent immunopathology<br />

in Gimap5-deficient mice<br />

12h30 - 13h00<br />

Claude Condé (Virology <strong>and</strong> immunology unit, GIGA-research, University of Liège,<br />

Belgium): Study of the anti-apoptotic role of SHIP-1 in T cells<br />

13h00 - 16h00 Lunch <strong>and</strong> visit of the expo<br />

14


Workshops 2 <strong>and</strong> Posters Session 2<br />

14h00 - 15h00 Sarah Payne (GE Healthcare Europe GmbH)<br />

15h00 - 16h00 Alex Sim (AMSBIO)<br />

14h00 - 15h30 Poster session (Odd numbers) <strong>and</strong> visit of the expo<br />

15h30 - 16h00 Coffee break <strong>and</strong> visit of the expo<br />

Session 5: Proteomics<br />

Chair: Gerry Melino (University of Rome Tor Vergata, Italy)<br />

16h00 - 16h30<br />

Antonio del Sol (Université de Luxembourg, Gr<strong>and</strong>-Duchy of Luxembourg):<br />

Protein population shift leading to disease-related changes in cellular networks<br />

16h30 - 17h00<br />

Bernd Wollscheid (Institute of Molecular Systems Biology, Zürich, Switzerl<strong>and</strong>):<br />

Proteomic phenotyping of the cell surface information gateway<br />

17h00 - 17h30<br />

Pascale Cossart (Institut Pasteur, Paris, France):<br />

De-SUMOylation: a new strategy used by Listeria to counteract the host defense<br />

17h30 - 18h00<br />

Christopher Overall (The University of British Columbia, Canada):<br />

To the Ends of the Proteome World: Multiplex Quantitative N <strong>and</strong> C-Terminomics<br />

18h00 - 18h15<br />

Margarida Fardilha (University of Aveiro, Portugal):<br />

Identification of the Human Testis Protein Phosphatase 1 Interactome<br />

Alfred C. O. Vertegaal (LUMC, Leiden, The Netherl<strong>and</strong>s):<br />

18h15 - 18h30 Site-Specific Identification of SUMO-2 Conjugation Sites in Cells Reveals Novel<br />

SUMOylation Motifs<br />

Angela Brieger (Goethe-University, Frankfurt a.M., Germany):<br />

18h30 - 18h45 Regulation of Lynch syndrome-related DNA mismatch repair heterodimer MutLalpha by<br />

phosphorylation?<br />

19h00 Shuttles leave to the hotels from the meeting center<br />

Friday January 28th, 2011<br />

Shuttle bus from the hotels to the meeting center (see map for details)<br />

8h00 - 19h00: Registration<br />

Session 6: Epigenetics<br />

Chair: Luciano Di Croce (Center for Genomic Regulation, Barcelona, Spain)<br />

9h00 - 9h30<br />

François Fuks (ULB, Brussels, Belgium):<br />

Mechanisms of Epigenetics in Health <strong>and</strong> Disease<br />

9h30 - 10h00<br />

Michael Bots (Peter MacCallum Cancer Centre, Victoria, Australia):<br />

Treating Cancer with drugs that target the epigenome<br />

10h00 - 10h30<br />

Roberto Gambari (University of Ferrara, Italy):<br />

MicroRNA <strong>and</strong> erythroid differentiation<br />

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

15


Session 7: Cancer signaling networks<br />

Chair: Dean W. Felsher (Stanford University, USA)<br />

11h00 - 11h30<br />

Andrei Zinovyev (Institut Curie, Paris, France):<br />

Computational Systems Biology of Cancer: modeling cell fate decision <strong>and</strong> disease outcome<br />

11h30 - 12h00<br />

Dean W. Felsher (Stanford University, USA):<br />

A Systems Approach to Modeling <strong>and</strong> Predicting Oncogene Addiction<br />

12h00 - 12h30<br />

Alex<strong>and</strong>er R. A. Anderson (H. Lee Moffitt Cancer Center, USA):<br />

Microenvironmental Independence In Tumor Progression: An Integrated Approach<br />

Evan Keller (University of Michigan, USA):<br />

12h30 - 13h00 Targeting the invasive phenotype using systemic evolution of lig<strong>and</strong>s by exponential<br />

enrichment creates anti-metastatic aptamers<br />

13h00 - 14h00 Lunch <strong>and</strong> visit of the expo<br />

Session 8: Gene expression networks in health <strong>and</strong> disease<br />

Chair: Guido Kroemer (Institut Gustave Roussy, Paris France)<br />

14h00 - 14h30 Guido Kroemer (Institut Gustave Roussy, Paris France):<br />

Autophagy - suicidal self-cannibalism or homeostatic recycling?<br />

14h30 - 15h00 Yong Sang Song (Cancer Research Institute, Seoul National University, Korea):<br />

Genes related to energy metabolism differ the visceral adipose-derived stem cells from the<br />

subcutaneous ones<br />

15h00 – 15h15 Andrew Koff (Memorial Sloan-Kettering Cancer Center, NY, USA):<br />

A bioinformatic approach to illuminate why a biomarker, p27, has prognostic value in<br />

human tumors<br />

15h15 – 15h30 Stefanie Kaempf (NonWoTecc Medical GmbH, Cologne, Germany):<br />

Pathological aspects of vascular aging <strong>and</strong> strategies for new biomimetic <strong>and</strong> biological<br />

active NonWoTecc biomaterials<br />

15h30 – 16h00 Coffee break <strong>and</strong> visit of the expo<br />

Session 9: Neurodegenerative diseases<br />

Chair: Vittorio Calabrese (University of Catania, Catania, Italy)<br />

16h00 - 16h30<br />

16h30 - 17h00<br />

17h00 - 17h30<br />

17h30 - 18h00<br />

Mark P. Mattson (Laboratory of Neurosciences, National Institute on Aging Intramural<br />

Research Program, Baltimore, MD. USA):<br />

Signaling Pathways that Help Neurons Help Themselves<br />

Junying Yuan (Harvard Medical School, USA):<br />

Genome-wide analysis reveals mechanisms modulating autophagy in normal brain aging <strong>and</strong><br />

in Alzheimer's disease<br />

Vittorio Calabrese (University of Catania, Catania, Italy):<br />

Vitagenes, metabolic stress <strong>and</strong> Hormesis in aging <strong>and</strong> Neurodegenerative disorders<br />

Xin-Fu Zhou (Flinders University, Adelaide, South Australia):<br />

p75NTR regulates Ab deposition by increasing Ab production but inhibiting Ab aggregation<br />

with its extracellular domain<br />

18h00 - 18h30<br />

Paul R. Fisher (La Trobe University, Melbourne, Australia):<br />

A “no-brainer” – mitochondrial <strong>and</strong> neurodegenerative disease in Dictyostelium.<br />

18h30 End of the meeting<br />

18h30 Shuttles leave to the hotels from the meeting center<br />

16


Oral presentations<br />

(In chronological order)<br />

17


Gene Targeting into the 21 st Century: Mouse Models of Human Disease from Cancer to<br />

Neuropsychiatric Disorders<br />

Mario Capecchi<br />

University of Utah School of Medicine, Salt Lake City, USA<br />

Gene targeting allows the designed modification of any gene in the mouse genome. Since<br />

genes impact all biological phenomena this methodology can be used to study any biological<br />

phenomena common to mammals in the mouse. We are using it to model human disease in<br />

the mouse. The models can be used to analyze the pathology of the disease at a level not<br />

feasible in humans <strong>and</strong> as a platform for the development of new therapeutic protocols. I will<br />

discuss modeling of a human cancer, synovial sarcoma, <strong>and</strong> a neuropsychiatric disorder,<br />

obsessive compulsive (OCD) spectrum disorder in the mouse. The former provides insight<br />

into the molecular biology of cancer. The latter provides the unexpected conclusion that<br />

microglia derived from bone marrow may be controlling behavior.<br />

18


Activated Ras Requires Autophagy to Maintain Oxidative Metabolism <strong>and</strong><br />

Tumorigenesis<br />

Eileen White<br />

The Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ<br />

08903<br />

Autophagy is a catabolic pathway utilized by cells to support metabolism in response to<br />

starvation <strong>and</strong> to clear damaged proteins <strong>and</strong> organelles in response to stress. We report here<br />

that expression of a H-ras V12 or K-ras V12 oncogene upregulates basal autophagy, which is<br />

required for tumor cell survival in starvation <strong>and</strong> in tumorigenesis. In Ras-expressing cells,<br />

defective autophagosome formation or cargo delivery causes accumulation of abnormal<br />

mitochondria <strong>and</strong> reduced oxygen consumption. Autophagy defects also lead to tricarboxylic<br />

acid (TCA) cycle metabolite <strong>and</strong> energy depletion in starvation. As mitochondria sustain<br />

viability of Ras-expressing cells in starvation, autophagy is required to maintain the pool of<br />

functional mitochondria necessary to support growth of Ras-driven tumors. Human cancer<br />

cell lines bearing activating mutations in Ras commonly have high levels of basal autophagy,<br />

<strong>and</strong> in a subset of these, down-regulating the expression of essential autophagy proteins<br />

impaired cell growth. As cancers with Ras mutations have a poor prognosis, this “autophagy<br />

addiction” suggests that targeting autophagy <strong>and</strong> mitochondrial metabolism are valuable new<br />

approaches to treat these aggressive cancers.<br />

19


p53 family, its involvement in neuronal development via miR-34a:<br />

Why we need them?<br />

Melino G,<br />

University Tor Vergata, Rome, Italy; Medical Research Council, Toxicology Unit,<br />

Leicester, UK<br />

In the last ten years, p63 <strong>and</strong> p73 have been identified as the ancestral members of the p53<br />

family. Despite the high sequence <strong>and</strong> structural similarity, the mouse knockouts revealed a<br />

crucial role in neural development for p73 <strong>and</strong> in epidermal formation for p63. We identified<br />

several transcriptional targets, the mechanisms of regulation of cell death, <strong>and</strong> the p63<br />

isoform involved in epithelial development. Both genes are involved in female infertility <strong>and</strong><br />

maternal reproduction as well as in cancer formation, although with distinct mechanisms.<br />

p73 steady state protein levels are kept low under normal physiological conditions through<br />

degradation by the 26S proteasome, mediated by the HECT-containing E3 ubiquitin ligase<br />

ITCH, for which we are developing an inhibitor. We have also described additional<br />

mechanisms of degradation: (1) the orphan F-box protein FBXO45 ; (2) the ring finger<br />

domain ubiquitin ligase PIR2 (p73-induced Ring Finger 2), <strong>and</strong> (3) the antizyme ubiquitinindependent,<br />

proteasome-dependent pathway, both specific for the !Np73 isoforms<br />

Here, we describe the involvement of p73 in neuronal development. TAp73 knockout mice<br />

(TW Mak G&D 2008) show hippocampal dysegensis. Conversely, !Np73 knockout mice<br />

(TW Mak G&D 2010) show sign of moderate neurodegeneration with a significant loss of<br />

cellularity in the cortex. TAp73 is able to drive the expression of miR-34a, acting on specific<br />

binding sites present on the miR-34a promoter. In agreement with these in vitro data, miR-<br />

34a transcript expression is significantly reduced in vivo both in the cortex <strong>and</strong> hippocampus<br />

of p73-/- mice. In keeping, we show a role for miR-34a, in parallel to TAp73 expression,<br />

during in vitro differentiation of ES cells. Expression of miR-34a increases during in vitro<br />

neuronal terminal differentiation, of ex vivo primary cortical neuronal cultures, in parallel<br />

with the expression of TAp73. Moreover, we also detect an increase ex vivo of miR-34a<br />

steady state expression during postnatal development of the brain <strong>and</strong> cerebellum, when<br />

synaptogenesis occurs. We further confirm a role for miR-34a in synaptogenesis, as<br />

overexpression or silencing of miR-34a results in an inverse expression of a number of<br />

synaptic genes, via their 3’-UTR. In particular, miR-34a overexpression decreases<br />

synaptotagmin I <strong>and</strong> syntaxin-1A expression, <strong>and</strong> the endogenous levels of miR-34a are able<br />

to regulate only synaptotagmin I expression. Our findings show that p73 drives the<br />

expression of miR-34a during terminal, synaptic differentiation.<br />

Finally, we will speculate on why we need the p53 family at evolutionary level: it is the<br />

guardian of maternal reproduction.<br />

20


DNA-damage stress response in female germ cells: is Abl a fine tuner or a dangerous<br />

amplifier?<br />

Claudia Di Bartolomeo 1 , Emiliano Maiani 1 , Flavio Maina § , Francesca Sacco 1 , Marc<br />

Diederich # , Gianni Cesareni 1 <strong>and</strong> Stefania Gonfloni 1<br />

1 Department of Biology, University of Rome, “Tor Vergata”, via della Ricerca<br />

Scientifica, I-00133 Rome, Italy; § Developmental Biology Institute of Marseille-Luminy<br />

(IBDML) CNRS UMR 6216- Campus de Luminy – case 907, F-13288 Marseille cedex 09<br />

- France; # Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Fondation de<br />

Recherche Cancer et Sang, Hôpital Kirchberg, 9 rue Edward Steichen, L-2540<br />

Luxembourg, Luxembourg<br />

Cells experiencing DNA damage undergo a complex response entailing cell cycle arrest,<br />

DNA repair <strong>and</strong> apoptosis, the relative importance of the three being modulated by the extent<br />

of the lesions. The observation that Abl interacts in the nucleus with several proteins involved<br />

in different aspects of DNA repair has lead to propose that this kinase is part of the damagesensing<br />

mechanism. However, the mechanistic details underlying the role of Abl in DNA<br />

repair are largely unclear. Here, we will discussed the evidences supporting our current<br />

underst<strong>and</strong>ing of Abl activation following DNA insults while focusing on the relevance of<br />

such mechanisms in protecting DNA injured germ cells. Recent findings have implicated Abl<br />

in a cisplatin-induced signaling pathway eliciting death of immature oocytes 1 . A p53-related<br />

protein, TAp63, is an important immediate downstream effector of this pathway.<br />

Pharmacological inhibition of Abl by Imatinib (STI571) protects the ovarian reserve from the<br />

toxic effect of cisplatin <strong>and</strong> has a long-term effect on fertility. Taken together these<br />

observations indicate that imatinib may act as a fertoprotective drug by counteracting the<br />

effects of chemotherapy on the follicle reserve 2 . Although the mechanistic details involved in<br />

such a process are not fully understood, it remains beyond doubt, that a fine-tuning of nuclear<br />

outcomes by pharmacological inhibition of c-Abl will provide the basis for the development<br />

of effective fertoprotective adjuvants.<br />

References<br />

1) Gonfloni S. et al Nat. Med 2009; 15:1179-1185.<br />

2) Woodruff T. Nat. Med. 2009; 15:1124-1125.<br />

21


Altered expression of Apoptotic Machinery genes explains the decrease of oocytes<br />

competence with aging.<br />

MR Guglielmino 1 , M Vento 2 , M Santonocito 1 , P Borzì 2 , M Ragusa 1 , D Barbagallo 1 , I<br />

Casciano 3 , L Statello 1 , P Scollo 2 , M Romani 3 , M Purrello 1 , C Di Pietro 1 .<br />

1<br />

Dipartimento Gian Filippo Ingrassia, Sezione di Biologia, Genetica, Genomica<br />

Cellulare e Molecolare Giovanni Sichel, Università degli Studi di Catania; E-mail:<br />

dipietro@unict.it<br />

2<br />

Servizio di PMA/Azienda Ospedaliera Cannizzaro, Catania; E-mail:<br />

mrln.vento@gmail.com<br />

3<br />

Tumor Genetics <strong>and</strong> Epigenetics, Istituto Nazionale per la Ricerca sul Cancro - IST<br />

Genova; E-mail: massimo.romani@istge.it<br />

To identify the biomolecular bases of reduced oocyte competence with aging, we compared<br />

the Apoptotic Machinery (AM) transcriptome of pooled MII oocytes from two cohorts of<br />

women, respectively younger than 35 (young) <strong>and</strong> older than 38 years (old). We found that<br />

the expression pro<strong>file</strong> of AM genes is strongly dependent on maternal age. In fact, 36 out of<br />

the 84 AM genes analyzed showed a specific age-dependent expression pro<strong>file</strong>: 16<br />

proapoptotic genes were considerably upregulated <strong>and</strong> 7 antiapoptotic genes were<br />

downregulated in oocytes from older women. Through Real-Time PCR on single MII oocytes<br />

(6 from young <strong>and</strong> 5 from older women), we confirmed that the mRNAs encoding prosurvival<br />

proteins Bcl2 <strong>and</strong> cFLAR are more abundant in oocytes from younger women (p


Towards underst<strong>and</strong>ing of life <strong>and</strong> death regulation at CD95/Fas<br />

Nicolai Fricker(1,3), Carina Pforr(1), Rol<strong>and</strong> Eils(2,3,4), Peter H. Krammer(1),<br />

Inna N. Lavrik(1,3)<br />

1Division of Immunogenetics, German Cancer Research Center (DKFZ), 69120<br />

Heidelberg, Germany<br />

2Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ),<br />

69120 Heidelberg, Germany<br />

3Bioquant, University of Heidelberg, 69120 Heidelberg, Germany<br />

4Department for Bioinformatics <strong>and</strong> Functional Genomics, Institute for Pharmacy <strong>and</strong><br />

Molecular Biotechnology, University of Heidelberg, 69120 Heidelberg, Germany<br />

CD95 (APO-1/Fas) is a member of the death receptor family. CD95 stimulation leads to the<br />

induction of apoptotic as well as non-apoptotic pathways. CD95 signaling starts with the<br />

formation of the death-inducing signaling complex (DISC). The DISC consists of CD95, the<br />

adaptor molecule FADD (Fas-Associated Death Domain), procaspase-8, procaspase-10 <strong>and</strong> c-<br />

FLIPL/S/R (the cellular FLICE-inhibitory proteins). The regulation of life <strong>and</strong> death decisions<br />

at the DISC is largely influenced by the ratio between pro- <strong>and</strong> anti-apoptotic DED (death<br />

effector domain)–containing proteins <strong>and</strong> is poorly understood. We have applied a systems<br />

biology approach to underst<strong>and</strong> the regulation of life/death decisions in CD95 signaling<br />

pathway in quantitative terms. In our experiments we have used HeLa cells overexpressing<br />

various components of the CD95 system. Stimulation of HeLa cells has led to the induction of<br />

both apoptotic <strong>and</strong> non-apoptotic pathways depending on the concentration of the CD95<br />

DISC components. We have quantitatively analyzed apoptotic <strong>and</strong> non-apoptotic responses<br />

applying single-cell <strong>and</strong> population-based measurements. Based on these experimental data a<br />

mathematical model of the CD95 DISC has been constructed. The model takes into account<br />

the complex interplay between various DED-containing proteins of the DISC, e.g.<br />

procaspase-8 <strong>and</strong> c-FLIP isoforms as well as their cleavage products. The application of<br />

systems biology allowed to identify new mechanisms of regulation of CD95 signaling on the<br />

quantitative level <strong>and</strong> has demonstrated how exact concentrations of c-FLIP <strong>and</strong> procaspase-8<br />

<strong>and</strong> their cleavage products at the DISC can determine life/death decisions.<br />

23


Computational Systems Biology in Cancer Research<br />

Ilya Shmulevich<br />

Shmulevich Group, Institute for Systems Biology, 1441 North 34th Street , Seattle 98103,<br />

USA<br />

Cancer is a complex disease of the genes caused by genetic aberrations that disrupt the normal<br />

functioning of molecular networks in cells. I will discuss our efforts within The Cancer<br />

Genome Atlas (TCGA) project to carry out integrated analyses of TCGA-derived highthroughput<br />

experimental data sets in order to: (a) identify mechanisms of genomictranscriptomic<br />

regulation associated with specific cancers <strong>and</strong> clinical parameters; (b) infer<br />

networks that explain cancer type-specific transcriptional pro<strong>file</strong>s, <strong>and</strong> identify molecules that<br />

may be important control nodes in these networks as a means to prioritize drug targets for<br />

therapeutic intervention; <strong>and</strong> (c) compare cancer-associated features across multiple cancer<br />

types within TCGA, to gain insights into the regulatory basis for cancer progression.<br />

24


Systems Analysis of Endocytosis by Quantitative Image Analysis: Insights into<br />

Pathogenetic Mechanisms<br />

Varadharajan Sundaramurthy, Claudio Collinet, Martin Stöter, Charles Bradshaw,<br />

Nikolay Samusik, Jochen C. Rink, Bianca Habermann, Eugenio Fava, Yannis<br />

Kalaidzidis <strong>and</strong> Marino Zerial<br />

Max Planck Institute of Molecular Cell Biology <strong>and</strong> Genetics MPI-CBG, Dresden,<br />

Germany<br />

Endocytosis is an essential process serving multiple key cellular functions, such as nutrient<br />

uptake, signal transduction, <strong>and</strong> defence against pathogens. We have undertaken a broad<br />

systems biology analysis of endocytosis. We systematically pro<strong>file</strong>d the activity of human<br />

genes with respect to Transferrin <strong>and</strong> EGF endocytosis by performing an image-based RNAi<br />

screening of HeLa cells in cooperation with the HT-TDS, the screening facility of the MPI-<br />

CBG. The genes were identified on the basis of a multi-parametric analysis quantitatively<br />

measuring uptake <strong>and</strong> intracellular cargo distribution. We uncovered novel regulators of<br />

endocytosis <strong>and</strong> endosome trafficking, including many signalling pathways (e.g. Wnt,<br />

Integrin, TGF-!, <strong>and</strong> Notch). A systems analysis by Bayesian networks further uncovered<br />

design principles regulating the number, size, concentration of cargo <strong>and</strong> intracellular position<br />

of endosomes. Mathematical modelling revealed reverse engineering principles governing the<br />

organization <strong>and</strong> transport properties of the endocytic pathway. These results have profound<br />

implications for our underst<strong>and</strong>ing of the mechanisms regulating organelle biogenesis <strong>and</strong><br />

signalling at the cellular, tissue <strong>and</strong> organism level. They further provide insights into the<br />

pathogenetic mechanisms underlying a spectrum of human diseases (e.g. infectious diseases,<br />

metabolic disorders, cancer <strong>and</strong> metastasis, neurodegeneration).<br />

25


Using single cell analysis tools to search for human breast cancer stem cells<br />

Anne Grosse-Wilde, Rolf E Kuestner, <strong>and</strong> Adrian Ozinsky<br />

Institute for Systems Biology, Seattle, WA, 98103 USA<br />

Studies suggest that chemotherapy resistance <strong>and</strong> tumor metastatic potential are dependent on<br />

a small sub-population of cancer stem cells. These cells seem to mimic many of the<br />

characteristics of embryonic or induced pluripotent stem cells, <strong>and</strong> are presumed to be<br />

regulated by stem cell-specific transcription factors. We have been examining whether these<br />

genes actually are co-expressed within individual cancer cells. We have focussed on<br />

measuring gene expression during cell lineage transitions between epithelial <strong>and</strong><br />

mesenchymal cells that are induced by in vitro assays that enrich for aggressive <strong>and</strong><br />

metastatic cell phenotypes.<br />

Grant acknowledgments: NIH (NIGMS P50GM076547) <strong>and</strong> the Luxembourg Centre for<br />

Systems Biomedicine <strong>and</strong> the University of Luxembourg<br />

26


Whole genome scans in drosophila to model human disease<br />

Josef Penninger<br />

IMBA<br />

A plethora of genes have been correlated with human diseases. Genetic animal models have<br />

proven to be extremely valuable to elucidate the essential functions of genes in normal<br />

physiology <strong>and</strong> the pathogenesis of disease. During the last years, my laboratory have done<br />

large scale Drosophila screens to map pain perception (thermal avoidance, Neely et al. Cell<br />

2010), stress induced heart failure (Neely et al. Cell, 2010), obesity (Pospisilik et al. Cell<br />

2010), <strong>and</strong> bacterial infections <strong>and</strong> gut immunity (Cronin et al. Science 2009). The idea is to<br />

mine the genome of one organism using systems genetics <strong>and</strong> then link <strong>and</strong> compare our<br />

results to large scale genome association studies in human disease populations. Based on such<br />

comparisons, we then choose new genes/pathways to generate knock-out mice for validation<br />

in mammals.<br />

27


Valproic Acid perturbs hematopoietic homeostasis by inhibition of erythroid<br />

differentiation <strong>and</strong> activation of the myelo-monocytic pathway<br />

Sébastien Chateauvieux 1,3 , Serge Eifes 1,3 , Franck Morceau 1 , Christina Grigorakaki 1 ,<br />

Michael Schnekenburger 1 , Estelle Henry 1 , Mario Dicato 2 , Marc Diederich 1 .<br />

1 Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, L-<br />

2540 Luxembourg; 2 Luxembourg Medical Center, Luxembourg, L-1210 Luxembourg;<br />

3 These authors contributed equally<br />

As a histone deacetylase inhibitor, Valproic acid (VPA) is a c<strong>and</strong>idate for anticancer therapy.<br />

Besides, VPA exhibits various mechanisms of action <strong>and</strong> its effects on the molecular basis of<br />

hematopoiesis remain unclear. To study the effects of VPA on the hematopoietic system, we<br />

performed microarray analysis using K562 cells treated with 1 mM VPA over a 72 h time<br />

course. The association between Gene Ontology (GO) terms <strong>and</strong> the lists of differentially<br />

expressed genes was tested using the Bioconductor package GOstats. Enrichment analysis for<br />

cellular differentiation pathways was performed based on manually curated gene lists. Results<br />

from microarray analysis were confirmed by studying cell differentiation features at the<br />

molecular <strong>and</strong> cellular levels using other hematopoietic cell lines as well as hematopoietic<br />

stem/progenitor CD34 + cells. Microarray analysis revealed 3440 modulated genes in the<br />

presence of VPA. Genes involved in the granulo-monocytic differentiation pathway were upregulated<br />

while genes of the erythroid pathway were down-regulated. This was confirmed by<br />

analyzing erythrocytic <strong>and</strong> myeloid membrane markers <strong>and</strong> lineage-related gene expression in<br />

HEL, MEG01, HL60 as well as CD34 + cells. Moreover, GATA-1 <strong>and</strong> its co-factors (FOG1,<br />

SP1) were down-regulated, while myelopoiesis activator PU.1 was up-regulated, in agreement<br />

with an inhibition of erythropoiesis. Our functional profiling <strong>and</strong> cell phenotyping approach<br />

demonstrates that VPA is able to alter hematopoietic homeostasis by modifying the cell<br />

population balance in the myeloid compartment. This may lead to a potential failure of<br />

erythropoiesis in patients with cancer or chronic inflammatory diseases having a welldescribed<br />

propensity to anemia.<br />

28


Search for the effects of anticancer drugs: A look to apoptosis but also to autophagy!<br />

Micol Tillhon 1 , Francesca Donà 1 , Michele Parks 1 , Vincenzo Giansanti 1 , Giuliano<br />

Mazzini 1 , Ennio Prosperi 1 , Paolo Lombardi 2 , A. Ivana Scovassi 1<br />

1 Istituto di Genetica Molecolare CNR, Via Abbiategrasso 207, I-27100 Pavia;<br />

2 Naxospharma, Novate Milanese, Italy scovassi@igm.cnr.it<br />

Drug resistance of cancer cells is often correlated to apoptosis evasion; however, an active<br />

involvement of autophagy in this scenario has been recently proposed, based on the evidence<br />

that this process could exert a protective role towards the activation of apoptosis in cancer<br />

cells. We addressed this question by analysing the effects of 2-methoxyestradiol (2-ME),<br />

which inhibits the growth of cancer cells, on cell proliferation <strong>and</strong> cell cycle of HeLa <strong>and</strong><br />

colon carcinoma cell lines, namely HCT116 <strong>and</strong> SW613-B3. We showed a net time- <strong>and</strong><br />

dose-dependent anti-proliferative effect of 2-ME on both cell lines, which is accompanied by<br />

cell cycle arrest leading to the accumulation of cells in G2/M. We also demonstrated that 2-<br />

ME induces caspase-dependent apoptosis characterized by mitochondria impairment. Of note,<br />

we detected autophagic markers in 2-ME-treated cells (Parks et al. 2-Methoxyestradiol: New<br />

perspectives in colon carcinoma treatment. Mol. Cell Endocrinol. 331:119-128 (2011). The<br />

simultaneous detection of apoptosis <strong>and</strong> autophagy prompted us to investigate in the same cell<br />

lines the effect of the naturally-occurring alkaloid berberine <strong>and</strong> of structurally related novel<br />

derivatives. In cells treated with berberine <strong>and</strong> analogues, we detected several apoptotic<br />

hallmarks (nuclear shrinkage, DNA degradation, caspase activation, PARP-1 proteolysis) as<br />

well as the evidence for autophagy markers (LC3 conversion, increased beclin-1 expression).<br />

On the whole, our results could contribute to the underst<strong>and</strong>ing of the role of autophagy in<br />

cancer treatment.<br />

29


Signaling by the Frizzled family of receptors: G protein-coupled mechanisms <strong>and</strong><br />

targets<br />

Vladimir L. Katanaev<br />

Department of Biology, University of Konstanz. Universitätsstrasse 10, Box 643.<br />

Konstanz 78457, Germany. E-mail vladimir.katanaev@uni-konstanz.de<br />

The Frizzled family of unconventional GPCRs transmits the signals from the developmentally<br />

<strong>and</strong> medically important group of Wnt-type growth factors. Despite the GPCR topology of<br />

Frizzled proteins, their functional belonging to the G protein-coupled type of receptors has<br />

been long questioned. In the recent years, the involvement of heterotrimeric G proteins in<br />

Frizzled signaling has been established in a number of model systems, as was the biochemical<br />

activity of Frizzled proteins as GPCRs. Frizzled proteins from flies to humans have been<br />

shown to couple to the Go/i type of heterotrimeric G proteins. A number of interesting targets<br />

of G proteins in Frizzled signaling, including the scaffolding protein Axin <strong>and</strong> the small<br />

GTPase Rab5 have been identified. These advances shed light on the mechanisms of this<br />

intricate signal transduction cascade <strong>and</strong> identify new approaches for the anti-cancer drug<br />

development.<br />

Recent papers:<br />

1. Purvanov V, Koval A, Katanaev VL. (2010) A direct <strong>and</strong> functional interaction between<br />

the trimeric G protein Go <strong>and</strong> Rab5 in G protein-coupled receptor signaling. Sci. Signal. 3:<br />

ra65.<br />

2. Koval A, Katanaev VL. (2010) Wnt3a Stimulation Elicits G Protein-Coupled Receptor<br />

Properties of Mammalian Frizzled Proteins. Biochem. J. DOI 10.1042/BJ20101878<br />

3. Egger-Adam D, Katanaev VL. (2010) The trimeric G protein Go inflicts a double impact<br />

on Axin in the Wnt/Frizzled signaling pathway. Dev. Dyn. 239:168–83.<br />

4. Kopein D, Katanaev VL. (2009) Drosophila GoLoco-protein Pins is a target of G!omediated<br />

G protein-coupled receptor signaling. Mol. Biol. Cell 20:3865-77.<br />

30


<strong>Redox</strong> regulation of transcription factors by electrophilic lipid mediators<br />

Young-Joon Surh<br />

WCU Department of Biopharmaceutical Sciences <strong>and</strong> Molecular Medicine,<br />

College of Pharmacy, Seoul National University, Seoul 151-742, South Korea<br />

The implication of inflammatory cell/tissue damage in carcinogenesis has been under intense<br />

investigation both at the research level <strong>and</strong> in clinical practice. Numerous studies have been<br />

reported with the global biochemical profiling technologies, such as DNA microarray,<br />

proteomics, metabolomics, lipidomics, etc., to identify <strong>and</strong> characterize a series of critical<br />

molecules/changes in the inflammatory signaling. It is by gaining this type of mechanistic<br />

underst<strong>and</strong>ing of a disease that researchers will unlock the keys to discovering new<br />

diagnostics as well as preventive <strong>and</strong> therapeutic strategies for the management of<br />

inflammation-associated cancer. Cyclopentenone prostagl<strong>and</strong>ins of J series as products of<br />

cyclooxygenase-2 (COX-2)-mediated arachidonic acid cascades have been reported to possess<br />

multifaceted cellular functions, including anti-inflammatory <strong>and</strong> cytoprotective effects. A<br />

typical example is 15-deoxy-! 12,14 -prostagl<strong>and</strong>in J2 (15d-PGJ2) that modulates the activities<br />

of various intracellular signaling molecules. Because of the electrophilic ",#-unsaturated<br />

carbonyl moiety present in its cyclopentenone ring structure, 15d-PGJ2 acts as a Michael<br />

addition acceptor <strong>and</strong> can readily interact with critical cellular nucleophiles, such as cysteine<br />

thiol groups of proteins. Many of the biological effects induced by 15d-PGJ2 are mediated by<br />

targeting redox-sensitive transcription factors <strong>and</strong> their regulators, including I$B kinase-NF-<br />

$B, AP-1, Nrf2-Keap1, HIF-1!, STAT3 <strong>and</strong> p53 tumor suppressor. We have reported that<br />

15d-PGJ2 treatment rescues PC12 cells from peroxynitrite- <strong>and</strong> hydrogen peroxide-induced<br />

apoptosis by upregulating the Nrf2-driven expression of glutamate cysteine ligase or heme<br />

oxygenase-1. Elevated production of 15d-PGJ2 has also been implicated in tumorigenesis<br />

through induction of angiogenesis <strong>and</strong> metastasis. Cysteine thiols present in various<br />

transcription factors <strong>and</strong> their regulators function as redox sensors in fine-tuning of<br />

transcriptional regulation of many genes essential for maintaining cellular homeostasis. Thus,<br />

oxidation or covalent modification of thiol groups present in some redox-sensitive<br />

transcription factors <strong>and</strong> their regulating molecules can provide a unique strategy for<br />

molecular target-based chemoprevention <strong>and</strong> cytoprotection.<br />

31


Novel mechanisms of transcriptional control in differentiation <strong>and</strong> cancer<br />

Luciano Di Croce<br />

Epigenetic events in Cancer, CRG/ICREA, Dr. Aiguader 88, Barcelona<br />

Underst<strong>and</strong>ing the genetic basis of cancers has been a topic of intense research, <strong>and</strong> hundreds<br />

of gene mutations have been identified that can cause carcenogenesis. However, in the past<br />

few years, increasing evidence has suggested that mutations are not the only genetic changes<br />

that lead to cancer. Indeed, perturbations of chromatin structure <strong>and</strong> of other epigenetic<br />

mechanisms can cause inappropriate gene expression <strong>and</strong> genomic instability, resulting in<br />

cellular transformation <strong>and</strong> malignant outgrowth.<br />

I will discuss the role of several protein complexes that are involved in chromatin dynamics<br />

<strong>and</strong> metabolism, which when altered could participate in the establishment <strong>and</strong> maintenance<br />

of the aberrant silencing of tumor suppressor genes during transformation. Our data suggested<br />

that the ZRF1, Polycomb group of proteins (PcG) <strong>and</strong> the histone variant macroH2A are -<br />

with different timing <strong>and</strong> kinetics - involved in setting up an altered chromatin structure with<br />

aberrant gene silencing in cancer cells. Furthermore, the role of ubiquitylated H2A, <strong>and</strong> its<br />

localization in the human genome will also be discussed.<br />

32


A dual role of JAK1: regulation of IFN-! dependent <strong>and</strong> independent IL-12p70<br />

production<br />

Andreas H. Wagner $ , Michael Conzelmann* # , Sascha Dietrich* # , Claudia Schäfer,<br />

Oliver Krämer, Michael Hess* # , Annika Zota*, Christine S. Falk ! , Markus Hecker $ , <strong>and</strong><br />

Thomas Luft* #<br />

*From the German Cancer Research Center, Dept. of Molecular Oncology/Hematology,<br />

Heidelberg, # Dept. of Medicine V, University of Heidelberg, $ Institute of Physiology <strong>and</strong><br />

Pathophysiology, University of Heidelberg, <strong>and</strong> ! National Center for Tumor diseases<br />

<strong>and</strong> Institute of Immunology, University of Heidelberg, Germany.<br />

Inhibition of JAK1 is an emerging clinical concept resulting in spleen size reduction due to<br />

cytokine inhibition in human myelofibrosis. Therefore, JAK1 inhibition might become a<br />

promising concept for a variety of autoimmune diseases, but similarly raises concerns<br />

regarding immunosuppressive side effects.<br />

At the example of IL-12p70 production we demonstrate that JAK1 has a dual role in<br />

differentially regulating the effects of weak <strong>and</strong> strong activation stimuli.<br />

Weak NF-"B-activating stimuli such as CD40L or LPS require complementary JAK1inducing<br />

cytokines such as IFN-! to induce IL-12p70. This pathway involves RELA, cREL,<br />

JAK1 <strong>and</strong>/or JAK2, STAT1, IRF1 <strong>and</strong> IRF8 <strong>and</strong> is inhibited by RELB <strong>and</strong> TYK2.<br />

Here we show evidence that an alternative pathway exists depending on strong NF-"B<br />

induction (e.g. via intact E.coli or LPS plus IL-1#) that is inhibited by JAK1, STAT3 <strong>and</strong><br />

RELB. STAT3 directly binds to a combined STAT/NF-"B site at the IL-12p35 promoter<br />

without altering access of RELA <strong>and</strong> cREL. Furthermore, a direct role of JAK1/STAT3 is<br />

demonstrated for RELB expression using siRNA. This pathway is independent of IFN-!,<br />

STAT1, IRF1 <strong>and</strong> IRF8.<br />

Our study suggests that in vivo blockade of JAK1 specifically inhibits pro-inflammatory<br />

effects of weak, IFN-! dependent, NF-"B activation stimuli (such as CD40L or LPS) whilst<br />

preserving or enhancing IL-12p70 induced by strong, IFN-! independent activation stimuli.<br />

33


Learning immunology from successful vaccines: innate immunity to systems<br />

Vaccinology<br />

Bali Pulendran<br />

Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA.<br />

bpulend@emory.edu<br />

Despite their great success, we underst<strong>and</strong> little about how effective vaccines stimulate<br />

protective immune responses. Two recent developments promise to yield such underst<strong>and</strong>ing:<br />

the appreciation of the crucial role of the innate immune system in sensing microorganisms<br />

<strong>and</strong> tuning immune responses, <strong>and</strong> advances in systems biology. In this presentation, I will<br />

discuss how these developments are yielding insights into the mechanism of some of the most<br />

successful vaccines ever developed. Furthermore, such developments promise to address a<br />

major challenge in vaccinology: that the efficacy of a vaccine can only be ascertained<br />

retrospectively, upon infection. The identification of molecular signatures induced rapidly<br />

after vaccination, which correlate with <strong>and</strong> predict the later development of protective<br />

immune responses, would represent a strategy to prospectively determine vaccine efficacy.<br />

Such a strategy would be particularly useful when evaluating the efficacy or immunogenicity<br />

of untested vaccines, or in identifying individuals with sub-optimal responses amongst high<br />

risk populations, such as infants or the elderly. We have recently used a systems biology<br />

approach to identify early gene signatures that correlate with, <strong>and</strong> predict the later immune<br />

responses in humans vaccinated with the live attenuated yellow fever vaccine YF-17D, or<br />

with the influenza vaccines. I will review these studies, <strong>and</strong> discuss their broader implications<br />

for vaccinology.<br />

34


Immunity <strong>and</strong> death in chronic viral infections<br />

Marie-Lise Gougeon<br />

Antiviral immunity, Biotherapy <strong>and</strong> Vaccine Unit,<br />

Institut Pasteur, 28 rue du Dr. Roux, 75015 Paris, France<br />

Persistent viruses are able to subvert innate host antiviral strategies. For example, HIV-1 has<br />

evolved ways to exploit dendritic cells (DCs), thereby facilitating viral dissemination.<br />

Dendritic cells (DCs) are the first targets for HIV upon primary mucosal infection. The fate of<br />

DCs is extremely dependent on the interaction with autologous natural killer (NK) cells <strong>and</strong><br />

once infected, they should be rapidly eliminated by these innate effectors. NK-DC interaction<br />

is bidirectional <strong>and</strong> may lead to DC maturation <strong>and</strong> priming for Th1 responses, or may<br />

enhance NK-cell cytotoxicity through IL-12 release. NK-mediated killing of infected DCs is<br />

believed to be an essential step for early control of viral replication <strong>and</strong> dissemination. We<br />

discovered that, once infected with HIV, DCs become resistant to NK-dependent lysis.<br />

Apoptosis resistance of infected DCs is dependent upon a crosstalk with NK cells that leads to<br />

the dramatic upregulation of two key inhibitors of apoptosis, cIAP-2 <strong>and</strong> c-FLIP, resulting in<br />

a lack of response to TRAIL released by NK cells. During this crosstalk, NK cells trigger HIV<br />

replication in DCs, thus contributing to the constitution of reservoirs in infected DCs.<br />

The molecules involved in DC resistance to NK-dependent apoptosis were identified <strong>and</strong> the<br />

essential role of HMGB1, an alarmin expressed at NK-DC synapse, was discovered. Indeed,<br />

HMGB1 is responsible for the upregulation of the two apoptosis inhibitors c-FLIP <strong>and</strong> c-IAP2<br />

in infected DCs, <strong>and</strong> blocking HMGB1 activity by glycyrrhizin or specific antibodies restored<br />

the susceptibility of HIV-infected DC to NK cell killing, <strong>and</strong> abrogated HIV replication in<br />

DCs. The pivotal role of HMGB1 was confirmed by the ability of exogenous rhHMGB1 both<br />

to upregulate the two apoptosis inhibitors <strong>and</strong> to trigger HIV-1 replication in infected DCs<br />

Overall, these observations provide new insights into how HIV hijacks DCs to promote viral<br />

dissemination <strong>and</strong> uses the NK-DC interaction to maintain viability of long-term reservoirs.<br />

In addition, they challenge the question of the in vivo involvement of HMGB1 in the<br />

establishment of viral persistence <strong>and</strong> identify potential therapeutic targets to eliminate viral<br />

reservoirs.<br />

35


Gimap5: a critical mediator of lymphocyte homeostasis <strong>and</strong> determinant of autoinflammatory<br />

disease.<br />

Halil Aksoylar, Michael J Barnes, <strong>and</strong> Kasper Hoebe<br />

Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, USA<br />

Homeostatic control of the immune system involves mechanisms that ensure the selftolerance,<br />

survival <strong>and</strong> quiescence of hematopoietic-derived cells. Previous reports linked<br />

genetic aberrancies in the GTPase the immunity-associated protein 5 (Gimap5) to autoimmune<br />

disorders in both humans <strong>and</strong> rats, suggesting that Gimap5 is an important mediator<br />

of immune homeostasis. Our laboratory recently identified a recessive ENU-induced germline<br />

mutation that destabilizes Gimap5. Homozygote mutant mice develop lymphopenia, hepatic<br />

extramedullary hematopoiesis, weight loss <strong>and</strong> intestinal inflammation. Although Gimap5deficient<br />

CD4 + T <strong>and</strong> B cells undergo normal development, they fail to proliferate upon<br />

antigen-receptor stimulation even though NF-!B, MAP kinase <strong>and</strong> Akt activation occur<br />

normally. Activation of CD4 + T cells results in normal cytokine secretion predominantly<br />

polarized towards TH17. In addition mutant mice contain reduced numbers of peripheral Treg<br />

cells that also exhibit a reduced suppressive capacity compared to wildtype Treg cells.<br />

Together, these data establish Gimap5 as a key regulator of hematopoietic integrity <strong>and</strong><br />

lymphocyte homeostasis.<br />

36


Study of the anti-apoptotic role of SHIP-1 in T cells<br />

Claude Condé 1 , Xavier Rambout 2 , Franck Dequiedt 2 , Geoffrey Gloire 1 <strong>and</strong> Jacques<br />

Piette 1<br />

1 Virology <strong>and</strong> immunology unit, GIGA-research (+2), University of Liège<br />

1, avenue de l’hôpital Bât B34 4000 Liège – claude.conde@ulg.ac.be<br />

2 Cellular <strong>and</strong> Molecular Biology Unit, Gembloux Agro Bio-Tech, University of Liège<br />

13, Avenue Maréchal Juin Bât 92 5030 Gembloux<br />

Background :<br />

SHIP-1 is an inositol 5’-phosphatase, principally expressed in hematopoietic cells. It acts by<br />

dephosphorylating phosphatidylinostol-3-phosphate thereby down-modulating PI3K pathway<br />

<strong>and</strong> cellular proliferation. From studies with SHIP-1 KO mice, it becomes apparent that<br />

SHIP-1 plays a dual role: in one h<strong>and</strong>, it has been demonstrated that SHIP-1 has a<br />

proapoptotic activity in myeloid <strong>and</strong> B cells, this properties is directly linked to its catalytic<br />

activity. In the other h<strong>and</strong>, SHIP-1 plays an anti-apoptotic role in T cells that is mediated by it<br />

adaptator function. For a better underst<strong>and</strong>ing of the anti-apoptotic activity of SHIP-1 in T<br />

cells, we searched for new protein interaction partners of SHIP-1.<br />

Methods:<br />

To identify new protein interaction partners of SHIP-1, we carried out a yeast two hybrid<br />

screening using different constructs of SHIP-1 as a bait <strong>and</strong> the hORFeome v5.1 pooled in<br />

preys. The advantage of this method is that the preys are not restricted to a specific tissue or a<br />

specific cDNA library.<br />

Results :<br />

Among the partner of SHIP-1, we identified two members of the IAP family : cIAP-1 <strong>and</strong><br />

XIAP. These proteins are well known for their anti-apoptotic activity but also for playing an<br />

important role in many cellular pathways. Our goal is first to demonstrate the interaction<br />

between SHIP-1 <strong>and</strong> XIAP or cIAP-1 in vivo, <strong>and</strong> then to characterise the role of this<br />

interaction.<br />

37


Protein population shift leading to disease-related changes in cellular networks<br />

Antonio del Sol 1 , Wiktor Jurkowski 1 , Kirsten Roomp 1 , Hiroaki Kitano 2<br />

1<br />

Luxembourg Center for Systems Biomedicine (LCSB), University of Luxembourg,<br />

Luxembourg<br />

2<br />

The Systems Biology Institute, Shinjuku-ku, Tokyo, Japan<br />

Proteins exist as dynamic ensembles of different structural <strong>and</strong> dynamic states including<br />

functionally relevant conformers as the most populated states, as well as malfunctioning<br />

conformers as less populated states. Despite a large number of cellular <strong>and</strong> environmental<br />

changes that proteins experience under normal physiological conditions, robustness of protein<br />

structures guarantees performance of their functional activities. Yet, specific perturbations<br />

such as mutations or binding to metabolites are capable of shifting a protein population<br />

towards a malfunctioning state. These protein population shifts may lead to distinct<br />

perturbations of cellular networks, <strong>and</strong> thus the population of cells shifts towards disease<br />

states as their internal networks are perturbed. Here we study prion disease as an illustrative<br />

example of protein population shift leading to disease-related network perturbations.<br />

Conversion of benign forms of prion protein (PrPC) to disease-specific isoforms (PrPSc),<br />

yields to the activation of bi-stable circuits enabling the transition of a steady stable healthy<br />

state of a cellular network towards a disease-related steady stable state. In addition we carry<br />

out a structural analysis of the effect of disease-causing mutations on proteins associated to<br />

different diseases. Furthermore, in each case we analyze the impact of the particular protein<br />

malfunction on the relevant cellular pathways that are associated with the disease outcome.<br />

Thus, this study shows that a molecular analysis of cellular network components can provide<br />

important insights into disease-related network perturbations, with potentially important<br />

consequences for molecular therapeutic strategies.<br />

38


Proteomic phenotyping of the cell surface information gateway<br />

Bernd Wollscheid<br />

Institute of Molecular Systems Biology (IMSB), NCCR Neuro Center for Proteomics,<br />

ETH Hönggerberg, HPT D77, Wolfgang Pauli-Str. 16, CH-8093 Zürich, Switzerl<strong>and</strong><br />

The classification of cell types has relied on the identification of cell surface proteins as<br />

differentiation markers. Currently, flow cytometry allows for the detection of up to a dozen<br />

differentiation markers in a single measurement. We have developed the cell surface<br />

capturing (CSC) technology for the multiplexed mass-spectrometric identification of several<br />

hundred N-glycosites specifically from cell surface exposed glycoproteins, which can be used<br />

to phenotype cells without antibodies in an unbiased fashion <strong>and</strong> without a priori knowledge.<br />

We show that the CSC technology allows for the parallel detection <strong>and</strong> relative quantitative<br />

comparison of the cell surface N-glycoproteome in various cell types, as well as for the<br />

monitoring of emerging <strong>and</strong> declining ES cell surface N-glycoprotein markers during their<br />

controlled differentiation towards the neuronal lineage. CSC technology enables a snapshot<br />

view of the cell surface N-glycoprotein l<strong>and</strong>scape <strong>and</strong> can detect panels of N-glycoproteins as<br />

potential differentiation markers that are currently not accessible by other means.<br />

39


De-SUMOylation: a new strategy used by Listeria to counteract the host defense<br />

David Ribet 1,2,3 <strong>and</strong> Pascale Cossart 1,2,3<br />

1 Institut Pasteur, Unité des Interactions Bactéries-Cellules, F-75015 Paris, France.<br />

2 Inserm, U604, F-75015 Paris, France.<br />

3 INRA, USC2020, F-75015 Paris, France.<br />

The effects of pathogenic bacteria on SUMOylation, a fundamental post-translational<br />

modification in eukaryotic cells, remain largely unknown. We have examined SUMOylation<br />

during infection by the intracellular bacterial pathogen Listeria monocytogenes. Our data<br />

reveal that Listeria monocytogenes infection leads to a decrease in the levels of host SUMOconjugated<br />

proteins. This event is triggered by the bacterial virulence factor listeriolysin O<br />

(LLO) which induces a degradation of Ubc9, an essential enzyme of the SUMOylation<br />

machinery, <strong>and</strong> a degradation of some SUMOylated proteins. The effect of LLO on Ubc9 is<br />

dependent on the pore-forming capacity of the toxin <strong>and</strong> is shared by other bacterial poreforming<br />

toxins like perfringolysin O (PFO) <strong>and</strong> pneumolysin (PLY). Ubc9 degradation was<br />

also observed in vivo in infected mice. Furthermore, we showed that SUMO overexpression<br />

impairs bacterial infection. Together, our results reveal that Listeria, <strong>and</strong> probably other<br />

pathogens, dampen the host response to infection by preventing SUMOylation of key<br />

regulatory proteins [1].<br />

1. Ribet, D., Hamon, M., Gouin, E., Nahori, M.-A., Impens, F., Neyret-Kahn, H.,<br />

Gevaert, K., V<strong>and</strong>ekerckhove, J., Dejean, A. <strong>and</strong> Cossart, P. Listeria monocytogenes impairs<br />

SUMOylation for efficient infection. Nature 2010; 464 :1192-5.<br />

40


Traveling to the Ends of the Proteome World. Positional N-terminal <strong>and</strong> C-terminal<br />

proteomics deciphers protein terminal <strong>and</strong> proteolytic post-translational modifications<br />

in complex proteomes in vivo.<br />

Christopher M. Overall.<br />

UBC Centre for Blood Research, University of British Columbia, 2350 Health Sciences<br />

Mall, Vancouver, B.C. V6T 1Z3 Canada. chris.overall@ubc.ca http://www.clip.ubc.ca.<br />

Specific degradomics techniques are needed to rapidly identify <strong>and</strong> quantify the N- <strong>and</strong> Cterminomes<br />

in order to reveal the extent of post-translational modifications of protein termini<br />

<strong>and</strong> therefore the functional state of key molecules, the extent of proteolysis in a system, <strong>and</strong><br />

to identify new protease substrates. Using positional proteomics we find that ~50% of<br />

proteins in skin are present as stable cleavage products <strong>and</strong> in E Coli ~40% of proteins have a<br />

truncated C-terminus. Broad coverage N-terminome analysis necessitates a negative selection<br />

procedure as the variety of original mature protein N-terminal blocked peptides each present<br />

individual chemical hurdles for their enrichment by positive selection strategies. We<br />

developed two combined terminomics <strong>and</strong> protease substrate discovery degradomics<br />

platforms for the simultaneous quantitative analysis of the N-terminome <strong>and</strong> proteolysis on a<br />

proteome-wide scale. A specific C-terminal analysis procedure was recently reported<br />

(Schilling et al 2010 Nature Methods 7, 508-511) as well as a N terminal procedure termed<br />

Terminal Amino Isotopic Labelling of Substrates (TAILS, Kleifeld et al Nature<br />

Biotechnology 28, 281-288; Prudova et al 2010 Mol Cell Proteomics; auf dem Keller et al<br />

2010 Mol Cell Proteomics). By using a novel polymer to deplete the internal tryptic peptides,<br />

TAILS suffers little from sample loss <strong>and</strong> low yields, so requiring only 100 microgram of<br />

sample <strong>and</strong> one MS/MS analysis per sample. By a two-day procedure with flexible labelling<br />

options, TAILS can be adapted to a variety of experimental situations including cell culture<br />

<strong>and</strong> complex biological sample analysis. Incorporating iTRAQ labelling iTRAQ-TAILS also<br />

provides wide coverage of all forms of naturally blocked N-terminal peptides <strong>and</strong> allows for<br />

their quantification through labelling of lysine side-chains in up to 8 samples. TAILS permits<br />

exploitation of the acetylated <strong>and</strong> other blocked mature protein N-terminal peptides as a<br />

statistical classifier that is then used to set isotope ratio cut offs that reveal protease activity.<br />

Being a quantitative procedure, TAILS can analyse the substrate degradome of a broad<br />

specificity protease or one with no known specificity without manual data parsing, in the<br />

same experiment, <strong>and</strong> also do this in vivo. We have applied TAILS to a variety of<br />

metalloproteases <strong>and</strong> compared protease knock out mice, analysed inflamed skin, breast<br />

cancer <strong>and</strong> arthritic tissues. Typical analyses identify over 3000 N-terminal peptides from<br />

which we found that the removal of the N-terminal methionine is dependent upon the amino<br />

acid at position 2 with distinct preferences found for valine, glycine, alanine <strong>and</strong> serine. In<br />

one experiment, acetylation occurred on 731 original mature protein N-terminal peptides but<br />

at the initiator methionine in only 153 of these instances. In 578 cases, acetylation was at<br />

position 2 in the protein after removal of 1 Met, with alanine, serine <strong>and</strong> methionine being the<br />

preferred acetylated residues. Finally N-terminal positional proteomics enables MS sample<br />

simplification with proteins identified in bronchoalvelar fluid having abundances spanning a<br />

range greater than six orders of magnitude.<br />

41


Identification of the Human Testis Protein Phosphatase 1 Interactome<br />

Margarida Fardilha 1 , Sara L.C. Esteves 1 , Luís Korrodi-Gregório 1 , Ana Paula Vintém 1 ,<br />

Sara C.T.S. Domingues 2 , S<strong>and</strong>ra Rebelo 2 , Nick Morrice 3 , Patricia T. W. Cohen 3 , Odete<br />

A.B. da Cruz e Silva 2 <strong>and</strong> Edgar F. da Cruz e Silva 1†<br />

1 Signal Transduction Laboratory, Centre for Cell Biology, Biology Department,<br />

University of Aveiro; <strong>and</strong> 2 Neuroscience Laboratory, Centre for Cell Biology, Health<br />

Sciences Department, University of Aveiro; <strong>and</strong> 3 Medical Research Council Protein<br />

Phosphorylation Unit, School of Life Sciences, University of Dundee; † deceased on the<br />

2 nd March 2010<br />

Protein phosphorylation is a critical regulatory mechanism in cellular signalling. To this end,<br />

PP1 is a major eukaryotic serine/threonine-specific phosphatase whose cellular functions, in<br />

turn, depend on complexes it forms with PP1 Interacting Proteins – PIPs. The importance of<br />

the testis/sperm-enriched variant, PP1!2, in sperm motility <strong>and</strong> spermatogenesis has<br />

previously been shown. Given the key role of PIPs, it is imperative to identify the<br />

physiologically relevant PIPs in testis <strong>and</strong> sperm. Hence, we performed Yeast Two-Hybrid<br />

screens of a human testis cDNA library using as baits the different PP1 isoforms <strong>and</strong> also a<br />

proteomic approach aimed at identifying PP1!2 binding proteins. To the best of our<br />

knowledge this is the largest data set of the human testis PP1 interactome. We report the<br />

identification of 77 proteins in human testis <strong>and</strong> 7 proteins in human sperm that bind PP1. The<br />

data obtained increased the known PP1 interactome by reporting 72 novel interactions.<br />

Confirmation of the interaction of PP1 with 5 different proteins was also further validated by<br />

co-immunoprecipitation or protein overlays. The data here presented provides important<br />

insights towards the function of these proteins <strong>and</strong> opens new possibilities for future research.<br />

In fact, such diversity in PP1 regulators makes them excellent targets for pharmacological<br />

intervention.<br />

Related publications:<br />

!" Fardilha M, Esteves SL, Korrodi LMG, da Cruz e Silva OA <strong>and</strong> da Cruz e Silva<br />

FF (2010) The physiological relevance of Protein Phosphatase 1 <strong>and</strong> its interacting<br />

proteins to health <strong>and</strong> disease. Current Medicinal Chemistry Oct 13. [Epub ahead of<br />

print]#<br />

2. Gareth J. Browne*, Margarida Fardilha*, Senga Oxenham*, Wenjuan Wu, Nick<br />

Helps, Odete A. B. da Cruz e Silva, Patricia T.W. Cohen <strong>and</strong> Edgar F. da Cruz e Silva<br />

(2007). SARP, a new alternatively spliced protein phosphatase 1 <strong>and</strong> DNA interacting<br />

protein. Biochemical Journal 402, 187-196. *, These authors contributed equally to this<br />

work.<br />

3. Wu W, Baxter JE, Wattam SL, Hayward DG, Fardilha M, Knebel A, Ford EM,<br />

da Cruz E Silva E, Fry AM (2007). Alternative splicing controls nuclear translocation<br />

of the cell cycle regulated nek2 kinase. J Biol Chem. Sep 7;282(36):26431-26440 (Jul 11;<br />

Epub ahead of print).<br />

42


Site-Specific Identification of SUMO-2 Conjugation Sites in Cells Reveals Novel<br />

SUMOylation Motifs<br />

Joost Schimmel 1 , Ivo A. Hendriks 1 , Ivan Matic 2 , Matthias Mann 2 <strong>and</strong> Alfred C.O.<br />

Vertegaal 1<br />

1 Molecular Cell Biology, Leiden University Medical Center, Leiden, 2300 RC, the<br />

Netherl<strong>and</strong>s. 2 Max-Planck Institute for Biochemistry, Martinsried, D-82152, Germany.<br />

Email: vertegaal@lumc.nl<br />

Post-translational modification by SUMO is essential for eukaryotic viability. Mass<br />

spectrometry-based proteomics projects have uncovered hundreds of potential SUMOylated<br />

proteins. However, direct identification by mass spectrometry of the SUMOylated lysines in<br />

endogenous target proteins is still very challenging. We have developed a novel method for<br />

selective enrichment of SUMOylated peptides from complex cellular proteomes. We have<br />

used this method to map 103 SUMO modification sites in endogenous target proteins purified<br />

from cell lysates. Our method is generic <strong>and</strong> can be adapted to study other ubiquitin-like<br />

proteins. Our results provide the following novel insights into protein SUMOylation:<br />

1. We have discovered an inverted SUMOylation consensus site E/DxKpsi.<br />

2. Several alternative amino acids were identified that precede the SUMOylated lysines in<br />

KxE type SUMOylation sites.<br />

3. We have identified SUMO attachment sites in sixteen proteins on lysines that were<br />

preceded by hydrophobic clusters of at least three hydrophobic residues <strong>and</strong> named this novel<br />

type of SUMOylation site the Hydrophobic Cluster SUMOylation Motif (HCSM). Follow up<br />

studies established the remarkable stoichiometric SUMOylation of target proteins via this<br />

HCSM.<br />

4. Virtually all identified SUMOylation sites that fit the classical SUMOylation consensus site<br />

psiKxE/D contain a glutamic acid instead of an aspartic acid.<br />

5. We have uncovered crosstalk between SUMOylation <strong>and</strong> phosphorylation with a preferred<br />

spacer of four amino acids between the SUMOylated lysine <strong>and</strong> the phosphorylated serine.<br />

6. SUMO-acceptor lysines in four proteins were previously identified as acetylated lysines.<br />

Since SUMOylation <strong>and</strong> acetylation are mutually exclusive, this indicates competition<br />

between acetylation <strong>and</strong> SUMOylation for the same lysines.<br />

43


Regulation of Lynch syndrome-related DNA mismatch repair heterodimer MutLalpha<br />

by phosphorylation?<br />

Angela Brieger, S<strong>and</strong>ra Passmann, Stefan Zeuzem <strong>and</strong> Jörg Trojan<br />

Medical Clinic I, Biomedical Research Laboratory, Goethe-University, Frankfurt a.M.,<br />

Germany<br />

Lynch syndrome, a hereditary disease associated with many different tumor types, is caused<br />

by mutations in DNA mismatch repair (MMR)-genes. 50% of these mutations are detected in<br />

the MLH1 protein. The heterodimer MutLalpha, which is formed by MLH1 <strong>and</strong> PMS2,<br />

coordinates a series of key events in the MMR mechanism. In addition to MMR, MutLalpha<br />

is involved in many other cellular processes as the regulation of cell cycle checkpoints <strong>and</strong><br />

apoptosis most likely signaling DNA damage to downstream pathways. A sophisticated<br />

regulation of the different MutLalpha functions might be important as well as reasonable. In<br />

order to focus on MutLalpha modulation, we analyzed MLH1 <strong>and</strong> PMS2 for phosphorylation<br />

sites using different computational systems <strong>and</strong> detected multiple potential loci <strong>and</strong><br />

corresponding kinases for both genes. Using phospho-specific purification columns we were<br />

able to isolate unphosphorylated <strong>and</strong> phosphorylated MLH1. In addition to this, Westernblot<br />

analysis of 2-D gels resolved MLH1 into several different distinct forms detected in lysates<br />

from HEK293 cells. Furthermore, we detected phosphorylation of purified MLH1 by<br />

PKCdelta, one of the computer-supported most probable MutLalpha kinases. Ongoing<br />

projects are directed towards the underst<strong>and</strong>ing how phosphorylation might be capable to<br />

switch MutLalpha function between MMR <strong>and</strong> DNA damage signaling.<br />

44


Mechanisms of Epigenetics in Health <strong>and</strong> Disease<br />

François Fuks<br />

Laboratory of Cancer Epigenetics, Free University of Brussels (ULB), 808 route de<br />

Lennik, 1070 Brussels, Belgium.<br />

!"#$%&'()*+',-.$/*+)0$+.$,%/-1'+.'$1-*&$,.$2+.2&1$+.3$,0$+00-2,+'&3$4,'($5&.&$0,*&.2,.5$<br />

-6$ '7%-71$ 07//1&00-1$ 5&.&08$ 9(&$ %&'()*+',-.$ -6$ :/;$ 0,'&0$ ,0$ &0'+


Molecular characterization of HDAC inhibitor-induced death in a mouse model of acute<br />

myeloid leukemia<br />

Michael Bots 1 , Inge Verbrugge 1 , Ben Martin 1 , Jessica Salmon 1 , Kym Stanley 1 , Johannes<br />

Zuber 2 , Scott Lowe 2 <strong>and</strong> Ricky Johnstone 1<br />

1 Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC,<br />

Australia; 2 Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States<br />

Underst<strong>and</strong>ing the molecular pathways targeted by chemotherapeutic drugs is essential<br />

information to decide on treatment modalities for patients with malignant disease. Histone<br />

deacetylase inhibitors (HDACi) are an interesting <strong>and</strong> encouraging group of anti-cancer<br />

drugs. However, the specific mechanism behind their anti-cancer effect remains largely<br />

unknown <strong>and</strong> may depend on the specific type of tumor <strong>and</strong>/or the oncogenic lesions that<br />

drive tumorigenesis.<br />

We have examined the therapeutic effect of HDACi in a mouse model of acute myeloid<br />

leukemia (AML). This model is based on the expression of both mutant Nras (co-expressed<br />

with luciferase) <strong>and</strong> the fusion protein AML1/ETO9a (co-expressed with GFP), in<br />

hematopoietic stem cells to drive tumorigenesis. Recruitment of HDAC-containing repressor<br />

complexes by this fusion protein is important for the onset <strong>and</strong> progression of AML1/ETOexpressing<br />

AML. This provides a strong rationale for using HDACi in the treatment of<br />

AML1/ETO9a-driven AML.<br />

Upon transplantation of primary leukemias into recipient mice, we found that treatment with<br />

the HDACi, panobinostat, indeed results in a good response, with clearance of leukemic blasts<br />

from the periphery. Importantly, this in vivo response translates into a significant overall<br />

survival benefit. In contrast to conventional chemotherapy, mice bearing p53 -/- leukemias still<br />

display survival benefit from treatment with panobinostat. To further characterize the<br />

molecular pathways utilized by panobinostat, we generated AML1/ETO9a leukemias with<br />

defined genetic alterations in the intrinsic as well as extrinsic apoptotic pathways. Similar to<br />

non-manipulated leukemias, we found that loss of TRAIL or DR5 <strong>and</strong> over-expression of Bcl-<br />

2 does not change the response to panobinostat. All these leukemias still displayed an overall<br />

survival benefit. This data suggest that induction of apoptosis is not required for therapeutic<br />

efficacy of panobinostat. Further mechanistic studies using additional genetically manipulated<br />

AML1/ETO9a tumors are currently ongoing.<br />

Our studies provide an important insight into the mechanism of HDACi-induced anti-tumor<br />

responses <strong>and</strong> demonstrate that HDACi are a rational choice for the treatment of human AML<br />

driven by expression of AML1/ETO.<br />

46


MicroRNAs <strong>and</strong> erythroid differentiation<br />

Roberto Gambari<br />

BioPharmaNet/ThalLab, Department of Biochemistry <strong>and</strong> Molecular Biology, Ferrara<br />

University, via Fossato di Mortara 74, 44121, Ferrara, Italy; e-mail:gam@unife.it.<br />

MicroRNAs (miRNAs, miRs) are a family of small noncoding RNAs that regulate gene<br />

expression by sequence-selective targeting of mRNAs, leading to translational repression or<br />

mRNA degradation. Considering that a single miRNA can target several mRNAs <strong>and</strong> the<br />

3’UTR sequence of a single mRNA might contain several signals for miRNA recognition, at<br />

least 10-40% of human mRNAs are potential targets of microRNAs, leading to control of<br />

highly regulated biological functions. In our laboratory we have analyzed by microarray the<br />

miR-pro<strong>file</strong> in erythroid precursor cells (ErPC) from normal <strong>and</strong> thalassemic patients<br />

expressing different levels of foetal haemoglobin (including patients exhibiting a HPFH<br />

phenotype). The microarray data were confirmed by RT-PCR analysis, <strong>and</strong> allowed us to<br />

identify miR-210 as highly expressed in the erythroid precursor cells from HPFH patients.<br />

When RT-PCR was performed on mithramycin (MTH)-induced K562 cells <strong>and</strong> erythroid<br />

precursor cells, we demonstrated that miR-210 is induced in time-dependent <strong>and</strong> dosedependent<br />

fashion, together with induction of !-globin genes. Molecular biology studies<br />

allowed to identify raptor mRNA <strong>and</strong> mTORC1 as putative miR-210 targets. As far as<br />

modulation of miR-210, peptide nucleic acids (PNAs) might be of interest. We have studied a<br />

PNA conjugated to a polyarginine peptide which (a) is efficiently internalized within target<br />

cells; (b) strongly inhibits miR-210 activity; (c) deeply alters the expression of raptor <strong>and</strong> !globin<br />

genes.<br />

Supported by Telethon (GGP10124), Fondazione CARIPARO <strong>and</strong> MIUR-PRIN-2007.<br />

47


MATHEMATICAL MODELLING OF CELLULAR DECISIONS BETWEEN LIFE<br />

AND DEATH IN CANCER<br />

Andrei Zinovyev 1 , Laurence Calzone 1 , Laurent Tournier 1 , Simon Fourquet 1 , Emmanuel<br />

Barillot 1 , Denis Thieffry 2 , Boris Zhivotovsky 3<br />

1 Institut Curie, INSERM U900, Paris, France<br />

2 Ecole Normale Superieur, Paris, France<br />

3 Karolinska Institutet, Stockholm, Sweden<br />

E-mail: <strong>and</strong>rei.zinovyev@curie.fr<br />

Compromised balance between cellular decisions towards survival <strong>and</strong> various modalities of<br />

programmed cell death is responsible for uncontrolled cell proliferation in tumors. In normal<br />

cells this balance is tightly controlled by a complex systems of molecular switches that<br />

irreversibly trigger a particular cellular fate. The complexity of this mechanism <strong>and</strong> the level<br />

of crosstalk between survival <strong>and</strong> cell death pathways are such that systems approach is<br />

needed to decipher its main features <strong>and</strong> predict possible intervention strategies to restore the<br />

violated balance.<br />

In order to formalize the existing knowledge in the field <strong>and</strong> to make it accessible for<br />

computer analysis, we reconstructed a comprehensive map of molecular interactions of the<br />

cell death machinery. For the moment this map contains 1587 chemical species involved in<br />

1133 reactions. This formal description is annotated by 563 relevant publications in the field.<br />

Formal analysis of the map together with high-throughput data allows to make general<br />

conclusions about involvement of particular molecular players in concrete cancers.<br />

In order to better underst<strong>and</strong> the main features of cell fate decision machinery, we have<br />

constructed a mathematical model of cell fate decision in response to triggering death<br />

receptors, based on logical (discrete) formalism. The model provides a generic high-level<br />

view of the interplays between NF!B pro-survival pathway, RIP1-dependent necrosis <strong>and</strong> the<br />

apoptosis pathway <strong>and</strong> constitutes a valuable reasoning tool that can suggest experiments for<br />

novel biological insights.<br />

48


A Systems Approach to Modeling <strong>and</strong> Predicting Oncogene Addiction<br />

Dean W Felsher<br />

Division of Oncology, Departments of Medicine <strong>and</strong> Pathology, Stanford, Medicine-<br />

Oncology, Stanford, CA, 94305<br />

The targeted inactivation of oncogenes can elicit sustained tumor regression, associated with<br />

the phenomenon that has been described as oncogene addiction. Utilizing conditional<br />

transgenic mouse models, we have gleaned some insight into the mechanisms of oncogene<br />

addiction illustrating that upon oncogene inactivation tumors undergo proliferative arrest,<br />

apoptosis, differentiation <strong>and</strong>/or senescence. The specific consequences of oncogene<br />

addiction appear to depend upon both cellular <strong>and</strong> genetic context <strong>and</strong> both tumor cell<br />

intrinsic <strong>and</strong> host-dependent mechanisms appear to be critical. Recently, we have utilized a<br />

conditional mouse model of MYC induced T-acute lymphoblastic lymphoma to dissect the<br />

role of proliferative <strong>and</strong> death signaling in tumor regression associated with MYC<br />

inactivation. By multi-scale modeling, we were able to show that characteristic changes in<br />

survival <strong>and</strong> death signaling can be used to predict oncogene addiction. We were able to<br />

directly test this model by interrogating the consequences of the mutation of specific key<br />

regulators of cellular signaling <strong>and</strong> demonstrating that using our model they had a predictable<br />

outcome on tumor regression. Finally, we were able to use our model to predict clinical<br />

outcome to erlotinib therapy in patients with lung cancer. Our results illustrate that using a<br />

systems approach we are able to derive models that may be useful to predict oncogene<br />

addition <strong>and</strong> thereby the clinical response to targeted oncogene inactivation.<br />

49


Microenvironmental Independence In Tumor Progression: An Integrated Approach<br />

Alex<strong>and</strong>er R. A. Anderson<br />

Integrated Mathematical Oncology (IMO), H. Lee Moffitt Cancer Center <strong>and</strong> Research<br />

Institute,Tampa, FL 33612.<br />

Mathematical <strong>and</strong> computational models are an ideal compliment to experimentation as they<br />

allow for the integration of multiple experimental results across a range of spatial <strong>and</strong><br />

temporal scales. In application to cancer they hold great promise for prediction of tumor<br />

outcomes; however their application has been limited by the lack of experimental data<br />

integration. Even though mathematical modeling of cancer is not new, in fact it goes back<br />

over half a century, it has largely been ignored - mostly due to insufficient communication<br />

between the mathematical <strong>and</strong> biological communities. These facts highlight the need for a<br />

new integrated approach to underst<strong>and</strong>ing cancer <strong>and</strong> biological systems in general. In this<br />

approach the dialogue between the modeler <strong>and</strong> the experimentalist drives the definition <strong>and</strong><br />

direction of both the model <strong>and</strong> the experiments that ultimately leads to fundamental insights<br />

that could not have been achieved independently. In this talk we discuss our recent efforts<br />

using mathematical modeling, computation <strong>and</strong> experimentation to study cancer progression,<br />

with a special emphasis on the role of the tumor microenvironment. Specifically, to<br />

underst<strong>and</strong> the competitive dynamics of tumor cells in diverse microenvironments, we<br />

experimentally parameterized a hybrid discrete-continuum mathematical model with<br />

phenotypic trait data from a set of related mammary cell lines with normal, transformed, or<br />

tumorigenic properties. Surprisingly, in a resource-rich microenvironment, with few<br />

limitations on proliferation or migration, transformed but not tumorigenic cells were most<br />

successful <strong>and</strong> outcompeted other cell types in heterogeneous tumor simulations. Conversely,<br />

constrained microenvironments with limitations on space <strong>and</strong>/or growth factors gave a<br />

selective advantage to phenotypes derived from tumorigenic cell lines. Analysis of the<br />

relative performance of each phenotype in constrained versus unconstrained<br />

microenvironments revealed that, although all cell types grew more slowly in resourceconstrained<br />

microenvironments, the most aggressive cells were least affected by<br />

microenvironmental constraints. Our results suggest that a critical feature of the process of<br />

tumor progression is selection of cells that can escape from resource limitations by achieving<br />

a relative microenvironmental independence.<br />

50


Targeting the invasive phenotype using systemic evolution of lig<strong>and</strong>s by exponential<br />

enrichment creates anti-metastatic aptamers.<br />

Greg Shelley, Xiaohua Zhang, Jinlu Dai, Chunyan Yu, Elisabeth A. Pedersen, Yusuke<br />

Shiozawa, June Escara-Wilke, Jill Keller, Jian Zhang, Russell S. Taichman, Evan T.<br />

Keller.<br />

University of Michigan, USA<br />

Multiple methods (e.g., small molecules <strong>and</strong> antibodies) have been designed to target specific<br />

proteins <strong>and</strong> signaling pathways in cancer. However, many mediators of the cancer<br />

phenotype are unknown <strong>and</strong> the ability to target these phenotypes would help mitigate cancer.<br />

Aptamers are small DNA or RNA molecules that are designed for therapeutic use. Design of<br />

aptamers to target cancers can be challenging. Accordingly, we used a modification of<br />

systemic evolution of lig<strong>and</strong>s by exponential enrichment (SELEX) to target a known<br />

phenotype of cancer metastasis, i.e. invasion. We call this method pheno-SELEX. A highly<br />

invasive prostate cancer (PCa) cell line was established <strong>and</strong> used to identify aptamers that<br />

bound to it with high affinity as opposed to a less invasive variant to the cell line. The antiinvasive<br />

aptamer (AIA1) was found to inhibit in vitro invasion of the original PCa cell line, as<br />

well as an additional PCa cell line <strong>and</strong> an osteosarcoma cell line. AIA also inhibited in vivo<br />

development of metastasis in both a PCa <strong>and</strong> osteosarcoma model of metastasis. These<br />

results indicate that pheno-SELEX can be successfully used to identify aptamers without<br />

knowledge of underlying molecular targets. This study establishes a new paradigm for<br />

identification of functional aptamers.<br />

51


Autophagy: suicidal self-cannibalism or homeostatic recycling?<br />

Guido Kroemer<br />

INSERM, U848, Villejuif, France; Metabolomics Platform, Institut Gustave Roussy, Villejuif,<br />

France; Centre de Recherche des Cordeliers, Paris, France; Pôle de Biologie, Hôpital Européen<br />

Georges Pompidou, AP-HP, Paris, France; Université Paris Descartes, Paris 5, Paris, France.<br />

Autophagy has been considered for some time as a mechanism of cellular self-destruction leading to<br />

cell death. We have tackled the question whether autophagy is cytoprotective or cytotoxic in two<br />

ways.<br />

First we have screened more than 1000 established or experimental anticancer agents for their capacity<br />

to induce autophagic LC3 puncta in human cancer cells, finding that some 100 were able to do so, but<br />

that none among these “autophagy inducers” killed tumor cells in an autophagy-dependent fashion.<br />

Rather autophagy inhibition sensitized the tumor cells to cell death induction.<br />

Second, we determined the impact of whole-body autophagy induction in experimental animals. We<br />

found that autophagy-inducing pharmacological agents such as resveratrol <strong>and</strong> spermidine enhanced<br />

life span in an autophagy-dependent fashion. Similarly genetic manipulations that increase longevity<br />

(such as inactivation of the C. elegans p53 ortholog Cep1 or overexpression of the SIRT1 ortholog<br />

sirt2.1) induce autophagy, <strong>and</strong> inhibition of autophagy abolishes their lifespan-extending effect. We<br />

found that induction of autophagy by resveratrol requires the NAD + -dependent deacetylase sirtuin 1<br />

(SIRT1). The acetylase inhibitor spermidine stimulates autophagy independent of SIRT1 in human<br />

<strong>and</strong> yeast cells as well as in nematodes. Although resveratrol <strong>and</strong> spermidine ignite autophagy through<br />

distinct mechanisms, these compounds stimulate convergent pathways that culminate in concordant<br />

modifications of the acetylproteome. Both agents favor convergent deacetylation <strong>and</strong> acetylation<br />

reactions in the cytosol <strong>and</strong> in the nucleus, respectively. At doses at which neither resveratrol nor<br />

spermidine did stimulate autophagy alone, these agents synergistically induced autophagy, both in<br />

vitro <strong>and</strong> in vivo. Altogether, these data underscore the importance of an autophagy-regulatory<br />

network of antagonistic deacetylases <strong>and</strong> acetylases that can be pharmacologically manipulated.<br />

Moreover, our data indicate that autophagy is mostly (always?) a cytoprotective event.<br />

References:<br />

Madeo F, Tavernarakis N, Kroemer G. Can autophagy promote longevity? Nat Cell Biol 2010 Sep;12(9):842-6<br />

Criollo A, Senovilla L, Kepp O, Authier H, Shen S, Maiuri MC, Tasdemir E, Tallier M, Morselli E, Galluzzi L, Delahaye N,<br />

Tesniere A, Commo F, Harper F, Vicencio JM, Ben Younes A, Pierron G, Lav<strong>and</strong>ero S, Zitvogel L, Israel A, Baud V,<br />

Kroemer G. The IKK complex contributes to the induction of autophagy. EMBO J. 2010 Feb 3;29(3):619-31<br />

Kroemer G, Marino G, Levine B. Autophagy <strong>and</strong> the integrated stress response. Mol Cell. 2010 Oct 22;40(2):280-93<br />

Morselli E, Maiuri MC, Markai M, Megalou E, Pasparaki A, Palikaras K, Galluzzi L, Criollo A, Malik SA, Madeo F,<br />

Tavernarakis N, Kroemer G. Caloric restriction <strong>and</strong> resveratrol prolong longevity via the sirtuin-1 dependent induction of<br />

autophagy. Cell Death Disease 1, e10<br />

Morselli E, Marino G, Bennetzen M, Eisenberg T, Megalou E, Schroeder S, Carbrera S, Bénit P, Rustin P, Criollo A, Shen S,<br />

Kepp O, Miauri C, Horio Y, Lopez-Otin C, Andersen JS, Tavernarakis N, Madeo F, Kroemer G. Spermidine <strong>and</strong> resveratrol<br />

induce autophagy by distinct yet convergent pathways affecting the acetylproteome. J Cell Biol in press<br />

Eisenberg T, Knauer H, Schauer A, Büttner S, Ruckenstuhl C, Carmona-Gutierrez D, Ring J, Schroeder S, Magnes C,<br />

Antonacci L, Fussi H, Deszcz L, Hartl R, Schraml E, Criollo A, Megalou E, Weiskopf D, Laun P, Heeren G, Breitenbach M,<br />

Grubeck-Loebenstein B, Herker E, Fahrenkrog B, Fröhlich KU, Sinner F, Tavernarakis N, Minois N, Kroemer G*, Madeo F.<br />

(*Co-corresponding author). Induction of autophagy by spermidine promotes longevity. Nat Cell Biol. 2009<br />

Nov;11(11):1305-14<br />

Tavernarakis N, Pasparaki A, Tasdemir E, Maiuri MC, Kroemer G. The effects of p53 on whole organism longevity are<br />

mediated by autophagy. Autophagy. 2008 Oct 1;4(7):870-3.<br />

Tasdemir E, Maiuri MC, Galluzzi L, Vitale I, Djavaheri-Mergny M, D'Amelio M, Criollo A, Morselli E, Zhu C, Harper F,<br />

Nannmark U, Samara C, Pinton P, Vicencio JM, Carnuccio R, Moll UM, Madeo F, Paterlini-Brechot P, Rizzuto R,<br />

Szabadkai G, Pierron G, Blomgren K, Tavernarakis N, Codogno P, Cecconi F, Kroemer G. Regulation of autophagy by<br />

cytoplasmic p53. Nat Cell Biol. 2008 Jun;10(6):676-87<br />

Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell 2008 Jan11; 132(1): 27-42<br />

Maiuri MC, Le Toumelin G, Criollo A, Rain JC, Gautier F, Juin P, Tasdemir E, Pierron G, Troulinaki K, Tavernarakis N,<br />

Hickman JA, Geneste O, Kroemer G. Functional <strong>and</strong> physical interaction between Bcl-X(L) <strong>and</strong> a BH3-like domain in<br />

Beclin-1. EMBO J. 2007 May 16;26(10):2527-39.<br />

52


Genes related to energy metabolism differentiate the visceral adipose-derived stem cells<br />

from the subcutaneous ones<br />

Yong Sang Song 1, 2, 3 , Boram Lee 1 , Seung Pyo Gong 4 , Aekyung Park 5 , Hee Seung Kim 2 ,<br />

Hyun Hoon Chung 2 , Sangchul Lee 6 , Woong-Yang Park 5 , Jeong Mook Lim 3, 7 , Noh Hyun<br />

Park 2 , Eunsik Lee 6<br />

1 Cancer Research Institute, College of Medicine, Seoul National University, Seoul 110-<br />

799, Republic of Korea (Song YS: yssong@snu.ac.kr; Lee B: kjkjmi@snu.ac.kr)<br />

2 Department of Obstetrics <strong>and</strong> Gynecology, Seoul National University College of<br />

Medicine, Seoul 110-744, Republic of Korea (Song YS: yssong@snu.ac.kr; Park NH:<br />

pnhkhr@snu.ac.kr; Chung HH; chhkmj@gmail.com; Kim HS; bboddi0311@snu.ac.kr)<br />

3 Major in Biomodulation, World Class University, Seoul National University, Seoul 151-<br />

921, Republic of Korea (Song YS: yssong@snu.ac.kr; Lim JM: limjm@snu.ac.kr)<br />

4 Department of Marine Bio-materials & Aquaculture, College of Fisheries Science,<br />

Pukyong National University, Busan 608-737, Republic of Korea (Gong SP:<br />

kongpyo2@snu.ac.kr)<br />

5 Department of Biomedical Sciences, Seoul National University College of Medicine,<br />

Seoul 110-744, Republic of Korea (Park WY: wypark@snu.ac.kr; Park A:<br />

parkak11@snu.ac.kr)<br />

6 Department of Urology, Seoul National University College of Medicine, Seoul 110-744,<br />

Republic of Korea (Lee E: eslee@snu.ac.kr; Lee S: uromedi@naver.com)<br />

7 Department of Agricultural Biotechnology, Research Institute for Agriculture <strong>and</strong> Life<br />

Science, Seoul National University, Seoul 151-742, Republic of Korea (Lim JM:<br />

limjm@snu.ac.kr)<br />

Central obesity is more strongly implicated in the pathogenesis of metabolic syndrome than<br />

peripheral obesity. The obesity, central or peripheral, has been presumed to be determined<br />

genetically. We investigated the expression pro<strong>file</strong>s of adipose-derived stem cells (ASC) from<br />

human subcutaneous (SC) <strong>and</strong> visceral (VIS) adipose tissues. First, we confirmed the<br />

characteristics of adipose derived stem cells (ASC) of isolated cells from SC <strong>and</strong> VIS fat<br />

depots by FACS using CD31, CD45, CD34, CD73 <strong>and</strong> CD105 antibody, <strong>and</strong> the<br />

differentiation of ASC into adipocyte, osteocyte <strong>and</strong> chondrocyte. Differential expression<br />

pattern was shown between S-ASC <strong>and</strong> V-ASC in more than 500 of 23700 genes. Genes<br />

related to lipid synthesis <strong>and</strong> oxidative phosphorylation were significantly up-regulated in V-<br />

ASC, while genes associated with insulin signaling were up-regulated in S-ASC. The<br />

expression patterns of adipocytes from subcutaneous <strong>and</strong> visceral fat tissue were similar. In<br />

inbred mice, the immunohistochemistry using antibody against each depot specific antigen<br />

showed the same pattern as shown in human samples. These data suggest that S-ASC may be<br />

different from V-ASC <strong>and</strong> this difference may explain the different risk of metabolic<br />

syndrome between central <strong>and</strong> peripheral obesity.<br />

53


A bioinformatic approach to illuminate why a biomarker, p27, has prognostic value in<br />

human tumors.<br />

Jeffrey P. Miller, Doruk Keskin, Andrew S. Goldstein, David Cobrinik, <strong>and</strong> Andrew<br />

Koff.<br />

Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021.<br />

Bioinformatics hold the promise to revolutionize discovery in basic science by constructing<br />

robust molecular networks. Thus, we decided to use a bioinformatic approach to address a<br />

problem originally raised by studies on cell transformation carried out by Hahn <strong>and</strong> his<br />

colleagues. Those investigators showed that SV40 small t-antigen (ST) or activated alleles of<br />

myc, akt, rac or ral could collaborate with SV40 large T-antigen (LT) <strong>and</strong> activated ras v12 to<br />

promote transformation in a variety of immortalized human cells. However, precisely what<br />

the relationship between these four signaling pathways was is unclear.<br />

Thus, we examined the connectivity of those oncogenes in silico. We found that the cdk<br />

inhibitor, p27, was connected either directly or through a single intermediary to every<br />

oncogene that would substitute for ST. p27 plays a role regulating the choice between<br />

proliferation <strong>and</strong> exit from the cell cycle, <strong>and</strong> a low p27 staining index is associated with poor<br />

outcome in a wide assortment of intermediate grade human cancers. Consistent with this, the<br />

loss of p27 will also accelerate tumor progression in a variety of mouse models regardless of<br />

the tissue of origin or the initiating oncogenic stimulus.<br />

Given that the oncogenes that replace ST were all linked to p27, <strong>and</strong> p27 loss cooperates with<br />

the expression of LT to accelerate tumor progression in mice, we tested the genetic<br />

relationship between ST <strong>and</strong> p27 as predicted by the bioinformatic model. We determined that<br />

p27 loss could substitute for the expression of ST during transformation of both rodent <strong>and</strong><br />

human cells. ST expression reduces p27 accumulation by accelerating phosphorylationdependent<br />

p27 turnover. ST mutants that lack the ability to inhibit PP2A activity do not<br />

cooperate with LT, nor was ST able to cooperate with LT to transform phosphorylationresistant<br />

S10A <strong>and</strong> T187A mutant cells to anchorage-independent growth. Thus, in silico<br />

pathway analysis developed a network that suggested that p27 levels were a direct reflection<br />

of the activation state of many oncogenic signaling pathways, which may account for its<br />

broad utility in human cancer prognosis.<br />

54


Pathological aspects of vascular aging <strong>and</strong> strategies for new biomimetic <strong>and</strong> biological<br />

active NonWoTecc biomaterials<br />

Stefanie Kaempf , Christoph Classen, Andreas Henseler, Frank Willems<br />

NonWoTecc Medical GmbH, Nattermannallee 1, Cologne 50829, Germany,<br />

s.kaempf@nonwotecc.de, c.classen@nonwotecc.de, a.henseler@nonwotecc.de,<br />

f.willens@nonwotecc.de, Tel: 0049 (0)221 992247-0, Fax: 0049 (0)221 992247-29<br />

The rising aging of the population <strong>and</strong> the typical lifestyle in the industrial nations are reasons<br />

for the increasing number of diabetes, arteriosclerosis <strong>and</strong> cardiovascular diseases. Hence<br />

there is a high medical need for new cardiovascular drugs <strong>and</strong> biomaterials in combination<br />

with stem/progenitor cell repair like endothelial progenitor cells (EPCs). Current vascular<br />

materials provide tolerable biocompatibility <strong>and</strong> average patency, except for small diameter<br />

coronary vessels. Especially for arteriosclerotic patients this situation often is a dilemma since<br />

there is no suitable autologous arteries or veins available for transplantation. In contrast to<br />

current bypasses the NonWoTecc graft is based on an entirely new biomimetic technology<br />

that mimics biomechanical properties <strong>and</strong> biological structures of arterial wall layers <strong>and</strong><br />

allows a long-term patency by autologous endothelial seeding via EPCs, adventitia sourced<br />

progenitor cells <strong>and</strong> cellular vascular wall reconstruction especially in aged patients. The EPC<br />

cell number is slightly reduced in individual aging patients but will sometimes increase<br />

because of disease stimuli like arteriosclerosis. Despite the age dependent changes of EPCs in<br />

older patients these cells will retain regenerative potential for vascular repair.<br />

As a result of the new cellular based autologous stem/progenitor cell concept, a significant<br />

reduced thrombosis <strong>and</strong> hyperplasia there is a need for new anti-inflammatory drugs for the<br />

NonWoTecc-technology which enable the endothel regeneration. COX-1 <strong>and</strong> COX-2<br />

inhibition by Aspirin is a good concept to inhibit thrombosis as well as neointimal<br />

hyperplasia, whereas Clopidogrel prevents thrombosis via P2Y12-receptor inhibition.<br />

The in vivo results in animals <strong>and</strong> clinial results in the first 30 humans after 10 months<br />

demonstrate the biomimetic concept <strong>and</strong> outst<strong>and</strong>ing biocompatibility of the of the<br />

NonWoTecc grafts by a reconstruction of a functional arterial wall scaffold with<br />

Neoendothel, -media <strong>and</strong> –adventitia, promising a long-term patency of NonWoTecc vascular<br />

grafts <strong>and</strong> an improvement of quality of life in aging, vascular diseased patients.<br />

55


Signaling Pathways that Help Neurons Help Themselves<br />

Mark P. Mattson<br />

Laboratory of Neurosciences, National Institute on Aging Intramural Research<br />

Program, Baltimore, MD. USA.<br />

It is important that neurons be stimulated regularly to activate adaptive stress response<br />

signaling pathways involving transcription factors that induce the expression of neurotrophic<br />

factors, protein chaperones <strong>and</strong> antioxidant enzymes. Such stimulation can be accomplished<br />

by exercise (both physical <strong>and</strong> cognitive), dietary energy restriction <strong>and</strong> ingestion of certain<br />

phytochemicals <strong>and</strong> drugs. The results of studies of animal models suggest that dietary<br />

energy restriction <strong>and</strong> exercise can enhance neural plasticity <strong>and</strong> reduce the vulnerability of<br />

the brain to disorders such as Alzheimer’s, Parkinson’s diseases, <strong>and</strong> stroke. Diabetes <strong>and</strong><br />

obesity may compromise the function of adaptive cellular stress response pathways <strong>and</strong><br />

thereby render neurons vulnerable to aging, injury <strong>and</strong> disease. Energy state-sensitive factors<br />

that are proving particularly important in regulating energy balance <strong>and</strong> improving or<br />

preserving cognitive function are brain-derived neurotrophic factor <strong>and</strong> glucagon-like peptide<br />

1. Alternate day calorie restriction, novel insulin-sensitizing <strong>and</strong> neuroprotective agents, <strong>and</strong><br />

drugs that activate adaptive stress response pathways, are examples of approaches for<br />

preserving brain function that show promise in preclinical studies.<br />

56


Control of class III PI3 kinase in autophagy <strong>and</strong> cancers<br />

Marta M. Lipinski, Minsu Kim, Greg Hoffman, Aylwin Ng, John Blenis, Ramnik J.<br />

Xavier <strong>and</strong> Junying Yuan. .<br />

Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston,<br />

MA 02115; Center for Computational <strong>and</strong> Integrative Biology, Massachusetts General<br />

Hospital, Harvard Medical School, 185 Cambridge Street, Boston, MA 02114<br />

Autophagy, a lysosome-dependent catabolic process involved in the turnover of cellular<br />

components, mediates normal homeostasis during development <strong>and</strong> protects multicellular<br />

eukaryotes from neurodegeneration, cancer <strong>and</strong> other diseases. However, the mechanisms<br />

regulating autophagy under normal nutritional conditions most frequently encountered by<br />

cells in organisms under physiological conditions remain unknown. Our recent studies<br />

highlight an important role of class PI3 kinase in regulating autophagy. First, we show that<br />

phosphorylation of Vps34 by Cdk1 <strong>and</strong> Cdk5 negatively regulate type III PI3 kinase <strong>and</strong><br />

production of PI3P in mitotic cells <strong>and</strong> in neurodegeneration. Second, we show that multiple<br />

growth factors negatively regulate the activity of type III PI3 kinase. In an image-based<br />

genome-wide human siRNA screen, we show that up regulation of autophagy by only 14<br />

(6.4%) of the identified hit genes was accompanied by down regulation of activity of<br />

mTORC1, an essential regulator of starvation induced autophagy. On the other h<strong>and</strong>, we<br />

found that 110 (50.2%) of the hit genes, including a group of growth factors <strong>and</strong> cytokines,<br />

regulate autophagy by targeting the type III PI3 kinase. These cytokines inhibit the type III<br />

PI3 kinase through multiple pathways. Third, we show that ROS may have a general signaling<br />

function in autophagy. We uncovered a large group of genes (117, or 54% of all genes tested)<br />

whose knock-down led to vesicular translocation of LC3-GFP in the absence but not the<br />

presence of anti-oxidant, suggesting that ROS are required for the induction of autophagy.<br />

Finally, using a small molecule inhibitor that promotes the degradation of class III PI3 kinase,<br />

we show that successful targeting this pathway can lead to selective cancer cell death. Our<br />

study suggests that the type III PI3 kinase integrates diverse signals to regulate cellular levels<br />

of autophagy. Thus, autophagy <strong>and</strong> cell proliferation may represent two alternative cell fates<br />

that are regulated in a mutually exclusive manner, implicating autophagy as a common tumor<br />

suppression mechanism downstream of multiple signaling pathways.<br />

57


VITAGENES, METABOLIC STRESS AND HORMESIS IN AGING AND<br />

NEURODEGENERATIVE DISORDERS<br />

Vittorio Calabrese<br />

Chairman <strong>and</strong> Professor of Clinical Biochemistry, Faculty of Medicine, University of<br />

Catania, Italy;<br />

Protein quality control is a critical feature of intracellular homeostasis. In particular, unfolded or<br />

misfolded proteins resulting from environmental stresses or free radicals are rapidly degraded via the<br />

ubiquitin–proteasome pathway 1 . In addition, modulation of endogenous cellular defense mechanisms<br />

via the stress response signaling represents an innovative approach to therapeutic intervention in<br />

diseases causing chronic tissue damage, such as neurodegeneration <strong>and</strong> cancer 1 . Protein thiols play a<br />

key role in redox sensing, <strong>and</strong> regulation of cellular redox state is crucial mediator of multiple<br />

metabolic, signalling <strong>and</strong> transcriptional processes. Under optimal conditions long-term health<br />

protection is accomplished by protein homeostasis, a highly complex network of molecular<br />

interactions that balances protein biosynthesis, folding, translocation, assembly/disassembly, <strong>and</strong><br />

clearance 2,3 . Efficient functioning of maintenance <strong>and</strong> repair processes is crucial for both survival <strong>and</strong><br />

physical quality of life. This is accomplished by a complex network of the so-called longevity<br />

assurance processes, which are composed of several genes termed vitagenes 4-8 . The term vitagenes<br />

refers to a group of genes which are strictly involved in preserving cellular homeostasis during<br />

stressful conditions. The vitagene family is actually composed of the heat shock proteins (Hsp) Hsp32,<br />

Hsp70, the thioredoxin system <strong>and</strong> the sirtuin system 9 . Dietary antioxidants, such as polyphenols <strong>and</strong><br />

L-carnitine/acetyl-L-carnitine, have recently been demonstrated in vitro to be neuroprotective through<br />

the activation of hormetic pathways, including vitagenes 10-12 . Over the past decade there has been a<br />

remarkable increase of interest in hormesis as a result of more significance being given to low dose<br />

effects <strong>and</strong> the use of more powerful study designs which have enabled to identify rational approaches<br />

to detect hormetic biphasic dose responses in the low dose zone. The hormetic dose–response,<br />

challenging long-st<strong>and</strong>ing beliefs about the nature of the dose–response in a lowdose zone, has the<br />

potential to affect significantly the design of pre-clinical studies <strong>and</strong> clinical trials as well as strategies<br />

for optimal patient dosing in the treatment of numerous diseases, including oxidant disorders. Given<br />

the broad cytoprotective properties of the heat shock response there is now strong interest in<br />

discovering <strong>and</strong> developing pharmacological agents capable of inducing stress responses. We have<br />

recently focused our research on the role of acetylcarnitine in the defense mechanisms against cellular<br />

stress <strong>and</strong> neurodegeneration. In addition, with a redox proteomics approach, we identified<br />

mitochondrial oxidatively modified proteins as a function of brain aging, specifically in those brain<br />

regions, such as cortex <strong>and</strong> hippocampus, that are commonly affected by the aging process. In all brain<br />

regions examined, many of the identified proteins were energy-related, such as pyruvate kinase, ATP<br />

synthase, aldolase, creatine kinase, <strong>and</strong> a-enolase. These alterations were associated with increased<br />

expression of Hsps, as well as carnosinase <strong>and</strong> thioredoxin reductase <strong>and</strong> with significant changes in<br />

both cytosolic <strong>and</strong> mitochondrial redox status in all brain regions analyzed. This findings are relevant<br />

to potential pharmacological interventions in healthy medicine strategy, pointing to maximize cellular<br />

stress resistance of the brain thus providing neuroprotection 9-14 , <strong>and</strong> will be extended to other systemic<br />

oxidant disorders such as diabetic nephropathy or cancer.<br />

References:<br />

1 Halliwell B. (2008) Arch Bioch Biophys 476:107; 2 Calabrese V. (2007) Nature Neurosci 8,766;<br />

3 Gutteridge JM (2010) BBRC 393:561; 4 Calabrese V. (2010) J Neurosi Res 88:3498;<br />

5 Calabrese V. (2010) Antiox <strong>Redox</strong> Signal 13:1763; 6 Calabrese V. (2009) Front Biosci. 14:376;<br />

7 Calabrese V. (2009) Antiox <strong>Redox</strong> Signal 11:2717; 8 Calabrese V. (2010) Neurochem Res 35:1880;<br />

9 Calabrese V. (2009) Antiox <strong>Redox</strong> Signal 11:2759; 10 Calabrese EJ (2010) Hum Exp Toxicol. 29:1034;<br />

11 Calabrese EJ (2010) Hum Exp Toxicol. 29:980; 12 Calabrese V. (2010) Neurochem Res 35:2184;<br />

13 Mattson MP. (2010) Neuron 67:900; 14 Min SW. (2010) Neuron 67:953.<br />

58


p75NTR regulates A! deposition by increasing A! production but inhibiting A!<br />

aggregation with its extracellular domain<br />

Xin-Fu Zhou, Xin Wang, Jian-Jun Lu, Qiao-Xin Li, Chang-Yue Gao, Xiao-Hong Liu,<br />

Yin Sun, Miao Yang, Yoon Lim, Genevieve Evin, Jin-Hua Zhong, Hua-Dong Zhou, Yan-<br />

Jiang Wang<br />

Department of Human Physiology <strong>and</strong> Centre for Neuroscience, Flinders University<br />

GPO Box 2100, Adelaide, South Australia<br />

Accumulation of toxic amyloid-beta (A!) in the cerebral cortex <strong>and</strong> hippocampus is a major<br />

pathological feature of Alzheimer’s disease (AD). The neurotrophin receptor, p75NTR, has<br />

been proposed to mediate A!- induced neurotoxicity; however its role in the development of<br />

AD remains to be clarified. The p75NTR/ExonIII-/- mice <strong>and</strong> APPSwe/PS1dE9 mice were<br />

crossed to generate transgenic AD mice with deletion of p75NTR gene. Study of p75NTR<br />

immunoreactivity in the AD model APPSwe/PS1dE9 transgenic mice, indicates its<br />

localization to the basal forebrain neurons <strong>and</strong> degenerative neurites in amyloid plaques of<br />

neocortex, increase with aging, <strong>and</strong> further activation by A! accumulation. Deletion of the<br />

p75NTR gene by crossing APPSwe/PS1dE9 mice with p75NTR knockout mice reduced<br />

soluble A! levels in the brain <strong>and</strong> plasma, but increased the accumulation of insoluble A! <strong>and</strong><br />

A! plaque formation. There was no change in the levels of amyloid precursor protein (APP)<br />

<strong>and</strong> its proteolytic derivatives, or ", ! <strong>and</strong> # secretase activities, or in levels of BACE1,<br />

neprilysin (NEP) <strong>and</strong> insulin degrading enzyme (IDE) proteins. Recombinant extracellular<br />

domain of p75NTR attenuated the oligomerization <strong>and</strong> fibrillation of synthetic A!42 peptide<br />

in vitro, <strong>and</strong> reduced local A! plaques after hippocampus injection in vivo. In addition,<br />

deletion of p75NTR attenuated microgliosis but increased the microhemorrhage pro<strong>file</strong>s in<br />

the brain. The deletion of p75NTR did not significantly change the cognitive function of the<br />

mice up to the age of 9 months. Our data suggest that p75NTR plays a critical role in<br />

regulating A! levels by both increasing A! production <strong>and</strong> attenuating its aggregation <strong>and</strong><br />

they caution that a therapeutic intervention simply reducing p75NTR may exacerbate AD<br />

pathology.<br />

59


A “no-brainer” – mitochondrial <strong>and</strong> neurodegenerative disease in Dictyostelium.<br />

Paul R. Fisher, Lisa M. Francione, Claire Y. Allan, Paige K. Smith, Sanjanie Fern<strong>and</strong>o,<br />

Shawn de Piazza, Jasmine Ilievska, Oana Sanislav, Anita Chavan, Suwei Chen, William<br />

J. Burrage <strong>and</strong> Sarah J. Annesley.<br />

Department of Microbiology, La Trobe University, Melbourne, AUSTRALIA.<br />

P.Fisher@latrobe.edu.au<br />

The major intracellular signalling pathways of eukaryotic cells were already present in the<br />

primeval ancestor of animals, fungi <strong>and</strong> Amoebozoa. These ancient regulatory networks are<br />

deranged in many human diseases, including mitochondrial <strong>and</strong> neurodegenerative diseases.<br />

The social amoebozoan Dictyostelium discoideum is an experimentally tractable<br />

microorganism whose unique life cycle offers diverse phenotypic “readouts” of conserved<br />

signalling pathways. We have created, in Dictyostelium, sublethal genetic disease models for<br />

mitochondrial disorders <strong>and</strong> various neurodegenerative diseases (Batten Disease,<br />

Mucolipidosis, Parkinson’s Disease). In all cases the disease leads to a chronic dysregulation<br />

of intracellular signalling networks, sublethal at the cellular level but causing whole organism<br />

pathology. We have begun to dissect these pathways genetically <strong>and</strong> to study the resulting<br />

disease phenotypes in growth <strong>and</strong> cell cycle progression, photo- <strong>and</strong> thermosensory signal<br />

transduction, multicellular development (including programmed autophagic cell death),<br />

endocytic nutrient uptake <strong>and</strong> susceptibility to the intracellular bacterial pathogen, Legionella<br />

pneumophila. In mitochondrial disease ATP generation is compromised, leading to chronic<br />

hyperactivity of an energy-sensing protein kinase, AMPK (AMP-activated protein kinase) <strong>and</strong><br />

thence to diverse downstream cytopathologies. As well as AMPK-mediated defects,<br />

additional disease pathways are activated in a specific Complex I deficiency caused by<br />

knocking out MidA, a novel Complex I assembly factor. AMPK is not involved at all in<br />

Neuronal Ceroid Lipofuscinosis (Batten Disease). Instead, one of its downstream targets,<br />

TOR Complex I (TORC1) is inhibited via the Rheb GTPase. Rheb also acts on TOR Complex<br />

2 (TORC2) so that lysosomal dysfunction in Batten Disease also misregulates TORC2 targets.<br />

In Mucolipidosis, decreased function of the lysosomal Ca 2+ channel, mucolipin, causes<br />

defects in intracellular Ca 2+ signalling that are superimposed on <strong>and</strong> modify the phenotypic<br />

outcomes of lysosomal dysfunction. Both wild type <strong>and</strong> disease-causing point mutant forms<br />

of the Parkinson’s Disease protein alpha-synuclein are cytotoxic but the pathways are distinct<br />

from those of mitochondrial <strong>and</strong> lysosomal disorders. However a disease-causing truncated<br />

form of the protein is mislocalized in the cell, forms cytoplasmic aggregates that associate<br />

with the mitochondria <strong>and</strong> causes additional phenotypes characteristic of mitochondrial<br />

dysfunction.<br />

60


Workshop presentations<br />

61


Underst<strong>and</strong>ing <strong>and</strong> performing DNA <strong>and</strong> siRNA transfection<br />

Geraldine Guerin-Peyrou<br />

Polyplus Transfection<br />

Transfection is widely used as a tool in research. It consists in introducing nucleic acids into<br />

mammalian cells. If inadequate, transfection can become a limiting step for any project. Thus<br />

it is critical to underst<strong>and</strong> the mechanism of transfection to choose the most appropriate<br />

method. This tutorial will present the various transfection mechanisms as well as currently<br />

available methods <strong>and</strong> reagents for DNA <strong>and</strong> siRNA transfection. We will explain why some<br />

methods are more effective at transporting <strong>and</strong> releasing DNA while others are better for<br />

siRNA. In addition the critical parameters for transfection <strong>and</strong> their optimisation will be<br />

described. Our goal is to give a solid basis in transfection in order to obtain the best results in<br />

the shortest timeframe.<br />

62


Agilent Technologies<br />

New enrichment tools with high flexibility for targeted sequencing of exones <strong>and</strong> cancer related<br />

regions.<br />

Winfried van Eyndhoven<br />

Agilent Technologies, Amstelveen, The Netherl<strong>and</strong>s.<br />

While next-generation sequencing has revolutionized the way genomes are sequenced, this technology<br />

possesses a fundamental weakness—the inability to easily target specific regions of a genome. To<br />

address this, Agilent has developed the SureSelect platform that allows next-generation sequencing<br />

researchers to focus their sequencing efforts on genomic targets of interest, greatly improving nextgeneration<br />

sequencing process efficiencies. The SureSelect platform of products allows nextgeneration<br />

sequencing researchers to increase study sample size <strong>and</strong> reduce overall experimental cost.<br />

During this presentation information will be given about the enrichment technology <strong>and</strong> its<br />

performance for capturing whole exomes, kinases or RNA transcripts.<br />

Opening the archives for state of the art tumor genetic research – FFPE based sample<br />

processing for array-CGH.<br />

Karoly Szuhai<br />

Leiden University Medical Center dept. Molecular Cell Biology, Leiden, The Netherl<strong>and</strong>s.<br />

Molecular genetic studies on rare tumor entities, such as bone tumors, often require the use of<br />

decalcified, formalin-fixed, paraffin-embedded tissue (dFFPE) samples. Regardless of which<br />

decalcification procedure is used, this introduces a vast breakdown of DNA that precludes the<br />

possibility of further molecular genetic testing. We set out to establish a robust protocol that would<br />

overcome these intrinsic hurdles for bone tumor research. This presentation will discuss the findings<br />

of these developments.<br />

Developing a kinome sequencing platform to identify key signaling pathways in breast cancer<br />

Ian J. Majewski PhD<br />

The Netherl<strong>and</strong>s Cancer Institute (NKI-AVL), Department of Molecular Carcinogenesis<br />

email: i.majewski@nki.nl<br />

Developing a kinome sequencing platform to identify key signaling pathways in breast cancer Breast<br />

cancer is a heterogeneous disease. Our work focuses on two very different types of breast cancer,<br />

invasive lobular carcinoma (ILC) <strong>and</strong> triple negative breast cancer (TN), which together represent<br />

25% of all breast cancers. New therapies are desperately required for the treatment of these cancers.<br />

Kinases are important mediators of cell signaling that are considered to be good therapeutic targets.<br />

We have developed an exon capture system for high-throughput mutation analysis that will allow us to<br />

identify kinases that are mutated in ILC <strong>and</strong> TN breast cancer. I will discuss our work to validate the<br />

kinome sequencing platform in cell lines models <strong>and</strong> xenografts. We aim to extend this work to survey<br />

several hundred cancers from patients with ILC <strong>and</strong> TN breast cancer.<br />

Speaker:<br />

63


An Introduction to High Content Screening of Intracellular Signaling Assays using<br />

the IN Cell Analyzer 2000<br />

Sarah Payne<br />

Modality Manager IN Cell <strong>and</strong> Cell Factory, GE Healthcare<br />

The IN Cell Analyzer 2000 from GE Healthcare represents a high performance, automated<br />

imaging system which is capable of managing the entire high content analysis workflow, from<br />

automated image acquisition through to data quantification, data mining <strong>and</strong> data<br />

visualisation. Designed with flexibility in mind, the IN Cell Analyzer 2000 is equipped<br />

to h<strong>and</strong>le a diverse range of research requirements, from investigative microscopy to<br />

automated high throughput screening. The scope of biological assays that can be performed<br />

using this system range from those on a sub-cellular <strong>and</strong> cellular level, through to the imaging<br />

of tissue sections <strong>and</strong> small organisms. This modular system may be equipped with<br />

transmitted light options for label-free analysis of cellular responses, plus environmental<br />

control (CO2, temperature control, humidity) <strong>and</strong> liquid h<strong>and</strong>ling for the monitoring of fast<br />

<strong>and</strong> slow kinetic effects within live cells.<br />

This presentation will provide an overview of the features <strong>and</strong> benefits of the IN Cell<br />

Analyzer 2000 for monitoring intracellular signalling events.<br />

64


MAPTrix TM biomimetics from amsbio: Innovation in cell culture for cancer, stem cell<br />

<strong>and</strong> regenerative medicine research<br />

Alex Sim<br />

AMSBIO<br />

The extracellular matrix (ECM) is a natural substrate/scaffold that has been widely used in cell<br />

culture to enhance cellular adhesion. Cellular adhesion to ECM proteins is a fundamental feature<br />

of cell development, maintenance of tissue organization, <strong>and</strong> many pathologicalb conditions. It<br />

is, therefore, crucial to underst<strong>and</strong> the adhesion process in order to study cellular functions or<br />

tissue engineering applications.<br />

Most experiments require cells to proliferate, differentiate <strong>and</strong> migrate on the substrate. Cell<br />

adhesion to ECM lig<strong>and</strong>s is of growing importance in studying invasion, migration <strong>and</strong> wound<br />

healing.<br />

The immobilization of short peptides such as RGD on synthetic or natural materials may<br />

produce biofunctional surfaces that bind adhesion receptors <strong>and</strong> promote cell adhesion,<br />

however, this conventional chemistry approach is time-consuming, inconvenient, <strong>and</strong> lacks<br />

reproducibility.<br />

To overcome these problems, AMSBIO will present MAPTrix TM , a series of “ECM-like”<br />

biomimetic surfactant polymers that exhibit quantitative control over the proliferation <strong>and</strong><br />

migrational properties of various human cells.<br />

MAPTrix TM technology has been used to produce:<br />

- Direct ECM substitutes,<br />

- A basement membrane non-animal protein incorporating peptides that mimic the biological<br />

activity of the large, natural growth factors,<br />

- A user-defined <strong>and</strong> in situ formable hydrogel system called MAPTrix HyGel TM which<br />

generates three-dimensional (3D) extracellular matrix environments for 3D cell culture<br />

applications.<br />

The Mussel Adhesive Protein based matrix (MAPTrix TM ) line of products are coating<br />

reagents with genetically incorporated bioactive peptides. They mimic the extracellular matrix<br />

(ECM) activity of collagen, fibronectin, laminin, <strong>and</strong> vitronectin proteins <strong>and</strong> promote cell<br />

attachment, spreading <strong>and</strong> growth. A MAPTrix TM surface coating offers an animal-free,<br />

chemically defined attachment <strong>and</strong> growth surface that provides a highly controlled environment<br />

for cell culture <strong>and</strong> tissue engineering applications.<br />

The presentation will focus on 3D cell culture without the need for Matrigel <strong>and</strong> similar animalderived<br />

substrates in cell culture experiments. Changing the way we consider cell culture is<br />

crucial to underst<strong>and</strong>ing the adhesion process in order to study cellular functions or tissue<br />

engineering applications. In particular, promotion of attachment <strong>and</strong> spreading of many normal<br />

<strong>and</strong> stem cells including epithelial, endothelial, hepatocyte <strong>and</strong> mesenchymal stem cells will be<br />

covered.<br />

65


j_A5p_11:BJ 17/1/11 11:14 Page 1<br />

Improved discovery » Increased visibility » Enhanced retrieval » Animations » Multimedia » 3D interactive structures<br />

NEW KNOWLEDGE ENVIRONMENTS!<br />

Be Seen<br />

Be Cited<br />

Biochemical<br />

Journal<br />

Chair Peter R. Shepherd – Auckl<strong>and</strong><br />

Vice Chair, The Americas Guy Salvesen – La Jolla, CA<br />

Vice Chair, Asia-Pacific Tao Xu – Beijing<br />

Vice Chair, Europe Dario Alessi – Dundee<br />

Vice Chair, Reviews Alex Toker – Boston<br />

● Nine knowledge environments<br />

● Fast decision<br />

● Immediate publication<br />

● No submission or page charges<br />

● – Fully compliant with funding<br />

body m<strong>and</strong>ates<br />

...submit your paper online at<br />

www.BiochemJ.org<br />

See us at st<strong>and</strong> #7<br />

Portl<strong>and</strong> Press » publishing innovation<br />

Impact<br />

factor up<br />

again!<br />

5.155<br />

Published in<br />

Central<br />

Reviews<br />

Cell<br />

ChemBio<br />

Disease<br />

Energy<br />

Gene<br />

Metabolism<br />

Plant<br />

Signal<br />

Structure<br />

Classics<br />

China<br />

Images courtesy of: Dr G. Beakes, Newcastle University | C. A. Mannella, Research & Technology, Wadsworth Center, Albany, NY | S. M. Stack, University of Missouri | Nottingham Arabidopsis Stock Centre (NASC) G. P. Côté, Kingston, Ont. | G. Salvesen, La Jolla<br />

– an innovative way of viewing articles online


&:;4"N]LL"<br />

LSM"_/80?)1."#X""<br />

LTM"K4+E;+)8P012-8`/394-""<br />

LUM"!@I#=O""<br />

LWM"(&"A/2+05321/"&)14;/"(


!"#$%&<br />

'()*+),-&.,+&&<br />

/$$0,-&1$,2$%&<br />

34)*4.%/",)$&<br />

5",1$%2&6.**&<br />

7.),&$,2%.,1$&


2<br />

1<br />

Map <strong>and</strong> bus stops<br />

3<br />

4<br />

Meeting Center


Map <strong>and</strong> bus stops (city center)<br />

8<br />

5<br />

6<br />

Meeting Center<br />

7


11<br />

5<br />

PARC DE L'EUROPE<br />

Dommeldange<br />

Walferdange<br />

WALFER-GARE<br />

19<br />

25<br />

DOMMELDANGE-GARE<br />

CHÂTEAU<br />

CYPRIEN<br />

MERJAI<br />

HELSEMER BARRIÄR<br />

SCHOUL<br />

10<br />

JEAN ENGLING<br />

HENRI<br />

DUNANT<br />

Beggen<br />

STESEL-MICHEL<br />

RODANGE PLAZ<br />

GEMENGE PLAZ<br />

Senningerberg<br />

VAN DER MEULEN<br />

TOMMYS<br />

BRUCH<br />

BOURGAASS<br />

KNUPP<br />

DOMMELDANGE-<br />

CONTOURNEMENT<br />

EMILE<br />

METZ<br />

BINNCHEN<br />

BASTOGNE<br />

BÄRELDENG-<br />

SCHOUL<br />

LAVAL<br />

BIECHELCHEN<br />

EECHER<br />

KLINIK<br />

HENRI<br />

HEYMANS<br />

BEGGEN-<br />

KIERCH<br />

BEGGEN-<br />

RUE DU PONT<br />

BÄRELDENG-<br />

POLICE<br />

GONNESCHHOF BÄRELDENG-<br />

ELTERSTRACHEN<br />

UM GRUEF<br />

9<br />

WEIMERSKIRCH<br />

PIERRE-ELOI<br />

DUERF SCHOUÉ ST VITH<br />

MËNSTERBËSCH<br />

CHARLYS STATIOUN<br />

LAANGSCHIB<br />

Weimerskirch<br />

MÄERTESBUR<br />

LAVALS<br />

PARK<br />

Eich<br />

Mühlenbach<br />

Steinsel<br />

Réseau des<br />

Autobus<br />

Municipaux<br />

RUE DU GOLF<br />

PAUL<br />

NOESEN<br />

VILLEROY & BOCH PEIFFESCHBIERG SIWEMUERGEN EECHER SCHMELZ ECOLE EICH EECHER PLAZ<br />

EICH-CENTRE CULTUREL<br />

21 13<br />

PRINCE<br />

FELIX<br />

KIRCHBERG-KIERCH<br />

4 2<br />

Rollingergrund<br />

22<br />

LÉON THYES MATHIAS TRESCH<br />

AVALON<br />

SCHMATT<br />

Pfaffenthal<br />

AVENIR<br />

SQUARE<br />

ANDRÉ<br />

3<br />

LYCÉE TECHNIQUE<br />

MICHEL LUCIUS<br />

LÉANDRE<br />

LACROIX<br />

CENTRE<br />

UNIVERSITAIRE<br />

NEUMANS<br />

PARK<br />

CENTRE HOSPITALIER<br />

SICHEGRONN<br />

UNIVERSITÉ<br />

KIRCHBERG<br />

OCTROI<br />

LEVÈVRE<br />

RÉIMERWEE<br />

BATTY<br />

WEBER<br />

19<br />

STRASSEN-PRIMEURS<br />

WAASSER<br />

TUERM<br />

KETTEN<br />

BOIS<br />

KANNERKLINIK<br />

ALPHONSE<br />

WEICKER<br />

ALBERT<br />

BORSCHETTE<br />

KONRAD<br />

ADENAUER<br />

ANTOINE DE<br />

ST EXUPÉRY<br />

JEAN MONNET<br />

ALEN<br />

TRAMS-<br />

ERMESINDE N. S. PIERRET<br />

ROUDEBIERG<br />

POUTTY STEIN<br />

G. SCHNEIDER<br />

SCHAPP<br />

STADE<br />

G. DIDERICH<br />

CRISPINUS<br />

VAL ST ANDRE<br />

CARLO HEMMER<br />

LYCÉE DES<br />

GARÇONS<br />

ALFRED<br />

DE MUSSET<br />

HENRI VII<br />

WANDMILLEN<br />

ARTS & MÉTIERS<br />

P+R<br />

PARKING FOIRE<br />

Kirchberg<br />

VAL FLEURI<br />

BELLE-VUE<br />

BEI<br />

SICHENHAFF<br />

ALEN<br />

EECHERBIERG<br />

LUXEXPO<br />

Limpe rts berg<br />

BEIM KLOMP<br />

STADE<br />

12 1<br />

LYCÉE ROBERT SCHUMAN<br />

ALLÉE<br />

SCHEFFER<br />

GLACIS/<br />

NIKLOS-<br />

KIERFECHT<br />

BEI DER FIELS<br />

Strassen<br />

BELLE-<br />

ÉTOILE GEESENECK BARBLÉ AURELIA HUORGARTEN<br />

8<br />

18<br />

COUDENHOVE-<br />

KALEGRI<br />

LÉON HENGEN<br />

ERASME/D'COQUE<br />

BIISCHTEFABRIK<br />

SERVICE D'INCENDIE<br />

SCHOENACHT KESSELER BENELUX HONDSECK<br />

WAASSERTUERM<br />

AUBÉPINES<br />

LES<br />

AUBÉPINES<br />

GRANDE-DUCHESSE<br />

CHAPELLE CHARLOTTE RÉIMERWEE<br />

MILLEWEE<br />

SENNÉNGERBIERG-<br />

AUTOBUNN<br />

HÔPITAL<br />

KIRCHBERG<br />

JOHN F. KENNEDY<br />

BRICHERHAFF<br />

MUGUETS<br />

PÂQUERETTES<br />

PARLEMENT<br />

EUROPÉEN<br />

PHILHARMONIE<br />

MUDAM<br />

THÉIWESBUR<br />

FOND. PESCATORE<br />

ARCHIDUCS<br />

RUE DE<br />

L'OUEST<br />

MICHEL<br />

ANGELO<br />

KIEM<br />

BECHEL<br />

RIEDGEN<br />

BLEUETS MARGUERITES<br />

VAUBAN<br />

LABOURS<br />

CLINIQUE<br />

SACRÉ-CŒUR<br />

AMSTERDAM<br />

BELAIR-<br />

KIERCH<br />

MERL-<br />

KIERFECHT<br />

JOSEPH<br />

HACKIN<br />

STE CUNÉGONDE<br />

7<br />

25<br />

BRICHER<br />

GÄSSEL<br />

Weimershof<br />

BADANSTALT<br />

REUTER<br />

UM<br />

BOCK<br />

VAL STE CROIX<br />

CLAUSENER BRÉCK<br />

ORADOUR<br />

GRÜNEWALD<br />

REHAZENTER<br />

CARRIÈRES<br />

Clausen<br />

ROYAL<br />

Belair<br />

PLATEAU<br />

ALTMÜNSTER<br />

FORUM<br />

ROYAL<br />

HAMILIUS<br />

PLACE DE LIÈGE<br />

Bertrange<br />

Findel<br />

NEIDUERF-KIERFECHT<br />

TOUR JACOB<br />

KASINOSGAASS<br />

MONTEREY<br />

WAMPACH<br />

FINDEL<br />

BUSINESS<br />

CENTER<br />

FIERSCHTER-<br />

HAUS<br />

NEIDUERF-<br />

KAPELL<br />

HUESE-<br />

GRËNDCHEN<br />

Neudorf<br />

FRANCISCAINES<br />

MERL-<br />

PARC<br />

PLACE DE<br />

FRANCE<br />

LAANG<br />

HECK<br />

GRUEF<br />

PLAAK<br />

BARTRÉNG-<br />

FLEURI<br />

BARTRÉNG-<br />

MONTEREALE<br />

BARTRÉNG-<br />

GEMENG<br />

CITÉ<br />

AÉROPORT<br />

KALCHES-<br />

BRÜCK<br />

LAANGE-<br />

GRONN<br />

RUMM<br />

CRÉCY<br />

16<br />

CHARLES DE<br />

TORNACO<br />

NEIDUERF-<br />

BRASSERIE<br />

JULES<br />

SALENTINY<br />

HIERZEKRËPP FIXMER NEIDUERF-<br />

KIERCH<br />

MALAKOFF<br />

Centre<br />

17<br />

RHEINSHEIM<br />

BRAGANCE<br />

GUILLAUME<br />

ORVAL<br />

ST HUBERT<br />

OP DER<br />

MILLEN<br />

BARTRÉNG-<br />

RICHTERWEE<br />

BARTRÉNG-<br />

KIERFECHT<br />

CATHÉDRALE<br />

7<br />

6<br />

AÉROPORT<br />

PAUL<br />

ALBRECHT<br />

FORT<br />

DUMOULIN<br />

KÉIBIERG-<br />

INS<br />

HELFENTERBRÜCK<br />

GABRIEL DE<br />

MAIRIE<br />

ARTHUR<br />

KNAFF<br />

TAWIOUN<br />

STADGRONN-<br />

BRÉCK<br />

HÄLLEPULL<br />

CENTS-<br />

WAASSERTUERM<br />

CENTS-<br />

KIERCH<br />

23<br />

MARTYRS<br />

STE ZITHE<br />

13<br />

F. D. ROOSEVELT<br />

NASSAU<br />

HELFENTERBRÜCK-PLETZER<br />

HELFENTERBRÜCK-<br />

CITY CONCORDE<br />

NIC<br />

ROLLINGER CENTS-HALTE CFL<br />

PARIS/ZITHA<br />

14<br />

TRÈVES CARMEL CAMILLE<br />

POLFER<br />

ROBERT<br />

BRUCH<br />

ÄPPELWEE-INS<br />

Grund<br />

MICHEL<br />

RODANGE<br />

Merl<br />

HELFENTERBRÜCK-<br />

AUTOMOBILE CLUB<br />

BARTRÉNG-<br />

DICKS<br />

POMMIERS<br />

GÉNÉRAL<br />

PATTON<br />

AL AVENUE<br />

Cents<br />

LABORATOIRE<br />

MARIE-<br />

ADÉLAÏDE<br />

ATHÉNÉE<br />

PULVERMÜHL-<br />

HAMMERDÄLLCHEN<br />

LAVOISIER<br />

Gare<br />

HEENTZE PARK<br />

WALLIS<br />

Hamm<br />

HUESE VIC<br />

GARE<br />

CENTRALE<br />

8<br />

15<br />

Pulvermühl<br />

VIRCHOW<br />

23 20<br />

9<br />

10 11<br />

COMMERCE<br />

Hol lerich<br />

CONSERVATOIRE<br />

NANCY<br />

RUE DE BITBOURG<br />

ETATS-UNIS<br />

RUE<br />

HAUTE<br />

HAMM-<br />

SCHOUL<br />

ENGLEBERT<br />

NEVEU<br />

KOFFER-<br />

FABRIK<br />

FRATERNITÉ<br />

POINCARE<br />

P+R<br />

ROTONDE<br />

JEAN-BAPT.<br />

MERKELS<br />

FONDERIE<br />

SALZHAFF<br />

P+R<br />

BARRIÈRE BOUILLON<br />

SCHLUECHTHAUS<br />

15<br />

KÄSCHTEWEE<br />

HAMM-<br />

KIERCH<br />

MONTMÉDY HAMM-<br />

COLONIE<br />

GABRIEL<br />

LIPPMANN<br />

CELTES<br />

MARCEL<br />

CAHEN<br />

JEAN<br />

JACOBY<br />

WISESTROOSS<br />

LÉON XIII<br />

DERNIER SOL<br />

ANATOLE<br />

FRANCE<br />

FORT<br />

WEDELL<br />

PIERRE &<br />

PAUL<br />

ASSURANCES SOCIALES<br />

FISCHERHAFF<br />

Bonnevoie<br />

DEMY<br />

SCHLECHTER<br />

ALSACE<br />

ARTISANS BARRÈS<br />

JEANNE<br />

D'ARC<br />

LASCOMBES<br />

HOLLERICH-GARE<br />

IZEGERKNUPP<br />

WILLIBRORD<br />

HIPPODROME<br />

DÉPORTATION<br />

12<br />

17<br />

1<br />

24<br />

5 6<br />

PONT REMY<br />

LYCÉE TECHNIQUE DE BONNEVOIE<br />

NEUFCHÂTEAU/LTB<br />

MILLEWEE<br />

ED. GRENIER<br />

AL<br />

GASPERICH<br />

KLENSCH<br />

GAASPERECHERBIERG<br />

CLEMENCEAU<br />

HOWALD-CIPA<br />

RANGWEE<br />

BEI DER<br />

AUER<br />

LOUIS DE FROMENT<br />

CESSANGE-KIERCH<br />

HOWALD-JHANGELI<br />

Cessange<br />

KALTREIS<br />

HOWALD-<br />

MOURESCHANZ<br />

NIC<br />

MARTHA<br />

JULES<br />

FISCHER<br />

RICHARD<br />

WAGNER<br />

RUE VERTE<br />

CESSANGE-DUERF<br />

BOY KONEN<br />

HOWALD-BEI DER KIERCH<br />

HOWALD-<br />

RONNEBËSCH<br />

Numéro de ligne<br />

10<br />

AM ECK<br />

4<br />

2<br />

HOWALD-MINSBËSCH<br />

HOWALD-<br />

OP DER STIRZEL<br />

Gasperich<br />

BEETHOVEN<br />

Terminus<br />

LUXEXPO<br />

KUELEBIERG<br />

TUBISHAFF<br />

ANTOINE<br />

HOWALD-WAASSERTUERM<br />

HESPER-<br />

CITÉ UM SCHLASS<br />

P+R LUX-SUD<br />

RUE DE<br />

LEUDELANGE<br />

Arrêt desservi en deux directions<br />

AM ECK<br />

3<br />

16<br />

22<br />

P+R<br />

14<br />

FRANÇOIS<br />

HOGENBERG<br />

STUMPER<br />

RAIFFEISEN<br />

PLANTIN<br />

Cloche d'Or<br />

Arrêt desservi uniquement dans<br />

le sens de la flèche<br />

MILLEWEE<br />

Howald<br />

CLOCHE D'OR<br />

24<br />

GUILLAUME KROLL<br />

Tronçon non desservi en permanence<br />

RUPPERT<br />

Kockelscheuer<br />

21<br />

P+R<br />

18<br />

Park <strong>and</strong> Ride<br />

P+R<br />

BIAN<br />

KOCKELSCHEUER-PATINOIRE<br />

SCHARFEN ECK<br />

KOCKELSCHEUER-<br />

CAMPING<br />

Gare CFL<br />

Aéroport<br />

Édition novembre 2009


Luxembourg,<br />

January 25th to 28th, 2012<br />

Luxembourg<br />

Natural compounds -<br />

Regulators of cell signaling<br />

pathways <strong>and</strong> novel therapeutic<br />

tools<br />

Contact : Marc Diederich<br />

Laboratoire de Biologie Moléculaire<br />

et Cellulaire du Cancer (LBMCC)<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

Fax: + 352 2468-4060<br />

Email: marc.diederich@lbmcc.lu<br />

Please register at:<br />

http://www.transduction-meeting.lu


A heart for cancer sick children<br />

Together we can make difference in the lives of children<br />

<strong>and</strong> their families living with childhood cancer


About geneXplain<br />

GeneXplain’s mission is to provide a comprehensive<br />

platform for bioinformatic, systems<br />

biological <strong>and</strong> cheminformatic tools. The raison<br />

d'être of this platform is to assist translational<br />

research in the life sciences, mainly in the context<br />

of personalized medicine <strong>and</strong> pharmacogenomics.<br />

We intend to make our expertise available to<br />

academic <strong>and</strong> commercial partners in collaborative<br />

research projects.<br />

Statistics<br />

Input: High-throughput<br />

data from patients<br />

(genomics,<br />

transcriptomics, ChIPseq,<br />

proteomics, etc.)<br />

Output: List of relevant<br />

genes or proteins<br />

The geneXplain platform<br />

Based on the BioUML technology created by<br />

the Institute of Systems Biology, a platform<br />

has been developed that allows to integrate a<br />

number of individual modules, “bricks”, each<br />

of which provides a well-defined function in<br />

the field of bioinformatics, systems biology or<br />

cheminformatics. Altogether, the whole<br />

system will provide a toolbox suitable to<br />

establish complete pipelines from<br />

experimental high-throughput (“-OMICS”)<br />

data to new drugs.<br />

The central platform is going to be made<br />

freely available, so that any community<br />

member can contribute own modules.<br />

In addition, geneXplain GmbH provides a<br />

number of state-of-the-art bricks on a<br />

commercial basis, implying a thorough<br />

quality control <strong>and</strong> maintenance guarantee.<br />

The workflow<br />

The incorporated statistical<br />

analyses help to identify relevant genes or<br />

proteins in the raw dataa,<br />

e.g. those that are differentially expressed.<br />

The Bioinformatics block<br />

allows to reveal potential regulation of genes by<br />

transcription factors or miRNAs.<br />

Directors: E. Wingender, A. Kel �Coommercial<br />

register:<br />

HRB 202564, Amtsger. Braunschweeig<br />

�VAT No.: DE271983408<br />

To achieve this, geneXplain offers:<br />

• The X-Platform to provide an integrated <strong>and</strong><br />

comprehensive workflow management of a large<br />

number of “bricks”, each providing a specific function<br />

in analyzing biological data, in particular OMICS<br />

results<br />

• In Silico Molecular Cloning (IMC) for h<strong>and</strong>ling largescale<br />

genome data<br />

• PASS <strong>and</strong>d Ph PharmaExpert E t ffor predicting di ti bi biological l i l<br />

activities of potential drugs<br />

Bioinformatics<br />

Search for regulatory<br />

Systems biology approaches analyze networks of<br />

molecular events, in particular gene regulatory <strong>and</strong><br />

signaling pathways, <strong>and</strong> suggest promising drug<br />

target molecules <strong>and</strong> their mechanisms of action.<br />

modules in any genomic Th The cheminformatics h i f i tools l enable bl to di direct<br />

regions<br />

Output: List of transcription<br />

factors potentially<br />

compound screening by pre-selection of chemicals<br />

with desirable <strong>and</strong> without adverse or toxic effects.<br />

responsible for the observed<br />

(co-)regulation of genes<br />

Systems Biology<br />

Toopological<br />

analysis of the<br />

networks upstream of<br />

transcription factors,<br />

simulation<br />

of the network<br />

behavior, patient<br />

stratification<br />

OOutput:<br />

List of potential<br />

master regulators<br />

Cheminformatics<br />

Prediction of biological<br />

activities of the<br />

compounds, d selection l ti of f<br />

compounds with required<br />

effects <strong>and</strong> without<br />

adverse or toxic effects.<br />

Output: List of potential<br />

lead structures for<br />

validation<br />

geneXplain GmbH<br />

Am Exer 10b<br />

D-38302 Wolfenbüttel, Germany<br />

info@genexplain.com<br />

www.genexplain.com


Wednesday January 26th<br />

Keynote session:<br />

9h30 - 10h30 Mario Capecchi<br />

10h30 - 11h00 Coffee break/expo<br />

Session 1: Cell death<br />

11h00 - 11h30 Eileen White<br />

11h30 - 12h00 Gerry Melino<br />

12h00 - 12h30 Stefania Gonfloni<br />

12h30 - 12h45 Cinzia Di Pietro<br />

12h45 – 13h00 Inna N. Lavrik<br />

13h00 - 14h00 Lunch/expo<br />

Workshops 1 <strong>and</strong> Poster Session 1<br />

14h00 - 15h00 Polyplus-transfection<br />

15h00 - 16h00 Agilent Technologies, Belgium<br />

14h00 - 15h30 Poster (Even numbers)/expo<br />

15h30 – 16h00 Coffee break/expo<br />

Session 2: Cell signaling<br />

16h00 - 16h30 Ilya Shmulevich<br />

16h30 - 17h00 Varadharajan Sundaramurthy<br />

17h00 - 17h30 Anne Grosse-Wilde<br />

17h30 - 18h00 Josef Penninger<br />

18h00 - 18h15 Sébastien Chateauvieux<br />

18h15 - 18h30 Ivana Scovassi<br />

18h30 - 18h45 Vladimir L. Katanaev<br />

19h00 - 19h30 Official talks<br />

19h30 - 20h30 Reception/expo<br />

20h30 - 21h00 Shuttles to the hotels<br />

------------------------------------------------<br />

Thursday January 27th<br />

Session 3: Transcriptional control<br />

9h00 - 9h30 Young-Joon Surh<br />

9h30 - 10h00 Luciano Di Croce<br />

10h00 - 10h30 Thomas Luft<br />

10h30 - 11h00 Coffee break/expo<br />

Session 4: Immunology<br />

11h00 - 11h30 Bali Pulendran<br />

11h30 - 12h00 Marie-Lise Gougeon<br />

12h00 - 12h30 Kasper Hoebe<br />

12h30 - 13h00 Claude Condé<br />

13h00 - 16h00 Lunch/expo<br />

Workshops 2 <strong>and</strong> Posters Session 2<br />

14h00 - 15h00 GE Healthcare Europe GmbH<br />

15h00 - 16h00 AMSBIO<br />

14h00 - 15h30 Posters (Odd numbers)/expo<br />

15h30 - 16h00 Coffee break/expo<br />

Session 5: Proteomics<br />

Cell Signal-omics 2011 on one page<br />

16h00 - 16h30 Antonio del Sol<br />

16h30 - 17h00 Bernd Wollscheid<br />

17h00 - 17h30 Pascale Cossart<br />

17h30 - 18h00 Christopher Overall<br />

18h00 - 18h15 Margarida Fardilha<br />

18h15 - 18h30 Alfred C. O. Vertegaal<br />

18h30 - 18h45 Angela Brieger<br />

19h00 Shuttles to the hotels<br />

------------------------------------------------<br />

Friday January 28th<br />

Session 6: Epigenetics<br />

9h00 - 9h30 François Fuks<br />

9h30 - 10h00 Michael Bots<br />

10h00 - 10h30 Roberto Gambari<br />

10h30 - 11h00 Coffee break/expo<br />

Session 7: Cancer signaling networks<br />

11h00 - 11h30 Andrei Zinovyev<br />

11h30 - 12h00 Dean W. Felsher<br />

12h00 - 12h30 Alex<strong>and</strong>er R. A. Anderson<br />

12h30 - 13h00 Evan Keller<br />

13h00 - 14h00 Lunch/expo<br />

Session 8: Gene expression networks<br />

14h00 - 14h30 Guido Kroemer<br />

14h30 - 15h00 Yong Sang Song<br />

15h00 – 15h15 Andrew Koff<br />

15h15 – 15h30 Stefanie Kaempf<br />

15h30 – 16h00 Coffee break/expo<br />

Session 9: Neurodegenerative diseases<br />

16h00 - 16h30 Mark P. Mattson<br />

16h30 - 17h00 Junying Yuan<br />

17h00 - 17h30 Vittorio Calabrese<br />

17h30 - 18h00 Xin-Fu Zhou<br />

18h00 - 18h30 Paul R. Fisher<br />

18h30 End of the meeting<br />

18h30 Shuttles to the hotels<br />

!<br />

!


Poster presentations<br />

Posters are classified by session<br />

<strong>and</strong> then in alphabetical order (PRESENTING AUTHOR)<br />

(Late breaking abstracts are at the end of the <strong>book</strong>)<br />

Session 1: Cell death<br />

66


Session 1: Cell death Poster 1<br />

The molecular chaperone HSP90 is cleaved by oxidative stress at a highly conserved Nterminal<br />

amino acid motif.<br />

Pedro Buc Calderon a , Raphael Beck a , Nicolas Dejeans a , Christophe Glorieux a , Mélanie<br />

Creton a , Edouard Delaive b , Marc Dieu b , Martine Raes b , Philippe Levêque c , Bernard<br />

Gallez c , Matthieu Depuydt d , Jean-François Collet d , <strong>and</strong> Julien Verrax a .<br />

a Toxicology <strong>and</strong> Cancer Biology Research Group, Louvain Drug Research Institute,<br />

Université Catholique de Louvain, Belgium; b Unité de Recherche en Biologie Cellulaire,<br />

FUNDP, Belgium; c Biomedical Magnetic Resonance Unit, Louvain Drug Research<br />

Institute, Université Catholique de Louvain, Belgium; d de Duve Institute, Université<br />

Catholique de Louvain, Belgium. Email: pedro.buccalderon@uclouvain.be<br />

Hsp90 is an essential chaperone which is necessary for the folding, stability <strong>and</strong> activity of<br />

numerous proteins. Here, it is shown that free radicals formed during oxidative stress<br />

conditions can cleave Hsp90. The cleavage is observed in intact cells, in cell lysates <strong>and</strong> in<br />

purified Hsp90 protein demonstrating that proteases are not required. In cells, Hsp90 cleavage<br />

leads to the degradation of its client proteins like Bcr-Abl, RIP, c-Raf, IKK! (NEMO) <strong>and</strong><br />

hTert. As this cleavage is dependent on ROS production <strong>and</strong> the availability of free redoxactive<br />

iron, we hypothesized that it occurs through a Fenton-type reaction. The formation of<br />

protein radicals upon the exposure of Hsp90 to oxidative stress was confirmed by immunospin<br />

trapping <strong>and</strong> EPR experiments. Using a proteomic analysis, we determined that the<br />

cleavage occurs in a conserved motif of the N-terminal nucleotide binding site, between Ile-<br />

126 <strong>and</strong> Gly-127 in Hsp90". Our results indicate that in situ formation of ROS by a Fentontype<br />

reaction at the N-terminal nucleotide binding pocket of Hsp90 forms a protein radical,<br />

which, by rearrangement, causes the rupture of the peptide backbone <strong>and</strong> leads to the<br />

degradation of client proteins that are critical for cells.<br />

67


Session 1: Cell death Poster 2<br />

Tumor GRP94 overexpression: a hallmark of aggressiveness <strong>and</strong> recurrence in breast<br />

cancer.<br />

Nicolas Dejeans a , Samuel Guenin b , Christophe Glorieux a , Raphael Beck a , Julien<br />

Verrax a , Brice Sid a , Bettina Bisig b , Philippe Delvenne b , Pedro Buc Calderon a .<br />

a Université Catholique de Louvain, Louvain Drug Research Institute, Toxicology <strong>and</strong><br />

Cancer Biology Research Group, PMNT Unit, Belgium. b Department of Pathology,<br />

GIGA Cancer, University of Liege, B23 CHU Sart Tilman, 4000 Liège. Email:<br />

nicolas.dejeans@uclouvain.be<br />

Targeting the altered redox status of cancer cells is emerging as an interesting approach to<br />

potentiate chemotherapy. However, to favor the effectiveness of this strategy <strong>and</strong> define the<br />

proper chemotherapeutic association, it is important to underst<strong>and</strong> the biological<br />

consequences of chronically exposing cancer cells to ROS. Using an H2O2 generating system,<br />

we have selected a ROS-resistant MCF-7 cell line, namely the Resox cells. By exploring<br />

different survival pathways that usually are induced during an oxidative stress, we identified<br />

in these cells a constitutive overexpression of the endoplasmic reticulum chaperone GRP94,<br />

while levels of either its cytoplasmic homologue HSP90 or GRP78 are not modified. Such an<br />

overexpression is not mediated by constitutive unfolded protein response (UPR) activation.<br />

The increase of GRP94 is tightly associated to an increase in proliferation <strong>and</strong> migration<br />

capacities, as shown by GRP94 silencing experiments. By doing immunohistochemistry on<br />

breast tumor biopsies, we showed that GRP94 expression was elevated in recurrent human<br />

breast cancers compared to their paired primary tissues. Likewise in the Resox cells, this<br />

increase was not associated with a higher phosphorylation of eIf2, demonstrating that it was<br />

not linked to an increase in UPR activation in recurrent tumors. In conclusion, this study<br />

suggests that GRP94 overexpression may be a hallmark of aggressiveness <strong>and</strong> recurrence in<br />

breast cancers.<br />

68


Session 1: Cell death Poster 3<br />

In vitro characterization of bovine neutrophil cell death following Escherichia coli<br />

phagocytosis<br />

Sofie Notebaert 1* , Kristel Demeyere 1 *, Dieter Demon 1 , Filip Boyen 2 , Chris Guerin 3 ,<br />

Peter V<strong>and</strong>enabeele 4 , Evelyne Meyer 1<br />

1 Laboratory of Biochemistry, Faculty of Veterinary Medicine, Ghent University, Salisburylaan<br />

133, 9820 Merelbeke, Belgium – sofie.notebaert@hotmail.com; kristel.demeyere@ugent.be;<br />

dieter.demon@ugent.be; evelyne.meyer@ugent.be<br />

2 Department of Pathology, Bacteriology <strong>and</strong> Avian Diseases, Faculty of Veterinary Medicine,<br />

Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium – filip.boyen@ugent.be<br />

3 Microscopy Core Facility, Department for Molecular Biomedical Research, VIB, Ghent<br />

University, 9052 Zwijnaarde, Belgium - chris.guerin@dmbr.vib-ugent.be<br />

4 Molecular Signaling <strong>and</strong> Cell Death Unit, Department for Molecular Biomedical Research,<br />

VIB, Ghent University, 9052 Zwijnaarde, Belgium - peter.v<strong>and</strong>enabeele@dmbr.vib-ugent.be<br />

* These authors contributed equally to this work.<br />

Neutrophils are known to play a key role in the early host defense towards coliform mastitis. The<br />

molecular events during phagocytosis-induced cell death (PICD) are rarely described, especially for<br />

bovine neutrophils. Therefore, our objective was to elucidate the type of cell death of bovine<br />

neutrophils after in vitro phagocytosis of Escherichia coli.<br />

Peripheral blood samples were collected from clinically healthy heifers. Neutrophils were isolated by<br />

density gradient centrifugation, resuspended in RPMI with 10 % fcs <strong>and</strong> incubated with E. coli strain<br />

P4:O32 (moi 5:1) at 37°C. PS exposure <strong>and</strong> cell membrane integrity loss was determined flow<br />

cytometrically by Annexin-V-FITC/PI. ROS were measured as luminol-amplified chemiluminescence.<br />

Equal amounts of total cell lysates were analyzed by western blotting using anti caspase-1 (C-1),<br />

cleaved C-3 <strong>and</strong> C-7 <strong>and</strong> X-IAP. C-3/-7 activity was determined using the caspase-Glo ® assay <strong>and</strong><br />

bovine IL-1! was quantified with ELISA.<br />

The higher rate of PS exposure in activated than in control neutrophils undergoing spontaneous<br />

apoptosis, indicates that bovine neutrophil cell death is accelerated following phagocytosis of E. coli,<br />

as recently described. In contrast to control cells, PICD occurred with significant ROS production but<br />

independent from C-3/-7 activation. The latter finding is in line with a recent report describing<br />

caspase-independent cell death of human neutrophils incubated with E. coli, although no ROS<br />

production was seen. In both human <strong>and</strong> mouse neutrophils, NADPH oxidase prevents caspase<br />

activation following phagocytosis of S. aureus. Nevertheless, all these data do not corroborate a study<br />

with human neutrophils incubated with E. coli where C-3 activation was demonstrated. This<br />

discrepancy could be either species <strong>and</strong>/or micro-organism related. Interestingly, the well-described<br />

X-IAP inhibitor for C-3 <strong>and</strong> C-7, was only present in control samples, suggesting that X-IAP may<br />

control C-3/-7 activity in spontaneous bovine neutrophil apoptosis but is superfluous in E. coli PICD<br />

in vitro. Life cell imaging using GFP-labeled E. coli <strong>and</strong> PI staining showed no clear morphological<br />

features of either netosis or autophagy, but late PI positivity <strong>and</strong> membrane blebbing without apoptotic<br />

bodies. Complementing these features with C-1 activation <strong>and</strong> intracellular IL-1! suggests that the<br />

PICD in our setup is executed by pyroptosis.<br />

Taken together, our in vitro data indicate that bovine neutrophils undergo C-3/-7-assisted spontaneous<br />

apoptosis <strong>and</strong> that E. coli stimulated PICD most likely can be characterized as C-3/-7-independent, but<br />

C-1- <strong>and</strong> ROS-dependent pyroptosis.<br />

69


Session 1: Cell death Poster 4<br />

The in vivo role of executioner Caspase-3 <strong>and</strong> -7 in the Escherichia coli-infected<br />

mammary gl<strong>and</strong><br />

Koen BREYNE 1,! , Dieter DEMON 1,! , Tom VANDEN BERGHE 2 , Peter<br />

VANDENABEELE 2 , <strong>and</strong> Evelyne MEYER 1<br />

1 Laboratory of Biochemistry, Department of Pharmacology, Toxicology <strong>and</strong><br />

Biochemistry, Ghent University, 9820 Merelbeke, Belgium, Koen.Breyne@Ugent.be,<br />

Dieter.Demon@Ugent.be, Evelyne.Meyer@Ugent.be<br />

2 Molecular Signaling <strong>and</strong> Cell Death Unit, Department for Molecular Biomedical<br />

Research, Technologiepark 927, 9052 Ghent, Belgium, Tom.V<strong>and</strong>enberghe@dmbr.vib-<br />

Ugent.be, Peter.V<strong>and</strong>enabeele@dmbr.vib-Ugent.be<br />

! These authors contributed equally to this work<br />

Bacterial infection of the mammary gl<strong>and</strong> can disturb lactation <strong>and</strong> involution through a process called<br />

mastitis. The pathogenesis of mastitis in humans <strong>and</strong> animals is insufficiently understood, resulting in<br />

excessive use of antibiotics. To reduce bacterial resistance due to the use of antibiotics, alternative<br />

therapeutic strategies are desirable. Mastitis is characterized by apoptosis <strong>and</strong> inflammation, two<br />

processes in which caspases are involved. Executioner Caspase-3 (C-3) <strong>and</strong> C-7 play central roles in<br />

coordinating the stereotypical events during apoptosis. Moreover, C-7 is also involved in<br />

inflammatory signaling. Underst<strong>and</strong>ing the pathways involving C-3 <strong>and</strong> -7 may identify targets<br />

towards a faster <strong>and</strong> more accurate diagnosis or treatment of mastitis. Therefore, we evaluated C-3 -/-<br />

<strong>and</strong> -7 -/- single knockout mice in the acute Escherichia coli (E. coli)-induced mastitis model.<br />

Lactating wild type (wt; n=10), C-3 -/- (n=5) <strong>and</strong> C-7 -/- (n=5) mice were intraductally inoculated with<br />

the bovine mastitis isolate E. coli P4:O32 in the right mammary gl<strong>and</strong> of the 4 th pair <strong>and</strong> with<br />

phosphate buffered saline (PBS) in the contralateral gl<strong>and</strong>. Serum was collected at 18h postinoculation<br />

<strong>and</strong> the inoculated gl<strong>and</strong>s were isolated, homogenized <strong>and</strong> plated to quantify colony<br />

forming units (CFU). Homogenates were lysed in 1% NP40 buffer for western blot <strong>and</strong> cytokine<br />

analysis. Two gl<strong>and</strong> pairs/group were paraffin-embedded for histopathological evaluation.<br />

Our data show significantly more proteolysis of C-7 in E. coli-infected than in control mammary<br />

gl<strong>and</strong>s. Interestingly, next to the classically detected p20 fragment of active C-7 we observed in both<br />

gl<strong>and</strong>s a 23 kDa fragment with unknown function thus far. E .coli infection also results in the loss of a<br />

C-3 cleavage fragment, a phenomenon which is typically seen with inhibition by X-IAP, the welldescribed<br />

inhibitor of C-3 <strong>and</strong> -7. Interestingly, more intense fragments of X-IAP are seen in E. coliinoculated<br />

gl<strong>and</strong>s, suggesting that it may indeed regulate C-3 <strong>and</strong> -7 activity in mastitis. However, the<br />

absence of C-3, which is typically linked with apoptosis, does not affect bacterial growth nor local <strong>and</strong><br />

systemic cytokine release during E. coli-induced intramammary infection, indicating no critical role<br />

for this caspase in mastitis. Ablation of C-7 seems to sensitize mice slightly to intramammary E. coli<br />

infection, as highlighted by the higher final CFU/g gl<strong>and</strong>, <strong>and</strong> local <strong>and</strong> systemic levels of the proinflammatory<br />

cytokines IL-1! <strong>and</strong> IL-6, respectively. Prototype inflammatory C-1 is also activated in<br />

E. coli-infected gl<strong>and</strong>s <strong>and</strong> could thus be the upstream activator of C-7. However, the PARP cleavage<br />

pattern suggests the functional contribution of calpain, cathepsin-B <strong>and</strong> granzyme-B, which have all<br />

been shown to activate C-7 in vitro. Analysis of the separate cell populations (macrophage, neutrophil<br />

or epithelial cell), instead of total homogenates will help to further elucidate the specific role of C-7 in<br />

mastitis.<br />

70


Session 1: Cell death Poster 5<br />

Functional crosstalk between p53 <strong>and</strong> HtrA2/Omi<br />

Eric Duplan, Frédéric Checler <strong>and</strong> Cristine Alves da Costa<br />

Institut de Pharmacologie Moléculaire et Cellulaire <strong>and</strong> Institut de NeuroMédecine<br />

Moléculaire, UMR 6097 CNRS/UNSA, 660 route des Lucioles, 06560 Valbonne, France.<br />

duplan@ipmc.cnrs.fr<br />

p53 is the product of a well known tumor-suppressor gene, mutations in which lead to its<br />

inactivation <strong>and</strong> increased risk to cancer. It is well established that p53 is a key regulator of<br />

the intrinsic apoptotic pathway via its transcriptional factor dependent <strong>and</strong> independent<br />

properties. It has been recently shown that p53 induces the transcription of HtrA2/Omi, a<br />

serine protease that is known to cleave (among other substrates) <strong>and</strong> inactivate the Inhibitor of<br />

Apoptosis Proteins, XIAP. Thus, p53 can regulate cell death processes via transcriptional<br />

activation of the protease Omi. Omi activity is also compromised in several cancer types <strong>and</strong><br />

recently it has been shown that following apoptotic stimuli, Omi accumulates in the nucleus<br />

where it cleaves the p53 family member p73alpha. The processed p73 fragment can then<br />

activate the apoptotic gene bax. These results suggest that the serine protease properties of<br />

Omi accounts for its pro-apoptotic function. Since p73 <strong>and</strong> p53 contain homologous<br />

transactivation, DNA-binding, <strong>and</strong> oligomerization domains, we thus decided to examine if<br />

Omi was capable to regulate the levels of p53 <strong>and</strong>, as a corollary, its pro-apoptotic function.<br />

We found that Omi controls the transcriptional activity, protein <strong>and</strong> mRNA levels of p53. The<br />

precise mechanism of this regulation will be discussed.<br />

71


Session 1: Cell death Poster 6<br />

Alpha-Synuclein cytotoxicity in Dictyostelium.<br />

Sanjanie G. Fern<strong>and</strong>o 1 , William A. Burrage 2 , Peter L. Beech 2 , Sarah J. Annesley 1 , Paul<br />

R. Fisher 1<br />

1 Department of Microbiology, Latrobe University, VIC 3086, Australia. 2 Centre for<br />

Cellular <strong>and</strong> Molecular Biology, Deakin University, Burwood, VIC 3125, Australia.<br />

Alpha-synuclein is implicated in the pathogenesis of Parkinson’s disease (PD) which is a<br />

neurodegenerative disorder. Hyperexpression or mutations in the alpha-synuclein gene are<br />

believed to be associated with mitochondrial abnormalities <strong>and</strong> onset of familial PD. To<br />

establish a Dictyostelium model for PD <strong>and</strong> to investigate the role of mitochondrial<br />

dysfunction in its cytopathology, we created stable, clonal Dictyostelium transformants<br />

hyperexpressing these normal <strong>and</strong> mutant forms of alpha-synuclein. All three forms were<br />

cytotoxic <strong>and</strong> impaired both phagocytosis <strong>and</strong> growth on bacterial lawns. We have also used<br />

immunofluorescence <strong>and</strong> confocal microscopy to show that alpha-synuclein is concentrated in<br />

the membrane particularly at the leading edge of the cell (resembling the presynaptic<br />

membrane in neurons) which is the same subcellular location as in human cells. However, it<br />

did not colocalize or associate with mitochondria. Further phenotypic characterisation showed<br />

that the aberrant phenotypes caused by alpha-synuclein are distinct from those observed in<br />

mitochondrial disorders in Dictyostelium. This indicates that there are distinct alphasynuclein<br />

cytotoxicity pathways that differ from those associated with mitochondrial<br />

dysfunction.<br />

72


Session 1: Cell death Poster 7<br />

Model-based dissection of CD95 signaling dynamics reveals both a pro- <strong>and</strong><br />

antiapoptotic role of c-FLIPL.<br />

Fricker N, Beaudouin J, Richter P, Eils R, Krammer PH, Lavrik IN.<br />

C-FLIP proteins (isoforms: c-FLIPL, c-FLIPS <strong>and</strong> c-FLIPR) regulate caspase-8 activation <strong>and</strong><br />

death receptor-induced apoptosis. The function of the two short c-FLIP isoforms, c-FLIPS <strong>and</strong><br />

c-FLIPR, has been well described. However there are contradictory reports with respect to the<br />

pro- or anti-apoptotic role of the long isoform, c-FLIPL. Here, using a combination of<br />

mathematical modeling, imaging <strong>and</strong> quantitative western blots we determine conditions<br />

leading to pro- or anti-apoptotic function of c-FLIPL. We present a mathematical model<br />

describing the regulation of caspase-8 activation by c-FLIP at the DISC (death inducing<br />

signalling complex). We show that c-FLIPL has a pro-apoptotic role only upon moderate<br />

expression in combination with strong receptor stimulation or in the presence of high amounts<br />

of one of the short c-FLIP isoforms: c-FLIPS or c-FLIPR. Our findings resolve the present<br />

controversial discussion on the function of c-FLIPL as a pro- or anti-apoptotic protein in death<br />

receptor-mediated apoptosis.<br />

73


Session 1: Cell death Poster 8<br />

Reverse effects of AMP-activated protein kinase on pro-apoptotic extracellular signalregulated<br />

kinase activation by inducing dual-specificity protein phosphatases under<br />

glucose deprivation in human colon carcinoma.<br />

Sooho Lee <strong>and</strong> Joohun Ha<br />

Department of Biochemistry <strong>and</strong> Molecular Biology, Medical Research Center <strong>and</strong><br />

Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul 130-701,<br />

Republic of Korea. E-mail: sooholee0827@gmail.com<br />

Mitogen-activated protein kinase (MAPK) pathways are involved in the regulation of cellular<br />

responses, including cell proliferation, differentiation, cell growth, <strong>and</strong> apoptosis. Because<br />

these responses are tightly related to cellular energy level, AMP-activated protein kinase<br />

(AMPK), which plays an essential role in energy homeostasis, has emerged as another key<br />

regulator. In the present study, we demonstrate a novel signal network between AMPK <strong>and</strong><br />

MAPK in HCT116 human colon carcinoma. Glucose deprivation activated AMPK <strong>and</strong> three<br />

MAPK subfamilies, extracellular signal-regulated kinase (ERK), c-Jun NH(2)-terminal kinase<br />

(JNK), <strong>and</strong> p38 MAPK. Under these conditions, inhibition of endogenous AMPK by<br />

expressing a dominant-negative form significantly potentiated ERK activation, indicating that<br />

glucose deprivation-induced AMPK is specifically antagonizing ERK activity in HCT116<br />

cells. Moreover, we provide novel evidence that AMPK activity is critical for p53-dependent<br />

expression of dual-specificity phosphatase (DUSP) 1 <strong>and</strong> 2, which are negative regulators of<br />

ERK. Notably, ERK exhibits pro-apoptotic effects in HCT116 cells under glucose<br />

deprivation. Collectively, our data suggest that AMPK protects HCT116 cancer cells from<br />

glucose deprivation, in part, via inducing DUSPs, which suppresses pro-apoptotic ERK,<br />

further implying that a signal network between AMPK <strong>and</strong> ERK is a critical regulatory point<br />

in coupling the energy status of the cell to the regulation of cell survival.<br />

74


Session 1: Cell death Poster 9<br />

Protective Effects of Marine Natural Products against UVB-induced Oxidative Damage<br />

<strong>and</strong> Cell Death in Human Skin Fibroblast<br />

Chan Lee, En-Joo Lee, <strong>and</strong> Chan-Ik Park<br />

Department of Cosmeceutical Science, Daegu Haany University, Gyeongsangbuk-do<br />

712-715, S. Korea (E-mail : cipark@dhu.ac.kr)<br />

Ultraviolet is the one of the main environmental factors promoting aging process via<br />

increased intracellular accumulation of reactive oxygen species (ROS) <strong>and</strong> decreased<br />

expression of endogenous antioxidant molecules. Therefore, in this study, we have tried to<br />

search for skin-protective antioxidant marine natural products (Porphyra thalli, Laminariae<br />

japonicae thallus, Ostreae concha, Sargassum thallus, Undaria thallus, Haliotidis concha,<br />

Codium thalli, Syngnathoides biaculeatus, Hippocampus, Stichopus stichopus, Thalli, Hizikia<br />

fusiforme thalli) which can protect against UVB-induced oxidative cell death in HS86 cells.<br />

Free radical scavenging activity was shown in the order of Undaria thallus, Sargassum thallus,<br />

Laminariae japonicae thallus, Hippocampus, Haliotidis concha, Ostreae concha, <strong>and</strong><br />

Syngnathoides biaculeatus as assessed by DPPH assay. In another experiment, UVB-induced<br />

cell death in HS68 cells were effectively suppressed by treatment with Sargassum thallus,<br />

Haliotidis concha, Codium thalli, or Hippocampus water extract by inhibition of intracellular<br />

accumulation of ROS. The protective effects of marine natural products against UVB-induced<br />

oxidative cell death seemed to be mediated by increased expression of antioxidant enzymes<br />

such as catalase, superoxide dismutase, <strong>and</strong> heme oxygenase-1. These results suggest that<br />

Sargassum thallus, Haliotidis concha, Codium thalli, <strong>and</strong> Hippocampus may have preventive<br />

<strong>and</strong>/or protective potentials against oxidative stress-mediated skin damages <strong>and</strong> aging with<br />

antioxidant properties.<br />

Keywords: Ultraviolet, marine natural products, antioxidant, oxidative stress, skin fibroblast<br />

75


Session 1: Cell death Poster 10<br />

Different response to UV-B irradiation of Niemann-Pick B-Lymphocytes cell line<br />

Barbara Canonico 1 , Erica Cesarini 1 , Marcella Arcangeletti 1 , Fulvio Palma 1 , Loris<br />

Zamai 1 , Stefano Papa 1 <strong>and</strong> Francesca Luchetti 1<br />

1<br />

Department of Earth, Life <strong>and</strong> Environmental Sciences. University of Urbino “ Carlo<br />

Bo”<br />

Via Cà le Suore 2 61029 Urbino. barbara.canonico@uniurb.it; erica.cesarini@uniurb.it;<br />

marcella.arcangeletti@uniurb.it; fulvio.palma@uniurb.it; loris.zamai@uniurb.it;<br />

stefano.papa@uniurb.it; francesca.luchetti@uniurb.it<br />

Autophagy is an intracellular lysosomal degradation process, characterized by the formation<br />

of double membrane-bounded vesicle (autophagosome). Recently, the autophagic pathway<br />

was coupled with lysosomal storage disorders (LSD), on the hypothesis that accumulation of<br />

undegraded substrates in lysosomes may impair the autophagic process. Niemann-Pick<br />

disease (NPD) type A <strong>and</strong> B are lysosomal storage disorders (LSD) resulting from the<br />

sphingomyelin accumulation in lysosomes relying on reduced or absent acid<br />

sphingomyelinase (ASM). The purpose of this study is to examine the autophagic pathway in<br />

a NPD B-Lymphocytes cell line, in response to UV-B irradiation, by means of flow cytometry<br />

<strong>and</strong> microscopic analysis. We evaluated cell viability, caspase activation, various<br />

mitochondrial parameter, acidic compartment (by Lysotracker-LT <strong>and</strong> Acridine Orange–AO<br />

fluorescence) <strong>and</strong> we performed preliminary test on endosomal network (by CD63 staining).<br />

Microscopic analysis was applied to evaluate morphological aspects. Cells were UVirradiated<br />

for 5 <strong>and</strong> 10 minutes <strong>and</strong> treated with nocodazole (NZ) <strong>and</strong> wortmannin (WM), two<br />

autophagy inhibitors. Furthermore we also used rapamicyn (RM), an autophagic inductor. Our<br />

preliminary data show a different trend between short (5’) <strong>and</strong> prolonged (10’) stress: a<br />

predominance of apoptotic <strong>and</strong> autophagic trend, respectively. A mitochondrial involvement<br />

is evident in both times of irradiation of cells, with a progressive decrease in mitochondrial<br />

integrity (NAO) <strong>and</strong> function (TMRE). Lymphocytes treated by NZ <strong>and</strong> WM highlighted<br />

immunofluorescence values (IF) for AO higher than control lymphocytes (+19% <strong>and</strong> +40%,<br />

respectively). We found a decrease in CD63+ cells, strong for RM (55%), WM+UV10’ (54%)<br />

<strong>and</strong> NZ+UV5’ (85%), weaker for WM+UV5’ (12%). Similarly UV irradiated cells highlight<br />

decrease in CD63+ events. On the contrary IF for CD63 exhibited an opposite trend: we<br />

found the highest values in RM samples <strong>and</strong> UV10’ treated samples with or without pretreatment<br />

with both inhibitors. Our preliminary results suggest a possible autophagy<br />

involvement in NPB cells oxidative stress-response.<br />

76


Session 1: Cell death Poster 11<br />

MAPK/ERK targeting by Sorafenib down-regulates Doxorubicin-induced death<br />

signaling in hepatocellular carcinoma Hep3B cells: the role of autophagy.<br />

Yulia Pollak, Rinata Broneshter, Theodore C. Iancu <strong>and</strong> Irena Manov<br />

Pediatric Research <strong>and</strong> Electron Microscopy Unit, Faculty of Medicine, Technion-Israel<br />

Institute of Technology, Haifa, Israel, e-mail: irmanov@tx.technion.ac.il<br />

A multikinase inhibitor of Raf/MEK/ERK pathway, sorafenib is increasingly used in the<br />

management of hepatocellular carcinoma (HCC) <strong>and</strong> its combination with doxorubicin<br />

(DOX) stimulates particular interest. Although the MEK/ERK pathway was identified as<br />

survival signaling in tumors, activation of these kinases mediates DOX-triggered death<br />

signaling. In the present study, we investigated the effect of sorafenib on DOX efficacy in<br />

HCC cells <strong>and</strong> possible mechanisms of drug interaction. Treatment of Hep3B cells with DOX<br />

arrested proliferation <strong>and</strong> reduced cell viability. Electron microscopy revealed intensive<br />

vacuolization of cytoplasm <strong>and</strong> formation of autophagosomes, while apoptotic changes were<br />

not conspicuous. Immunofluorescence demonstrated induction of LC3B in DOX-exposed<br />

Hep3B cells. Sorafenib alone affected viability <strong>and</strong> cell cycle progression <strong>and</strong> induced a<br />

rapid, massive mitochondrial degradation. In contrast, sorafenib combined with DOX<br />

increased survival, facilitated cell cycle progression <strong>and</strong> reduced autophagy. To elucidate the<br />

mechanisms by which sorafenib reduced DOX efficacy, we examined the implication of<br />

ERK1/2 <strong>and</strong> AKT, as well as molecular sensors of autophagy (MCL-1) <strong>and</strong> cell cycle<br />

progression (cyclin D1). The increased phospho-ERK in DOX-exposed cells was reduced by<br />

sorafenib. Similarly to sorafenib, MEK/ERK targeting by U0126 or PD98059 deregulated<br />

DOX-induced death signaling <strong>and</strong> increased survival. DOX had no effect on phospho-Akt,<br />

whereas combined treatment with sorafenib resulted in AKT activation. Sorafenib-induced<br />

degradations of Cyclin D1 <strong>and</strong> MCL1 were amplified by co-treatments with DOX. Thus,<br />

MEK/ERK counter-effective regulation <strong>and</strong> stimulation of cell survival via AKT, may<br />

produce the paradoxical effect of DOX plus sorafenib co-treatment. The escape from<br />

autophagy through MCL1 <strong>and</strong> cyclin D1 down regulation is discussed. The use of<br />

MEK/ERK inhibitors in combination with chemotherapeutics, meant to enhance anti-cancer<br />

efficacy, requires considering possible antagonistic effects.<br />

77


Session 1: Cell death Poster 12<br />

MOUSE MODELS OF ODONTOGENESIS – FOCUSED ON CELL DEATH<br />

Eva Matalová (1,2), Tom V<strong>and</strong>en Berghe (3), Ivana Chlastáková (1,2), Eva !v<strong>and</strong>ová<br />

(1), Ivan Mí"ek (1), Jaroslav Doubek (2), Abigail S. Tucker (4)<br />

(1) Department of Physiology, University of Veterinary <strong>and</strong> Pharmaceutical Sciences,<br />

Palackého 1-3, 612 42, Brno, Czech Republic, (2) Institute of Physiology <strong>and</strong> Genetics<br />

CAS, v.v.i, Veve#í 97, 602 00 Brno, Czech Republic, (3) Department for Molecular<br />

Biomedical Research, Ghent University, Technologiepark 927B, 9052 Ghent, (4)<br />

Department of Craniofacial Development, King´s College London, Guy´s Hospital, SE1<br />

9RT London, UK<br />

matalova@iach.cz, Tom.V<strong>and</strong>enberghe@dmbr.VIB-UGent.be,<br />

ivanachlastakova@seznam.cz, 184576@mail.muni.cz, misek@iach.cz, doubekj@vfu.cz,<br />

abigail.tucker@kcl.ac.uk<br />

Abnormal number or shape of teeth is the most common congenital malformation in humans.<br />

Dentition defects may occur as isolated disorder, or as components of many hereditary syndromes.<br />

Molecular networks underlying tooth formation are mostly studied in the mouse, the classical<br />

experimental model is the mouse first molar (M1). Knowledge obtained in the mouse has been<br />

successfully extrapolated to human odontogenesis, <strong>and</strong> related disorders connected with altered tooth<br />

number, size, structure <strong>and</strong> shape found in human dentistry (1). Over 20 % of human population miss<br />

one or more third molars; 5 % lack other permanent tooth.<br />

Here, we studied morphogenesis of the mouse third molar (M3) with a special interest in signalling<br />

centres, the enamel knots. These specific populations are known to release mitotic signals related to<br />

tooth shaping, particularly transition from the bud to the cap stage (primary enamel knot, PEK). The<br />

PEK cells do not proliferate <strong>and</strong> undergo apoptosis after fulfilling their missing. PEK in M3 was found<br />

as a small cluster of concentrically arranged cells in the posterior part of the enamel organ at the<br />

postnatal day 3. The presence of the PEK was confirmed by in situ hybridisation for Fgf-4 <strong>and</strong> Shh.<br />

Apoptotic bodies were evident, accompanying the gradual elimination of the PEK. PEK cells were<br />

PCNA (proliferating cell nuclear antigen) negative.<br />

PEK apoptosis was shown to be caspase dependent. In our previous studies (2,3), caspase-9 (along<br />

with Apaf-1) <strong>and</strong> caspase-3 were demonstrated to be essential for PEK apoptosis. In this work, another<br />

member of so called death trio of execution caspases (3-6-7), caspase-7, was investigated.<br />

Immunohistochemistry (IHC) of active caspase-7 was used in serial histological sections of mouse<br />

heads of the caspase-7 knock-out <strong>and</strong> wild type mice. Caspase-7 activation was evaluated along with<br />

morphology (apoptotic bodies), TUNEL test (apoptotic DNA breaks), caspase-3 activation (IHC) <strong>and</strong><br />

cell proliferation (IHC of PCNA). Even when caspase-3 <strong>and</strong> caspase-7 are often considered as<br />

functionally redundant, this does not seem to apply for PEK apoptosis. As caspase-3 deficiency<br />

inhibited PEK apoptosis, lack of caspase-7 did not cause any alterations. However, interestingly,<br />

active caspase-7 was found in osteoclasts in areas forming the bony socket of developing molar tooth,<br />

moreover, also in osteoclasts not undergoing apoptosis (TUNEL negative). These findings suggest<br />

possible involvement of caspase-7 in osteoclast differentiation as reported for caspase-3 (4).<br />

(1) Fleischmannova et al. (2008) Eur J Oral Sci 116, 1-10.<br />

(2) Setkova et al. (2007) Arch Oral Biol 52: 15-19<br />

(3) Matalová et al. (2006) Int J Dev Biol 50: 491-497<br />

(4) Szymczyk et al. (2006) J Cell Physiol 209: 836-844<br />

The research was funded by the GAAV project IAA600450904.<br />

78


Session 1: Cell death Poster 13<br />

Nitric oxide, P-glycoprotein <strong>and</strong> calreticulin: three mediators of<br />

chemoimmunoresistance in human tumors.<br />

Chiara Riganti, Joanna Kopecka, Dario Ghigo, Amalia Bosia<br />

Dep. of Genetics, Biology <strong>and</strong> Biochemistry, University of Turin, via Santena 5/bis,<br />

10126, Turin, Italy; emails: chiara.riganti@unito.it; joanna.kopecka@unito.it;<br />

dario.ghigo@unito.it; amalia.bosia@unito.it<br />

A great obstacle in the successful treatment of tumors is multidrug resistance (MDR), due to<br />

the overexpression of membrane pumps, like P-glycoprotein (Pgp) <strong>and</strong> MDR-related proteins<br />

(MRPs), which mediate the efflux of several anticancer drugs. Doxorubicin is a versatile<br />

chemotherapeutic agent, because exerts both direct cytotoxic <strong>and</strong> indirect pro-immunogenic<br />

cell death; unfortunately it is a substrate of Pgp <strong>and</strong> MRPs.<br />

We show that in chemosensitive cancer cells doxorubicin up-regulates the inducible nitric<br />

oxide (NO) synthase (iNOS) gene <strong>and</strong> the synthesis of NO. The latter mediates part of the<br />

doxorubicin cytotoxicity; induces chemosensitization (by reducing the efflux activity of Pgp<br />

<strong>and</strong> MRP3, through a direct nitration of critical tyrosines on the transporters); plays a role in<br />

the doxorubicin pro-immunogenic effects, promoting - via the cGMP/protein kinase G<br />

pathway <strong>and</strong> the actin cytoskeleton remodelling - the surface translocation of calreticulin<br />

(CRT) <strong>and</strong> triggering the tumor cells phagocytosis by dendritic cells (DCs). The lack of NO<br />

synthesis is responsible for a chemo-immunoresistant phenotype: in drug-resistant cells, as<br />

well as in drug-sensitive cells stably silenced for iNOS, doxorubicin is extruded <strong>and</strong> does not<br />

induce direct tumor cell death nor translocation of CRT. These events however are restored<br />

by using NO donors, iNOS inducers other than doxorubicin or doxorubicins synthetically<br />

conjugated with NO-releasing groups. Unexpectedly, despite high levels of surface CRT, the<br />

DCs-mediated phagocytosis does not occur in chemoresistant cells, where CRT is bound by<br />

Pgp in endoplasmic reticulum <strong>and</strong> translocates with it on plasma-membrane. Since the<br />

phagocytosis is restored only after silencing Pgp, we hypothesize that CRT-Pgp interaction<br />

impairs the functional activity of CRT <strong>and</strong> that Pgp may represent not only a marker of<br />

chemoresistance, but also an immunosuppressive signal.<br />

Our findings show that chemo- <strong>and</strong> immunoresistance are associated in human tumors <strong>and</strong><br />

could rely on low levels of NO, low levels of CRT <strong>and</strong> high levels of Pgp. The modulation of<br />

their amounts may lead to new strategies in chemo-immunotherapy protocols.<br />

79


Session 1: Cell death Poster 14<br />

Pathways of cell death induced by nanoparticles<br />

Fengjuan Wang, Nikolai Slavov, Iseult Lynch, , Anna Salvati, Kenneth A. Dawson<br />

Centre for BioNano Interactions, University College Dublin, Belfield, Dublin 4, Irel<strong>and</strong><br />

Fengjuan.Wang@cbni.ucd.ie, nslavov.princeton@gmail.com, Iseult.Lynch@cbni.ucd.ie,<br />

Anna.Salvati@cbni.ucd.ie, Kenneth.A.Dawson@cbni.ucd.ie<br />

Nanoparticles composed of very different materials can enter most cells easily, <strong>and</strong> those<br />

without a targeting moiety are often seen to accumulate in lysosomes, with no clear evidence<br />

of their subsequent export or degradation. 1, 2 Very different outcomes for the cell can be<br />

observed, depending on the nature of the material: in many cases no alteration of cellular<br />

functions can be detected, whereas in other specific cases, nanoparticles can induce<br />

programmed cell death.<br />

The capability of activating very different pathways, including cell death, together with their<br />

small size which allows them to enter almost everywhere in living organisms <strong>and</strong> inside the<br />

cells, makes nanoparticles a powerful tool for targeted therapeutics.<br />

Our hypothesis is that nanoparticle uptake, trafficking <strong>and</strong> impact on cellular functions are<br />

governed by the properties of the nanoparticles (such as surface charge) together with the<br />

nature of the adsorbed proteins, as once in contact with biological fluids, such as serum,<br />

nanoparticles get coated with a very specific layer of proteins, the so-called protein corona,<br />

which constitutes a new biological identity interacting with the cellular machinery.<br />

Where cell death is induced following uptake of specific nanoparticles, a time resolved study<br />

of the pathways <strong>and</strong> signals activated by the nanoparticles as they travel to the lysosomes has<br />

been obtained, which includes mitochondrial <strong>and</strong> lysosomal damage, with activation of<br />

apoptosis <strong>and</strong> autophagy. Gene arrays over the same time frame allow us to begin to untangle<br />

<strong>and</strong> describe the activated pathways at system level.<br />

1. Shapero K, Fenaroli F, Lynch I, Cottell DC, Salvati A, Dawson KA. Time <strong>and</strong> space<br />

resolved uptake study of silica nanoparticles by human cells. Molecul BioSyst 2010; DOI:<br />

10.1039/C0MB00109K:<br />

2. Lesniak A, Campbell A, Monopoli M, Lynch I, Salvati A, Dawson KA. Serum Heat<br />

Inactivation Affects Protein Corona Composition And Nanoparticle Uptake. Biomater 2010;<br />

31: 9511-9518.<br />

3. Cedervall T, Lynch I, Lindman S, Berggard T, Thulin E, Nilsson H, Dawson KA,<br />

Linse S. Underst<strong>and</strong>ing the nanoparticle-protein corona using methods to quantify exchange<br />

rates <strong>and</strong> affinities of proteins for nanoparticles. Proc Natl Acad Sci USA 2007; 104: 2050-<br />

2055.<br />

4. Lynch I, Salvati A, Dawson KA. Protein-nanoparticle interactions: What does the cell<br />

see? Nat Nanotech 2009; 4: 546-547.<br />

80<br />

3, 4


Session 1: Cell death Poster 15<br />

�<br />

ATP regulated Apoptosis versus Autophagy in Cultured Dendritic Cells.<br />

Sarah Flacke 1 , Patricia Schilling 1 , Fengguang Liu 1 , Karl J. Foehr 1 , Manfred Weiss, Paul<br />

Walther 2 , E. Marion Schneider 1<br />

1 Sektion Experimentelle Anaesthesiologie, University Hospital Ulm, Steinhoevelstr. 9, 89075<br />

Ulm, Germany; 2 Electron Microscopy Facility, Ulm University, Ulm, Germany<br />

Cell stress may lead to the release of nucleotides in many tissues. Specifically, fluid shear, cell<br />

stretching, hypoxia, osmotic swelling, but also temperature-related stimuli induce ATP release in<br />

addition to other nucleotides. Extracellular nucleotides bind to cell surface P2 receptors, either P2X,<br />

which are ATP-gated nonselective cation channels, or P2Y, which are G-protein-coupled receptors.<br />

There are seven subtypes of P2X (P2X1–7) <strong>and</strong> eight subtypes of P2Y (P2Y1, -2, -4, -6, <strong>and</strong> -11–14)<br />

receptors currently identified in mammals. A major effect occurs by ATP following stimulation of the<br />

P2RX7 receptor leading to extensive cell blebbing in addition to pore formation, facilitating the<br />

transport of molecules as large as 900 kD. Amongst cells of the hematopoietic system, macrophages<br />

<strong>and</strong> dendritic cells express P2RX7 receptors at high density. The expression of P2RX7 as well as its<br />

functional activity can be further increased by in vitro culture in the absence of exogenous growth<br />

factors <strong>and</strong> cytokines. Using cell biology methods, patch clamping, caspase 3/7 <strong>and</strong> caspase 8<br />

measurements in lysates, <strong>and</strong> or LC-3 staining by flow cytometry, we determined the effect of<br />

extracellular ATP on cultured dendritic cells from patients with macrophage activation syndromes<br />

(MAS). In contrast to healthy donors, MAS patients were unique by their increased precursor<br />

frequencies of P2RX7 high expressing dendritic cells in peripheral blood. We found two types of<br />

ATP- responses in selected donors: One which induced apoptosis following ATP-stimulation <strong>and</strong><br />

another mediated increased LC-3 expression <strong>and</strong> autophagy by the same ATP stimulation protocol.<br />

These differential results corresponded to a donor-specifically smaller (about 10pA/pF) or larger<br />

(about 100pA/pF) electrophysiological response as determined by patch clamping. Pyrosequencing<br />

was applied to study functional single nucleotide polymorphisms of the P2RX7 receptor in different<br />

donors. Out of 5 SNPs investigated*, the P2RX7 signal intensity as well as pro- vs. anti-apoptosis <strong>and</strong><br />

the corresponding induction of autophagy in these dendritic cells, was largely related to a 1513A!C<br />

polymorphism, responsible for a Glu496!Ala exchange in the intracellular C-terminus of the P2RX7<br />

receptor. We therefore conclude that functional SNPs of the P2RX7 receptor are responsible for<br />

dichotomous stress-induced <strong>and</strong> P2RX7-mediated response leading to either apoptosis or increased<br />

survival of immature dendritic cells. Moreover, this observation may explain a differential traumainduced<br />

stress response in vivo.<br />

*Geistlinger et al. Clin Chim Acta, in press.<br />

81


Session 1: Cell death Poster 16<br />

Involvement of AMPK signaling in liver xenobiotic-metabolizing enzymes regulation.<br />

Brice SID, Nicolas DEJEAN, Christophe GLORIEUX, Raphael BECK, Julien<br />

VERRAX <strong>and</strong> Pedro Buc CALDERON<br />

Toxicology <strong>and</strong> Cancer Biology Research Group, Louvain Drug Research Institute,<br />

Université catholique de Louvain, 1200-Brussels. BELGIUM. Email:<br />

brice.sid@uclouvain.be<br />

AMP-activated protein kinase (AMPK) is a phylogenetically conserved serine/threonine<br />

protein kinase, which has been proposed as a metabolic master switch mediating the cellular<br />

adaption to environmental or nutritional stress factors. AMPK responds to any cellular stress<br />

that threatens to lower ATP levels by arresting nonessential ATP-using functions <strong>and</strong><br />

stimulating ATP-generating pathways. Cytochrome P450 (CYPs) <strong>and</strong> Transferases are<br />

responsible for metabolism of most xenobiotics <strong>and</strong> required for the efficient elimination of<br />

foreign chemicals from the body. Paradoxically, theses enzymes may activate biologically<br />

inert compounds to eletrophilic derivatives that can cause toxicity, cell death <strong>and</strong> sometimes<br />

cellular transformation resulting in cancer. Because the regulation of theses enzymes can be<br />

affected by metabolic <strong>and</strong> nutritional stress <strong>and</strong> some studies showed that AMPK is involved<br />

in the induction of some CYPs in human hepatoma-derived cells <strong>and</strong> in primary cultures of<br />

human <strong>and</strong> mouse hepatocytes, we investigated the role of AMPK in rat liver xenobiotic<br />

metabolism regulation. In our study, we used rat precision-cut liver slices (PCLS), which are<br />

described as valuable tools for in vitro metabolic studies of drug c<strong>and</strong>idates. As compared to<br />

isolated hepatocytes, this model offers the advantages of preserving the tissue architecture <strong>and</strong><br />

the proportion of the different cell types. We showed that treatment of the slices with<br />

A769662 AMPK activator during 6h induces AMPK phosphorylation on Thr172 residue,<br />

which is essential for its activity as well as downstream Ser79 phosphorylation of acetyl-<br />

CoA-carboxylase (ACC), a metabolic target of AMPK. In parallel, A769662 treatment<br />

significantly decreases CYP2E1 <strong>and</strong> glucuronosyltransferases (UGT) activities without<br />

altering ATP content <strong>and</strong> lactate deshydrogenase leakage. These data suggest an essential role<br />

of AMPK in the liver xenobiotic metabolism regulation.<br />

82


Session 1: Cell death Poster 17<br />

Increase in p21 CDKN1A protein levels by protesome inhibitors does not affect the<br />

apoptotic response after DNA damage<br />

Micol Tillhon 1 , Nicoletta Pieri 1 , Ornella Cazzalini 2 , Lucia A. Stivala 2 , A. Ivana Scovassi 1 ,<br />

Ennio Prosperi 1<br />

1 Istituto di Genetica Molecolare CNR, Pavia; 2 Dipartimento di Medicina Sperimentale,<br />

Sez. Patologia generale, Università di Pavia, Via Ferrata 1, 27100 Pavia, Italy -<br />

tillhon@igm.cnr.it<br />

p21 CDKN1A is a well known cell cycle inhibitor playing also important roles in transcription<br />

regulation, apoptosis, <strong>and</strong> DNA repair. Pharmacological approaches have been developed to<br />

contrast tumor cell proliferation, by exploiting the p21 ability to induce cell cycle arrest. In<br />

this regard, both inhibitors of histone deacetylases (HDAC) <strong>and</strong> of the proteasomal<br />

machinery, have been shown to increase p21 protein levels, thereby impairing tumor cell<br />

growth. However, this type of intervention has been regarded as potentially dangerous, when<br />

applied in concomitance with typical anticancer drugs inducing DNA damage. In fact, high<br />

p21 levels have been shown to inhibit the apoptotic program <strong>and</strong> to allow the repair of DNA<br />

damage, thereby rendering tumor cells less susceptible to the killing activity of the latter<br />

drugs. Thus, p21 protein levels may greatly influence the outcome of chemotherapy. We have<br />

previously found that HDAC inhibitors did not significantly affect initiation of nucleotide<br />

excision repair (NER) in the presence of p21 protein. In contrast, no clear information is<br />

available on the effect of proteasome inhibitors on DNA repair. Here, we have investigated<br />

whether the presence of elevated p21 protein levels induced by the proteasome inhibitor<br />

MG132, may affect the NER <strong>and</strong> the apoptotic process. We have analyzed the recruitment of<br />

NER proteins <strong>and</strong> p21 to localized DNA damage sites, <strong>and</strong> determined NER efficiency in<br />

normal, as well as in p21-null human fibroblasts. The results have shown that MG132 induced<br />

the persistence of XPC, PCNA <strong>and</strong> p21 protein, at local DNA damage sites. The persistence<br />

of p21 at DNA damage sites did not significantly affect the recruitment of other PCNAinteracting<br />

NER factors, like DNA polymerase delta. However, NER efficiency was reduced<br />

<strong>and</strong> cell death was increased by the proteasome inhibitor. These results suggest that MG132<br />

affects NER efficiency <strong>and</strong> induce apototic cell death, independently of the presence of p21<br />

protein. [Work supported by AIRC].<br />

83


Session 1: Cell death Poster 18<br />

Mechanistic study on protective activity of humanins.<br />

Zapa!a B 1 , Góralska J 1 , Knapp A 1 , "liwa A 1 , Awsiuk M 1 , Tjelle TE 2 , Wybra#ska I 1 ,<br />

Polus A 1 ; Rustan AC 3 , Blomhoff R 2 , Berge RK 4 , Dembi#ska-Kie$ A 1 .<br />

1<br />

Department of Clinical Biochemistry Jagiellonian University Medical College, Krakow,<br />

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

2<br />

Department of Nutrition Institute of Basic Medical Sciences University of Oslo, Norway<br />

3<br />

Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo,<br />

Norway<br />

4<br />

Institute of Medicine, Section of Medical Biochemistry, University of Bergen, Norway<br />

Humanin (HN), the 24-amino acid peptide, known as a potent intracellular inhibitor of<br />

apoptosis, interacts with Bax, the member of the Bcl2 proapoptotic protein family. Increased<br />

expression of HN peptide observed in the AD brain <strong>and</strong> skeletal muscles cells from patients<br />

with mitochondrial disorders suggests that HN plays a role in modification of pathological<br />

events such as neurodegeneration, inflammation, or energy deficiency.<br />

The aim of the study was to examine the influence of exogenous HN <strong>and</strong> its derivatives:<br />

HNG, 13ThrHN10b <strong>and</strong> 13IleHN10b on mitochondria-dependent antiapoptotic mechanisms<br />

in cellular (LN-18, Daoy, C8-D1A HUVEC,) as well as animal transgenic models of<br />

inflammatory response (NFkß-luciferase reporter mice <strong>and</strong> the human TNF-! overexpressed<br />

mice).<br />

HN <strong>and</strong> its synthetic isoforms did not influence directly the changes in mitochondrial<br />

permeability (BD Bioimager 855 microscopy <strong>and</strong> Canto II flow cytometry), oxygen<br />

consumption (Oxygraph-2k, OROBOROS), mitochondrial ATP generation (ATPliteTM<br />

Luminescence ATP Detection Assay System, Perkin Elmer) or oxidation of energy substrates<br />

( 14 C-glucose <strong>and</strong> 14 C-oleic acid) (Microbeta, PerkinElmer) in cells. The mitochondrial<br />

megachannel activation (Hewlett Packard 8452A) <strong>and</strong> calcium retention capacity<br />

(extramitochondrial calcium ([Ca 2+ ]e) of isolated rat liver mitochondria was also not<br />

influenced by HN peptides.<br />

The apoptotic gene expression (TaqMan Applied Microarray system, TLDA) in the brain cell<br />

lines pointed to the higher than endothelial cells antiapoptotic potential. HN peptides<br />

modulated the antiapoptotic potential not only by influencing Bcl/Bax genes expression ratio.<br />

Results from microarray analyses in brain from mice treated with HNG <strong>and</strong> challenged with<br />

LPS showed that HNG down regulated numerous gene sets associated with apoptosis <strong>and</strong><br />

neurogenerative processes (GSEA analyses). In NFkß-luciferase reporter mouse model there<br />

were a tendency that HNG inhibited NFkB-regulated luciferase activity, although not<br />

significant, in heart <strong>and</strong> brain. Moreover in TNF-! overexpressed mice the reduction of<br />

arthritis development (confirmed by forelimb grip test) pointed to the possible<br />

antiinflammatory activity of HN. However, the observed effects seem to be stressordependent,<br />

cell-dependent as well as dependent on humanin peptide structure.<br />

The study was supported by Polish-Norwegian grant no. PNRF-104-AI-1/07 <strong>and</strong> Polish grant<br />

no. k/pbp/000318<br />

84


Posters are classified by session<br />

<strong>and</strong> then in alphabetical order (PRESENTING AUTHOR)<br />

(Late breaking abstracts are at the end of the <strong>book</strong>)<br />

Session 2: Cell signaling<br />

85


Session 2: Cell signaling Poster 1<br />

NT-S100A8 INHIBITS INSULIN RELEASE AND ACTIVATES AKT AND NF-KB<br />

CELL SIGNALLING IN PANCREATIC CANCER (PC) CELLS<br />

Daniela Basso, Paola Fogar, Andrea Padoan,Stefania Moz, Dania Bozzato, Elisa Fadi,<br />

Eliana Greco, Filippo Navaglia, Carlo-Federico Zambon <strong>and</strong> Mario Plebani.<br />

Department of Laboratory Medicine, University of Padua, Via Giustiniani 2, 35128<br />

Padova, Italy. E-mail: biolmol@unipd.it<br />

Objectives: to verify whether NT-S100A8, a peptide isolated from PC tissue of diabetic<br />

patients:1) alters Akt <strong>and</strong> NFkB signalling in PC cells, 2) interferes with insulin (Ins)<br />

signalling, 3) alters Ins response to glucose stimulation.<br />

Methods: PC cell lines (BxPC3,Capan1,MiaPaCa2) remained unstimulated or were stimulated<br />

with 50 mU Ins for 10 min with/without 50 <strong>and</strong> 500 nM NT-S100A8 for 5,10,15 <strong>and</strong> 30 min.<br />

Cell lysates immunoblots were performed with pIkBa, pAkt (Ser 473 ,Thr 308 ), Akt, pPDK1<br />

(Ser 241 ), p-mTOR (Ser 2448 ), b-actin antibodies. bTC6 rat insulinoma cells were untreated or<br />

treated 30 min daily for 1 week with 50, 200 <strong>and</strong> 500 nM NT-S100A8 plus 20 mM glucose or<br />

with 20 mM glucose alone. Cells were then stimulated with 20 mM glucose <strong>and</strong> Ins was<br />

measured at 2,3,5,10,15,30 min.<br />

Results: both Ins <strong>and</strong> NT-S100A8 independently induced Akt Ser 473 phosphorylation in<br />

BxPC3 <strong>and</strong> MiaPaCa2, not in Capan1. Akt Thr 308 phosphorylation in all PC cell lines was<br />

induced by Ins, not by NT-S100A8. NT-S100A8 time <strong>and</strong> dose-dependently induced pmTOR<br />

in BxPC3, but it did not induce pPDK1. To study NFkB signalling we assessed the<br />

phosphorylation of its cytoplasmic inhibitor IkBa. In all cell lines IkBa was constitutively<br />

phosphorylated. Ins determined a significant reduction of pIkBa in Capan1 <strong>and</strong> MiaPaCa2<br />

<strong>and</strong> an enhancement in BxPC3, effects not counteracted by NT-S100A8. In BxPC3 <strong>and</strong><br />

Capan1, NT-S100A8 caused an increase in pIkBa after 5min, followed by a reduction at 10<br />

<strong>and</strong> 15min <strong>and</strong> a recovery at 30 min. pIkBa in MiaPaCa2 cells was not modified. Glucose<br />

induced early (2min) <strong>and</strong> late (15-30min) Ins release in control bTC6 cells. The amount of Ins<br />

release was progressively reduced when cells were stimulated with glucose for one week <strong>and</strong><br />

almost completely abolished when glucose was given with NT-S100A8 (Repeated measures<br />

ANOVA: p


Session 2: Cell signaling Poster 2<br />

NPHS2 GENE MUTATION ANALYSIS RESULTS OF 631 TURKISH PATIENTS<br />

WITH STEROID RESISTANT NEPHROTIC SYNDROME<br />

AFIG BERDELI 1 , SINEM NALBANTOGLU 1 , DEMET TIGLI 1 , MERVE ATAN 1 ,<br />

PERVIN TOPARLAK 1 , AYBEN PASOLAR 1 , ILKAY DEMIREL 1 , FATMA<br />

AKYIGIT 1 , SEVGI MIR 2<br />

1. Ege University Faculty of Medicine, Child Hospital Molecular Medicine Laboratory,<br />

35100, Bornova, izmir, Turkey.<br />

2. Ege University Faculty of Medicine, Department of Pediatrics, Pediatric Nephrology<br />

Division, 35100, Bornova, izmir, Turkey.<br />

NPHS2 gene, encoding the 24-KDa slit diaphragm localized glomerular protein podocin, is<br />

mutated in autosomal recessive steroid-resistant nephrotic syndrome (SRNS) which is<br />

characterized by the presence of the central findings of proteinuria, hypoalbuminemia, <strong>and</strong><br />

edema. Mutations in NPHS2 encoding podocin are found as the prevalent cause of steroidresistant<br />

nephrotic syndrome.<br />

In this respect, we performed mutation analysis by DNA sequencing analysis of all coding<br />

exons <strong>and</strong> intron/exon boundaries of the NPHS2 gene in a total of 631 children patient group<br />

clinically diagnosed with SRNS (of which 66 of them were familial <strong>and</strong> the remaining 565<br />

were sporadic cases). A total of 100 healthy children were involved in the mutation scanning<br />

analysis for healthy control group.<br />

According to the mutation analysis results of NPHS2 gene; 18% of the patients were<br />

evaluated as mutation positive NPHS2 group (n=112) in which this rate corresponds to 31%<br />

of the familial cases, <strong>and</strong> to 17% of the sporadic cases group. In the mutation positive NPHS2<br />

group, most of the mutations were found out to harbor in exons 1, 4, <strong>and</strong> 5 while no mutation<br />

were found in exon 6 of the respective gene. A total of 46 mutations were determined in the<br />

NPHS2 gene, <strong>and</strong> 32 of which were characterized as a novel mutation. The mutation rate of<br />

R138Q was determined as 3.7% in our study group contrary to the findings of Europe <strong>and</strong><br />

Israil populations in which the observed frequency was 50%. Additionally, P118L, R138X,<br />

R168H, R138Q, IVS7+5G>A, A212T, c.460-467insT, c.503delG, H228D, S211A, <strong>and</strong><br />

V218G mutations were typically found as the cause of end-stage renal disease (ESRD). For<br />

the remaining 519 mutation negative patients, effects of possible modifier loci involving other<br />

podocin interacting proteins may be considered.<br />

In brief, as a causative SRNS gene, we should consider NPHS2 gene mutation<br />

screening in early diagnosis <strong>and</strong> the follow-up of the clinical course. In relation to<br />

homozygous or compound heterozygous NPHS2 mutated patients who have the lack of<br />

response to st<strong>and</strong>ard steroid therapy we suggest to perform NPHS2 gene mutation analysis for<br />

every child (if consent can be obtained) soon after the first episode of SRNS. For the newly<br />

diagnosed patients, the crucial certain determination of the causative disease gene mutation<br />

will enable clinicians to avoid redundant immunosuppressive therapeutic trials.<br />

87


Session 2: Cell signaling Poster 3<br />

Laminin 5 promotes epithelial mesenchymal transition (EMT) through phosphorylation<br />

– dependent integrin linked kinase activation.<br />

Dorota Gil, Joanna Dulinska-Litewka <strong>and</strong> Piotr Laidler<br />

Chair of Medical Biochemistry Jagiellonian University Medical College, Kraków,<br />

Pol<strong>and</strong>; ul.Kopernika 7, 31-034 Kraków, e-mail: dorotabeata.gil@uj.edu.pl<br />

Integrin linked kinase (ILK) is a serine/threonine protein kinase that regulates physiological<br />

number of cellular function. ILK is a binding partner of the beta1 <strong>and</strong> beta3 integrin subunit<br />

as a cytoplasmic effector of integrin receptors. Upon activation by different stimuli ILK<br />

phosphorylates Akt <strong>and</strong> GSK-3beta <strong>and</strong> triggers in the related signaling cascades. One of their<br />

major roles is to regulate beta-catenin transcriptional activity. In addition Akt <strong>and</strong> GSK-3<br />

have been reported to activate the NF-kB p65 subunit, increasing the binding of the NF-kB<br />

complex to DNA or inactivates the NF-kB signaling, respectively. We postulate that ILK is a<br />

major signaling mediator involved in EMT, the critical event in the process of cancer<br />

progression. Integrin-mediated cell adhesion is known to regulate the expression of number of<br />

genes. There is no integrin alpha chain associated with beta1 that is as yet known to be<br />

specifically involved in EMT <strong>and</strong> the molecular mechanism through which ECM regulates<br />

EMT are undefined. Some studies suggests however, that alpha3beta1 intergin (laminin 5<br />

receptor) is required for accumulation of beta-catenin in the nucleus of cells undergoing EMT.<br />

We demonstrated that ILK is in human melanoma cells involved in signal transduction from<br />

integrin alpha3beta1 as its activation by laminin5 induced translocation of beta-catenin <strong>and</strong><br />

NF-kB (p65) to nucleus <strong>and</strong> this phenomenon was mediated through ILK phosporylation at S-<br />

343 <strong>and</strong> T -173 <strong>and</strong> by phosphorylation of IKKB. We were also able to show that silencing<br />

of ILK expression by siRNA significantly abolished the presence of molecular <strong>and</strong><br />

phenotypic markers for EMT. In addition NF-kB (p65) nuclear translocation <strong>and</strong> beta-catenin<br />

activation were completely abolished upon ILK silencing in melanoma cells.<br />

It is a first report that shows regulation of ILK by direct phosphorylation.<br />

This work is supported by: MNiSW through Jagiellonian University Medical College<br />

K/ZDS/001464<br />

88


Session 2: Cell signaling Poster 4<br />

Establishing a pipeline for the determination of cell line <strong>and</strong> experiment specific gene<br />

regulatory networks<br />

Thorben Kaetzel, Fabrice Tolle, Jean-Luc Bueb, Thomas Sauter.<br />

<strong>Inflammation</strong> group/Systems Biology group; Life Sciences Research Unit; Faculty of<br />

Science, Technology <strong>and</strong> Communication; University of Luxembourg, 162A, Avenue de<br />

la Faïencerie, L-1511 Luxembourg. Contact: thorben.kaetzel@uni.lu<br />

A bioinformatics pipeline was established for the fast <strong>and</strong> convenient extraction of cell line-<br />

<strong>and</strong> experiment specific c<strong>and</strong>idate networks based on public <strong>and</strong> own microarray data sets <strong>and</strong><br />

by integration of additional available interaction data. This automated workflow allows for<br />

obtaining such networks in a few steps <strong>and</strong> helps to identify nodes, respectively genes which<br />

play significant roles in signalling pathways of interest. Two filtering steps are applied<br />

therefore: 1) cell type specific array data available from public databases is used for coexpression<br />

analysis (assuming that co-expressed genes are also co-regulated) to identify cell<br />

type specific clusters <strong>and</strong> interactions in general; 2) additional (own) stimulation data of<br />

specific expression pro<strong>file</strong>s is applied then to further filter the co-expression network. Only<br />

genes with a fold change over <strong>and</strong> under a certain threshold were depicted therefore <strong>and</strong><br />

included in the c<strong>and</strong>idate networks. To avoid the exclusion of important central nodes of the<br />

network during this filtering step, IPA Knowledge Base (Ingenuity) was used to ensure the<br />

significance <strong>and</strong> completeness of the networks. The curated <strong>and</strong> filtered c<strong>and</strong>idate networks<br />

can then be subjected to mathematical modelling <strong>and</strong> further analysis using Probabilistic<br />

Boolean Modelling. The pipeline was applied to atherosclerosis related microarray data from<br />

two cell lines (THP-1 monocytes <strong>and</strong> HUVEC endothelial cells) which were chosen to mimic<br />

monocytic <strong>and</strong> endothelial behaviour during the process of development of atherosclerosis.<br />

Both cells were stimulated during 1, 4 <strong>and</strong> 24 hours with the cytokine TNF-alpha <strong>and</strong>/or the<br />

micronutrient 1alpha,25(OH)2 vitamin D3, looking for interactions between stress-sensing<br />

nuclear factors <strong>and</strong> nutrient-sensing nuclear receptors which could, by a lifetime exposure, be<br />

part of a misbalanced molecular process towards atherosclerosis.<br />

In a first attempt, two co-expression networks could be extracted for THP-1 <strong>and</strong> HUVEC. In<br />

case of HUVEC, a 1500 node co-expression network could be filtered as described above <strong>and</strong><br />

resulted in a final core network of 84 genes.<br />

89


Session 2: Cell signaling Poster 5<br />

Cooperativity of the MUC1 <strong>and</strong> HGF/c-Met Pathway in HCC<br />

Giray BOZKAYA 1* , Peyda KORHAN 1* , Murat COKAKLI 1 , Esra ERDAL 1 , Ozgul<br />

SAGOL 2 , Sedat KARADEMIR 3 , Cristopher KORCH 4 , Nese ATABEY 1**<br />

1 Dokuz Eylul University, School of Medicine, Department of Medical Biology <strong>and</strong> Genetics, 35340,<br />

Balcova-Izmir, Turkey, korhanpeyda@hotmail.com<br />

2 Dokuz Eylul University, School of Medicine, Department of Pathology, 35340, Balcova-Izmir, Turkey<br />

3 Dokuz Eylul University, School of Medicine, Department of Surgery, 35340, Balcova-Izmir, Turkey<br />

4 University of Colorado Cancer Center-DNA Sequencing <strong>and</strong> Analysis Core Aurora, CO, USA<br />

* Equal contribution **Corresponding author<br />

Hepatocyte growth factor (HGF) induced c-Met activation is known as the main stimulus for<br />

hepatocyte proliferation <strong>and</strong> essential for liver development <strong>and</strong> regeneration. Inappropriate<br />

HGF/c-Met signaling has been correlated with aggressive phenotype <strong>and</strong> poor prognosis in<br />

hepatocellular carcinoma (HCC). MUC1 is a transmembrane mucin whose overexpression is<br />

also correlated with poor prognosis in HCC. However, involvement of MUC1 in HGF/c-Met<br />

signaling is unknown in HCC. In this study we aimed to elucidate possible cooperation of<br />

MUC1 <strong>and</strong> c-Met expressions <strong>and</strong> their association in HCC. We first analyzed the expression<br />

of MUC1 <strong>and</strong> c-Met receptor in poorly differentiated (SNU-475, SNU-449, Mahlavu) <strong>and</strong><br />

well differentiated (HuH-7, Hep3B, Hep G2) HCC cell lines by immunoblotting. We showed<br />

that poorly differentiated HCC cell lines had high expression of both MUC1 <strong>and</strong> c-Met,<br />

whereas well differentiated ones revealed no MUC1 expression <strong>and</strong> lower level of c-Met<br />

expression compared to poorly differentiated ones. We then demonstrated a physical<br />

interaction between c-Met <strong>and</strong> MUC1 in Mahlavu cells by immunoprecipitation.<br />

Interestingly, c-Met activation reduced MUC1 protein level which resulted in decreased<br />

MUC1/c-Met interaction in a time dependent manner. Using SU11274, we restored HGF<br />

induced MUC1 downregulation <strong>and</strong> established that MUC1 downregulation was mediated by<br />

c-Met activation in our experimental system. Intriguingly, siRNA silencing of MUC1 resulted<br />

in enhanced c-Met activation. Lastly we demonstrated that c-Met inhibited tumor cells<br />

exhibited poor migrative <strong>and</strong> invasive ability in response to HGF, whereas MUC1 silencing<br />

increased HGF induced cell motility <strong>and</strong> invasion. Overall, these results suggest that coexpression<br />

<strong>and</strong>/or association of MUC1 <strong>and</strong> c-Met might affect c-Met mediated signaling in<br />

HCC.<br />

90


Session 2: Cell signaling Poster 6<br />

LGR5 has been regulated by Wnt/!-catenin signaling in HCC cells<br />

Imge Kunter 1 , Ne"e Atabey 1 , Esra Erdal 1<br />

1 Dokuz Eylul University, Faculty of Medicine, Department of Medical Biology <strong>and</strong><br />

Genetics, #zmir, Turkey.<br />

Leucine-rich repeat containing G-protein coupled receptor 5 (LGR5) recently has been<br />

described as a stem cell marker in gastrointestinal organs <strong>and</strong> it has been defined as a Wnt/!catenin<br />

target in colon carcinoma. Although overexpression of LGR5 has been observed in<br />

the hepatocellular carcinoma containing !-catenin mutations, there is no enough data<br />

to elucidate the role of LGR5 in liver cancer. In this study expression of LGR5 has<br />

been examined in HCC cell lines (Huh7, Hep3B, HepG2, SNU398, PLC, SNU 475, SNU449,<br />

SKHep1, SNU387, SNU 182) at both transcriptional <strong>and</strong> protein levels. Out of ten cell lines,<br />

HepG2 <strong>and</strong> SNU398 only have overexpression for LGR5 at protein level as expected since<br />

both cell lines contain distinct !-catenin overexpression mutations. When wnt/ !-catenin<br />

signaling has been activated by lithium treatment, expression of LGR5 has been increased<br />

significantly in Huh7, Hep3B <strong>and</strong> PLC cell lines. Additionally, the induction of the Wnt/!catenin<br />

signaling through transfection of the BCAT overexpression plasmid vector leads to<br />

increase in the LGR5 expression, contrarily, diminishing !-catenin activity on Tcf/Lef<br />

transcription through transfection of "NTCF plasmid vector leads to decrease in the LGR5<br />

expression in Huh7 cells. This data provides us evidence that LGR5 may be a target gene of<br />

Wnt/!-catenin signaling pathway in HCC. LGR5 might be very important gene to describe the<br />

initiation mechanisms in the development of HCC.<br />

91


Session 2: Cell signaling Poster 7<br />

Double suppression of the Galpha activity by RGS proteins<br />

Chen Lin 1, 2 1, 3<br />

<strong>and</strong> Vladimir.Katanaev<br />

1 University of Konstanz, Department of Biology, Universitätsstrasse 10, Box 643, 78457<br />

Konstanz, Germany.<br />

konstanz.de<br />

2 Chen.Lin@uni-konstanz.de<br />

92<br />

3 Vladimir.Katanaev@uni-<br />

Signaling molecules such as neurotransmitters, hormones <strong>and</strong> other extracellular lig<strong>and</strong>s act<br />

via G-protein-coupled receptors to regulate crucial functions in animal physiology <strong>and</strong><br />

development. Regulator of G-protein signaling (RGS) proteins are a large <strong>and</strong> diverse<br />

superfamily characteristic by the conserved RGS domain which possesses the GAP (GTPase<br />

activating protein) activity towards G!-subunits of heterotrimeric G proteins, resulting in<br />

acceleration of the GTP hydrolysis <strong>and</strong> faster deactivation of G protein signaling. By using<br />

the yeast two-hybrid screening, we found CG5036, a Drosophila member of the RGS protein<br />

RZ subgroup, as the binding partner of Drosophila G!o. Surprisingly, both in the yeast twohybrid<br />

system as well as in the biochemical pull-down experiments, CG5036 interacted with<br />

both the GTP - <strong>and</strong> GDP - loaded forms of G!o. Furthermore, in addition to GAP activity,<br />

CG5036 possessed the GDI (guanine nucleotide dissociation inhibitor) activity towards G!o<br />

in vitro. These findings were recapitulated using mammalian G!o <strong>and</strong> G!i, as well as RGS19<br />

proteins. To confirm the biological significance of this unexpected GDI activity, we began to<br />

downregulate CG5036 function during the asymmetric cell divisions leading to the<br />

development of the peripheral sensory bristles of Drosophila - the developmental program<br />

coordinately regulated by the G!o <strong>and</strong> G!i proteins. We found that downregulation of<br />

CG5036 could enhance the dominant phenotypes induced not only by expression of the wildtype<br />

form of the G proteins, but also by the GTPase-deficient mutant form of G!o. Thus, our<br />

results provide the first in vitro <strong>and</strong> in vivo data for the double mechanism of action of an<br />

RGS protein towards G!-subunits, involving both inhibition of GTP incorporation by G! <strong>and</strong><br />

the acceleration of GTP hydrolysis by it.


Session 2: Cell signaling Poster 8<br />

The trimeric Go protein transduces the Wingless-Frizzled2 signal <strong>and</strong> interacts with<br />

Anykrin2 to regulate microtubule stability in Drosophila neuromuscular junction<br />

Anne-Marie Lüchtenborg 1,2 , Diane Egger-Adam 1,3 , &Vladimir L. Katanaev 1,4<br />

1<br />

University of Konstanz, Department of Biology, Universitätsstrasse 10, Box 643, 78457<br />

2 3<br />

Konstanz, Germany. anne-marie.luechtenborg@uni-konstanz.de, diane.eggeradam@uni-konstanz.de,<br />

4 vladimir.katanaev@uni-konstanz.de<br />

The Drosophila neuromuscular junction is a powerful model system to investigate<br />

glutamatergic synapse formation <strong>and</strong> stability. In the fruit fly the correct formation of the<br />

neuromuscular junction depends among others on components of the Wingless signal<br />

transduction pathway which acts in the transcription-independent manner in the synapse.<br />

Giant isoforms of Ankyrin2, a neuronal protein that links cytoskeleton <strong>and</strong> the membrane,<br />

have also been shown important for synapse formation. We could show that the trimeric G<br />

protein Go is heavily expressed in the neuromuscular junction <strong>and</strong> its presynaptic expression<br />

is crucial for formation of this structure. We demonstrate that Go is a transducer of the<br />

Wingless signal acting downstream from Frizzled2. In addition, the alpha subunit of Go<br />

physically <strong>and</strong> genetically interacts with Ankyrin2. Our genetic studies suggest that the<br />

Wingless signal bifurcates downstream from Frizzled2 into the Ankyrin2-dependent branch<br />

<strong>and</strong> the signaling branch involving the Axin-based destruction complex. We provide evidence<br />

that both pathways converge on the microtubule binding protein Futsch, the Drosophila<br />

homolog of the MAP1B. This elaborate mechanism results in coordinated regulation of<br />

neuromuscular junction formation.<br />

93


Session 2: Cell signaling Poster 9<br />

Microvesicles Harbouring Glycosylphosphatidylinositol-Anchored Proteins <strong>and</strong> Derived<br />

From Rat Adipocytes <strong>and</strong> Serum Transfer RNA Coding For Lipid Synthesis Into<br />

Adipocytes<br />

Günter Müller 1 , Eva-Maria Wetekam 1 , Susanne Wied 1 <strong>and</strong> Elisabeth-Ann Dearey 1<br />

1 Sanofi-Aventis Deutschl<strong>and</strong> GmbH, Research & Development, Diabetes Division,<br />

Industrial Park Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany.<br />

guenter.mueller@sanofi-aventis.com<br />

Small vesicles, such as microvesicles <strong>and</strong> exosomes, have been amply documented to transfer<br />

proteins <strong>and</strong> nucleic acids from a variety of donor to acceptor cells with corresponding<br />

physiological <strong>and</strong> pathological consequences. Recently the in vitro transfer of<br />

glycosylphosphatidylinositol-anchored proteins (GPI-proteins) from vesicles released from<br />

large rodent adipocytes to intracellular lipid droplets of small adipocytes has been shown to<br />

be stimulated by certain physiological (palmitate, H2O2) <strong>and</strong> pharmacological stimuli (antidiabetic<br />

sulfonylurea drug glimepiride) <strong>and</strong> to induce the stimulation of esterification into <strong>and</strong><br />

inhibition of the release of fatty acids from triacylglycerol. Here the analysis of small vesicles<br />

derived from rat adipocytes or plasma revealed that those harbouring the GPI-proteins, Gce1<br />

<strong>and</strong> CD73, contain RNA species which are transferred into acceptor adipocytes <strong>and</strong> trigger<br />

upregulation of fatty acid esterification. The transferred RNA species are specific for fatty<br />

acid esterification (e.g. glycerol-3-phosphate acyltransferase) <strong>and</strong> lipid droplet biogenesis<br />

(perilipin-A, caveolin-1). The transfer is more efficient for small rather than large adipocytes<br />

<strong>and</strong> is significantly upregulated by palmitate, glimepiride <strong>and</strong> H2O2. Together the data<br />

suggest, that vesicles released from large adipocytes stimulate triacylglycerol storage in small<br />

adipocytes by mediating the transfer of the required information encoded by either relevant<br />

RNA <strong>and</strong> GPI-protein species. Paracrine <strong>and</strong> endocrine regulation of triacylglycerol storage<br />

<strong>and</strong> in parallel cell size between large <strong>and</strong> small adipocytes by RNA- <strong>and</strong> GPI-proteinharbouring<br />

vesicles may represent a novel target for interference with metabolic diseases,<br />

such as obesity <strong>and</strong> type II diabetes.<br />

94


Session 2: Cell signaling Poster 10<br />

MAPK signaling pathway is involved in the proliferative stimulus triggered by early<br />

weaning in the gastric mucosa<br />

Luciana H Osaki, Patricia Gama<br />

Laboratory of Gastrointestinal Epithelium Biology, Department of Cell <strong>and</strong><br />

Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Av.<br />

Prof. Lineu Prestes 1524, 05508-900, Sao Paulo, SP, Brazil, luciana.osaki@usp.br<br />

Epidermal Growth Factor Receptor (EGFR) regulates cell proliferation <strong>and</strong> differentiation by<br />

activating several signaling pathways, such as MAPK <strong>and</strong> Src. The dietary pattern is an<br />

important regulator of the growth <strong>and</strong> maturation of the stomach. Early weaning (EW), which<br />

means an abrupt change from milk to solid food ingestion, stimulates gastric cell proliferation<br />

<strong>and</strong> differentiation, in parallel with the overexpression of EGFR. ERK <strong>and</strong> Src are also more<br />

activated in EW animals, suggesting their involvement in the modifications of the gastric<br />

growth triggered by EGFR. Here, we investigated whether ERK pathway is involved in<br />

gastric cell proliferation stimulus after EW. Early weaning was performed by separating 15-dold<br />

Wistar rat pups from the dam <strong>and</strong> feeding them with powdered chow. One group of<br />

animals was injected with PD98059 at 300 !g/kg to test the effects of MAPK inhibition <strong>and</strong><br />

the control group received 0.5% DMSO. Samples were collected 2 <strong>and</strong> 3 days after the onset<br />

of treatment. We confirmed that p-ERK was decreased in the gastric mucosa after PD98059<br />

treatment (p


Session 2: Cell signaling Poster 11<br />

The Role of MT1-MMP in FGF Signaling<br />

Hoi Leong Xavier Wong, Kui Ming Chan, Zhongjun Zhou<br />

Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong<br />

Kong, Hong Kong SAR, China. xavier _1110@hotmail.com<br />

Craniosynostosis is a craniofacial defect resulting from the premature fusion of cranial<br />

sutures. It is often linked to the deregulated Fibroblast Growth Factor (FGF) Receptor<br />

signaling. Membrane Type 1-Matrix Metalloproteinase, MT1-MMP, is a membrane tethered<br />

enzyme capable of remodeling extracellular matrix. MT1-MMP knockout mice display severe<br />

craniofacial phenotypes including delayed suture closure which resembles FGF signaling<br />

mutant mice. To underst<strong>and</strong> the defects in craniofacial development in MT1-MMP deficient<br />

mice <strong>and</strong> to elucidate the molecular mechanism behind, we have examined the embryonic<br />

development of calvarial bone in MT1-MMP deficient mice. We found that MT1-MMP<br />

deficient mice exhibit defects in calvarial bone development, initiating at 15.5dpc. MT1-<br />

MMP co-expresses with FGF lig<strong>and</strong>s <strong>and</strong> FGF receptors during parietal bone development at<br />

the embryonic stage. Moreover, proliferation rate of primary calvarial osteoblast deficient in<br />

MT1-MMP is significantly reduced <strong>and</strong> FGF2 induced accelerated bone growth is also<br />

reduced in MT1-MMP -/- calvaria explants in ex vivo culture. More importantly, MT1-<br />

MMP/FGFR1 compound heterozygous mice exhibit abnormal bone phenotypes, such as<br />

shortened long bone <strong>and</strong> reduced body length, which clearly indicates the genetic interaction<br />

between mmp14 <strong>and</strong> fgfr1. Our results suggest the importance of MT1-MMP in calvarial<br />

osteogenesis via regulation of FGF signaling.<br />

96


Posters are classified by session<br />

<strong>and</strong> then in alphabetical order (PRESENTING AUTHOR)<br />

(Late breaking abstracts are at the end of the <strong>book</strong>)<br />

Session 3: Transcriptional control<br />

97


Session 3: Transcriptional control Poster 1<br />

Mithramycin-mediated regulation of raptor gene transcription during erythroid<br />

differentiation of K562 cells<br />

Alessia Finotti 1 , Nicoletta Bianchi 1 , Cristina Zuccato 1 , Enrica Fabbri 1 ,<br />

Monica Borgatti 1 <strong>and</strong> Roberto Gambari 1<br />

1 Laboratory for the Development of Pharmacological <strong>and</strong> Pharmacogenomic Therapy<br />

of Thalassemia, Biotechnology Center, Ferrara University, via Fossato di Mortara 64,<br />

44121, Ferrara, Italy; e-mail:gam@unife.it.<br />

The mTOR pathway might be associated with erythroid differentiation, as rapamycin (a<br />

strong <strong>and</strong> highly selective mTOR inhibitor) is able to induce erythroid differentiation of<br />

K562 cells. Here we have analyzed the effects of erythroid-inducer mithramycin (MTH) on<br />

raptor gene transcription, in consideration of the fact that raptor is an important companion of<br />

mTOR. We have found that raptor gene promoter contains at least 10 binding sites for the<br />

transcription factor Sp1. By gel retardation assay, we have demonstrated that three of these<br />

sites are bound very efficiently by Sp1. Interestingly, MTH interferes with these Sp1/DNA<br />

interactions. Moreover, chromatin immunoprecipitation assays fully support the concept that<br />

MTH inhibits in intact K562 cells the recruitment of Sp1 to the raptor gene promoter. This is<br />

associated with a strong effect of MTH on raptor gene expression. The results obtained by<br />

quantitative RT-PCR clearly indicate that content of raptor mRNA decreases during erythroid<br />

induction of human leukemia K562 cells. These data have been confirmed by western blotting<br />

also using erythroid precursor cells (ErPC) isolated from beta-thalassemic patients <strong>and</strong> treated<br />

with MTH. In this case, we always observed a reduction of raptor mRNA accumulation <strong>and</strong><br />

raptor production. Accordingly, inhibition of phosphorilated p70S6K <strong>and</strong> of ribosomal S6<br />

protein phosphorilation were found following MTH treatment.<br />

Roberto Gambari has received grants from Fondazione CARIPARO (Cassa di Risparmio di<br />

Padova e Rovigo) <strong>and</strong> Telethon (contract GGP10124). This research is also supported by The<br />

Associazione Veneta per la Lotta alla Talassemia, Rovigo.<br />

98


Session 3: Transcriptional control Poster 2<br />

Regulation of the catalase expression in a model of breast cancer MCF-7 resistant to<br />

oxidative stress.<br />

Christophe Glorieux, Julien Verrax, Nicolas Dejeans, Raphaël Beck, Brice Sid <strong>and</strong><br />

Pedro Buc Calderon.<br />

Toxicology <strong>and</strong> Cancer Biology research group, Louvain Drug Research Institute,<br />

Université catholique de Louvain, Brussels, Belgium. Email :<br />

christophe.glorieux@uclouvain.be<br />

Cancer cells are more sensitive to oxidative stress due a low level of antioxidant enzymes<br />

(catalase, glutathion peroxidase, superoxide dismutase,...). Given the increasing interest to<br />

develop pro-oxidant therapies in clinical studies by targeting intracellular redox alteration, we<br />

decided to study the molecular consequences of chronically exposing cancer cells to oxidative<br />

stress. To this end, we used an H2O2-generating system, namely the association<br />

ascorbate/menadione (asc/men) that generates an oxidative stress which ultimately causes<br />

cancer cell death. We generate breast cancer MCF-7 cells resistant to oxidative stress<br />

(asc/men) in order to underst<strong>and</strong> the mechanism of resistance acquisition. In these resistant<br />

cells we founded an increased expression of catalase. We decided to explore how the catalase<br />

gene is regulated in these resistant cell lines. We showed a two fold increase of the catalase<br />

mRNA in the resistant cell lines <strong>and</strong> transcriptional activity by using a reporter plasmid. At<br />

present time, no evidence of signalling pathways (Foxo3a, PPAR gamma,...) were identified.<br />

Interestingly, we found a 2,5 fold increase of the protein level <strong>and</strong> the enzymatic activity of<br />

the catalase but the post-translational modifications appeared to be unchanged as compared to<br />

control MCF-7 cells. Finally, we observed an effect of both HDACs <strong>and</strong> DNMTs inhibitors<br />

suggesting a possible role of epigenetic events to explain the overexpression of catalase level<br />

in resistant cells.<br />

99


Session 3: Transcriptional control Poster 3<br />

The NF-kappaB signaling pathway is inhibited by heat shock independently of active<br />

transcription factor HSF1 <strong>and</strong> increased levels of HSP70i.<br />

Patryk Janus 1 , Magdalena Kalinowska-Herok 1 , Ma!gorzta Paku!a 2 , Katarzyna<br />

Szo!tysek 1 , Wojciech Pig!owski 1 , Marek Kimmel 3 , Piotr Wid!ak 1<br />

1) Maria Sklodowska-Curie Memorial Cancer Center <strong>and</strong> Institute of Oncology; Gliwice,<br />

Pol<strong>and</strong>; e-mail: patrykjanus@gmail.com, mkalinowska@io.gliwice.pl, kszoltysek@io.<br />

gliwice.pl, wpiglowski@io.gliwice.pl, widlak@io.gliwice.pl; 2) University of Aarhus;<br />

Aarhus, Denmark; e-mail: mpakula@inano.dk; 3) Silesian University of Technology;<br />

Gliwice, Pol<strong>and</strong>; e-mail: mkimmel@polsl.pl<br />

NF-!B transcription factor regulates numerous genes important for inflammation, immune<br />

response <strong>and</strong> cell survival. HSF1 is the primary transcription factor activated under stress<br />

conditions that is responsible for induction of genes encoding heat shock proteins. The NF-<br />

!B activation pathway is blocked by heat shock. Here we investigated whether active<br />

HSF1 inhibited this pathway in the absence of stress conditions. Activation of the NF-!B<br />

pathway <strong>and</strong> expression of NF-!B-dependent genes was analyzed in TNF"-stimulated U-2<br />

OS human osteosarcoma cells that were either preconditioned with hyperthermia or<br />

engineered to express a constitutively active form of HSF1 in the absence of heat shock. The<br />

hyperthermia resulted in a general blockade in the degradation of the I!B" inhibitor, nuclear<br />

translocation of NF-!B <strong>and</strong> expression of NF-!B-dependent target genes. In marked contrast,<br />

the presence of constitutively active HSF1 did not block TNF"-induced activation of the NF-<br />

!B pathway or general expression of the NF-!B-dependent genes. We concluded that in the<br />

absence of heat shock the NF-!B activation pathway is not inhibited by active HSF1<br />

transcription factor or by increased level of HSF1-induced HSP70i.<br />

100


Session 3: Transcriptional control Poster 4<br />

Leflunomide overcomes CD40L/IL-4-mediated resistance to fludarabine-induced<br />

apoptosis <strong>and</strong> effectively blocks proliferation of CLL cells<br />

S. Dietrich, A. Zota, M. Hess, M. Rieger, C. Schweizer, A.D. Ho, P. Dreger, T. Luft,<br />

Dept. Medicine V, University of Heidelberg (Heidelberg, DE)<br />

BACKGOUND AND AIM: Chronic lymphocytic leukemia (CLL) has a poor prognosis if resistance<br />

to purine analogues, such as fludarabine, occurs. Therefore strategies to overcome fludarabine<br />

resistance in poor-risk CLL are highly warranted. Proliferation <strong>and</strong> resistance to fludarabine-induced<br />

apoptosis of CLL cells can be induced in vitro with CD40L <strong>and</strong> IL-4. This experimental setting may<br />

mimic the anti-apoptotic environment protecting CLL cells in lymph nodes in vivo. CD40 signalling<br />

involves NF-kB activation, whereas IL-4 acts through the JAK/STAT pathway. Leflunomide is an oral<br />

drug approved for treatment of rheumatoid <strong>and</strong> psoriasis arthritis. Leflunomide exerts its<br />

immunosuppressive effects by directly inhibiting dihydroorotate dehydrogenase (DHODH), an<br />

enzyme involved in pyrimidine synthesis, but has also been reported to interfere with NF-kB. Serum<br />

concentrations of more than 100 !g/ml of the active metabolite (A771726) can be achieved in patients<br />

with a 20 mg tablet per day.<br />

METHODS: We established a long-term culture system in order to study proliferation <strong>and</strong> apoptosis<br />

resistance of human CLL cells in vitro. This system involves stimulation of CLL cells with a CD40Ltransfected<br />

baby hamster kidney cell line (BHK) <strong>and</strong> IL-4 (50 U/ml). Thymidine incorporation assays<br />

<strong>and</strong> intracellular KI-67 FACS analyses were used to measure proliferation. Apoptosis was quantified<br />

using AnnexinV/7-AAD FACS analyses. Intracellular nuclear FACS analyses for phospho-p65,<br />

phospho-STAT1, phospho-STAT3, survivin <strong>and</strong> bcl-xL as well as quantitative Western Blot analyses<br />

were used to examine the mechanism of apoptosis resistance in CLL cells. Fludarabine (10!g/ml), the<br />

pan-JAK inhibitor Pyridone 6 (0.15 !g/ml) <strong>and</strong>/or Leflunomide (A771726: 0-100 !g/ml) were<br />

investigated in the context of a CD40L/IL-4 signal.<br />

RESULTS: Proliferation of CLL cells required a complementary CD40 <strong>and</strong> JAK/STAT signal <strong>and</strong><br />

could be blocked by the JAK inhibitor. In contrast, resistance to fludarabine-mediated apoptosis could<br />

be induced by CD40 activation alone. This coincided with persistently high levels of intracellular<br />

phospho-p65, survivin <strong>and</strong> bcl-xL. Apoptosis resistance was further enhanced by a complementary<br />

JAK/STAT signal but could not be blocked by the JAK inhibitor. Leflunomide inhibited proliferation<br />

at very low concentrations (IC50: 4 !g/ml), but induced apoptosis of CD40L+IL-4-activated<br />

(“resistant”) CLL cells only at higher concentrations (IC50: 72 !g/ml). Furthermore leflunomide in<br />

combination with fludarabine showed synergistic effects on induction of apoptosis of CLL cells (IC50:<br />

50 !g/ml). The anti-proliferative effect of low Leflunomide concentrations was likely due to inhibition<br />

of DHODH, as inhibition of STAT1/3 phosphorylation was observed at higher concentrations only<br />

(50-100 !g/ml). Similarly, in concentrations which induced apoptosis of CD40L/IL4-activated CLL<br />

cells, leflunomide efficiently inhibited phosphorylation of RelA resulting in low expression levels of<br />

the anti-apoptotic proteins bcl-xL <strong>and</strong> survivin. In contrast, fludarabine alone was inefficient to block<br />

bcl-xL <strong>and</strong> survivin expression in CD40L/IL-4 stimulated CLL cells.<br />

CONCLUSIONS: Leflunomide can overcome CD40L/IL4-mediated fludarabine resistance at<br />

clinically achievable concentrations by inhibiting phosphorylation of STAT1/3 <strong>and</strong> NF-kB-induced<br />

expression of survivin <strong>and</strong> bcl-xL. Furthermore leflunomide has a strong, JAK/STAT-independent,<br />

anti-proliferative effect on CD40L/IL4-activated CLL cells already at very low concentrations.<br />

Therefore leflunomide might be a promising c<strong>and</strong>idate drug to attack CLL cells in chemoresistant<br />

niches.<br />

101


Session 3: Transcriptional control Poster 5<br />

Role of MDC1 in NF-kappaB activation by DNA double-str<strong>and</strong> breaks.<br />

Helene Sabatel, Jacques Piette <strong>and</strong> Yvette Habraken.<br />

Laboratory of Virology <strong>and</strong> Immunology, Signal Transduction Unit, GIGA-R,<br />

University of Liège, 4000 Liège, Belgium. helene.sabatel@student.ulg.ac.be,<br />

jpiette@ulg.ac.be, yvette.habraken@ulg.ac.be<br />

Many anticancer treatments, such as ionizing radiation (IR) <strong>and</strong> DNA topoisomerase I <strong>and</strong> II,<br />

induce apoptosis in tumor cells by generating DNA double str<strong>and</strong> breaks (DSB). ATM kinase,<br />

rapidly activated by DSB, orchestrates different aspects of the DNA Damage Response: cell<br />

cycle arrest, DSB repair <strong>and</strong> transcription factors activation. Both NF-kappaB <strong>and</strong> p53 are<br />

activated by DSB, controlling anti- <strong>and</strong> pro-apoptotic signals <strong>and</strong> therefore affecting cell<br />

survival <strong>and</strong> the outcome of the cancer treatment.<br />

In this work, the importance of MDC1 (Mediator DNA damage checkpoint 1) in the DNA<br />

damage signalling to NF-!B is studied. As expected, MDC1 reduction by siRNAs resulted in<br />

a smaller number of nuclear foci after IR <strong>and</strong> Camptothecin treatments. We observed that<br />

reduced MDC1 level leads to a reduction of NF-!B nuclear translocation after both<br />

treatments. NF-!B transactivation potential was assessed with two reporter systems; either a<br />

transiently transfected !B-Luc reporter plasmid or a stably integrated !B-GFP reporter. In<br />

both cases, we noticed a large reduction of the transcriptionnal activity in cells treated with<br />

siRNA-MDC1. The reduction was however less important than the one observed in cells<br />

treated with an siRNA-ATM. Mefs WT <strong>and</strong> KO for MDC1 were tested for NF-!B activation<br />

by IR <strong>and</strong> Camptothecin. However, as Mefs WT did not activate NF-!B following DNA<br />

damage we could not use this model. The effect of the deletion of different domains of MDC1<br />

is also analysed to determine by which way this protein interferes with NF-kappaB signalling<br />

pathway (foci dependent or independent manner).<br />

In summary, our data indicate that deletion of MDC1 has an impact on NF-!B activation after<br />

both IR <strong>and</strong> Camptothecin treatments contrarily to the disruption of NBS/Mre/Rad50 complex<br />

that affects only IR signalling under the tested conditions.<br />

Financial supports: IAP 6/18, Centre anti-cancéreux de l’ULg, FRS-FNRS, F.R.S.M<br />

102


Session 3: Transcriptional control Poster 6<br />

Regulation of NFkappaB-dependent <strong>and</strong> p53-dependent genes – crosstalk between<br />

signaling pathways.<br />

Katarzyna Szo!tysek 1 , Patryk Janus 1 , Adam Makuchowski 2 , Marek Kimmel 2 , Piotr<br />

Wid!ak 1<br />

1) Maria Sklodowska-Curie Memorial Cancer Center <strong>and</strong> Institute of Oncology; Gliwice,<br />

Pol<strong>and</strong>; e-mail: kszoltysek@io.gliwice.pl, patrykjanus@gmail.com,<br />

widlak@io.gliwice.pl;<br />

2) Silesian University of Technology; Gliwice, Pol<strong>and</strong>; e-mail:<br />

adam.makuchowski@polsl.pl, mkimmel@polsl.pl;<br />

Background: Signaling pathways that depend on p53 or NFkappaB transcription factors are<br />

essential components of cellular responses to stress. Both proteins participate in regulation of<br />

the expression of numerous genes that are involved in cell cycle arrest, DNA repair,<br />

apoptosis, immune response <strong>and</strong> inflammation. Here we analyzed the interference between<br />

both signaling pathways at the level of expression of p53- <strong>and</strong> NFkappaB-dependent genes.<br />

Methods: Colon carcinoma HCT116 cell line was used in two congenic variants either<br />

containing or lacking transcriptionally competent p53. Cells were stimulated with TNF-alpha<br />

cytokine to activate the NFkappaB pathway, <strong>and</strong>/or irradiated with UV to activate the p53<br />

pathway; both stimuli were used in two different combinations – cells were treated with TNF<br />

either 3 hrs before or 6 hrs after irradiation. Activation of the NFkappaB <strong>and</strong> p53 pathways<br />

was monitored by Western-blotting. Expression levels of selected p53-dependent genes<br />

(MDM2, p21/WAF1, PTEN, NOXA) were assessed by QRT-PCR at different time points<br />

after irradiation. Expression pro<strong>file</strong>s of 84 NFkappaB dependent genes were analyzed by the<br />

Human NFkappaB Signaling Pathway RT!Pro<strong>file</strong>r PCR Array at 1 hr time point after the<br />

TNF stimulation.<br />

Results: We observed that radiation-induced activation of p53-dependent genes was affected<br />

in cells stimulated with TNF: UV-induced expression of MDM2, p21/WAF1 <strong>and</strong> NOXA<br />

genes was further up-regulated by either type of TNF treatment. Analysis of NFkappaB<br />

dependent genes revealed 7 genes, namely BCL3, NFKBIA, REL, IL1A, IL8, TNFA,<br />

TNFAIP3, which expression levels differed between cells with different status of p53.<br />

Notably, irradiation of p53-competent cells before activation of the NFkappaB pathway<br />

resulted in down-regulation of all these 7 genes. The data indicated crosstalk between<br />

activation of NFkappaB or p53 pathways <strong>and</strong> expression of genes dependent on the opposite<br />

factor.<br />

103


Session 3: Transcriptional control Poster 7<br />

The NAD dependent deacetylase Sirt1 suppresses pituitary GH synthesis through a<br />

GSK3beta/PP1/ CREB pathway.<br />

Marily Theodoropoulou, Maria A. Tichomirowa, Jose Luis Monteserin Garcia, Günter<br />

K. Stalla<br />

The energy sensor silent information regulator 2 (Sir2) /Sirtuin 1 (Sirt1) plays a pivotal role in<br />

regulating organism metabolic homeostasis <strong>and</strong> lifespan in response to nutrient availability,<br />

by sensing perturbations in NAD + /NADH ratio. Sirt1 activation (by overexpression or<br />

resveratrol treatment) <strong>and</strong> inhibition (by siRNA, sirtinol or nicotinamide treatment)<br />

respectively suppressed <strong>and</strong> upregulated pituitary GH synthesis. GH transcription is primarily<br />

regulated by the cAMP/CREB pathway. Sirt1 significantly suppressed CREB DNA binding<br />

<strong>and</strong> transcriptional activity. CREB transcription <strong>and</strong> protein stability were not altered, but its<br />

dephosphorylation rate was accelerated in pituitary cells overexpressing Sirt1 or treated with<br />

resveratrol. In contrast, overexpression of the deacetylase dead Sirt1-H363Y completely<br />

abolished this effect; in these cells the physiological attenuation phase of CREB<br />

phosphorylation normally observed after three hours of forskolin treatment did not take place<br />

<strong>and</strong> CREB remained phosphorylated. Sirt1 was found to physically associate with CREB <strong>and</strong><br />

to decrease its acetylation levels, indicating a link between CREB acetylation <strong>and</strong><br />

phosphorylation status. CREB dephosphorylation is dependent on the protein phosphatase 1<br />

(PP1), which we found to physically associate with <strong>and</strong> be activated by Sirt1. PP1 is activated<br />

when its inhibitor I-2 gets phosphorylated at threonine residues <strong>and</strong> dissociates from PP1<br />

leaving it in an active form. Resveratrol treatment increased I-2 threonine phosphorylation at<br />

the site phosphorylated by GSK3beta <strong>and</strong> decreased the phosphorylated GSK3beta-Ser9<br />

levels indicative of kinase activation. All these events needed a deacetylase intact Sirt1. Their<br />

importance is evident by the fact that treatment with phosphatase inhibitor okadaic acid <strong>and</strong><br />

with the GSK3beta inhibitor SB-415286 abolished resveratrol’s suppressive action on<br />

forskolin-induced CREB phosphorylation levels. In summary, the present study shows that<br />

the energy sensor Sirt1 regulates GH synthesis by activating GSK3beta <strong>and</strong> PP1 <strong>and</strong><br />

suppressing CREB transcriptional activity. This may be a novel mechanism through which<br />

pituitary somatotroph cells sense changes in energy levels <strong>and</strong> subsequently adjust GH<br />

synthesis to meet the metabolic needs of the organism.<br />

104


Session 3: Transcriptional control Poster 8<br />

Stress- <strong>and</strong> nutrition-sensing transcription factors: looking for biomarkers in<br />

atherosclerosis<br />

Fabrice Tolle, Amit Kumar, Thomas Sauter, Jean-Luc Bueb.<br />

<strong>Inflammation</strong> group/Systems biology group; Life Sciences Research Unit; Faculty of<br />

Science, Technology <strong>and</strong> Communication; University of Luxembourg, 162A, Avenue de<br />

la Faïencerie,<br />

L-1511 Luxembourg - Contact: fabrice.tolle@uni.lu<br />

A misbalance of functional interactions between nutrient-sensing nuclear receptors (e.g.<br />

Vitamin D receptor, VDR) <strong>and</strong> stress-sensing transcription factors (e.g. NF-kappaB) induced<br />

by lifetime exposure to micronutrients <strong>and</strong> cytokines, may be a central molecular process<br />

towards atherosclerosis. Our objectives are to highlight potential interactions between VDR<br />

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

NF-kappaB signalling in vascular endothelial cells, the first cell population implicated in this<br />

disease. Microarray analyses were done on total RNA which was extracted from EA.hy926<br />

cells (human umbilical vein endothelial cells line) after stimulation with TNFalpha <strong>and</strong>/or<br />

1alpha25(OH)2 vitamin D3 (VD) up to 24 hours. The raw data were transformed <strong>and</strong><br />

normalised using Bioconductor software <strong>and</strong> then analysed using IPA Knowledge Base<br />

(Ingenuity) with a special focus on chemokines molecules. As expected, most of the CCL <strong>and</strong><br />

CXCL chemokine families are up-regulated by the TNFalpha treatment <strong>and</strong> interestingly,<br />

these up-regulations are decreased by VD. To confirm these results, we focused our attention<br />

on a group of genes that appear to be implicated in macrophage chemo-attraction (Chemokine<br />

(C-C motif) lig<strong>and</strong> 2, 4 <strong>and</strong> 5 (CCL2, 4 <strong>and</strong> 5)). The microarray data were verified <strong>and</strong><br />

confirmed by real time RT-PCR for CCL2, 4 <strong>and</strong> 5. Moreover, the effect of VD was<br />

abolished when the VDR expression was switched off by siRNA. The same observation was<br />

also done when cycloheximide was added to the TNFalpha <strong>and</strong> VD treatment. We confirmed<br />

on EA.hy926 cells that CCL2, 4 <strong>and</strong> 5 genes are up-regulated by TNFalpha. We showed that<br />

these up-regulations are decreased in presence of VD <strong>and</strong> dependent on VDR expression. The<br />

implication of VDR seems however to be indirect <strong>and</strong> to involve a de novo synthesis of<br />

protein(s), which we are now trying to identify, as well as their way of interaction with NFkappaB<br />

signalling <strong>and</strong> CCL2, 4 <strong>and</strong> 5 genes.<br />

105


Session 3: Transcriptional control Poster 9<br />

Control of the response to chemotherapy by the Nrf2-antioxidant response element<br />

signaling pathway<br />

Julien Verrax, Nicolas Dejeans, Raphaël Beck, Christophe Glorieux <strong>and</strong> Pedro Buc<br />

Calderon.<br />

Toxicology <strong>and</strong> Cancer Biology research group, Louvain Drug Research Institute,<br />

Université catholique de Louvain, Brussels, Belgium. Email :<br />

julien.verrax@uclouvain.be<br />

The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is a major regulator<br />

of the cellular antioxidant response. Nrf2, which is activated in response to either electrophilic<br />

or oxidative stress, controls detoxifying enzymes <strong>and</strong> transporters that protect cells from<br />

reactive oxygen species (ROS) <strong>and</strong> toxic chemicals.<br />

Although Nrf2 plays an important cytoprotective role in healthy individuals, mutations of the<br />

genes encoding for the components of its regulatory pathway can lead to constitutive<br />

activation of Nrf2, a phenomenon that is suspected to promote the resistance of cancer cells to<br />

chemotherapy.<br />

We have therefore explored the putative impact of Nrf2 activation on the efficacy of different<br />

chemotherapies. Our results show that Nrf2 activation (achieved by either using chemical<br />

inducers or Keap1 silencing) leads to the up-regulation of several antioxidant genes <strong>and</strong> a<br />

two-fold increase in glutathione content. Cancer cells in which Nrf2 was activated were also<br />

more resistant to both alkylating <strong>and</strong> intercalating agents. Taken together, our results support<br />

the view that Nrf2 can be either a friend or a foe, depending on the context.<br />

J.Verrax is a FNRS postdoctoral researcher.<br />

106


Posters are classified by session<br />

<strong>and</strong> then in alphabetical order (PRESENTING AUTHOR)<br />

(Late breaking abstracts are at the end of the <strong>book</strong>)<br />

Session 4: Immunology<br />

107


Session 4: Immunology Poster 1<br />

Detection of extracellular matrix proteins <strong>and</strong> apoptotic cells in varicose veins wall of<br />

patients of different age groups<br />

Marina Aunapuu, Andres Arend<br />

Department of Anatomy, University of Tartu Ravila 19, 50411 Tartu, Estonia; e-mail:<br />

marina.aunapuu@ut.ee<br />

Varicose veins of lower extremities represent the most common pathology of peripheral blood<br />

vessels. Recently, in connection with the introduction of new immunological, molecular<br />

biological <strong>and</strong> histological methods, the underst<strong>and</strong>ing of the mechanism of progression of<br />

varicose vein disease has significantly improved. The aim of our study was to investigate<br />

morphological changes <strong>and</strong> differences in the protein expression of collagen I, II, IV <strong>and</strong><br />

laminin in the wall of great saphenous vein from male <strong>and</strong> female patients of different age. In<br />

addition, apoptotic endothelial <strong>and</strong> smooth muscle cells were detected. The patients were<br />

divided into 3 age groups – younger than 35 years (Group I, 8 males <strong>and</strong> 28 females), from 36<br />

to 50 years (Group II; 12 males <strong>and</strong> 35 females) <strong>and</strong> older than 50 years (Group III; 5 males<br />

<strong>and</strong> 31 females). Collagen I, II, IV <strong>and</strong> laminin were determined by means of<br />

immunohistochemistry, apoptotic cells were detected by the transferase-mediated dUTP nickend<br />

labeling (TUNEL) method using In Situ Cell Death Detection Kit (Roche Diagnostics,<br />

Roche, Germany). In most patients veins were stretched out with highly varying wall<br />

thickness. The amount of connective tissue was increased. The immunohistochemistry<br />

showed that expression of laminin was increased in varicose veins media <strong>and</strong> adventitia,<br />

especially in III group female patients. The expression of collagen IV in all layers of varicose<br />

veins wall <strong>and</strong> collagen II in the adventitia of older female patients was higher compared with<br />

younger patients. In the expression of collagen I differences between male <strong>and</strong> female patients<br />

groups were found. The percentage of apoptotic endothelial <strong>and</strong> smooth muscle cells<br />

increased in the female group III compared with younger patients, but in case of male patients<br />

no differences between age groups were found. The results demonstrate that morphological<br />

alterations in varicose veins become more pronounced with advancing age, especially in the<br />

female patients.<br />

108


Session 4: Immunology Poster 2<br />

Immunophenotyping of mammary gl<strong>and</strong> cells in an acute mouse mastitis model<br />

Koen BREYNE 1, Dieter DEMON 1, Kristel DEMEYERE 1, <strong>and</strong> Evelyne MEYER 1<br />

1 Laboratory of Biochemistry, Department of Pharmacology, Toxicology <strong>and</strong><br />

Biochemistry, Ghent University, 9820 Merelbeke, Belgium, Koen.Breyne@Ugent.be,<br />

Dieter.Demon@Ugent.be, Kristel.Demeyere@Ugent.be, Evelyne.Meyer@Ugent.be<br />

Mammary gl<strong>and</strong> infection (mastitis) is one of the major concerns for the dairy industry. A<br />

variety of bacteria can colonize the milk-producing alveoli <strong>and</strong> provoke local <strong>and</strong> systemic<br />

inflammation. Different immune <strong>and</strong> non-immune mammary cells participate in the innate<br />

defense against invading pathogens by the secretion of specific cytokines <strong>and</strong> bactericidal<br />

molecules. However, the contribution of each cell type in clearance of the bacteria remains<br />

largely unknown. To address the participation of the key cell types in mastitis, we propose<br />

flow cytometric identification of neutrophils, macrophages <strong>and</strong> epithelial cells. As an initial<br />

approach, we compare Escherichia coli (E.coli)-infected mouse mammary gl<strong>and</strong>s with<br />

lactating <strong>and</strong> involuting gl<strong>and</strong>s.<br />

For the acute E. coli-induced mouse mastitis model, 12 to 14 days lactating wild type<br />

C57BL/6 mice were intraductally inoculated with bovine mastitis isolate E. coli P4:O32 in the<br />

right mammary gl<strong>and</strong> of the 4 th pair (mastitis gl<strong>and</strong>) <strong>and</strong> with phosphate buffered saline (PBS)<br />

in the contralateral gl<strong>and</strong> (involuting control gl<strong>and</strong>). Mammary gl<strong>and</strong>s were isolated either<br />

from 12 to 14 days lactating females (lactating gl<strong>and</strong>) or at about 20h post-infection <strong>and</strong><br />

manually minced in small pieces. The remaining clumps were enzymatically digested <strong>and</strong><br />

filtered to obtain a single cell suspension. For multi-parametric flow cytometric analysis, cells<br />

were stained with a panel of fluorochrome-conjugated monoclonal antibodies. In addition to<br />

their forward <strong>and</strong> side scatter characteristics, leukocytes <strong>and</strong> epithelial cells were identified by<br />

the expression of the cell surface antigens CD45 <strong>and</strong> CD24, respectively. Leukocytes were<br />

subdivided in alveolar neutrophils <strong>and</strong> macrophages based on the expression of ly6G or<br />

F4/80, respectively. Myoepithelial <strong>and</strong> luminal epithelial cells were gated according to their<br />

difference in CD24 expression, as previously described.<br />

Our results indicate a significantly higher influx of neutrophils (Ly6G + /CD45 + ) in the mastitis<br />

gl<strong>and</strong> than in both the involuting control <strong>and</strong> fully lactating gl<strong>and</strong>. As expected, the lactating<br />

gl<strong>and</strong>s contain the most epithelial cells (CD24 + /CD45 - ). The proportion of macrophages<br />

(F4/80 + /CD45 + ) in the three different conditions is not significantly altered. Microscopic<br />

evaluation after sorting of CD45 + /Ly6G + <strong>and</strong> CD45 + /F4/80 + -cells confirmed that these sorted<br />

populations only consists of neutrophils <strong>and</strong> macrophages, respectively.<br />

In conclusion, a set of cell type-specific markers enabled the immunophenotyping of different<br />

cell populations in the infected murine mammary gl<strong>and</strong>. We confirmed the diapedesis of<br />

neutrophils in response to an intramammary E. coli invasion <strong>and</strong> the reduction of epithelial<br />

cells in the mammary gl<strong>and</strong> during early involution to decrease the milk production. In the<br />

future, this elegant tool will be used to elucidate the participation of each cell type in relevant<br />

knock-out mice or infections with diverse bacterial strains.<br />

109


Session 4: Immunology Poster 3<br />

The adaptor Lnk (SH2B3) is a key regulator of endothelial cell signaling <strong>and</strong> a potential<br />

molecular target to control vascular injury<br />

Julie DEVALLIERE, Juliette FITAU, Mathias CHATELAIS, Nathalie GERARD <strong>and</strong><br />

Béatrice CHARREAU<br />

INSERM UMR 643 <strong>and</strong> ITUN, Nantes University Hospital, University of Nantes, France<br />

Lnk (SH2B3) belongs to a family of SH2-containing proteins <strong>and</strong> is a signaling adaptor<br />

molecule that plays regulatory functions in the homeostasis <strong>and</strong> proliferation of immune cells.<br />

We previously showed that Lnk is expressed in endothelial cells (EC) <strong>and</strong> phosphorylated in<br />

response to TNF (1, 2). We further demonstrated that, in human EC, Lnk is a negative<br />

regulator of TNF signaling that inhibits CAM (E-selectin <strong>and</strong> VCAM-1) expression in<br />

response to TNF (3). Mechanistically, NFkappaB/p65 <strong>and</strong> IkappaBalpha phosphorylation<br />

were unchanged, suggesting that Lnk may not modulate canonical NF!B activity. However,<br />

Lnk activates PI3-kinase through Akt phosphorylation. Sustained Lnk-mediated activation of<br />

PI3-kinase in TNF"-activated ECs correlated with the inhibition of ERK1/2 phosphorylation,<br />

while phosphorylation of p38 <strong>and</strong> JNK MAPKs was unchanged. ERK1/2 inhibition decreases<br />

VCAM-1 expression in TNF"-treated ECs. In the present study we show that integrin<br />

ligation, using anti-CD29 Abs, also promotes Lnk activation <strong>and</strong> regulation in EC whereas<br />

integrin-mediated signaling (Akt <strong>and</strong> GSK3# phosphorylation) consecutive to CD29 ligation<br />

is abrogated in EC from Lnk-/- mice. We found that Lnk binds to ILK <strong>and</strong> activates FAK <strong>and</strong><br />

paxillin. Overall, Lnk causes F-actin stress fibers, cytoskeleton reorganization <strong>and</strong> increases<br />

both the number <strong>and</strong> density of focal adhesions. Functionally, Lnk expression dramatically<br />

increases EC adhesion, slows down EC migration <strong>and</strong> inhibits apoptosis induced by anoïkis.<br />

Transfection of mutants <strong>and</strong> SiRNA identified alpha-parvin as a molecular partner of Lnk<br />

mediating the negative regulation of EC migration (4). Thus, the adaptor Lnk is an effective<br />

key regulator of inflammatory- <strong>and</strong> integrin-signaling controlling EC activation, migration<br />

<strong>and</strong> death, all critical in vascular remodeling <strong>and</strong> regeneration. Our current work in animal<br />

models also proposes to modulate Lnk in graft EC to provide a cytoprotective signal in organ<br />

transplantation (5). (1) Boulday G. et al., Circ. Res. (2001) 2;88:430; (2) Boulday et al.<br />

Transplantation (2002) 4(9):1352; (3) Fitau J. et al., J. Biol. Chem. (2006), 281:20148; (4)<br />

Devallière J. et al. (2010) Submitted ; (5) Chatelais M. et al. (2010) in revision.<br />

110


Session 4: Immunology Poster 4<br />

Immunostimulatory <strong>and</strong> anti-tumor effects of 22-nt double str<strong>and</strong>ed RNA<br />

L.V.Kovtonyuk, T.O.Kabilova, E.I. Ryabchikova, N.A. Popova, V.P. Nikolin, V.I.<br />

Kaledin, M.A. Zenkova, V.V. Vlassov, E.L. Chernolovskaya<br />

Institute of Chemical Biology <strong>and</strong> Fundamental Medicine, SB RAS, Novosibirsk, Russia.<br />

Institute of Cytology <strong>and</strong> Genetics, SB RAS, Novosibirsk, Russia.<br />

We identified a set of immunostimulatory 22 b.p. double-str<strong>and</strong>ed RNAs (isRNAs) with<br />

original sequence, which has no specific homology with human mRNAs. In vitro experiments<br />

showed that enzymatically synthesized isRNA effectively inhibit growth of human cancer<br />

cells (KB-3-1, SK-N-MC, HEK 293). We found that the pretreatment of KB-3-1 with 2aminopurine<br />

(inhibitor of dsRNA-binding protein kinase R (PKR) ) abolished the<br />

antiproliferative activity of isRNA-U/A. These results indicate the involvement of PKRmediated<br />

pathway in isRNA intracellular signaling in carcinoma KB-3-1 cells. Transfection<br />

of isRNAs induce synthesis of IFN-! <strong>and</strong> pro-inflammatory cytokines IL-6 <strong>and</strong> TNF-! in<br />

adherent mononuclear cells from human peripheral blood. To determine the<br />

immunostimulatory consensus motif in the studied isRNA we investigated antiproliferative<br />

<strong>and</strong> immunostimulatory activities of isRNA with different substitutions in the sequence. We<br />

have found that introduction of mismatches in the middle part of isRNA sequence (10 – 16<br />

positions) do not decrease significantly its antiproliferative activity, while mismatches at the<br />

3’-end of isRNA sequence substantially reduce antiproliferative action of investigated<br />

isRNAs.<br />

In vivo investigation of immunostimulatory properties of the studied isRNAs revealed that<br />

single intravenous injection of isRNA in complex with transfection reagent (Lipofectamine TM<br />

2000) increased the level of IFN-!, but did not change significantly the level of proinflammatory<br />

cytokines IL-6 <strong>and</strong> TNF-! in mice blood serum.<br />

We found, that isRNAs (3 injections, 10 "g per each mouse) effectively inhibited metastasis<br />

spreading of melanoma in C57BL/6-mice: the average number of the metastasis in the lungs<br />

of treated animals was 5 times lower than those in untreated animals. The data demonstrate<br />

that isRNAs caused reliable 25% inhibition of tumor growth of hepatoma in CBA-mice.<br />

Microscopic analysis of liver cross sections also revealed antimetastatic effects of isRNAs on<br />

hepatoma: the number of the metastasis found in the livers of treated animals <strong>and</strong> their<br />

average size were significantly reduced.<br />

The obtained results revealed the pronounced immunostimulatory, anti-tumor <strong>and</strong><br />

antimetastatic properties of the studied isRNAs. This short double str<strong>and</strong>ed isRNAs can be<br />

evaluated as potential agents for the immunotherapy of oncological <strong>and</strong> viral diseases.<br />

This work was supported by RAS programs “Molecular <strong>and</strong> Cellular Biology” <strong>and</strong> “Basic<br />

sciences for medicine”, grant from SB RAS No. 41.<br />

111


Session 4: Immunology Poster 5<br />

Identification of new partners of the protein Nod2 <strong>and</strong> evaluation of their potential role.<br />

Aurore Lecat 1 , E. Di Valentin 1 , M. Fillet 2 , J. Piette 1 , Sylvie Legr<strong>and</strong> 1<br />

1. GIGA Research, Unit of Virology <strong>and</strong> Immunology, B34, University of Liege, Belgium<br />

– aurore.lecat@ulg.ac.be 2. GIGA Research, Unit of Chemical Chemistry, B34,<br />

University of Liege, Belgium<br />

The mammalian innate immune system has evolved to detect pathogens-associated<br />

molecular patterns (PAMPS). Recognition of PAMPS involves membrane-spanning proteins<br />

like the Toll-like receptors (TLR) <strong>and</strong> cytosolic proteins such as the recently identified NODlike<br />

receptors (NLR). Best studied are mutations in Nod2 that are linked to Crohn’disease<br />

(CD) <strong>and</strong> Blau syndrome. Nod2 was shown to sense the bacterial peptidoglycan subunit<br />

muramyl-dipeptide (MDP) <strong>and</strong> to subsequently mediate inflammatory responses in cells by<br />

activating the NF-kB <strong>and</strong> MAPKS signalling pathways.<br />

This project consists in the identification of new Nod2 partners <strong>and</strong> in the evaluation<br />

of their role in the Nod2 signalling pathway. We chose a proteomic strategy consisting in the<br />

purification of Nod2-containing complexes in stably Nod2-expressing HEK293 cells after<br />

MDP treatment or infection by Listeria monocytogenes followed by Nod2 partners<br />

identification by mass spectrometry.<br />

Among the c<strong>and</strong>idates, we focused on a Jun N-terminal kinase-binding protein, termed<br />

JNKBP1. This protein is ubiquitously expressed <strong>and</strong> contains twelve WD40 repeats. We<br />

checked interaction between Nod2 <strong>and</strong> JNKBP1 <strong>and</strong> showed that both the N-terminal CARDs<br />

<strong>and</strong> C-terminal LRRs cooperate in the wild type protein for efficient interaction with<br />

JNKBP1. SiRNA-mediated knockdown of endogenous JNKBP1 significantly upregulated the<br />

phosphorylation of IkB-alpha induced by MDP in stably Nod2-expressing HEK293 cells.<br />

These preliminary results suggest that JNKBP1, through Nod2 interaction, could act as a<br />

suppressor of Nod2-mediated NF-kB activation. In the context of the CD, where loss-offunction<br />

variants of Nod2 have been shown to play a causative role, a suppressor of Nod2<br />

signalling could be of special interest.<br />

We plan to determine the modulator effect of JNKBP1 on Nod2-mediated MAPKs activation<br />

as well as on target gene expression. The molecular mechanism by which JNKBP1<br />

modulates Nod2 signalling could be investigated. All of these experiments should be also<br />

performed in a more relevant cellular model expressing both endogenous proteins such as<br />

intestinal epithelial cells. Finally, we will determine whether JNKBP1 expression is<br />

modulated in pro-inflammatory conditions such as in the mucosa of CD patients.<br />

112


Session 4: Immunology Poster 6<br />

Identification of new partners of the protein Nod2 <strong>and</strong> evaluation of their potential role.<br />

Aurore Lecat 1 , E. Di Valentin 1 , M. Fillet 2 , J. Piette 1 , Sylvie Legr<strong>and</strong> 1<br />

1. GIGA Research, Unit of Virology <strong>and</strong> Immunology, B34, University of Liege, Belgium<br />

– aurore.lecat@ulg.ac.be 2. GIGA Research, Unit of Chemical Chemistry, B34,<br />

University of Liege, Belgium<br />

The mammalian innate immune system has evolved to detect pathogens-associated<br />

molecular patterns (PAMPS). Recognition of PAMPS involves membrane-spanning proteins<br />

like the Toll-like receptors (TLR) <strong>and</strong> cytosolic proteins such as the recently identified NODlike<br />

receptors (NLR). Best studied are mutations in Nod2 that are linked to Crohn’disease<br />

(CD) <strong>and</strong> Blau syndrome. Nod2 was shown to sense the bacterial peptidoglycan subunit<br />

muramyl-dipeptide (MDP) <strong>and</strong> to subsequently mediate inflammatory responses in cells by<br />

activating the NF-kB <strong>and</strong> MAPKS signalling pathways.<br />

This project consists in the identification of new Nod2 partners <strong>and</strong> in the evaluation<br />

of their role in the Nod2 signalling pathway. We chose a proteomic strategy consisting in the<br />

purification of Nod2-containing complexes in stably Nod2-expressing HEK293 cells after<br />

MDP treatment or infection by Listeria monocytogenes followed by Nod2 partners<br />

identification by mass spectrometry.<br />

Among the c<strong>and</strong>idates, we focused on a Jun N-terminal kinase-binding protein, termed<br />

JNKBP1. This protein is ubiquitously expressed <strong>and</strong> contains twelve WD40 repeats. We<br />

checked interaction between Nod2 <strong>and</strong> JNKBP1 <strong>and</strong> showed that both the N-terminal CARDs<br />

<strong>and</strong> C-terminal LRRs cooperate in the wild type protein for efficient interaction with<br />

JNKBP1. SiRNA-mediated knockdown of endogenous JNKBP1 significantly upregulated the<br />

phosphorylation of IkB-alpha induced by MDP in stably Nod2-expressing HEK293 cells.<br />

These preliminary results suggest that JNKBP1, through Nod2 interaction, could act as a<br />

suppressor of Nod2-mediated NF-kB activation. In the context of the CD, where loss-offunction<br />

variants of Nod2 have been shown to play a causative role, a suppressor of Nod2<br />

signalling could be of special interest.<br />

We plan to determine the modulator effect of JNKBP1 on Nod2-mediated MAPKs activation<br />

as well as on target gene expression. The molecular mechanism by which JNKBP1<br />

modulates Nod2 signalling could be investigated. All of these experiments should be also<br />

performed in a more relevant cellular model expressing both endogenous proteins such as<br />

intestinal epithelial cells. Finally, we will determine whether JNKBP1 expression is<br />

modulated in pro-inflammatory conditions such as in the mucosa of CD patients.<br />

113


Session 4: Immunology Poster 7<br />

Protective Effects of Korean Medicinal Herbs against Lipopolysaccharide- <strong>and</strong><br />

Dinitrochlorobenzene-induced Pro-inflammatory Responses in HaCaT Cells <strong>and</strong><br />

NC/Nga Mice<br />

Chan Lee, En-Joo Lee, <strong>and</strong> Chan-Ik Park<br />

Department of Cosmeceutical Science, Daegu Haany University, Gyeongsangbuk-do<br />

712-715, S. Korea (E-mail : cipark@dhu.ac.kr)<br />

Atopic dermatitis (AD) is a complex skin disorder accompanied by severe itching <strong>and</strong><br />

inflammation with frequently repeated episodes. The purpose of this study is to search for<br />

naturally occurring medicinal herbs which can protect against lipopolysaccharide (LPS)induced<br />

pro-inflammatory responses in HaCaT cells <strong>and</strong> improve dinitrochlorobenzene<br />

(DNCB)-induced AD-like skin lesions in NC/Nga Mice. Houttuynia Cordata (HC), Scutella<br />

Baicalensis (SB), <strong>and</strong> Coptis Chinensis (CC) extracts inhibited LPS-induced expression of<br />

inducible nitric oxide synthase (iNOS) <strong>and</strong> cyclooxygenase-2 (COX-2) <strong>and</strong> subsequent<br />

generation of nitric oxide (NO) <strong>and</strong> prostagl<strong>and</strong>in E2 (PGE2). In another experiments, topical<br />

application of HC, SB, <strong>and</strong> CC extracts to NC/Nga mice suppressed the development of<br />

scratching <strong>and</strong> AD-like dermatitis. Moreover, expression of inflammatory enzymes,<br />

production of pro-inflammatory mediators, activation of mast cells, <strong>and</strong> elevated serum levels<br />

of IgE were effectively attenuated by HC, SB, <strong>and</strong> CC extracts. These findings suggest that<br />

Korean medicinal herbs including HC, SB, <strong>and</strong> CC may have preventive <strong>and</strong> therapeutic<br />

potential against inflammatory skin damages in AD.<br />

Keywords : Houttuynia Cordata, Scutella Baicalensis, Coptis Chinensis,<br />

atopic dermatitis, inflammation<br />

114


Session 4: Immunology Poster 8<br />

Small GTPase RJL promotes breast cancer progression by inducing expansion <strong>and</strong><br />

accumulation of MDSCs via IL-6 <strong>and</strong> PGE2<br />

Qiuyan Liu, Taoyong Chen, Chaoxiong Zhang, <strong>and</strong> Xuetao Cao<br />

Institute of Immunology <strong>and</strong> National Key Laboratory of Medical Immunology, Second<br />

Military Medical University, Shanghai 200433, China, Email:lqy1969@yahoo.com.cn<br />

The expansion <strong>and</strong> accumulation of myeloid-derived suppressor cells (MDSCs) have been<br />

found in cancer patients <strong>and</strong> tumor-bearing mice, which are potent suppressors of adaptive<br />

<strong>and</strong> innate immunity <strong>and</strong> can promote tumor progression. RJLs are a family of Ras-related<br />

GTP-binding proteins characterized by the N-terminal GTP-binding domain (small GTPase<br />

domain) <strong>and</strong> the C-terminal J domain with unknown functions. Here, we report that RJL is<br />

overexpressed in most of the tumor cells <strong>and</strong> tumor tissues, <strong>and</strong> RJL overexpression promotes<br />

breast cancer growth, invasion <strong>and</strong> metastasis in vitro <strong>and</strong> in vivo. More expansion <strong>and</strong><br />

accumulation of MDSCs is found in RJL-overexpressing 4T1 tumor-bearing mice.<br />

Furthermore, RJL overexpression induces 4T1 cancer cells to produce more proinflammatory<br />

factor IL-6 <strong>and</strong> PGE2 by activating p38 MAPK, PI-3K/Akt, <strong>and</strong> NF!B signal pathways.<br />

Accordingly, silencing of RJL expression significantly decreases breast cancer progression<br />

<strong>and</strong> reduces MDSCs expansion <strong>and</strong> accumulation. Administration of COX2 specific inhibitor<br />

SC58125 can significantly reduce MDSCs expansion <strong>and</strong> accumulation in spleen, <strong>and</strong> tumor<br />

tissues, <strong>and</strong> administration of anti-IL-6 neutralization antibody reduces MDSCs accumulation<br />

in tumor tissue. Therefore, our results demonstrate that RJL can induce IL-6 <strong>and</strong> PGE2<br />

production, which contributes to the RJL-induced MDSCs expansion <strong>and</strong> recruitment<br />

promoting breast cancer progression. The results suggest that RJL maybe a novel oncogenic<br />

therapeutic target.<br />

Keywords:RJL; Myeloid-derived suppressor cells; IL-6; PGE2; immune escape<br />

This work was supported by grants from the National Natural Science Foundation of China<br />

(30771984, 30972688)<br />

115


Session 4: Immunology Poster 9<br />

Premature aging of immune system: regulatory T cells as a therapeutic target<br />

Alex<strong>and</strong>er Pukhalsky 1,2 , Galina Shmarina 1,2 <strong>and</strong> Vladimir Alioshkin 2<br />

1 Research Centre for Medical Genetics, Moscow 115478, 2 Gabrichevsky Institute of<br />

Epidemilogy <strong>and</strong> Microbilogy, Moscow 125212, Russia. E-mail: osugariver@yahoo.com<br />

It is well known that the brain <strong>and</strong> immune system are the two principal adaptive systems in the body,<br />

which permanently swap for the signals. Two major pathways are involved in such interaction:<br />

hypothalamic-pituitary-adrenal (HPA) axis <strong>and</strong> the sympathetic nervous system. During the immune<br />

response glucocorticoids (GCs) <strong>and</strong> catecholamines, the major stress hormones stimulate a negative<br />

feedback mechanism, which protects the organism from a surplus activity of pro-inflammatory<br />

cytokines <strong>and</strong> other products with tissue-damaging potential. Simultaneously regulatory T cells<br />

(Tregs) lose their suppressive activity. As immune response develops Treg number increases <strong>and</strong> their<br />

activity is restored. Augmentation of active Treg number results in gradual attenuation of the immune<br />

response. Normally, both mechanisms of inflammation control prompt (GCs <strong>and</strong> catecholamines) <strong>and</strong><br />

delayed (Tregs), are well equalized. However, the balance may be disturbed by age due to repeated<br />

episodes of stress <strong>and</strong> HPA axis activation. In time HPA axis activation replaces by its depletion <strong>and</strong><br />

Tregs become a principal mechanism of anti-inflammatory machinery. This mode of regulation of<br />

inflammatory reaction being more slow <strong>and</strong> rough does not allow to fine tune immune system <strong>and</strong><br />

each new environmental challenge promotes further Treg accumulation that leads to stable<br />

immunosuppression. Permanent stress <strong>and</strong>/or severe systemic disease may accelerate such aging<br />

process. Different methods of Treg depletion have been proposed. In our opinion alkylating drugs<br />

(ADs) are most promising among them. ADs belonging to nitrogen mustard family are commonly<br />

used as cytostatic <strong>and</strong> immunosuppressive agents. The effect of the drugs is mainly associated with<br />

cross-linking of DNA double str<strong>and</strong>s <strong>and</strong>, at higher concentrations, with induction of DNA str<strong>and</strong><br />

breaks. Although DNA is not a unique target for alkylation in the cell the others do not play any role<br />

in the cytostatic effect realization if the drug is used at a DNA-altering dose. However, when the dose<br />

is gradually decreased, the number of targets for alkylation will also be reduced. Studies in in vitro<br />

lymphocyte proliferation model show that the scenario varies with the ADs concentration decrease. If<br />

the concentration of ADs is high (100 µg/ml or more), cells die within few hours due to irreversible<br />

DNA damage. If the concentrations of ADs vary in the range from 30 to 100 µg/ml numerous sites of<br />

DNA are also alkylated, but DNA damaged segments restored during DNA repair. Nevertheless, the<br />

affected cells are died due to apoptosis induction. Moderate concentrations of ADs (1-10 µg/ml) do<br />

not kill the cells but prevent IL-2 production making the cells resistant to proliferative stimuli. Ultralow<br />

concentrations of ADs (0.3 µg/ml <strong>and</strong> lower) selectively inhibit Tregs due to disruption of the<br />

signal transduction by IL-2R. Among a large variety of T cell subsets only Tregs constitutively<br />

express high affinity receptor for IL-2, the cytokine, which is the factor of their growth <strong>and</strong> survival.<br />

IL-2R is not a unique receptor, which may be blocked with ADs. In addition similar effect has been<br />

shown for at least two other surface receptors: TNFR <strong>and</strong> Fas. So, ADs have a unique capacity for<br />

selective elimination of Tregs. Such elimination may be achieved with low doses of the drug, which<br />

do not affect other cell subsets. Optimal effect may be obtained when ADs are used in the form of<br />

pulse-therapy: repeated dose after one month recovery. The concept of such therapy may be<br />

formulated as “hit <strong>and</strong> run away”.<br />

116


Session 4: Immunology Poster 10<br />

INJURY INDUCED INFLAMMASOME ACTIVATION LEADING TO IL-1b AND IL-<br />

17 MEDIATED LUNG INFLAMMATION IS IL-22 INDEPENDENT<br />

Valérie Quesniaux, Isabelle Couillin, François Erard, Bernhard Ryffel<br />

UMR6218, CNRS <strong>and</strong> University, UMR6218 Orléans, France<br />

To underst<strong>and</strong> the molecular mechanisms of acute lung injury induced inflammation <strong>and</strong><br />

fibrosis we investigated <strong>and</strong> found a critical role for signaling though the IL-1 receptor 1 (IL-<br />

1R1) (Gasse et al 2007, 2009). Here we show that bleomycin induced injury results in<br />

expression of IL-1b, IL-23, IL-17 <strong>and</strong> IL-22 expression in the lung which depends on the<br />

activation of the NLPR3 inflammasome complex. To test the role of IL-17 <strong>and</strong> IL-22 in the<br />

pathogenesis we used neutralizing antibodies <strong>and</strong> gene deficient mice. We report here that IL-<br />

17A, but not IL-22, is critical to develop lung inflammation <strong>and</strong> fibrosis. The source of IL-17<br />

<strong>and</strong> IL-22 in the lung <strong>and</strong> the molecular pathways are under investigations.<br />

Therefore the data suggest that injury induced inflammasome activation leads to the activation<br />

the activation ofIL-17 while IL-22 is not involved in this process.<br />

117


Session 4: Immunology Poster 11<br />

T cell signaling in the development <strong>and</strong> treatment of cognitive disorders in mice<br />

Galina Shmarina 1,2 , Alex<strong>and</strong>er Pukhalsky 1,2 , <strong>and</strong> Vladimir Alioshkin 2<br />

1 Research Centre for Medical Genetics, Moscow 115478, 2 Gabrichevsky Institute of<br />

Epidemilogy <strong>and</strong> Microbilogy, Moscow 125212, Russia. E-mail: osugariver@yahoo.com<br />

Surplus accumulation of regulatory T cells (Tregs) is known to be at the bottom of many<br />

morbid conditions among them being neuropsychiatric diseases. In particular, Tregs may<br />

inhibit Th1 cells, including brain autoimmune lymphocytes, controlling the local microglial<br />

response <strong>and</strong> brain tissue homeostasis. CNS-specific T cells were also found to be required<br />

for special learning <strong>and</strong> memory <strong>and</strong> for the expression of brain-derived neurotrophic factors.<br />

Malfunction of these cells leads to the appearance of neurodegenerative foci. The present<br />

study was undertaken in an attempt to suggest a novel approach for the treatment of<br />

maladaptation to mental stress associated with excessive Treg accumulation. Recently it was<br />

shown that alkylating agents in the dose 100 fold lower than cytostatic one are capable to<br />

disturb signal transduction by various cell surface receptors, such as IL-2R, TNFR <strong>and</strong> Fas.<br />

Molecular mechanisms of the receptor blockage were investigated on the model of TNF<br />

signaling. We succeeded in demonstration that low concentrations of alkylating agent<br />

melphalan (Mp) protect murine fibroblastoid cells against TNF!-induced cytotoxicity. The<br />

protection did not depend on de novo protein synthesis. Moreover, no increase in NF-kB in<br />

nuclear extracts of Mp-treated cells was observed. At the same time, 1-hour treatment with<br />

Mp markedly reduced NF-kB activity in nuclear extracts of the cells challenged with TNF!.<br />

These data support the suggestion that specific alkylation of components in the cytoplasm or<br />

cell membrane by Mp interferes with surface receptor signaling pathway. As Tregs are a<br />

unique lymphocyte subset, which permanently expresses high affinity receptor for IL-2 (the<br />

cytokine, which is the factor of their growth <strong>and</strong> survival), low Mp concentrations might<br />

selectively eliminate these cells without affecting other T cell subsets. The animal model of<br />

Treg accumulation has been used. BALB/c mice were chronically treated with dexamethasone<br />

<strong>and</strong> IL-2. Spatial learning/memory was assessed in the Morris Water Maze Behavioral Test. It<br />

was shown that the pharmacologically-induced Treg accumulation leads to cognitive <strong>and</strong><br />

behavioral abnormalities, which may be prevented by Mp administration. Indeed, already on<br />

the 2 nd day of training the percentage of Mp-treated mice, which were able to find a<br />

submerged platform, was significantly higher (p=0.04; Wilcoxon paired test) than in untreated<br />

animals after Treg induction. In conclusion, disturbance of IL-2 signaling with alkylating<br />

agents is promised to be a new safe method for the treatment of cognitive disorders.<br />

118


Session 4: Immunology Poster 12<br />

The level of inducible nitric oxide (NO) synthesis in immune system of mice as the aging<br />

risk marker.<br />

Svetlana V. Vasilieva<br />

Institute of Biochemical Physics RAS, Russia 119334, Moscow, 4 Kosygin str.<br />

svasilieva@polymer.chph.ras.ru<br />

Nitric oxide (NO) which generates in mammals by NO- synthases (NOSs) has been<br />

proven to possess cyto- <strong>and</strong> genotoxicity associated with the immune response, thus<br />

decreasing resistance of bacteria, viruses <strong>and</strong> other invasive organisms. The present study<br />

examines changes in NO levels induced in two organs of mice by LPS intra abdominal<br />

injection. The experiments have been performed using a pair of isogenic lines of mice of the<br />

same age of 5 months, differ by the single mutation in the “hairless” gene (hr mutation): B10<br />

(wild type) <strong>and</strong> "Rhino" mutant line with hr mutation. The mutant animals had some defects<br />

in immune system due to over expression of the hr gene. They characterized by a specific<br />

phenotype, suffered from erythroleukaemia <strong>and</strong> died normally at the age of 6-8 months. There<br />

is the information that the hr mutation impairs a DNA-repair capacity, as well. Additionally<br />

we studied mice of NZB line at the ages of 5 <strong>and</strong> 8,5 months which are used frequently as the<br />

model of autoimmune hemolytic anemia, developing with aging. To control the levels of NO<br />

generated the EPR spectroscopy has been used. Diethyl dithiocarbamate (DDC, “Sigma”)<br />

formed [Fe 2+ -DDC] complex with endogenous or exogenous iron when injected into the<br />

animals. Acting as NO scavenger this complex bound NO <strong>and</strong> gave rise to paramagnetic<br />

mononitrosyl iron complex (MNIC-DDC). This process is accompanied by the appearance of<br />

protein-bound mononuclear dinitrosyl -iron complexes with SH groups of proteins, which are<br />

characterized by the EPR signal with g!=0,35 <strong>and</strong> g"=2,02. While initially, until 2 months of<br />

age, the experimental mice resembled each other in phenotype <strong>and</strong> the levels of inducible NO<br />

were more or less the same. During aging the animals diverged significantly in phenotype <strong>and</strong><br />

differed greatly in NO levels from organ to organ. This was the case for the animals of NZB<br />

line at the ages of 5 <strong>and</strong> 8, 5 months when we observed a significant decreasing in NO levels<br />

in liver <strong>and</strong> intestines of the elder suffering animals. NO was found to accumulate in the<br />

organs of B10 mice (w.t.) - 4 <strong>and</strong> 10 fold over the initial levels in liver <strong>and</strong> intestines,<br />

respectively. But this was not the case for the isogenic Rhino line: only 0 <strong>and</strong> 3, 5 fold over<br />

the initial NO level in liver <strong>and</strong> intestines, respectively. These differences in NO inducible<br />

levels in liver <strong>and</strong> intestines were due to the single hr mutation. So, the level of inducible<br />

nitric oxide synthesis is a specific "aging risk marker" in mice.<br />

119


Posters are classified by session<br />

<strong>and</strong> then in alphabetical order (PRESENTING AUTHOR)<br />

(Late breaking abstracts are at the end of the <strong>book</strong>)<br />

Session 5: Proteomics<br />

120


Session 5: Proteomics Poster 1<br />

PhosphoSitePlus ® : a Resource for the Proteome-wide Study of Protein Modifications in<br />

Cancer<br />

Peter V. Hornbeck, Elzbieta Skrzypek, Bin Zhang, Jon M. Kornhauser, Beth L.<br />

Murray, Vaughan M. Latham, <strong>and</strong> Sasha Tkachev.<br />

Cell Signaling Technology, Danvers, MA 01923 phornbeck@cellsignal.com,<br />

eskrzypek@cellsignal.com, bzhang@cellsignal.com, jornhauser@cellsignal.com,<br />

bmurray@cellsignal.com, vlatham@cellsignal.com, <strong>and</strong> stkachev@cellsignal.com.<br />

PhosphoSitePlus ® (PSP) is an open, dynamic, continuously curated, <strong>and</strong> highly interactive<br />

systems biology resource for studying experimentally observed post-translational<br />

modifications (PTMs). PSP has recently exp<strong>and</strong>ed its content <strong>and</strong> interfaces to enable the<br />

systematic investigation of protein modifications associated with various cancers. PSP<br />

includes critical structural <strong>and</strong> functional information about the topology, biological function<br />

<strong>and</strong> regulatory significance of specific modification sites, <strong>and</strong> powerful tools for mining <strong>and</strong><br />

interpreting this data in the context of diseases, tissues, <strong>and</strong> cell lines. PSP integrates both<br />

low- <strong>and</strong> high-throughput (LTP <strong>and</strong> HTP) data sources into a single reliable <strong>and</strong><br />

comprehensive resource. PSP has recently exp<strong>and</strong>ed. Information in PSP is shared with the<br />

research community in multiple ways. For example, users can access specific information<br />

about a single protein <strong>and</strong> each of its modification sites, or browse <strong>and</strong> <strong>download</strong><br />

comprehensive datasets of modification sites observed in specified diseases, cell lines, <strong>and</strong><br />

tissues. Metaanalyses of protein modifications in non-small cell lung cancer <strong>and</strong> chronic<br />

myelogenous leukemia suggest that distinct molecular signatures of different cancers may be<br />

identified using PSP. It is our hope that PSP, by providing a reliable <strong>and</strong> powerful resource<br />

focused on the roles of protein modifications in biological control, will accelerate the pace of<br />

discovery of basic mechanisms of cellular signaling, further our underst<strong>and</strong>ing of cellular<br />

regulation in health <strong>and</strong> disease, <strong>and</strong> facilitate the discovery of critical disease biomarkers <strong>and</strong><br />

potential drug targets.<br />

121


Session 5: Proteomics Poster 2<br />

Automated identification of Bcl-2 homologues using structure-aided HMM framework<br />

Valentine Rech de Laval 1 , Christophe Combet 1 , Gilbert Deléage 1 , Abdel Aouacheria 2<br />

1 Unité Bases Moléculaires et Structurales des Systèmes Infectieux; UMR 5086 CNRS –<br />

UCBL – IBCP, IFR128 Biosciences Lyon-Gerl<strong>and</strong>, 7 passage du Vercors, 69367 Lyon<br />

Cedex 07, France ; FR 3302 ; e-mail: c.combet@ibcp.fr<br />

2 Laboratoire de Biologie Moléculaire de la Cellule of Ecole Normale Supérieure de<br />

Lyon, UMR 5239 CNRS – UCBL – ENS Lyon, IFR128 Biosciences Lyon-Gerl<strong>and</strong>, 46<br />

Allée d’Italie, 69364 Lyon Cedex 07, France ; e-mail: a.aouacheria@ens-lyon.fr<br />

The Bcl-2 family controls induction of apoptosis (programmed cell death) at the mitochondria<br />

via opposing functions of prosurvival <strong>and</strong> proapoptotic regulators. Members of this family are<br />

classified, based on the presence of one or more Bcl-2 Homology (BH) domains, as<br />

antiapoptotic molecules (four BH domains: BH1–4) (such as Bcl-2), proapoptotic homologs<br />

(two or three BH domains: BH1-3) (such as Bax) or BH3-only death proteins (only the BH3<br />

domain) (including Bid or Bim). However, recent data highlighted the extent of sequence<br />

variation between Bcl-2 family proteins, including at the level of the BH domains, pointing to<br />

the need for a computational redefinition of the family. This goal can be achieved by the<br />

development of alignment pro<strong>file</strong>s combining sequence <strong>and</strong> structure information, namely<br />

structure-based hidden Markov models (HMMs). Here, we present a set of HMM-pro<strong>file</strong>s<br />

specific for cellular <strong>and</strong>/or viral-coded proteins sharing the same structural fold as Bcl-2.<br />

These models were checked for sensitivity <strong>and</strong> compared to st<strong>and</strong>ard domain signatures for<br />

Bcl-2 family recognition. Our results indicate that these novel pro<strong>file</strong>s are useful in<br />

identifying cellular <strong>and</strong> viral Bcl-2 homologs, including those with low sequence similarity.<br />

Indeed, we cover all known homology groups <strong>and</strong> we extend them by finding proteins<br />

belonging to new homology groups. Analysis of the full set of known <strong>and</strong> novel protein<br />

sequences retrieved by the HMM pro<strong>file</strong>s will be used for automatic update of the Bcl-2<br />

Family Database (http://bcl2db.ibcp.fr/) <strong>and</strong> for integrated systems biology <strong>and</strong> phylogenetic<br />

approaches.<br />

122


Session 5: Proteomics Poster 3<br />

Proteomic Profiling of Human Melanoma Metastatic Cell Line Secretomes<br />

Micaela Rocco 1* , Livia Malorni 2* , Giuseppe Palmieri 3 , Carla Rozzo 3 , Augusto Parente 1<br />

<strong>and</strong> Angela Chambery 1 .<br />

1<br />

Dipartimento di Scienze della Vita, Via Vivaldi 43, Seconda Università di Napoli, I-<br />

81100 Caserta, Italy<br />

micaela.rocco@unina2.it; augusto.parente@unina2.it; angela.chambery@unina2.it<br />

2<br />

Proteomic <strong>and</strong> Biomolecular Mass Spectrometry Center, Institute of Food Science <strong>and</strong><br />

Technology, National Research Council (CNR), Via Roma 64, I-83100 Avellino, Italy<br />

lmalorni@isa.cnr.it<br />

3<br />

Unit of Cancer Genetics, Institute of Biomolecular Chemistry, National Research<br />

Council (CNR), Traversa La Crucca 3, Baldinca Li Punti, I-07100 Sassari, Italy<br />

palmierig@icb.cnr.it; carla.rozzo@icb.cnr.it<br />

* These Authors contributed equally to this work.<br />

During the last years the incidence <strong>and</strong> mortality of melanoma have rapidly increased.<br />

Metastatic spread of malignant melanoma is often associated to cancer progression with poor<br />

prognosis <strong>and</strong> survival. These processes are controlled by dynamic interactions between<br />

tumor melanocytes <strong>and</strong> neighbouring stromal cells, whose deregulation leads to the<br />

acquisition of cell proliferation capabilities <strong>and</strong> invasiveness. It is increasingly clear that a key<br />

role in carcinogenesis is played by secreted molecules either by tumor <strong>and</strong> surrounding<br />

stromal cells. To address the issue of the proteins secreted by different metastases of the same<br />

patient, the proteomic profiling of secretomes of human metastatic cell lines (i.e. PES 41 <strong>and</strong><br />

PES 47 from two subcutaneous metastases <strong>and</strong> PES 43 from a lung metastasis) was<br />

performed by applying a shotgun LC-MS/MS-based approach. The results provide a list of<br />

c<strong>and</strong>idates associated to the metastatic potential of PES human melanoma cell lines. Among<br />

them, several matricellular proteins previously reported as involved in melanoma<br />

aggressiveness were identified (i.e. SPARC, osteopontin, galectins). In addition, the<br />

extracellular matrix protein 1 that stimulates the proliferation <strong>and</strong> angiogenesis of endothelial<br />

cells <strong>and</strong> fibronectin that is involved in cell adhesion <strong>and</strong> motility were identified. The<br />

presented work provides the basis to clarify the complex extracellular protein networks<br />

implicated in human melanoma cell invasion, migration <strong>and</strong> motility.<br />

123


Session 5: Proteomics Poster 4<br />

Proteomic analysis of signaling processes induced in cultured glioblastoma cells after<br />

sub-lethal photodynamic treatment<br />

A. Uzdensky 1 , A. Juzeniene 2 , J. Moan 2<br />

1<br />

Southern Federal University, 194/1, Stachky ave., NIINK, Rostov-on-Don, 344090,<br />

Russia, auzd@y<strong>and</strong>ex.ru<br />

2<br />

Institute for Cancer Research, Montebello, N-0310, Oslo, Norway<br />

Photodynamic therapy (PDT) is currently used for killing of malignant cells in oncology. To<br />

characterize initial signaling processes involved in response of glioblastoma D54Mg cells to<br />

PDT treatment with 5-aminolevulinic acid, we used small PDT doses that killed


Posters are classified by session<br />

<strong>and</strong> then in alphabetical order (PRESENTING AUTHOR)<br />

(Late breaking abstracts are at the end of the <strong>book</strong>)<br />

Session 6: Epigenetics<br />

125


Session 6: Epigenetics Poster 1<br />

Effects of Lithium-Induced Cell Cycle Arrest on The Histone 3 Lysine 27 Methylation in<br />

Huh7, Hepatocellular Carcinoma Cell Line.<br />

Sanem TERCAN AVCI 1 , Ne!e ATABEY 1 , Esra ERDAL 1<br />

1 Dokuz Eylul University School of Medicine, Department of Medical Biology <strong>and</strong><br />

Genetics, 35340-Inciralti, Izmir/TURKEY<br />

Exposure of normal liver cell with a viral or chemical agent triggers telomere shortening<br />

associated with repetitive necrosis/proliferation rotation. It causes changes in<br />

microenvironment of liver, activation of stem cells <strong>and</strong>/or satellite cells, after that, activation<br />

of cirrhosis, displastic nodule <strong>and</strong> hepatocellular carcinoma (HCC) depending on the aberrant<br />

cell division <strong>and</strong> resistance to apoptosis. It was demonstrated that during<br />

hepatocarcinogenesis most of genes silenced by methylation on their promoter, even though<br />

the roles of histone code alterations have not been known yet.<br />

In our previous study, it has been shown that lithium cause G1 cell cycle arrest on Huh7 cells,<br />

HCC cell line <strong>and</strong> microarray analysis further demonstrated that transcripts of histone<br />

modifier enzymes changed significantly in lithium treated cells in compared to untreated ones<br />

(Erdal E., 2005). So, we hypothesized that histone modifications on the chromatin might<br />

control cell cycle of HCC cells. In this study, it has been first shown that expression of EZH2<br />

histone methyl transferase decreased in both transcript <strong>and</strong> protein level under the effect of<br />

lithium treatment in Huh7 cells <strong>and</strong> expression of JMJD3 histone demethylase increased in<br />

transcript level as expected. Since both enzymes are known as responsible for the regulation<br />

of histone 3 lysine 27 methylation, we inquired whether H3K27 is affected in lithium treated<br />

Huh7. Unexpectedly, level of H3K27me3 modification has been shown as significant<br />

increase. This result makes us thinking lithium affects by a different mechanism. Recently, it<br />

was shown that GSK3! phosphorylates EZH2 <strong>and</strong> phosphorylated form of EZH2 in<br />

Polycomb Repressive Complex 2 (PRC2) do not catalyzed H3K27 methylation. Since lithium<br />

is very well known inhibitor of GSK3!, we can explain the unexpected result regarding<br />

decline in H3K27me3 although EZH2 decreases in protein level. To test this hypothesis, we<br />

analyzed level of phosphorylated form of EZH2 under the treatment of lithium. It has been<br />

found that lithium causes decrease on the phosphorylated form of EZH2 at 2nd <strong>and</strong> 6th hours<br />

after treatment.<br />

As a conclusion, these analyses showed us the alterations of histone modification patterns,<br />

especially H3K27me3 modification, might affect cell proliferation on HCC.<br />

Key Words: HCC, histone modifications, EZH2<br />

126


Session 6: Epigenetics Poster 2<br />

Reexpression of the hic1 tumorsuppressor gene after pharmacological unmasking<br />

results in increased radio-sensitivity of head <strong>and</strong> neck squamous cell carcinoma<br />

Jürgen Brieger, Sylvia Mann, Wolf Mann<br />

Laboratory of Molecular Tumor Biology, Department of Otolaryngology, Head <strong>and</strong><br />

Neck Surgery, University Medical Center of the Johannes Gutenberg University, 55101<br />

Mainz, Germany, eMail: juergen.brieger@unimedizin-mainz.de<br />

Hypermethylation of the genome <strong>and</strong> especially of tumor suppressor genes have been<br />

recognized as causative for the generation of several types of tumours. Here we analysed 5-<br />

Azacytidin (5-Aza) for demethylation treatment of the tumor suppressor hic1<br />

(hypermethylated in cancer 1) <strong>and</strong> as a radio-sensitizing agent in head <strong>and</strong> neck squamous cell<br />

carcinoma (HNSCC). To this end we treated an established HNSCC cell line with different<br />

concentrations of 5-Aza for 72h, followed by an single irradiation with 4 or 50 Gy.<br />

Methylation status <strong>and</strong> re-expression of hic1 were analysed by methylation specific PCR,<br />

quantitative PCR <strong>and</strong> Western blot before <strong>and</strong> after treatment. Survival, apoptosis,<br />

proliferation, <strong>and</strong> migration were analysed as functional parameters. We found hic1<br />

frequently hypermethylated in HNSCC. After 5-Aza treatment hic1 expression was restored<br />

<strong>and</strong> cellular survival reduced. The combined application of 5-Aza <strong>and</strong> irradiation further<br />

decreased survival of the cells. Additionally, migration was impaired as well as apoptosis<br />

increased.<br />

The data show the relevance of hypermethylation for radiation resistance <strong>and</strong> suggest that the<br />

reactivation of relevant tumor suppressor genes by demethylating active drugs might be<br />

beneficial to overcome irradiation resistance.<br />

127


Session 6: Epigenetics Poster 3<br />

Polyarginine peptide nucleic acids inibit biological activity of microRNA 210<br />

Enrica Fabbri 1 , Nicoletta Bianchi 1 , Eleonora Brognara 1 ,Alessia Finotti 1 , Giulia<br />

Breveglieri 1 , Monica Borgatti 1 , Alex Manicardi 2 , Roberto Corradini 2 ,<br />

Rosangela Marchelli 2 <strong>and</strong> Roberto Gambari 1<br />

1 BioPharmaNet, Department of Biochemistry <strong>and</strong> Molecular Biology, Ferrara<br />

University, via Fossato di Mortara, 74, 44121, Ferrara, Italy, e-mail:gam@unife.it;<br />

2 Department of Organic Chemistry, Parma University, Parco Area delle Scienze, 17/A,<br />

43124 Parma, Italy.<br />

Peptide nucleic acids are DNA mimics extensively used for pharmacological regulation of<br />

gene expression in antisense <strong>and</strong> anti-gene therapies. At present, very few data are available<br />

on the use of PNAs as molecules targeting microRNAs. MicroRNAs are a family of small (19<br />

to 25 nucleotide in length) noncoding RNAs that regulate gene expression by sequenceselective<br />

targeting of mRNAs, leading to a translational repression or mRNA degradation,<br />

depending on the degree of complementarity between microRNAs <strong>and</strong> the target sequences.<br />

MicroRNAs are deeply involved in the control of highly regulated biological functions, such<br />

as differentiation, cell cycle <strong>and</strong> apoptosis. The aim of the present study was to determine the<br />

activity of a PNA conjugated to polyarginine peptide <strong>and</strong> designed against microRNA 210, a<br />

microRNA associated to hypoxia <strong>and</strong> involved in the erythroid differentiation modulation.<br />

Our studies demonstrated that this PNA is efficiently internalized within target cells <strong>and</strong><br />

strongly inhibits microRNA 210 activity with an alteration of the raptor <strong>and</strong> !-globin gene<br />

expression. Unlike commercially available antagomiRs, which need continous<br />

administrations, a single administration of this PNA (without using transfection reagents like<br />

lipofectin or lipofectamine) is sufficient to obtain these biological effects. Our results<br />

demostrate PNA-based molecules are very promising reagents to modulate the biological<br />

activity of micro RNAs.<br />

128


Session 6: Epigenetics Poster 4<br />

Dataset integration identifies transcriptional regulation of microRNA genes by PPARG<br />

in mouse adipogenesis<br />

Elisabeth John 1 , Anke Wienecke-Baldacchino 1 , Merja Heinäniemi 1 , Carsten Carlberg 1,2#<br />

<strong>and</strong> Lasse Sinkkonen 1<br />

1<br />

Life Sciences Research Unit, University of Luxembourg, L-1511 Luxembourg,<br />

Luxembourg<br />

2<br />

Department of Biosciences, University of Eastern Finl<strong>and</strong>, FIN-70211 Kuopio, Finl<strong>and</strong><br />

E-mail: elisabeth.john@uni.lu, anke.wienecke@uni.lu, merja.heinäniemi@uni.lu,<br />

carsten.carlberg@uni.lu, lasse.sinkkonen@uni.lu<br />

Peroxisome proliferator-activated receptor G (PPARG) is a key transcription factor in<br />

mammalian adipogenesis. Genome-wide approaches identified thous<strong>and</strong>s of PPARG binding<br />

sites in mature mouse 3T3-L1 adipocytes <strong>and</strong> PPARG up-regulates hundreds of proteincoding<br />

genes during adipogenesis. However, so far no microRNA (miRNA) genes were<br />

identified as primary PPARG targets. By integration of four separate datasets of genome-wide<br />

PPARG binding sites in 3T3-L1 adipocytes we identified 98 miRNA clusters with PPARG<br />

binding sites within 50 kb from miRNA gene transcription start sites. Nineteen of these<br />

putative PPARG-regulated mature miRNAs were up-regulated above 2-fold during 3T3-L1<br />

adipogenesis. Focusing on miRNA loci with PPARG binding sites confirmed by at least three<br />

datasets resulted in six high confidence miRNA target loci. The up-regulation of five miRNA<br />

genes miR-103-1 (host gene Pank3), miR-148b (Copz1), miR-182/96/183, miR-205 <strong>and</strong> miR-<br />

378 (Ppargc1b) followed that of Pparg. The PPARG dependence of four of these miRNA loci<br />

was demonstrated by PPARG knock-down. The loci of miR-103-1 (Pank3) <strong>and</strong> miR-378<br />

(Ppargc1b) were also responsive to the PPARG lig<strong>and</strong> rosiglitazone. Finally, chromatin<br />

immunoprecipitation analysis validated three in silico predicted PPARG binding sites at the<br />

miR-103-1 locus <strong>and</strong> two at the miR-378 locus. In conclusion, we identified 22 putative<br />

PPARG target miRNAs genes, showed the PPARG dependence of four of these genes <strong>and</strong><br />

demonstrated two as functional PPARG target genes in mouse adipogenesis.<br />

129


Session 6: Epigenetics Poster 5<br />

Histone deacetylases 1, 6 <strong>and</strong> 8 are critical for invasion in breast cancer<br />

Soon Young Park, Ji Ae Jun, Kang Jin Jeong, Hoi Jeong Heo, Jang Sihn Sohn, Hoi<br />

Young Lee, Chang Gyo Park, <strong>and</strong> Jaeku Kang<br />

Myunggok Medical Research Institute, College of Medicine, Konyang University,<br />

Daejeon 302-718, Republic of Korea; E-mail: jaeku@konyang.ac.kr<br />

Histone deacetylases (HDACs) are associated with the development <strong>and</strong> progression of<br />

cancer, but which HDAC isoforms play important roles in breast cancer metastasis is not<br />

known. Here, we identify the specific HDAC isoforms that are necessary for invasion <strong>and</strong>/or<br />

migration in human breast cancer cell lines. MDA-MB-231 cells were significantly more<br />

invasive <strong>and</strong> expressed higher levels of matrix metalloproteinase-9 (MMP-9) than MCF-7<br />

cells. We compared the expression of HDAC isoforms between MCF-7 <strong>and</strong> MDA-MB-231<br />

cells <strong>and</strong> found greater expression of HDAC4, 6 <strong>and</strong> 8 in MDA-MB-231 by RT-PCR <strong>and</strong><br />

Western blot analyses. In addition, apicidin, a histone deacetylase inhibitor, was shown to<br />

attenuate the invasion, migration <strong>and</strong> MMP-9 expression in MDA-MB-231 cells. Using<br />

specific siRNAs directed against HDAC1, 4, 6 <strong>and</strong> 8, we show that inhibition of HDAC1, 6<br />

<strong>and</strong> 8, but not HDAC4, are responsible for invasion <strong>and</strong> MMP-9 expression in MDA-MB-231<br />

cells. We analyzed the invasiveness of MCF-7 cells overexpressing HDAC1, 4, 6 or 8 <strong>and</strong><br />

found that overexpression of HDAC1, 6 or 8 increased invasion <strong>and</strong> MMP-9 expression. By<br />

developing HDAC isoforms as potential biomarkers for breast cancer metastasis, the present<br />

study can be extended to developing therapies for breast cancer invasion.<br />

130


Session 6: Epigenetics Poster 6<br />

Functional studies of CBP/TDG complexes reveal a close link between DNA repair,<br />

transcription, <strong>and</strong> epigenetic signaling.<br />

Hélène Léger1, Caroline Smet-Nocca2, Sebastian Eilebrecht1, <strong>and</strong> Arndt Benecke1<br />

1 Institut des Hautes Études Scientifiques & CNRS USR3078, 35 route de Chartres,<br />

91440 Bures sur Yvette, France<br />

2 Unité de glycobiologie structurale et fonctionnelle CNRS UMR8576, Université des<br />

Sciences et Technologies de Lille, 59655 Villeneuve d'Ascq, France<br />

The packaging of eukaryotic DNA into chromatin represents an essential organizational <strong>and</strong><br />

an important regulatory event. Chromatin structure can specifically contribute positively or<br />

negatively to the correct assembly of transcription factors (TFs) <strong>and</strong> their activity. A local<br />

<strong>and</strong>/or global loss of epigenetic information is often a significant factor in genetic disorders<br />

<strong>and</strong> cancers because it leads to a deregulation of gene expression. Unfortunately the<br />

modifications occurring at the level of the chromosome, are ample <strong>and</strong> only in part<br />

understood. We have highlighted direct coupling between mechanisms of transcription by<br />

retinoic acid receptor (RAR) <strong>and</strong> its coregulator, the CREB Binding Protein (CBP) <strong>and</strong><br />

mechanisms of base excision repair (BER) by the Thymine DNA Glycosylase (TDG). TDG<br />

also acts as a coactivator of the family of RAR transcription factors <strong>and</strong> thus plays an<br />

essential role during differentiation <strong>and</strong> development. Furthermore, TDG has the ability to<br />

repair G: T mismatches produced by deamination of G: meC (methylated cytosine) in CpG<br />

isl<strong>and</strong>s in order to restore a G: C pair. Thus TDG deregulation might be an important<br />

contributor to demethylation of DNA. The CBP-RAR-TDG complex that we have shown to<br />

exist in living cells is the first link between DNA repair by base excision, transcription <strong>and</strong><br />

epigenetics <strong>and</strong> thus is a new way of regulating the integrity of the epigenome <strong>and</strong> gene<br />

expression. The aim of our work was to characterize functional CBP-TDG-RAR complexes in<br />

transcriptional regulation <strong>and</strong> epigenetics. The ultimate aim of this work is the underst<strong>and</strong>ing<br />

of genomic plasticity induced by the CBP-TDG complex <strong>and</strong> its role in oncogenesis.<br />

Transcriptome analyses with different mutant forms of TDG demonstrate altered levels of<br />

expression of transcription factors, developmental genes, migration factors, genes with<br />

functions in cell localization <strong>and</strong> cell proliferation. Mutations of the sumoylation site stabilize<br />

the CBP-TDG complex, whereas a mutation of TDG, which is not capable for acetylation by<br />

CBP prevent formation of CBP-TDG complex. We have also carried out transcriptome<br />

analyses of CBP/TDG complexes in the presence of different chemical agents to induce<br />

nuclear receptor driven transcription with retinoic acid <strong>and</strong> beta-estradiol, or to inhibit de<br />

novo methylation using 5-aza-2 deoxycytidine, <strong>and</strong> finally to massively induce DNA<br />

mismatches in vivo using 5-fluorouracil. In parallel, we have characterized in vitro the<br />

dynamics of the CBP-TDG complex analyzing the kinetics of the cleavage of<br />

oligonucleotides containing G / T or G / U, G / FU mismatches in the presence or absence of<br />

CBP <strong>and</strong> / or SUMO-1 for various forms of TDG. The results of these studies will be<br />

discussed here. Keywords : CREB binding protein (CBP), Thymine DNA glycosylase (TDG),<br />

Epigenetic regulation, Transcriptional regulation, Base Excision Repair (BER).<br />

131


Session 6: Epigenetics Poster 7<br />

Combinatorial microRNA regulation during adipogenesis: a systems biology approach.<br />

Maria Liivr<strong>and</strong> 1 , Merja Heinäniemi 2 , Carsten Carlberg 2 , Lasse Sinkkonen 2 <strong>and</strong> Thomas<br />

Sauter 1 .<br />

1 University of Luxembourg, LSRU, Systems Biology Group<br />

2 University of Luxembourg, LSRU, Computational Biology Group<br />

maria.liivr<strong>and</strong>@uni.lu; merja.heinäniemi@uni.lu; carsten.carlberg@uni.lu;<br />

lasse.sinkkonen@uni.lu; thomas.sauter@uni.lu.<br />

http://wwwen.uni.lu/research/fstc/life_sciences_research_unit/systems_biology<br />

Integration of knowledge <strong>and</strong> data between various fields such as biology, informatics <strong>and</strong><br />

medicine is the key to successful research. This thesis aims to create a new holistic<br />

perspective on adipogenesis by connecting molecular biology, computational modelling <strong>and</strong><br />

high-throughput datasets. The main focus will be on identifying miRNA families regulating<br />

single target genes in a synergistic, combinatorial manner. Relying on our previous modelling<br />

experience of a small adipogenic miRNA-target network as well as using new experimental<br />

data <strong>and</strong> computational approaches we aim first at constructing <strong>and</strong> validating a small detailed<br />

model of one core transcription factor (e.g. PPARG), two or three microRNAs (miRNAs) <strong>and</strong><br />

their shared targets’ interactions in an interconnected network. This will allow studying<br />

endogenous real-life examples of combinatorial regulation <strong>and</strong> help to draw general rules<br />

about miRNA function.<br />

The kinetic reconstruction will provide predictions on in silico stimulated changes to<br />

investigate the intrinsic stability <strong>and</strong> concentrational boundaries of molecular constituents of<br />

that system. In the next step, a larger network of molecular interactions will be created using<br />

computational databases <strong>and</strong> applications. We aim to identify more genes that are coregulated<br />

by multiple miRNAs from different families. Both networks will also be<br />

investigated for network motifs, common components of genetic interconnected circuits. This<br />

information will be further incorporated into an integrated model reflecting ratios between<br />

miRNAs <strong>and</strong> their targeted mRNAs. Finally, all these distinct relations will be organized into<br />

a larger model reflecting relations between core transcription factors, miRNAs <strong>and</strong> their target<br />

mRNAs.<br />

In this conceptual project we aim to reflect the effect of singular <strong>and</strong> combined molecular<br />

changes on a significant biological process – pre-adipocyte differentiation; find balancing<br />

scenarios in environmentally or genetically perturbated situations during adipogenesis <strong>and</strong><br />

provide significant insight into combinatorial miRNA regulation.<br />

132


Session 6: Epigenetics 8<br />

Switching Genes Promote Novel Pathway Response to Porphyromonas gingivalis<br />

Infection in Lean <strong>and</strong> Obese Macrophages<br />

Salomon Amar* x , Niraj S. Trivedi* <strong>and</strong> Calin Belta*.<br />

*Department of Bioinformatics, x Department of Periodontology <strong>and</strong> Oral Biology,<br />

Boston University, Boston, MA.<br />

!<br />

Obesity is a world epidemic problem affecting approximately 33% of US <strong>and</strong> 15% of the EU<br />

adult population. Obesity increases the risk of several chronic diseases including a diminished<br />

immune response to infection. To determine the underlying molecular dysfunction involved in<br />

the response of obese subjects to infection, we extracted Bone Marrow Macrophages (BMM)<br />

from lean <strong>and</strong> obese mice. BMM were exposed to Porphyromonas gingivalis (P. g.) for three<br />

incubation times (1h, 4h <strong>and</strong> 24h). Using a novel computational approach in conjunction with<br />

microarray data, we identified a special kind of differentially expressed genes, that we named<br />

switching genes. Our computation suggested that these genes controlled the behavior of<br />

different pathways by activating them in one condition, e.g. the lean macrophage, <strong>and</strong><br />

deactivating them in the second condition, e.g. the obese macrophage, <strong>and</strong> vice versa. The<br />

two most prominent switching genes that we found were proinflammatory factors:<br />

thrombospondin 1 <strong>and</strong> arginase 1. Our data, validated by PCR, showed that the inflammatory<br />

response was repressed in obese macrophage compared to lean counterparts. Our data suggest<br />

that, in order to produce a response to the P. g. infection, obese macrophages use switching<br />

genes to promote pathways associated with macrophage proliferation of <strong>and</strong> angiogenesis.<br />

The recruitment of these pathways is suggested to be the cause of the delay in the response to<br />

infection of obese subjects.<br />

133


Session 6: Epigenetics Poster 9<br />

Genome-wide DNA methylation Signatures of Cholangiocarcinoma<br />

Ruethairat Sriraksa 1,3 , Constanze Zeller 2 , Wei Dai 2 , Afshan Siddiq 4 , Andrew J Walley 5 ,<br />

Siobhan C McKay 7 , Naoya Kobayashi 6 , Geraldine Thomas 7 , Temduang Limpaiboon 3 ,<br />

Robert Brown 2 .<br />

1<br />

Graduate School, Khon Kaen University, Khon Kaen 40002, Thail<strong>and</strong>;<br />

r.sriraksa@gmail.com;<br />

2<br />

Epigenetics Unit, Department of Surgery <strong>and</strong> Oncology, Hammersmith Hospital,<br />

Imperial College, Du Cane Road, London W12 0NN, UK; c.zeller@imperial.ac.uk;<br />

w.dai@imperial.ac.uk; b.brown@imperial.ac.uk;<br />

3<br />

Centre for Research <strong>and</strong> Development of Medical Diagnostic Laboratories, Faculty of<br />

Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thail<strong>and</strong>;<br />

temduang@kku.ac.th;<br />

4<br />

Department of Epidemiology <strong>and</strong> Biostatistics, School of Public Health, Burlington-<br />

Danes Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK;<br />

a.siddiq@imperial.ac.uk;<br />

5<br />

Department of Genomics of Common Disease, School of Public Health, Burlington-<br />

Danes Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK;<br />

a.walley@imperial.ac.uk;<br />

6<br />

Department of Gastroenterological Surgery, Transplant <strong>and</strong> Surgical Oncology,<br />

Okayama University Graduate School of Medicine, Dentistry <strong>and</strong> Pharmaceutical<br />

Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan; immortal@md.okayamau.ac.jp;<br />

7<br />

Molecular Pathology, Hammersmith Hospital, Imperial College, Du Cane Road,<br />

London W12 0NN, UK; siobhan.mckay08@imperial.ac.uk;<br />

geraldine.thomas@imperial.ac.uk<br />

To address genome-wide DNA methylation changes in cholangiocarcinoma (CCA), we performed<br />

genome-wide DNA methylation profiling of 32 primary CCA, 6 matched adjacent normal samples, 5<br />

CCA cell lines <strong>and</strong> a normal biliary cell line using HumanMethylation27 BeadChips. We identified<br />

718 <strong>and</strong> 594 CG sites as being hypermethylated <strong>and</strong> hypomethylated in CCA compared to adjacent<br />

normal samples, respectively (False Discovery Rate; FDR85% of primary CCA as compared to adjacent normal samples using bisulfite<br />

pyrosequencing. Moreover, detailed gene set enrichment analysis (GSEA) showed overrepresentation<br />

of hypermethylated CpG sites associated with homeobox genes, PRC2 targets, EED targets, SUZ12<br />

targets <strong>and</strong> H3K27 targets. In contrast, hypomethylation of NOS targets <strong>and</strong> OCT4 targets was<br />

significantly enriched in CCA. Epigenetic aberrations of these genes have been proposed to play roles<br />

in the stem cell origin of cancers <strong>and</strong> they might provide new therapeutic targets for CCA. To our<br />

knowledge, this is the first report of genome-wide DNA methylation study in cholangiocarcinoma<br />

which provides a useful resource of epigenetic signature that could serve as the biomarkers. However,<br />

the biological functions <strong>and</strong> clinical application of these c<strong>and</strong>idate DNA methylation changes should<br />

be further clarified.<br />

134


Session 6: Epigenetics Poster 10<br />

Epigenetic regulation of proMMP-1, -2 <strong>and</strong> -9 expression in HT1080 human<br />

fibrosarcoma cell line.<br />

M. Poplineau 1 , J. Dufer 1 , F. Antonicelli 1 <strong>and</strong> A. Trussardi-Regnier 1 .<br />

1 Facultés de Médecine et Pharmacie, Unité MEDyC, UMR CNRS/URCA n°6237 :1,<br />

avenue du Maréchal Juin, 51096 REIMS Cedex, France.<br />

E-mail address: aurelie.trussardi@univ-reims.fr, frank.antonicelli@univ-reims.fr,<br />

jean.dufer@univ-reims.fr, mathilde.poplineau@etudiant.univ-reims.fr.<br />

The matrix metalloproteinase (MMP) family members play an important role in various<br />

physiological <strong>and</strong> pathological processes. The collagenase MMP-1 <strong>and</strong> the gelatinases MMP-<br />

2 <strong>and</strong> MMP-9 are involved in tumor invasiveness but the regulation of their expression is not<br />

fully elucidated <strong>and</strong> could implicate epigenetic mechanisms (DNA methylation <strong>and</strong>/or histone<br />

posttranslational modifications). The aim of this study was to analyze the effects of the<br />

histone deacetylase inhibitor trichostatine A (TSA) <strong>and</strong> the inhibitor of DNA methylation 5aza-2’-deoxycytidine<br />

(5-azadC) on the MMP-1,-2 <strong>and</strong> -9 expressions in human HT1080<br />

fibrosarcoma cell line. Real time RT-PCR revealed that 5-azadC or 5-azadC + TSA but not<br />

TSA alone (despite histone H4 hyperacetylation) increase mRNA levels. This transcription<br />

activation is correlated with chromatin decondensation observed by nuclear texture image<br />

analysis. Western blot <strong>and</strong> gelatin zymography analysis of cell culture media revealed<br />

significant secretion of proMMP-1 <strong>and</strong> high activity of MMP-2 <strong>and</strong> MMP-9, after 5-azadC or<br />

5-azadC + TSA treatment. These results suggest that epigenetic mechanism could be involved<br />

in MMP-1, MMP-2, MMP-9 gene <strong>and</strong> protein expression <strong>and</strong> could thereby modulate the<br />

invasive behavior of tumor cells.<br />

135


Session 6: Epigenetics Poster 11<br />

Epigenetic regulation of CIITA expression in T cells<br />

M.C.J.A. van Eggermond a , R. J. Wierda a , T. M. Holling a <strong>and</strong> P.J. van den Elsen a,b .<br />

a<br />

Department of Immunohematology <strong>and</strong> Blood Transfusion, LUMC, Leiden, The<br />

Netherl<strong>and</strong>s.<br />

b<br />

Department of Pathology, VU University medical center, Amsterdam, The<br />

Netherl<strong>and</strong>s.<br />

Previously we have shown that activated T cells accomplish expression of MHC-II molecules<br />

through induction of the class II transactivator (CIITA) by employment of CIITA promoter III<br />

(CIITA-PIII). In this study we show that epigenetic regulation controls CIITA-PIII promoter<br />

accessibility in normal T cells during activation. In unstimulated T cells, lacking CIITA <strong>and</strong><br />

MHC-II molecule expression, CIITA-PIII chromatin displays relative high levels of<br />

repressive histone methylation marks (3Me-K27-H3 <strong>and</strong> 3Me-K20-H4) <strong>and</strong> low levels of<br />

acetylated histones H3 (Ac-H3) <strong>and</strong> H4 (Ac-H4). These repressive histone marks are<br />

downregulated <strong>and</strong> replaced by histone methylation marks (3Me-K4-H3) associated with<br />

transcriptional active genes <strong>and</strong> high levels of Ac-H3 <strong>and</strong> Ac-H4 in activated T cells<br />

expressing CIITA. This is associated with concomitant recruitment of RNA polymerase II.<br />

Furthermore, both unstimulated <strong>and</strong> activated T cells lacked DNA methylation of CIITA-PIII.<br />

In T leukemia cells, devoid of CIITA expression, similar repressive histone methylation<br />

marks <strong>and</strong> low levels of acetylated histone H3 correlated with lack of CIITA expression. This<br />

in contrast to CIITA expressing T lymphoma cells, which display high levels of Ac-H3 <strong>and</strong><br />

3Me-K4-H3, <strong>and</strong> relative low levels of the repressive 3Me-K27-H3 <strong>and</strong> 3Me-K20-H4 marks.<br />

Of interest was the observation that the levels of histone acetylation <strong>and</strong> methylation<br />

modifications in histones H3 <strong>and</strong> H4 were also noted in chromatin of the downstream CIITA-<br />

PIV promoter as well as the upstream CIITA-PI <strong>and</strong> CIITA-PII promoters both in normal T<br />

cells <strong>and</strong> in malignant T cells. Together our data show that CIITA-PIII chromatin in activated<br />

T cells <strong>and</strong> in T lymphoma cells expressing CIITA display similar histone acetylation <strong>and</strong><br />

methylation characteristics associated with an open chromatin structure allowing RNA<br />

polymerase II recruitment. The opposite is true for unstimulated T cells <strong>and</strong> in T-leukemia<br />

cells lacking CIITA expression, which display histone modifications characteristic of<br />

condensed chromatin affecting RNA polymerase II recruitment.<br />

136


Posters are classified by session<br />

<strong>and</strong> then in alphabetical order (PRESENTING AUTHOR)<br />

(Late breaking abstracts are at the end of the <strong>book</strong>)<br />

Session 7: Cancer signaling networks<br />

137


Session 7: Cancer signaling networks Poster 1<br />

Retardation of human cancer cells proliferation by siRNAs targeted to the c-myc,<br />

N-myc, Her2, Cyclin B1 <strong>and</strong> PKC genes<br />

Ivan A. Akimov, Tatyana O. Kabilova, Marina A. Zenkova, Valentin V. Vlassov, Elena<br />

L. Chernolovskaya.<br />

Institute of Chemical Biology <strong>and</strong> Fundamental Medicine SB RAS,<br />

Novosibirsk 630090, Russia. E-mail: akimov@niboch.nsc.ru<br />

Elevated expression of genes encoding cell cycle regulatory proteins is a frequent cause of<br />

uncontrolled human cells proliferation leading to cancer. The aim of the research was to<br />

design effective inhibitors of proliferation of human cancer cells of different origin <strong>and</strong> to<br />

identify the effective therapeutic targets in each cell line. We investigated the impact of<br />

specific inhibition of c-myc, N-myc, Her2, Cyclin B1 <strong>and</strong> PKC genes expression by siRNAs<br />

(50-200 nM) on proliferation rate of KB-3-1, SK-N-MC, IMR-32, MCF-7 <strong>and</strong> HL-60 human<br />

cancer cells. The average level of gene expression was reduced to 20% compared to control.<br />

The maximum inhibition effect was achieved 72h after transfection <strong>and</strong> the initial targeting<br />

RNA level restored to 7th day. The obtained data demonstrate that inhibition of this gene<br />

expression reduces of proliferation rate differently depending on the cell type. The most<br />

effective reduction of proliferation rate induced in !"-3-1 cells (down to 10%) by inhibition<br />

of c-myc gene expression, in SK-N-MC cells (down to 4%) by inhibition of Cyclin B1 gene<br />

expression, in IMR-32 cells (down to 30%) by inhibition of N-myc gene expression <strong>and</strong> in<br />

MCF-7 cells (down to 10%) by inhibition of PKC gene expression. We investigated the<br />

proliferation of the cells after recovery of the baseline expression of target genes. We found<br />

that the proliferation rate restored after reactivation of Her2, Cyclin B1 <strong>and</strong> PKC genes in<br />

KB-3-1 <strong>and</strong> MCF-7 cells, while proliferation rate of neuroblastoma cells SK-N-MC remained<br />

reduced (to 35% <strong>and</strong> 15% respectively) after restoration of Her2 <strong>and</strong> Cyclin B1 genes<br />

expression. A selective staining of live, dead <strong>and</strong> cells in apoptotic stage showed that there are<br />

no increase in the percentage of dead <strong>and</strong> apoptotic cells in population, therefore retardation<br />

of cell division but not cell death is the reason of observed antiproliferative action. The<br />

mechanisms of cell differentiation may also be involved. Thus the obtained data suggest that<br />

Her2 <strong>and</strong> Cyclin B1 genes are the promising targets for the treatment of neuroblastomas. This<br />

work was supported by RAS programs “Molecular <strong>and</strong> cellular biology” <strong>and</strong> “Fundamental<br />

science for medicine”, president`s program SS-7101.2010.4 <strong>and</strong> SB RAS gr<strong>and</strong> No.41.<br />

138


Session 7: Cancer signaling networks Poster 2<br />

Control of CDC25 phosphatases splicing by genotoxic agents in MCF-7 breast cancer<br />

cell line.<br />

Hélène Albert 1 , Eric Battaglia 1 , Susana Santos 2 , Carolino Monteiro 2 , Denyse Bagrel 1<br />

1 Laboratoire d’Ingénierie Moléculaire et Biochimie Pharmacologique, EA 3940, FR<br />

CNRS 2843, Université Paul Verlaine-Metz, Rue du Général Delestraint, 57070, Metz,<br />

France.<br />

2 Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003<br />

Lisboa, Portugal.<br />

Email : bagrel@univ-metz.fr<br />

CDC25 phosphatases play a crucial role in cell cycle progression <strong>and</strong> are overexpressed in a<br />

large number of cancers. CDC25 are represented by three isoforms: CDC25A, CDC25B <strong>and</strong><br />

CDC25C, encoded by three different genes, all of them subjected to an alternative splicing<br />

mechanism. Alternative splicing is an essential process concerning 75 % of human genes <strong>and</strong><br />

thus contributes to proteomic diversity. Many studies are currently focused on alternative<br />

splicing regulation in cellular stress conditions. However, none of them have been conducted<br />

so far on CDC25. Using semi-quantitative <strong>and</strong> quantitative RT-PCR, we showed that<br />

treatment of MCF-7 human mammary adenocarcinoma cells with doxorubicin (1 !M) does<br />

not affect CDC25A <strong>and</strong> CDC25B splicing, while a modification of CDC25C splicing pro<strong>file</strong><br />

was observed after 6-12 hours. CDC25C5 splice variant proportion was increased 10-fold in<br />

comparison to that of CDC25C1 variant at both mRNA <strong>and</strong> protein levels. A similar<br />

alteration of CDC25C alternative splicing was observed with other genotoxic agents,<br />

including topoisomerases inhibitors camptothecin <strong>and</strong> etoposide, the electrophilic agent<br />

cisplatin <strong>and</strong> the reactive oxygen species generating agent tert-butyl hydroperoxide, but not<br />

with cyclophosphamide <strong>and</strong> vinblastine. These observations suggest that CDC25C alternative<br />

splicing, but not that of CDC25A <strong>and</strong> CDC25B, is modulated under conditions associated to<br />

DNA damage. Further studies are needed to elucidate the regulatory pathways involved in this<br />

mechanism.<br />

139


Session 7: Cancer signaling networks Poster 3<br />

Novel CDC25 small coumarin-based inhibitors: biological evaluation.<br />

E. Bana, S. Valente, E. Viry, G. Kirsch, D. Bagrel<br />

Laboratoire d’Ingénierie Moléculaire et Biochimie Pharmacologique, LIMBP, avenue<br />

du général Delestraint, 57070 Metz, France - bana@univ-metz.fr<br />

Cell division cycle 25 (CDC25) phosphatases (A, B, <strong>and</strong> C) are key cell cycle control proteins<br />

in eukaryotic cells <strong>and</strong> numerous studies rise up the correlation between their overexpression<br />

<strong>and</strong> cancer aggressiveness, high grade tumors <strong>and</strong> low vital prognosis. CDC25s are attractive<br />

c<strong>and</strong>idates for new cancer therapy targets as their inhibition could be able to slow down tumor<br />

growth. To date, about one hundred chemical compounds have been reported in literature as<br />

inhibitors of CDC25s. Most are quinonoid, phosphate surrogates or electrophilic inhibitors.<br />

Several silybin derivatives showed a cell cycle arrest <strong>and</strong> induced level variations of the three<br />

CDC25s in PC3 cells, <strong>and</strong> several flavones are able to inhibit CDC-25.1 phosphatase activity<br />

in C. elegans. These findings prompted us to project the design, synthesis <strong>and</strong> biological<br />

validation of novel coumarin-based (like flavon isomer) derivatives. Our study started with<br />

insertion of 4-methoxy(hydroxy)phenyl <strong>and</strong> styryl groups in C3 position of the coumarin<br />

nucleus. We then explored the C4 position with the chalcone-coumarin (benzoylvinyl)<br />

derivatives <strong>and</strong> the reverse chalcone-coumarin (cinnamoyl). At last we investigated the<br />

insertion of a sulfur atom in the spacer-chain between coumarin <strong>and</strong> benzoyl moiety,<br />

obtaining final thio-analogs.<br />

The CDC25 inhibitory potential of 16 new compounds was tested using the three human<br />

(glutathion-S-transferase)-CDC25s recombinant enzymes. This screening highlighted two<br />

chalcon-coumarin derivatives, namely 6a <strong>and</strong> 6d, which showed an IC50 value in the level of<br />

25 !M against CDC25A <strong>and</strong> C. 6a behaves as an irreversible inhibitor of CDC25A. Using<br />

breast cancer cell lines (MCF7 <strong>and</strong> derivatives), the study of a potential correlation between<br />

in vitro <strong>and</strong> in cellulo CDC25 inhibitions as well as cytostatic effect <strong>and</strong> cell cycle blocking is<br />

going on.<br />

We consider 6a <strong>and</strong> 6d as two new lead compounds worthy of note for models in developing<br />

new CDC25 inhibitors, <strong>and</strong> thus new potential anticancer drugs.<br />

140


Session 7: Cancer signaling networks Poster 4<br />

The inhibition of poly(ADP-ribosylation) affects cancer cell proliferation <strong>and</strong> telomere<br />

elongation<br />

Cristina Belgiovine, Francesca Donà, Ilaria Chiodi, Tatiana Raineri, Roberta Ricotti,<br />

Chiara Mondello <strong>and</strong> A. Ivana Scovassi<br />

Istituto di Genetica Molecolare CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy<br />

belgiovine@igm.cnr.it<br />

Poly(ADP-ribosylation) plays a central role in Base Excision Repair (BER) <strong>and</strong> also in<br />

neoplastic transformation <strong>and</strong> telomere length regulation. It is catalyzed by a family of<br />

poly(ADP-ribose) polymerases (PARPs), of which the best characterized are PARP-1, PARP-<br />

2, TANK-1 <strong>and</strong> TANK-2, which convert NAD into ADP-ribose further used to form<br />

polymers. We investigated their expression levels during neoplastic transformation of cen3tel<br />

cells, which derive from human fibroblasts immortalized by ectopic hTERT expression <strong>and</strong>,<br />

during propagation in vitro, acquired the ability to grow in agar <strong>and</strong> subsequently to form<br />

tumors in nude mice. Upon further propagation, tumorigenic cells became more aggressive,<br />

showing a reduction in the time required to induce tumors; moreover, they reached telomere<br />

lengths >100 Kb <strong>and</strong> increased telomerase activity. We found a notable increase of PARP-1<br />

protein (<strong>and</strong> mRNA) in parallel to the acquisition of the tumorigenic potential; PARP-2 levels<br />

showed the same trend, while tankyrase expression did not show significant changes. As<br />

supported by poly(ADP-ribose) accumulation, overexpressed PARP-1 <strong>and</strong> -2 were very<br />

active, thus suggesting that poly(ADP-ribosylation) is modulated during neoplastic<br />

transformation <strong>and</strong> could be a target for contrasting tumor development. To address this point,<br />

we analyzed the effect of the known PARP inhibitor, 3-aminobenzamide (3-AB), on cell<br />

proliferation <strong>and</strong> telomere length. Increasing concentrations of 3-AB inhibited the<br />

proliferation of neoplastic cells in a dose-dependent manner. Remarkably, prolonged 3-AB<br />

administration to early tumorigenic cells also affected telomere length. On the whole, our data<br />

support an active involvement of poly(ADP-ribosylation) in neoplastic transformation <strong>and</strong><br />

telomere length maintenance. Thus, our work provides additional evidence in favor of the use<br />

of PARP inhibitors for the treatment of human cancer [Donà et al., Curr. Pharmaceut.<br />

Biotechnol. (2010), in press].<br />

141


Session 7: Cancer signaling networks Poster 5<br />

Development of MIMOKINES, a New Class of Short CXCR4 Antagonists for Tumor<br />

<strong>and</strong> HIV-1 Treatment.<br />

Andy Chevigné1, Virginie Fievez1, Julie Mathu1, Jean-Claude Schmit1,2 <strong>and</strong> Sabrina<br />

Deroo1<br />

1Laboratory of Retrovirology, Centre de Recherche Public-Santé, 84, Val Fleuri, L-1526<br />

Luxembourg. 2Service National des Maladies Infectieuses, Centre Hospitalier<br />

Luxembourg, 4, Rue E. Barblé, L-1210 Luxembourg. <strong>and</strong>y.chevigne@crp-sante.lu<br />

The chemokine receptor CXCR4 is a G protein coupled receptors (GPCR) playing a crucial<br />

role in cancer <strong>and</strong> tumor cell biology by interacting with its unique lig<strong>and</strong> the chemokine<br />

CXCL12. CXCR4 also acts as an HIV-1 coreceptor for T cell infection. To date, no approved<br />

drugs targeting metastasis development or HIV-1 entry via CXCR4 are yet available.<br />

Therefore, the development of new molecules blocking the interactions between CXCR4 <strong>and</strong><br />

CXCL12 or viral gp120 protein is of great importance. In this study, we developed a new<br />

class of short CXCR4 antagonists based on peptides mimicking the N-terminus of CXCL12.<br />

We minimized the size of the chemokine to a peptide level (17-mer) <strong>and</strong> generated a large<br />

number of variants called Mimokines. Five phage displayed Mimokines libraries of high<br />

complexity (107 to 108 clones), characterized by complementary <strong>and</strong> progressive<br />

r<strong>and</strong>omization were engineered. Ten selection campaigns using either competitive or acid<br />

elution were performed on a simplified CXCR4 model. For acid elution campaigns<br />

enrichments up to 10 were obtained <strong>and</strong> 11 different positive clones with higher affinity (ratio<br />

1.3 to 8.5) for CXCR4 than the wild type peptide (wt) were identified. For competitive elution<br />

higher enrichments (up to 76) were obtained including clones with affinity ratio’s ranging<br />

from 3 to 14 compared to wt peptide. Sequence analysis revealed the presence of clones<br />

isolated at various frequencies (1/22 to 6/22). These peptides will be further analyzed for their<br />

antagonist properties towards CXCR4-CXCL12 axis <strong>and</strong> HIV-1 entry inhibition <strong>and</strong> may<br />

potentially provide new anti-metastasis <strong>and</strong> anti-HIV-1 lead molecules.<br />

142


Session 7: Cancer signaling networks Poster 6<br />

Study of the combined effect of 5-ALA-based photodynamic therapy <strong>and</strong> NF-kappaB<br />

inhibition on human glioblastoma cell survival<br />

Isabelle Coupienne, Grégory Fettweis <strong>and</strong> Jacques Piette<br />

University of Liege, GIGA-Research, Virology <strong>and</strong> Immunology Unit, B34, 1 avenue de<br />

l’Hopital, 4000 Liege, Belgium<br />

icoupienne@ulg.ac.be<br />

Glioblastoma constitute the most frequent <strong>and</strong> deadliest type of brain tumors in human adults.<br />

They are very resistant to all current therapies <strong>and</strong> are associated with a huge rate of<br />

recurrence. In most cases, this type of tumor is characterized by a constitutive activation of<br />

the nuclear factor-kappaB (NF-kappaB). This factor is known to be a key regulator of various<br />

physiological processes such as inflammation, immune response, cell growth or apoptosis. In<br />

the present study, we explored the role of NF-kappaB activation in the sensitivity of human<br />

glioblastoma cells to a treatment by 5-aminolevulinic acid (5-ALA)–based photodynamic<br />

therapy (PDT). Our results show that inhibition of NF-kappaB improves glioblastoma cell<br />

death in response to 5-ALA-PDT. We then studied the molecular mechanisms underlying the<br />

cell death induced by PDT combined or not with NF-kappaB inhibition. We found that PDT<br />

mainly induced necrosis in glioblastoma cells <strong>and</strong> NF-kappaB was found to have anti-necrotic<br />

functions in this context. In the second part of this study, we examined the role of the kinase<br />

RIP3, recently identified as a key effector of the necrotic pathway, in 5-ALA-PDT-induced<br />

necrosis <strong>and</strong> studied whether NF-kappaB interfered in RIP3-dependent necrosis induction.<br />

143


Session 7: Cancer signaling networks Poster 7<br />

The Anti Proliferative Effect of Aloin <strong>and</strong> Aloe Emodin on MCF-7 <strong>and</strong> MDA-MB-231<br />

Breast Cancer Cells<br />

Melor M. Daud, Muhammad J. Ibrahim, Gabriele R. A. Froemming <strong>and</strong> Narimah A.<br />

H. Hasani<br />

Institute of Medical Molecular Biotechnology, Universiti Teknologi MARA Sungai<br />

Buloh Medical Campus, Jalan Hospital, Sungai Buloh, 47 000 Selangor, Malaysia<br />

- impaxmelor@gmail.com<br />

Aloin <strong>and</strong> Aloe Emodin are active components in Aloe Barbadensis Miller, commonly known<br />

as Aloe vera. Earlier studies had shown both anthraquinones display anti cancer activity. We<br />

were investigating their possible anti tumor properties, upon oestrogen receptor positive (ER + )<br />

MCF-7 cells <strong>and</strong> oestrogen receptor negative (ER - ) MDA-MB-231 breast cancer cells.<br />

Normal breast cell line, MCF-10A represented as negative control <strong>and</strong> Tamoxifen as positive<br />

control. Cell proliferation was measured using WST-1 Reagent at 450nm between 0!M to<br />

500!M.<br />

For MCF-7, we found IC50 of Tamoxifen at 64!M, 78!M for Aloe Emodin <strong>and</strong> no inhibitory<br />

concentration by Aloin up to 150!M. In MDA-MB-231, Tamoxifen induced anti proliferative<br />

effect at IC50 of 62!M, whereas below 150!M both Aloin <strong>and</strong> Aloe Emodin does not reach<br />

the IC50 level respectively. For MCF-10A, IC50 for Tamoxifen is 38!M, 410!M for Aloin <strong>and</strong><br />

no IC50 by Aloe Emodin up to 150!M.<br />

These results indicated that Aloe Emodin can be an effective inhibitor to the hormonal<br />

dependence MCF-7 breast cancer cells at low concentration without being cytotoxic to normal<br />

breast cells. The therapeutic properties can further be investigated for apoptotic effect <strong>and</strong><br />

signaling pathways. However, Aloe Emodin does not inhibit proliferation of MDA-MB-231<br />

cancer cells. Aloin shows no effect to both cancer <strong>and</strong> normal mammary cells up to 500!M,<br />

suggesting that it might not have anti carcinogenic properties. In conclusion, Aloe Emodin<br />

shows anti proliferative effect selectively to breast cancer cells with significant probability to<br />

demonstrate chemotherapeutic activities in breast cancer in vitro studies.<br />

144


Session 7: Cancer signaling networks Poster 8<br />

Phosphatidylinositol 3-kinase inhibitor - LY294002 in Regulation of Androgen<br />

Receptors in LNCaP prostate cancer cells.<br />

Joanna Duli!ska-Litewka, Dorota Gil, Paulina Dudzik <strong>and</strong> Piotr Laidler<br />

Chair of Medical Biochemistry, Jagiellonian University Medical College, 31-034<br />

Kraków, ul. Kopernika 7, Pol<strong>and</strong>; e-mail: mblitewk@cyf-kr.edu.pl<br />

Prostate cancer is one of males’ most serious health problem <strong>and</strong> has a high prevalence to<br />

metastasize making it a very difficult to treat if not detected early. Androgen receptor (AR)<br />

controls the expression of genes involved in cell proliferation, migration, differentiation, <strong>and</strong><br />

cell death. Increasing level of the AR not only intensify <strong>and</strong>rogen-induced cell proliferation<br />

but also increases the sensitivity to non<strong>and</strong>rogenous mediators, allowing prostate cancer cells<br />

to grow in a low <strong>and</strong>rogen environment. We observed that knockout of AR functions by<br />

siRNA leads to meaningful inhibition of LNCaP cells’ proliferation. In addition we noticed<br />

that under normal growth conditions, inhibition of PI3K/Akt signaling by inhibitor of PI3K -<br />

LY294002 - causes LNCaP cell arrest in G1 phase rather than apoptosis. Akt can modulate<br />

<strong>and</strong>rogen signaling since beta-catenin acts as AR coactivator, however how these two<br />

regulators cooperate to control prostate cancer cell growth <strong>and</strong> death remains as yet unclear.<br />

Inactivation of PI3K in LNCaP cell has been demonstrated to lead to the decreased<br />

phosphorylation of Akt in prostate cancer cell lines. In parallel treatment of LNCaP cells with<br />

LY294002 results in the increase of prostate-specific antigen (PSA) <strong>and</strong> prostatic acid<br />

phosphates (PAP) mRNA expression without testosterone or DHT treatment. This increase<br />

suggests that blockade of PI3K/Akt signaling pathway enhances AR transactivation. To find<br />

out the mechanism that explains their interaction we treated LNCaP cells with LY294002 or<br />

knocked out their AR <strong>and</strong> studied mRNA (RT-PCR) <strong>and</strong> protein (Western blot) expression of<br />

Akt/(pAkt), PSA, PAP, beta-catenin, cyclin D1, bax, bcl-2 <strong>and</strong> AR. pAkt was inhibited by<br />

LY294002 treatment but PSA, PAP, beta-catenin <strong>and</strong> AR protein levels were increased<br />

significantly, suggesting that blockade of PI3K/Akt pathway may enhance AR activity by the<br />

increase of AR protein expression. Activation of Akt in effect of AR silencing has been<br />

implicated in protection of cells from apoptosis throughout stimulation of c-Myc, cyclinD1<br />

expression <strong>and</strong> phosphorylation of the Bcl-2, thereby promoting cell survival. This important<br />

finding implies that combined inhibition of both PI3K/Akt <strong>and</strong> AR signaling may become a<br />

powerful approach to treat the prostate cancer.<br />

This work was supported by MNiSzW grants: K/ZDS/001003 <strong>and</strong> K/PBW/ 000561 -<br />

Jagiellonian University Medical College, Krakow, Pol<strong>and</strong><br />

145


Session 7: Cancer signaling networks Poster 9<br />

HGF/c-Met signaling pathway regulates TXNIP, major actor of oxidative stress, in SK-<br />

Hep-1, HCC cell line.<br />

Gozukizil A., Ozen E., Erdal E., Atabey N.<br />

Dokuz Eylul University, School of Medicine Depertmant of Medical Biology <strong>and</strong><br />

Genetics, 35340, Inciralti/Izmir, Turkey a.gkizil@hotmail.com<br />

In the development of Hepatocellular carcinoma (HCC), imbalance between oxygen <strong>and</strong><br />

glucose homeostasis contributes to the production of the reactive oxygen species following<br />

induction of oxidative stress. It is very well known that chronic inflammation, one of the<br />

hallmarks of development of HCC, has been triggered by oxidative stress after occurring<br />

continuous rounds of hepatocyte injury <strong>and</strong> following regeneration. Additionally, TXNIP,<br />

transcription factor, regulates cellular responses under stress conditions, such as hypoxia,<br />

serum starvation <strong>and</strong> heat-shock. It is recently shown that over-expression of TXNIP results<br />

in activation of oxidative stress <strong>and</strong> apoptotic responses of cells <strong>and</strong> it behaves as tumor<br />

suppressor. However, the role of TXNIP in HCC is not defined properly.<br />

In this study, expression levels of TXNIP have been analysed 12 HCC cell lines (Huh7,<br />

Hep3B, HepG2, PLC/PRF/5, SNU398, SNU387, SNU423, SNU449, Snu475, SNU182, SK-<br />

Hep-1, Mahlavu) by using western blotting <strong>and</strong> RT-PCR. Interestingly TXNIP expression<br />

was significantly high in HCC cell lines which have mesenchymal-like phenotype <strong>and</strong> high<br />

basal motility ability such as SNU182, SNU387 <strong>and</strong> SNU423. In contrast there was no<br />

expression of TXNIP in HCC cell lines that have epithelial-like phenotype such as Huh7,<br />

Hep3B <strong>and</strong> PLC/PRF/5. To underst<strong>and</strong> the molecular mechanism of TXNIP up-regulation in<br />

HCC, we analyzed the regulation of its expression by HGF/c-Met signaling which is known<br />

as important protector against to oxidative stress during development of HCC. Here, we have<br />

demonstrated for the first time, TXNIP expression is up-regulated by HGF treatment via<br />

activation of the both HGF/c-Met <strong>and</strong> Erk1/2 mediated pathways in SK-Hep-1 cell line. In<br />

addition, when activated form of c-Met was inhibited by SU11274, TXNIP expression was<br />

blocked, in parallel, with the decreased in activation of Erk1/2. Also, TXNIP up-regulation<br />

has been observed as in parallel with activation of HGF/c-Met signaling under stress<br />

conditions induced by high glucose <strong>and</strong> hypoxia. When we classified these HCC cells based<br />

on basal HGF/c-Met activation levels, it is notably important that invasive <strong>and</strong> metastatic<br />

HCC cell lines which have also c-Met tyrosine kinase over-expression, becomes TXNIP upregulation.<br />

As a conclusion, HGF/c-Met signaling regulates TXNIP expression at both transcript <strong>and</strong><br />

protein levels, in parallel, it protects cells against oxidative stress regulated by elevated<br />

TXNIP expression in the first steps of hepatocarcinogenesis. Following, these cells become<br />

more motile <strong>and</strong> aggressive in time. Thus, we may propose TXNIP behaves as an oncogene in<br />

development of HCC.<br />

Key words: TXNIP, HCC, HGF/c-Met<br />

146


Session 7: Cancer signaling networks Poster 10<br />

A new Ca 2+ /calmodulin antagonist HBC exhibits anti-angiogenic effect by suppressing<br />

signal transduction to hypoxia-inducible factor<br />

Hye Jin Jung, Jong Hyeon Kim, Joong Sup Shim, <strong>and</strong> Ho Jeong Kwon*<br />

Chemical Genomics National Research Laboratory, Department of Biotechnology,<br />

Translational Research Center for Protein Function Control, College of Life Science <strong>and</strong><br />

Biotechnology, Yonsei University, Seoul 120-749, Korea<br />

* Corresponding author: kwonhj@yonsei.ac.kr<br />

Targeting hypoxia-inducible factor-1 (HIF-1) signaling is a promising strategy to treat cancer<br />

growth <strong>and</strong> metastasis through an anti-angiogenic effect. Recent reports have shown that<br />

Ca 2+ /calmodulin (Ca 2+ /CaM) activates HIF-1 <strong>and</strong> consequently induces the expression of proangiogenic<br />

factors such as vascular endothelial growth factor (VEGF). Here, we demonstrated<br />

the anti-angiogenic effect of a new Ca 2+ /CaM antagonist HBC (4-{3,5-bis-[2-(4-hydroxy-3methoxyphenyl)ethyl]-4,5-dihydropyrazol-1-yl}benzoic<br />

acid). HBC potently inhibited the<br />

proliferation of human umbilical vascular endothelial cells with no cytotoxicity. Furthermore,<br />

HBC blocked in vitro angiogenesis such as tube formation <strong>and</strong> chemoinvasion as well as<br />

neovascularization of the chorioallantoic membrane of growing chick embryo in vivo. We<br />

also evaluated the effect of HBC on the activity of HIF-1. The compound markedly inhibited<br />

HIF-1! expression at the translational level during hypoxia, thereby reducing HIF-1<br />

transcriptional activity <strong>and</strong> expression of its major target gene VEGF. In addition,<br />

combination treatment with HBC <strong>and</strong> various HIF-1 inhibitors had greater anti-angiogenic<br />

activity than treatment with each single agent. Collectively, HBC is a promising antiangiogenic<br />

agent targeting HIF <strong>and</strong> might be applied in combination therapy to overcome the<br />

chemoresistance of solid tumors.<br />

147


Session 7: Cancer signaling networks Poster 11<br />

Increased expression of delta ex2TFF2 mRNA associates with prolonged survival of<br />

cholangiocarcinoma patients<br />

Surasee Kamlua 1,2,3 , Siriporn Patrakitkomjorn 2,3 , Patcharee Jearanaikoon 2,3 , Trevelyan<br />

R. Menheniott 4 , Andrew S. Giraud 4 , Temduang Limpaiboon 2,3 .<br />

1 Graduate School, Khon Kaen University, Khon Kaen 40002, Thail<strong>and</strong>; !<br />

2 Department of Clinical Chemistry, Centre for Research <strong>and</strong> Development of Medical<br />

Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University,<br />

Khon Kaen 40002, Thail<strong>and</strong>;<br />

3 Liver Fluke <strong>and</strong> Cholangiocarcinoma Research Center, Faculty of Medicine, Khon<br />

Kaen University, Khon Kaen 40002, Thail<strong>and</strong>;<br />

4 Murdoch Children’s Research Institute, Royal children’s Hospital, Parkville, VIC<br />

3052, Australia.<br />

E-mail: kamikaze_me@hotmail.com<br />

Trefoil factor 2 (TFF2) is a member of trefoil factor family that plays the role in epithelial<br />

restitution in the gastrointestinal tract. Recently, we demonstrated the overexpression of TFF2<br />

in liver fluke-related cholangiocarcinoma (CCA). Moreover, we found a novel alternative<br />

splice variant of TFF2 mRNA, delta ex2TFF2, in CCA cell lines. Delta ex2TFF2 mRNA<br />

sequence revealed an out-of-frame deletion of exon 2 of TFF2 mRNA resulting in a stop<br />

codon (TAG) at exon 1 resulting in the translation of a truncated protein lacking the paired<br />

trefoil domains of the canonical TFF2 protein. The clinical significance of delta ex2TFF2 is<br />

unclear. Here, we studied the expression of delta ex2TFF2 mRNA <strong>and</strong> its association with<br />

clinical parameters of CCA patients. Seventy eight tumors <strong>and</strong> 15 normal adjacent tissues<br />

were quantified for the expression of delta ex2TFF2 <strong>and</strong> wild type TFF2 (wtTFF2) mRNA<br />

against the internal reference gene GAPDH. The relative expression of delta ex2TFF2 mRNA<br />

<strong>and</strong> delta ex2TFF2/wtTFF2 ratio in tumor tissues were significantly higher than in normal<br />

(P


Session 7: Cancer signaling networks Poster 12<br />

Collagen I Regulates Cell Motility through Integrin Alpha 4 in Hepatocellular<br />

Carcinoma Cell Lines<br />

Emine K<strong>and</strong>emis 1 , Nese Atabey 1 , Esra Erdal 1<br />

1 Dokuz Eylul University, Faculty of Medicine, Department of Medical Biology <strong>and</strong><br />

Genetics, 35340-Inciralti, Izmir, Turkey. eminecelik@gmail.com<br />

Hepatocellular carcinoma (HCC) abundantly arises on the viral <strong>and</strong>/or chemicalinduced<br />

cirrhosis in liver. One of the most prominent feature of liver cirrhosis is the<br />

uncontrolled collagen I production. It is also known that increasing in collagen I affects cell<br />

behavior such as cell motility <strong>and</strong> invasion through activation of gene expression in key<br />

regulators on the preneoplastic stage of HCC such as TNF-alpha, integrins, MMP2 <strong>and</strong> MT1-<br />

MMP. However, there is no enough information about in which mechanisms collagen I<br />

affects the cell motility in HCC cells. In our study we investigated the effect of collagen I on<br />

HCC cell lines (PLC/PRF/5, Hep3B, SNU-423, Sk-Hep-1 <strong>and</strong> SNU-449) by using wound<br />

assay <strong>and</strong>/or boyden-chamber motility methods. Cellular motility of PLC/PRF/5, Hep3B,<br />

SNU-423, SNU-449 <strong>and</strong> Sk-Hep-1 cells grown on the collagen I significantly decreased in<br />

different levels. Further we analyzed expression of integrins (alpha 1, alpha 2, alpha 3, alpha<br />

4, alpha 5, alpha 6, alpha v, beta 1 <strong>and</strong> beta 4) on the effect of collagen I by using reverse<br />

transcriptase PCR in SNU-449 cells. Unexpectedly, integrin alpha 1, alpha 2 <strong>and</strong> alpha 3<br />

which are collagen I receptors did not significantly change in the presence of collagen I,<br />

contrarily, the expression of integrin alpha 4 decreased in both mRNA <strong>and</strong> protein levels in<br />

SNU-449. Interestingly, we observed that HCC cell lines, SNU-423, SNU-449 <strong>and</strong> Sk-Hep-1,<br />

having high ability in cell motility expressed integrin alpha 4 in a much more level than HCC<br />

cell lines, Hep3B <strong>and</strong> PLC/PRF/5, which have low basal motility. These data suggest that<br />

reducing effect of collagen I on HCC cell motility might be regulated by integrin alpha 4<br />

expression. It may be an unique mechanism to explain the role of collagen I induced<br />

mechanisms during hepatocarcinogenesis.<br />

149


Session 7: Cancer signaling networks Poster 13<br />

Butein sensitizes human hepatoma cells to TRAIL-induced apoptosis via ERK/Sp1dependent<br />

DR5 up-regulation <strong>and</strong> NF-!B inactivation<br />

Chang-Hee Kang, 1 Jang-Yeon Jeong, 1 R.G.P.T. Jayasooriya, 1 Sang-Hyuck Kang, 1 Yung<br />

Hyun Choi, 2 <strong>and</strong> Gi-Young Kim 1,*<br />

1 Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National<br />

University, Jeju 690-756, Republic of Korea<br />

2 Department of Biochemistry, Dongeui University College of Oriental Medicine, Busan<br />

614-051, Republic of Korea<br />

Tumor necrosis factor (TNF)-related apoptosis-inducing lig<strong>and</strong> (TRAIL) induces cell death in<br />

various types of cancer cells but has little or no effect on normal cells. Human hepatoma cells<br />

are resistant to TRAIL-induced apoptosis. Although butein is known to mediate anti-cancer,<br />

anti-inflammatory, <strong>and</strong> anti-oxidant activities, little is known about the mechanism of butein<br />

in terms of TRAIL-induced apoptosis of human hepatoma cells. In this study, we determined<br />

that butein enhances TRAIL-induced apoptosis in hepatoma cells through up-regulation of<br />

DR5. Luciferase analysis showed that a 5’-flanking region containing four Sp1 binding sites<br />

within the DR5 promoter was enhanced by butein (-305/-300). Electrophoretic mobility shift<br />

assays <strong>and</strong> chromatin immunoprecipitation studies were used to analyze the elevation of Sp1<br />

binding to DR5 promoter sites by butein. Point mutations of the Sp1 binding site also<br />

attenuated promoter activity. Furthermore, pre-treatment of the blocking chimeric antibody<br />

<strong>and</strong> small interfering RNS for DR5 significantly suppressed TRAIL-mediated apoptosis by<br />

butein in Hep3B cells. Butein also stimulated ERK activation, <strong>and</strong> the ERK inhibitor<br />

PD98059 blocked butein-induced DR5 expression <strong>and</strong> suppressed binding of Sp1 to the DR5<br />

promoter. Additionally, generation of reactive oxygen species (ROS) had no effect on cell<br />

viability, although pre-treatment with N-acetyl-L-cystein or glutathione inhibited combined<br />

treatment-induced ROS. Indeed, butein repressed the TRAIL-mediated activation of NF-!B<br />

<strong>and</strong> decreased its transcriptional activity. Our results suggest that butein could sensitize<br />

certain human hepatoma cells to TRAIL-induced apoptosis through stimulating its death<br />

signaling <strong>and</strong> by repressing the survival function in these cells.<br />

150


Session 7: Cancer signaling networks Poster 14<br />

Sulforaphane sensitizes tumor necrosis factor-related apoptosis inducing lig<strong>and</strong><br />

(TRAIL)-mediated apoptosis through downregulation of ERK <strong>and</strong> Akt in lung<br />

adenocarcinoma A549 cells<br />

Chang-Hee Kang, 1 Yeon-Jeong Jang, 1 R.G.P.T. Jayasooriya, 1 Sang-Hyuck Kang, 1<br />

Cheung-Yun Jin, 2 Yung Hyun Choi, 2 <strong>and</strong> Gi-Young Kim 1,*<br />

1 Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National<br />

University, Jeju 690-756, Republic of Korea<br />

2 Department of Biochemistry, Dongeui University College of Oriental Medicine, Busan<br />

614-051, Republic of Korea<br />

The cytotoxic effect of the tumor necrosis factor-related apoptosis-inducing lig<strong>and</strong> (TRAIL)<br />

is limited in some cancer cells, including A549 lung adenocarcinoma cells. However,<br />

treatment with TRAIL in combination with subtoxic concentrations of sulforaphane (SFN)<br />

sensitizes TRAIL-resistant A549 cells to TRAIL-mediated apoptosis. Combined treatment<br />

with SFN <strong>and</strong> TRAIL induced chromatin condensation, DNA fragmentation, annexin V<br />

staining <strong>and</strong> sub-G1 phase DNA content. These indicators of apoptosis correlate with the<br />

induction of caspase-3 activity, which results in the cleavage of poly(ADP-ribose)polymerase<br />

(PARP) <strong>and</strong> the release of lactate dehydrogenase (LDH). Both the cytotoxic effect <strong>and</strong><br />

apoptotic characteristics induced by combined treatment were significantly inhibited by z-<br />

DEVD-fmk, a caspase-3 inhibitor, demonstrating the important role of caspase-3 in the<br />

observed cytotoxic effect. Combined treatment also triggered the activation of p38 MAPK<br />

<strong>and</strong> JNK, <strong>and</strong> downregulation of ERK <strong>and</strong> Akt. Inhibitors of ERK (PD98059) or Akt<br />

(LY294002), but not p38 MAPK, resulted in significantly decreased cell viability. Although<br />

the activation of JNK was increased in response to combined treatment, inhibition of the JNK<br />

pathway significantly attenuated cell viability. These results indicate that caspase-3 is a key<br />

regulator of apoptosis in response to combined SFN <strong>and</strong> TRAIL in human lung<br />

adenocarcinoma A549 cells through downregulation of ERK <strong>and</strong> Akt.<br />

151


Session 7: Cancer signaling networks Poster 15<br />

Heat shock proteins in oncology: Diagnostic biomarkers <strong>and</strong> therapeutic targets<br />

Ashraf A. Khalil 1 , Nihal F. Kabapy 1 , Sahar F. Deraz 1 , Christopher Smith 2<br />

1 Department of Protein Technology, Institute of Genetic Engineering <strong>and</strong><br />

Biotechnology, Mubarak City for Scientific Research, New Borg Elarab, Alex<strong>and</strong>ria,<br />

Egypt.<br />

2 Department of Biological Sciences, University of Chester, Parkgate Road Chester, UK<br />

Heat shock proteins (HSP) are a family of proteins induced in cells exposed to different<br />

insults allowing cells to survive stress conditions. Mammalian HSPs have been classified into<br />

six families according to their molecular size: HSP100, HSP90, HSP70, HSP60, HSP40 <strong>and</strong><br />

small HSPs (15 to 30 kDa) including HSP27. These proteins act as molecular chaperones<br />

either helping in the refolding of misfolded proteins or assisting in their elimination if they<br />

become irreversibly damaged. In recent years, proteomic studies have characterized several<br />

different HSPs in various tumour types which may be putative clinical biomarkers or<br />

molecular targets for cancer therapy. This has led to the development of a series of molecules<br />

capable of inhibiting HSPs. Furthermore, protein profiling of several models involving<br />

administered HSP90 inhibitors have revealed new biomarkers that can help in the evaluation<br />

of the degree of HSP90 inhibition as well as predicting the therapeutic response. Numerous<br />

studies speculated that over expression of HSP is in part responsible for resistance to many<br />

anti-tumours <strong>and</strong> chemotherapeutics. Hence, from a pharmacological point of view, the coadministration<br />

of HSP inhibitors along with other anti-tumour agents is of major importance<br />

to overcome therapeutic resistance. We provide an overview of the current status of the HSPs<br />

in diseases with special emphasis on cancer.<br />

152


Session 7: Cancer signaling networks Poster 16<br />

Endometriosis-related Ovarian Cancer Risk <strong>and</strong> Prognosis: A Meta-analysis<br />

Yong Beom Kim 1 , Hee Seung Kim 2 , Chae Hyeong Lee 3 , Yong-Tark Jeon 1 , Yong Sang<br />

Song 2<br />

1 Department of Obstetrics <strong>and</strong> Gynecology, Seoul National University Bundang<br />

Hospital, Seoungnam 463-707, Republic of Korea (Kim YB, ybkimlh@snubh.org; Jeon<br />

YT, asidof@snubh.org)<br />

2 Department of Obstetrics <strong>and</strong> Gynecology, Seoul National University College of<br />

Medicine, Seoul 110-744, Republic of Korea (Kim HS, bboddi0311@snu.ac.kr; Song YS,<br />

yssong@snu.ac.kr)<br />

3 Department of Obstetrics <strong>and</strong> Gynecology, Dongguk University Medical Center,<br />

Goyang 410-773, Republic of Korea (Lee CH, gynelee@dumc.or.kr)<br />

Background: The association between ovarian cancer <strong>and</strong> endometriosis has not been well<br />

established in previous studies. We investigated the impact of endometriosis on the<br />

development of ovarian cancer (OC) <strong>and</strong> prognosis in patients with OC using a meta-analysis.<br />

Methods: After we performed a MEDLINE search to identify all relevant studies between<br />

March 1945 <strong>and</strong> December 2010, we found 4 studies (3 case-control, 1 cohort; n=157,292)<br />

related with OC risk <strong>and</strong> 4 studies (1 case-control, 3 cohort; n=46,716) associated with<br />

prognosis in patients with OC among 2,390 potentially relevant studies. This meta-analysis<br />

was performed using the fixed-effect model because of no heterogeneity.<br />

Results: Endometriosis increased the risk of OC (OR, 1.42; 95% CI, 1.22-1.65). Although<br />

endometriosis did not influence the risk of OC in one cohort study (OR, 0.78; 95% CI, 0.25-<br />

2.44), 3 case-control studies showed that endometriosis was associated with the increased of<br />

OC (OR, 1.43; 95% CI, 1.23-1.67). On the other h<strong>and</strong>, endometriosis-associated OC (EAOC)<br />

showed improved overall survival (OS) when compared with non-EAOC (HR, 0.77; 95% CI,<br />

0.64-0.92). In sub-analyses based on the design of study, one case-control study demonstrated<br />

that EAOC was associated with improved OS (HR, 0.56; 95% CI, 0.36-0.87), <strong>and</strong> EAOC was<br />

also related with improved OS with marginal significance when compared with non-EAOC<br />

(HR, 0.82; 95% CI, 0.67-1.00) in 3 cohort studies.<br />

Conclusions: These findings suggest that women with endometriosis may have a higher risk<br />

of OC than those without endometriosis, whereas coexistent endometriosis may be associated<br />

with improved OS in patients with OC.<br />

153


Session 7: Cancer signaling networks Poster 17<br />

Impact of Body Mass Index on Overall Survival in Endometrial or Ovarian Cancer<br />

Survivors: A Meta-analysis<br />

Hee Seung Kim 1 , Chae Hyeong Lee 2 , Yong-Tark Jeon 3 , Yong Beom Kim 3 , Yong Sang<br />

Song 1<br />

1 Department of Obstetrics <strong>and</strong> Gynecology, Seoul National University College of<br />

Medicine, Seoul 110-744, Republic of Korea (Kim HS, bboddi0311@snu.ac.kr; Song YS,<br />

yssong@snu.ac.kr)<br />

2 Department of Obstetrics <strong>and</strong> Gynecology, Dongguk University Medical Center,<br />

Goyang 410-773, Republic of Korea (Lee CH, gynelee@dumc.or.kr)<br />

3 Department of Obstetrics <strong>and</strong> Gynecology, Seoul National University Bundang<br />

Hospital, Seoungnam 463-707, Republic of Korea (Kim YB, ybkimlh@snubh.org; Jeon<br />

YT, asidof@snubh.org)<br />

Background: The role of body mass index (BMI) has not been well-established for overall<br />

survival (OS) in patients with endometrial or ovarian cancer. To evaluated the impact of BMI<br />

in the patients, we performed a meta-analysis for comparing OS between (1) BMI


Session 7: Cancer signaling networks Poster 18<br />

Metformin inhibits gynaecological cancer cell growth through reduction of Dvl3 in<br />

Wnt/b-catenin signalling<br />

Virginia HT Kwan, David W Chan, Vincent WS Liu <strong>and</strong> Hextan YS Ngan<br />

Department of Obs. & Gyn., The University of Hong Kong, h0660139@hkusua.hku.hk<br />

Aberrant activation of Wnt/!-catenin signaling pathway is frequently associated with many<br />

human cancers. Dishevelled 3 (Dvl 3) is a key mediator of this pathway <strong>and</strong> functions as<br />

oncogenic factors in promoting cell proliferation.<br />

In this study, we found that the protein but not the mRNA level of Dvl3 is up-regulated in<br />

cervical cancer cell lines. Interestingly, treatment of Metformin can remarkably reduce the<br />

aberrant upregulation of Dvl3 in gynaecological cancer cell lines. Based on such discrepancy<br />

between basal mRNA <strong>and</strong> protein level, we hypothesized that the reduction of Dvl3 by<br />

metformin is due to the inhibition of protein synthesis through changes in AMPK/mTOR<br />

signaling pathway <strong>and</strong>/or promotion of proteasomal degradation machinery in these<br />

gynaecological cancer cells. On the other h<strong>and</strong>, treatment of proteosomal inhibitor (AM114)<br />

in cervical cancer <strong>and</strong> endometrial cancer cell lines can alleviate the metformin-mediated<br />

down-regulation of Dvl3, which strongly shows the involvement of proteasome degradation<br />

in Dvl3 down-regulation. Taken together, these data indicate that the upregulation of Dvl3 in<br />

gynaecological cancer cells is due to abnormal translational control <strong>and</strong> protein turnover.<br />

Meanwhile, metformin is also able to inhibit gynaecological cancer cell growth. In Colony<br />

formation <strong>and</strong> XTT assay, there was significant reduction of cell viability under the treatment<br />

of metformin in a dose-dependent manner. However, further biochemical studies on the<br />

responses of these pathways to Metformin in gynaecological cancer cells are warranted.<br />

155


Session 7: Cancer signaling networks Poster 19<br />

The effect of hCaMKIINalpha on TLR4-triggered cytokine production of colon cancer<br />

cells<br />

Nan Li, Chunmei Wang, Xingguang Liu, Qinghua, Zhang, Xuetao Cao<br />

National Key Laboratory of Medical Immunology <strong>and</strong> Institute of Immunology, Second<br />

Military Medical University, Shanghai 200433, P.R.China. Email: linan@immunol.org<br />

Increasing evidences suggest that chronic inflammation contributes to cancer development<br />

<strong>and</strong> progression. One of the underlying mechanisms is proposed that tumor cell-derived<br />

inflammatory <strong>and</strong> immunosuppressive cytokines contribute to tumor immune escape <strong>and</strong><br />

resistance to immunotherapy. Toll-like receptors (TLRs) have been implicated in tumor<br />

progression <strong>and</strong> metastasis. Our previous study showed that calcium/calmodulin-dependent<br />

protein kinase II (CaMKII)� promoted TLR-triggered proinflammatory cytokine in<br />

macrophages. hCaMKIINalpha, a novel CaMKII inhibitory protein identified by us,<br />

suppressed the growth of colon cancer cell by inducing cell cycle arrest in vitro <strong>and</strong> in vivo.<br />

Thus we wonder whether hCaMKIINalpha-mediated CaMKII inhibition affects TLR4triggered<br />

cytokine production of colon cancer cells for immune escape. In this study, we<br />

demonstrate that TLR4 is expressed on human colon cancer cell lines. TLR4 ligation<br />

promotes production of immunosuppressive cytokines IL-8 <strong>and</strong> VEGF. Overexpression of<br />

hCaMKIINalpha inhibits TLR4-triggered production of IL-8 <strong>and</strong> VEGF; H282R, constitutive<br />

activated CaMKII, significantly promotes TLR4-triggered IL-8 <strong>and</strong> VEGF secretion. In<br />

addition, we also observe that hCaMKIINalpha inhibits LPS-mediated activation of p-<br />

ERK1/2 <strong>and</strong> LPS-mediated TLR4 expression in SW620 cells. Furthermore, hCaMKIINalphamediated<br />

inhibition of ERK1/2 is necessary for suppression of TLR4-triggered IL-8 <strong>and</strong><br />

VEGF secretion. These results suggest that hCaMKIINalpha-mediated CaMKII inhibition<br />

might play important roles in the suppression TLR4-triggered metastasis <strong>and</strong> immune escape<br />

of human colon cancer cells by inhibiting immunosuppressive cytokine production.<br />

156


Session 7: Cancer signaling networks Poster 20<br />

Mechanism of action <strong>and</strong> anti-cancer activity of novel derivative of glycyrrhetic acid<br />

Logashenko E.B. 1 , Markov A.V. 1 , Salomatina O.V. 2 , Salakhutdinov N.F. 2 , Popova N.A. 3 ,<br />

Kaledin V.I. 3 , Nikolin V.P. 3 , Zenkova M.A. 1 .<br />

1 Institute of Chemical Biology <strong>and</strong> Fundamental Medicine SB RAS,<br />

Novosibirsk, Russian Federation, evg_log@niboch.nsc.ru<br />

2 Novosibirsk Institute of Organic Chemistry SB RAS, Novosibirsk, Russian Federation,<br />

3 Institute of Cytology <strong>and</strong> Genetics SB RAS, Novosibirsk, Russian Federation<br />

Glycyrrhetinic acid, aglycon of glycyrrhizin is abundant component of licorice root.<br />

Glycyrrhizin content in the triterpene extract of licorice root amounts to 90%. Recent reviews<br />

described the wide spectrum of glycyrrhetinic acid bioactivities, such as anti-inflammatory,<br />

antiviral, hepatoprotective, antitumor <strong>and</strong> immunomodulatory activity. Several studies<br />

reported that glycyrrhizin <strong>and</strong> glycyrrhetinic acid cause moderate cytotoxic <strong>and</strong> apoptotic<br />

effects on cancer cells, most of the results confirmed only moderate or low potency of the<br />

compounds. In attempt to prepare more potent analog we synthesized by directed<br />

modification of A, C <strong>and</strong> E rings of glycyrrhetic acid the methyl 2–cyano–3,12–dioxo–11–<br />

deoxo–18!H–glycyrrhet–1(2),9(11)–dienoate derivative (SG).<br />

The antitumor activity of the novel derivative was determined in vitro for different cancer cell<br />

lines by effects on cell growth by MTT assay. It was shown that SG is cytotoxic for cancer<br />

cells. IC50 varied from 5"10 -5 M (MCF-7 cell line) to 0,6x10 -6 M (#$-3-1cell line). Detailed<br />

study of mechanism of cells death showed that SG induces caspase–dependent apoptosis with<br />

disruption of mitochondrial potential. Exposure of the cells to glutathione that is an<br />

antioxidant completely prevents activity of SG.<br />

The antitumor activity in vivo was evaluated using Ehrlich's ascite tumor developed in<br />

%%57BR mice. Mice were twice intraperitoneally injected with SG (20 mg/kg). It was shown<br />

that SG administration leads to two fold decreasing of number of cancer cells in ascite as<br />

compared to control. Intraperitoneal injections of SG in mice with lymphosarcoma increase<br />

their live duration to 24 %.<br />

To increase solubility of SG we used Tween-80 <strong>and</strong> Cremophore–EL. Intraperitoneal<br />

injections of SG emulsions (50 mg/kg) with 10% Tween-80 <strong>and</strong> Cremophore–EL in mice<br />

with ascite tumor Krebs-2 supress tumor growth to 76 <strong>and</strong> 57%, respectively.<br />

Obtained data indicated that novel derivative of glycyrrhetic acid SG exhibits pronounced<br />

anti-cancer activity in vitro <strong>and</strong> in vivo.<br />

This work was supported by RAS programs “Molecular <strong>and</strong> cellular biology” <strong>and</strong><br />

“Fundamental sciences to medicine”, <strong>and</strong> by SB RAS (Interdisciplinary grant & 104).<br />

157


Session 7: Cancer signaling networks Poster 21<br />

Dendritic cells-based antitumor vaccines: opposite effects depending on application<br />

scheme<br />

Mironova N.L. 1 , Markov O.A. 1 , Popova N.A. 2 , Kaledin V.I. 2 , Nikolin V.P. 2 , Vlassov<br />

V.V. 1 , Zenkova M.A. 1<br />

1 Institute of Chemical Biology <strong>and</strong> Fundamental Medicine SB RAS, Lavrentieva ave. 8,<br />

Novosibirsk, 630090, mironova@niboch.nsc.ru<br />

2 Institute of Cytology <strong>and</strong> Genetics SB RAS, Lavrentieva ave. 11, Novosibirsk, 630090<br />

Dendritic cells (DC) are specialized antigen presenting cells that acquire, process, <strong>and</strong> present<br />

tumor-associated antigens to T cells for the induction of antigen-specific tumor immune<br />

responses. Triggering of effective antitumor immune response by ex vivo generated mature<br />

DC pulsed with tumor-associated antigens is the perspective approach to enhance the<br />

efficiency of anticancer therapy.<br />

Here the tumoricidal activity of DC depending on single or multiple antigen sources was<br />

studied in vitro <strong>and</strong> in vivo. We show that the efficacy of cytotoxic T lymphocytes (CTL)<br />

activated by DC pulsed with source of multiple antigens (further DC-MA) is 1.5-times higher<br />

than the efficacy of CTL generated by DC pulsed with source of a single antigen (further, DC<br />

SA).<br />

Anticancer potential of DC pulsed with tumor antigens was evaluated in mice model with<br />

Krebs-2 tumor. Administration of DC-MA as a vaccine prior to tumor transplantation resulted<br />

in the drastic suppression of CD8+ <strong>and</strong> CD4+ T cells proliferation <strong>and</strong> total absence of any<br />

antitumor effects. Administration of the same DC-MA after tumor transplantation resulted in<br />

2-fold retardation of primary tumor growth <strong>and</strong> was accompanied by enhanced proliferation<br />

of CD8+ T cells as well as significant increase of IFN-gamma production by factor 2.8. Thus,<br />

we revealed that DC can cause the opposite therapeutic effects depending on the time of DC<br />

administration. Attempts of DC application as a pre-exposure vaccine resulted in depletion of<br />

T-cell pool followed by break-off the cytotoxic immune response during tumor progression<br />

whereas therapy with DC upon tumor development had considerable antitumor effect.<br />

This work was supported by RAS programs “Molecular <strong>and</strong> cellular biology” <strong>and</strong><br />

“Fundamental sciences to medicine”, by RFBR grant 08-04-00753 <strong>and</strong> by Interdisciplinary<br />

grant of SB RAS (No. 15).<br />

158


Session 7: Cancer signaling networks Poster 22<br />

Glucose transport in Saccharomyces cerevisiae reveals heterogeneity of cell population<br />

Maria Nardelli 1 , Malkhey Verma 1 , Kathryn Blount 2 , Hans V. Westerhoff 1,2<br />

1<br />

Manchester Centre for Integrative Systems Biology, EPS, University of Manchester,<br />

131 Princess Street, M1 7DN, Manchester (UK)<br />

2<br />

DTC for Integrative Systems Biology, EPS, University of Manchester, 131 Princess<br />

Street, M1 7DN, Manchester (UK)<br />

Maria.Nardelli@manchester.ac.uk, Malkhey.Verma@manchester.ac.uk,<br />

Kathryn.Blount@postgrad.manchester.ac.uk, Hans.Westerhoff@manchester.ac.uk<br />

Saccharomyces cerevisiae is able to adjust to a wide range of glucose availability. Yeasts can<br />

modulate the expression of 17 different transporters to adapt <strong>and</strong> optimize the uptake of<br />

glucose. In our study we tested the ability of CEN-PK strain to express specific subsets of<br />

glucose transporters in response to the availability of glucose in their medium. To follow the<br />

uptake of glucose a fluorescent analogue, the 2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4yl)amino)-2-deoxyglucose<br />

(2NBDG), has been used. Yeast adapted to different amount of<br />

glucose <strong>and</strong> exposed to different concentrations of 2NBDG were analysed.<br />

The uptake of the molecule was followed measuring the fluorescence associated with the<br />

cells. The analysis of yeast cells via flow cytometry revealed the presence of heterogeneity in<br />

the yeast population depending on the growth condition <strong>and</strong> concentration of 2NBDG used.<br />

The presence of different yeast subpopulations indicates that the glucose transporter<br />

expression <strong>and</strong>/or activity require fine tuning. Underst<strong>and</strong>ing this aspect of the glucose uptake<br />

in yeast may provide further information to target the glucose transporters for cancer<br />

therapeutics.<br />

159


Session 7: Cancer signaling networks Poster 23<br />

Cinnamic acid effects on cultured human melanocytes <strong>and</strong> melanoma derived cells:<br />

evaluation of the specificity of its antitumor potential<br />

Ev<strong>and</strong>ro L O Niero, Camila Lau<strong>and</strong>, Gláucia M Machado-Santelli<br />

Laboratory of Cell <strong>and</strong> Molecular Biology, Department of Cell <strong>and</strong> Developmental<br />

Biology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu<br />

Prestes 1524, 05508-900, São Paulo, SP, Brazil, eloniero@usp.br<br />

Phenolic acids, commonly found in plants, <strong>and</strong> their derivatives have been largely studied<br />

because of their antitumor activities <strong>and</strong> low cytotoxicity in normal cells. Thus, the present<br />

work aimed to evaluate the specificity of the cinnamic acid (CA) (a phenolic acid)<br />

cytotoxicity in human melanocytes of blue nevus (NGM) <strong>and</strong> human melanoma derived cells<br />

(HT-144) just following cell cycle arrest, cell death <strong>and</strong> formation of nuclear aberration. CA<br />

was very efficient in inhibiting cell growth of melanoma cells compared to melanocytes<br />

according to MTT assay. The inhibition was probably associated with DNA damage leading<br />

to DNA synthesis inhibition as showed by BrdU incorporation assay, induction of nuclear<br />

aberrations (micronucleation, binucleation <strong>and</strong> multinucleation) leading to apoptosis in both<br />

cell lines. BrdU incorporation assay indicates cell cycle arrest in both cell lines induced by<br />

0.4mM of CA treatment. We also identified induction of cell death after 24 hours of<br />

treatment, especially in HT-144 cells, according to activated caspase-9 assay. CA at 3.2 mM<br />

also induced formation of nuclear buds <strong>and</strong> micronuclei in NGM cells but the effects were<br />

milder than in melanoma cells. This data could be associated with higher genetic instability of<br />

HT-144 cells. We identified, for example, many copies of chromosome 9 as showed by the<br />

presence of several positive signals to centromeric regions in melanoma cells. Nevertheless,<br />

we observed two or less signals to p16 in HT-144 cell line while NGM cells showed signals<br />

to two copies of both regions. High concentration of CA (3.2 mM) induced formation of<br />

nuclear buds <strong>and</strong> micronuclei in NGM cells. Together, our results indicate a high cytotoxic<br />

potential of CA to HT-144 cells compared to NGM. The genotoxic effects showed by both<br />

cells after exposure to 3.2 mM of CA indicates that the mechanisms of action of CA must be<br />

further investigated, but it could be related to its antioxidant activity <strong>and</strong> capacity to induce<br />

DNA damage.<br />

160


Session 7: Cancer signaling networks Poster 24<br />

Regulation of HGF-induced cell invasion through EGR1 in Hepatocellular Carcinoma<br />

Ozen E., Gozukizil A., Erdal E., Atabey N.<br />

Dokuz Eylul University School of Medicine Depertmant of Medical Biology <strong>and</strong><br />

Genetics, Inciralti Izmir , Turkey evinozen@hotmail.com<br />

Hepatocyte growth factor (HGF) is a mesenchyme-derived pleotrophic growth factor<br />

that elicits mitogenic, morphogenic <strong>and</strong> motogenic responses in wide variety of cells by<br />

binding to its receptor, c-Met receptor tyrosine kinase. HGF/cMet pathway induces activation<br />

of several downstream signaling cascades, such as mitogen-activated protein kinase (MAPK)<br />

<strong>and</strong> phosphatidyl-inositol 3-kinase (PI3K) pathways then regulates tissue regeneration as well<br />

as invasion <strong>and</strong> metastasis in hepatocellular carcinoma (HCC). Furthermore, it is known that<br />

matrix metalloproteinases (MMP) activation is important step in HGF-induced, invasion <strong>and</strong><br />

metastasis in different models. However, the molecular mechanism behind the regulation of<br />

MMP activation on the HGF-induced invasion has not been well understood yet in HCC.<br />

Recently, it has been reported that Early Growth Response Factor 1 (EGR1) plays a key role<br />

in HGF-induced angiogenesis <strong>and</strong> cell scattering in HCC. So we hypothesized that EGR1<br />

might be a downstream transcription factor that regulates HGF-induced invasion through<br />

activation of MMPs during hepatocarcinogenesis.<br />

In this study it has been first shown that activation of HGF/c-Met signaling by treatment of<br />

HGF induces significantly transcriptional activity of EGR1 in a time dependent manner <strong>and</strong><br />

inhibition of HGF/c-Met signaling by treatment of SU11274 decreases expression level of<br />

EGR1 in SKHep1, HCC cell line. Also, it has been demonstrated that activation of HGF/c-<br />

Met signaling activates MAPK signaling molecules <strong>and</strong> cell invasion in this cells as expected.<br />

Expression of EGR1 has been further stably knock-down by using inducible pSUPERIOR<br />

system in SKHep1. Silencing of EGR1 caused significant decrease in HGF-mediated<br />

invasion. It also resulted in decrease in HGF-mediated activation of both MMP-2 <strong>and</strong> MMP-<br />

9, in parallel, decrease in expression of MT1-MMP in SKHep1 cell.<br />

As a conclusion, we found that EGR1 activates HGF-induced cell invasion possibly through<br />

the regulation of MMPs in HCC. In future, this mechanism may provide a way to develop<br />

new drug targets in the treatment of HCC.<br />

In this study, we found that HGF induces invasion of SK-Hep-1 cell lines by activation of<br />

MMP’s through EGR1.<br />

Key words: HGF, invasion, EGR1, MMPs, HCC.<br />

161


Session 7: Cancer signaling networks Poster 25<br />

Tumor infiltrating lymphocytes as a prognostic factor after the radical surgery of<br />

colorectal carcinoma<br />

Authors: Pitule P.(1,2), Vycital O.(1), Cedikova M.(2), Bruha J.(1), Liska V(1), Treska<br />

V.(1), Daum O.(3)<br />

(1) Department of Surgery, (2) Department of Histology <strong>and</strong> Embryology, (3)<br />

Department of Pathological Anathomy, Faculty of Medicine in Pilsen, Charles<br />

University<br />

email adress of the first author: pitulep@seznam.cz<br />

Introduction: Tumor infiltrating lymphocytes (TIL) are important factor which corresponds<br />

with the ability of the organism to attenuate the development of the malignity.<br />

Aim of the study: We correlated the broadly used clinical <strong>and</strong> histopathological factors with<br />

the presence of TIL in the tumor. The goal of the study was to identify patients with the high<br />

risk of the poor overall survival (OS) <strong>and</strong> early reccurence (short disease-free interval, DFI)<br />

of the malignity after the radical surgical treatment.<br />

Methods: The set of 150 tumor samples was analysed for several factors – pre-operational<br />

leukocytosis, radicality of the surgery, post-operational complications, oncological treatment,<br />

tumor grading <strong>and</strong> staging, morphological features of the tumor, characterisation <strong>and</strong><br />

quantification of the TIL <strong>and</strong> the reactive changes in the lymp nodes (LN). All of these factors<br />

were statistically correlated with the OS <strong>and</strong> DFI of the patients. We have also evaluated<br />

immunohistochemical positivity of TIL (CD4 + ,CD8 + ).<br />

Results: We have included in the study 93 men <strong>and</strong> 57 women, with the 1, 3 <strong>and</strong> 5 years OS<br />

92,2%, 76,5% <strong>and</strong> 70,2% respectivelly, <strong>and</strong> 1, 3 <strong>and</strong> 5 years DFI 85,3%, 64,3% <strong>and</strong> 49,4%<br />

respectivelly. Endolyphatic infiltration <strong>and</strong> metastatic inflitration of the LN were confirmed as<br />

a negative prognostic factor for OS. N2 stage descrease the OS 9,3 times <strong>and</strong> DFI 5 times.<br />

Crohn-like peritumoral lymphocytic infiltration <strong>and</strong> follicular hyperplasia were confirmed as<br />

a protective factor. CD8 + <strong>and</strong> CD4 + TIL were confirmed as a positive prognostic factor <strong>and</strong><br />

their presence leads to the increase in OS <strong>and</strong> DFI. The CD8 + /CD4 + ratio was not significantly<br />

confirmed as a prognostic factor. Perineural infiltration was confirmed as a prognostic factor<br />

of early reccurence. Higher infiltration by CD8 + TIL was found to be positive prognostic<br />

factor which increase the DFI.<br />

The study was supported by grants VZ MSM 0021620819 <strong>and</strong> IGA MZ CR 10230.<br />

162


Session 7: Cancer signaling networks Poster 26<br />

Selective cytotoxic activity of a bifunctional chimeric protein containing a ribosome<br />

inactivating protein (PD-L4) <strong>and</strong> a serine protease inhibitor (WSCI).<br />

Rachele Tamburino 1 , Elio Pizzo 2 , Elia Poerio 3 <strong>and</strong> Antimo Di Maro 1<br />

1<br />

Dipartimento di Scienze della Vita, Seconda Università di Napoli, via Vivaldi, 43, I-<br />

81100 Caserta, Italy<br />

rachele.tamburino@unina2.it; antimo.dimaro@unina2.it<br />

2<br />

Dipartimento di Biologia Strutturale e Funzionale, Università “Federico II”, via<br />

Cinthia, I-80126 Napoli, Italy elipizzo@unina.it<br />

3<br />

Dipartimento di Agrobiologia e Agrochimica, Università della Tuscia, via S. Camillo de<br />

Lellis, I-01100 Viterbo, Italy poerio@unitus.it<br />

A bifunctional chimeric protein, potentially able to biotechnological applications, has been<br />

designed <strong>and</strong> expressed in E. coli cells. The first domain corresponds to the toxic/antiviral<br />

protein PD-L4 type 1 RIP, firstly isolated from P. dioica L. leaves. The second domain<br />

corresponds to the wheat protein inhibitor WSCI which is able to interfere with subtilisin,<br />

pancreatic chymotrypsin <strong>and</strong> chymotrypsin-like activities isolated from the midgut of a<br />

number phytophagous insect larvae. The chimeric construct, pd-l4-cDNA-oligonucleotide<br />

linker-wsci-cDNA, was cloned in the expression vector pET22b <strong>and</strong> employed in trasforming<br />

E. coli cells (strain BL21-DE3). The chimeric protein PD-L4/WSCI was expressed in E. coli<br />

<strong>and</strong> recovered from the inclusion bodies. Both t<strong>and</strong>em domains (PD-L4 <strong>and</strong> WSCI) retained<br />

their original activities. Cytotoxicity assays on SVT2 cells revealed that the chimeric protein<br />

strongly affected their viability; the recorded levels of toxicity were greater than those<br />

observed for PD-L4. Somehow, the presence of the non-toxic C-terminal domain WSCI<br />

contributed to enhance cytotoxicity of the bifunctional chimeric product. Surprisingly,<br />

cytotoxicity assays revealed that the chimera did not affect 3T3 cells viability.<br />

163


Session 7: Cancer signaling networks Poster 27<br />

New benzophenone polyphenols with potential chemoprotective effects<br />

Tzvetomira Tzanova a,c , Mariana Gerova b , Ognyan Petrov b , Margarita Karaivanova a ,<br />

Denyse Bagrel c<br />

a Medical University of Sofia, Faculty of Pharmacy, Department of Pharmacology <strong>and</strong><br />

Toxicology, 2 Dunav street, Sofia 1000, Bulgaria, tztzanova@gmail.com,<br />

mkaraivanova2001@yahoo.com<br />

b University of Sofia St. Kliment Ohridski, Faculty of Chemistry, Department of Applied<br />

Organic Chemistry, 1 James Bourchier Avenue, 1164 Sofia, Bulgaria,<br />

OPetrov@chem.uni-sofia.bg, ohtmg@chem.uni-sofia.bg<br />

c Laboratoire d'Ingénierie Moléculaire et Biochimie Pharmacologique, Université Paul<br />

Verlaine – Metz, FR CNRS 2843, UFR SciFA, Campus Bridoux, rue du Général<br />

Delestraint, 57070 Metz, France<br />

bagrel@univ-metz.fr<br />

Among phenolics, natural hydroxybenzophenones are biologically active metabolites present<br />

in plants <strong>and</strong> especially in Guttiferae family. Therefore we tested the antioxidant properties of<br />

novel natural benzophenones isolated from Hypericum annulatum. One of them –<br />

hypericophenonoside (Hd19) displayed a high in vitro antioxidant <strong>and</strong> antiradical activity.<br />

The cytoprotective effects of Hd19 <strong>and</strong> silymarine, our natural reference compound, were<br />

tested on breast cell lines – ie the cancerous MCF7 <strong>and</strong> the non-cancerous hTERT-HME1.<br />

The hypericophenonoside decreases the intracellular oxidative stress induced by cisplatin <strong>and</strong><br />

adriamycin, without affecting viability, apoptosis, necrosis <strong>and</strong> cell cycle progression<br />

modulated by these chemotherapeutic agents. A very promising fact was Hd19 <strong>and</strong> silymarine<br />

protection on the proximal renal tubular cells against cisplatin toxicity, its chief dose-limiting<br />

side effect.<br />

These interesting biological effects led us to design <strong>and</strong> synthesize novel benzophenones<br />

containing a benzothiazolone moiety which could adjoin important pharmacological<br />

activities. The protective potential of three novel synthetic benzophenones was characterized.<br />

One of them named 6c exerted a higher antioxidant activity than Trolox, our synthetic<br />

reference compound. An important increase in cell viability <strong>and</strong> GSH level was observed in<br />

hTERT-HME1 cells. 6c showed high protective potential against oxidative stress <strong>and</strong> cell<br />

mortality induced by tBHP. Moreover, we observed a significant protection in<br />

cardiomyoblastic H9c2 cells, which may be of great interest in the frame of novel antioxidant<br />

strategies for the limitation of anthracycline cardiac toxicity.<br />

164


Posters are classified by session<br />

<strong>and</strong> then in alphabetical order (PRESENTING AUTHOR)<br />

(Late breaking abstracts are at the end of the <strong>book</strong>)<br />

Session 8: Gene expression networks in health<br />

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

166


Session 8: Gene expression networks in health <strong>and</strong> disease Poster 1<br />

Dictyostelium discoideum as a model for Mucolipidosis Type IV<br />

Claire Y Allan 1 , Paul R Fisher 1<br />

Department of Microbiology, La Trobe University, Melbourne, AUSTRALIA<br />

cyallan@students.latrobe.edu.au<br />

Mucolipidosis Type IV (ML4) is a neurological lysosomal disease, which results in severe<br />

mental retardation, retinal degeneration <strong>and</strong> achlorhydria. Loss of function mutations in<br />

Mucolipin-1 (TRPML1), are responsible for ML4. TRPML1 is a member of the Transient<br />

Receptor Potential family of cation channels, <strong>and</strong> is involved in membrane trafficking along<br />

the endocytic pathway. The phenotypes associated with ML4 could result from<br />

endosomal/lysosomal dysfunction, as enlarged acidic compartments are formed as a result of<br />

defective lysosomal biogenesis <strong>and</strong> trafficking. Altered Ca2+ signalling associated with lack<br />

of function of TRPML1 may contribute to the pathogenesis of the disease, however the<br />

mechanisms of TRPML’s function in the endocytic pathway are not well understood. For this<br />

study, a model for ML4 has been created in the eukaryotic slime mould Dictyostelium<br />

discoideum. The D. discoideum mucolipin homologue was cloned to over-express <strong>and</strong> knock<br />

down the protein, <strong>and</strong> the resulting phenotypes showed similarities to those of ML4 patients.<br />

Immunofluorescence indicates that the protein is localised to acidic compartments <strong>and</strong> when<br />

the protein was both over- <strong>and</strong> underexpressed a build up of acidic compartments was<br />

observed. Observation of fruiting body morphologies showed a thickening of the basal disk<br />

<strong>and</strong> stalk. Since the stalk <strong>and</strong> basal disk in Dictyostelium are the endpoints of a form of<br />

programmed, autophagic cell death, this increased stalk <strong>and</strong> disk differentiation suggests<br />

increased autophagy, another phenotype associated with the human disease. The involvement<br />

of mucolipin in the endocytic pathway was demonstrated, as uptake of nutrients via<br />

phagocytosis, <strong>and</strong> subsequently growth rates, are increased in strains both overexpressing <strong>and</strong><br />

underexpressing mucolipin. Rates of pinocytosis in knockdown strains were also increased,<br />

however, interestingly, the growth in liquid medium was slightly slower, indicating<br />

differences in mucolipin involvement in the processing of macropinosomes <strong>and</strong> phagosomes.<br />

Alterations in Ca2+ signalling, have been observed in both over <strong>and</strong> underexpressing strains.<br />

These variations in signalling may be associated with the presenting phenotypes, as Ca2+<br />

signalling is known to be involved in fusion <strong>and</strong> biogenesis of endocytic vesicles, <strong>and</strong><br />

especially with formation of the phagocytic cup. Combined, all observations suggest that D.<br />

discoideum may be a useful model to study potential therapies for ML4.<br />

167


Session 8: Gene expression networks in health <strong>and</strong> disease Poster 3<br />

Mediterranean Fever Gene Mutation Profiling of 3051 Turkish Patients with Periodic<br />

Fever<br />

AFIG BERDELI 1 , SINEM NALBANTOGLU 1 , DEMET TIGLI 1 , MERVE ATAN 1 ,<br />

PERVIN TOPARLAK 1 , AYBEN PASOLAR 1 , ILKAY DEMIREL 1 , FATMA AKYIGIT 1<br />

1. Ege University Faculty of Medicine, Child Hospital Molecular Medicine Laboratory,<br />

35100, Bornova, izmir, Turkey.<br />

Familial Mediterranean Fever (FMF, MIM249100) is an autoinflammatory disorder<br />

characterized by seemingly unprovoked episodes of inflammation in the absence of high-titer<br />

autoantibodies or antigen-specific T cells. 16p13.3 chromosomally located MEFV<br />

(Mediterranean Fever) gene encoding the 781 amino acid protein “pyrin” is the responsible<br />

gene for this monogenic Mendelian disease. Pyrin is a component of inflammasome complex<br />

whose exact function on inflammatory stimuli is still unknown. To date, mutations were<br />

found in exons 1, 2, 3, 5, 9 <strong>and</strong> 10 of the MEFV gene, <strong>and</strong> exons 2 <strong>and</strong> 10 constitutes the socalled<br />

mutational `hot spots'. The objective of this study was to perform the MEFV<br />

(Mediterranean Fever) gene sequencing <strong>and</strong> to establish the genotype-phenotype relation in<br />

Turkish patients with definitive, posssible <strong>and</strong> suspicious FMF diagnosis. Mutation analysis<br />

of exons 2, 3, 5, <strong>and</strong> 10 of MEFV gene was carried out in 3051 patients <strong>and</strong> relatives of<br />

unrelated families by DNA sequencing analysis. Additionally, for patients who do not have<br />

mutations in exons 2, 3, 5, <strong>and</strong> 10; DNA sequencing was performed in exons 1, 7, 8 <strong>and</strong> 9, as<br />

well. In the DNA sequencing analysis, 67 different genotypes involving 10 novel mutations<br />

<strong>and</strong> 1 novel SNP in exons 2, 3, 5, 9, <strong>and</strong> 10 including R151S, S166L, G340R, P350R, G456A,<br />

Y471X, S503C, I506V, K695N, L709R, <strong>and</strong> P588P, G219G, were characterized <strong>and</strong><br />

registered in INFEVERS (database of hereditary autoinflammatory disorders mutations).<br />

Missense/nonsense <strong>and</strong> synonymous amino acid variations accounted for 37% <strong>and</strong> 55.8% of<br />

MEFV chromosomes (7.1% without any mutation or polymorphism), respectively. Through<br />

the missense mutations; 114 were homozygous, 257 were compound <strong>and</strong> complex<br />

heterozygous, <strong>and</strong> 760 were heterozygous for one mutation. Allelic frequencies of M694V,<br />

E148Q, V726A, <strong>and</strong> M680I accounted for 38,3; 21,3; 11,1; <strong>and</strong> 10,9%, respectively; followed<br />

by P369S, R408Q, K695R, M694I, R761H accounting for 4,3; 3,5; 2,8; <strong>and</strong> 1,6%.<br />

Significantly, 760 patients of the 1131 mutation positive group (67%), have only one single<br />

mutation (only one mutated MEFV) receiving colchicine therapy with whom several SNPs<br />

including D102D, D103D, P124P, G138G, A165A, P180P, R202Q, R314R, S363S, E474E,<br />

Q476Q, D510D, R501R, I506I, P588P, S683S, <strong>and</strong> P706P accompany.<br />

In conclusion, if screening for the most common mutations does not reveal any known<br />

common disease causing mutations <strong>and</strong> for asymptomatic individuals in particularly for the at<br />

risk populations, spesific DNA sequence mutation screening covering entire coding exons<br />

should need to be considered.<br />

169


Session 8: Gene expression networks in health <strong>and</strong> disease Poster 4<br />

Regulation of Lynch syndrome-related DNA mismatch repair heterodimer MutLalpha<br />

by phosphorylation?<br />

Angela Brieger, S<strong>and</strong>ra Passmann, Stefan Zeuzem <strong>and</strong> Jörg Trojan<br />

Medical Clinic I, Biomedical Research Laboratory, Goethe-University, Frankfurt a.M.,<br />

Germany<br />

Lynch syndrome, a hereditary disease associated with many different tumor types, is caused<br />

by mutations in DNA mismatch repair (MMR)-genes. 50% of these mutations are detected in<br />

the MLH1 protein. The heterodimer MutLalpha, which is formed by MLH1 <strong>and</strong> PMS2,<br />

coordinates a series of key events in the MMR mechanism. In addition to MMR, MutLalpha<br />

is involved in many other cellular processes as the regulation of cell cycle checkpoints <strong>and</strong><br />

apoptosis most likely signaling DNA damage to downstream pathways. A sophisticated<br />

regulation of the different MutLalpha functions might be important as well as reasonable. In<br />

order to focus on MutLalpha modulation, we analyzed MLH1 <strong>and</strong> PMS2 for phosphorylation<br />

sites using different computational systems <strong>and</strong> detected multiple potential loci <strong>and</strong><br />

corresponding kinases for both genes. Using phospho-specific purification columns we were<br />

able to isolate unphosphorylated <strong>and</strong> phosphorylated MLH1. In addition to this, Westernblot<br />

analysis of 2-D gels resolved MLH1 into several different distinct forms detected in lysates<br />

from HEK293 cells. Furthermore, we detected phosphorylation of purified MLH1 by<br />

PKCdelta, one of the computer-supported most probable MutLalpha kinases. Ongoing<br />

projects are directed towards the underst<strong>and</strong>ing how phosphorylation might be capable to<br />

switch MutLalpha function between MMR <strong>and</strong> DNA damage signaling.<br />

170


Session 8: Gene expression networks in health <strong>and</strong> disease Poster 5<br />

Interferences between Protein S <strong>and</strong> angiogenic factors signalling pathway in<br />

endothelial cells<br />

Sylvain Fraineau, Julie Talbot, Michel Philippe, Arnaud Monvoisin <strong>and</strong> Omar<br />

Benzakour<br />

Institut de Physiologie et de Biologie Cellulaires UMR CNRS 6187, Université de<br />

Poitiers, Poitiers, France<br />

Protein S is a plasma vitamin K-dependent glycoprotein, mainly known as a negative<br />

regulator of blood coagulation. Protein S heterozygous mice have been shown to display<br />

defects in vessel development <strong>and</strong> function 1 . Protein S secretion by endothelial cells <strong>and</strong> its<br />

substantial contribution to circulating protein S has been previously described. Expression of<br />

the putative tyrosine kinase receptors for both protein S <strong>and</strong> its structural homolog Gas6,<br />

termed TAM 2 (Tyro3, Axl <strong>and</strong> Mer) in endothelial cells was reported 3 . TAM receptors are<br />

involved in many cellular processes such as endothelial cells survival <strong>and</strong> morphogenesis 4 .<br />

Inhibition of VEGF/VEGFR-2 pathway by Axl/Gas6 signalling, involving the tyrosine<br />

phosphatase SHP-2 (SH-2 domain-containing tyrosine phosphatase), has been described 5 The<br />

major aim of our study was to assess if Gas6 homolog, protein S may also be involved in<br />

inhibiting VEGF/VEGFR-2-induced endothelial cells activation.<br />

We show that Mer stimulation by protein S inhibits vascular endothelial growth factor<br />

(VEGF)- <strong>and</strong> fibroblast growth factor 2 (FGF-2)-induced endothelial cells proliferation.<br />

Protein S also inhibits VEGF-inducedVEGFR-2. Proliferation assays using NSC 87877, a<br />

pharmacological inhibitor of tyrosine phosphatases SHP-1 <strong>and</strong> 2, suggested their implication<br />

in the inhibition by protein S/Mer complex of VEGFR-2 activation. Protein S also inhibited<br />

VEGF <strong>and</strong> FGF-2-activated downstream signalling pathways including inhibition of VEGF<br />

<strong>and</strong> FGF-2-induced MAP kinase Erk 1/2 phosphorylation. Interestingly, on its own, Protein S<br />

induced in HUVECs MAP kinase Erk 1/2 activation <strong>and</strong> cell proliferation which was<br />

suppressed in the presence of Erk 1/2 pathway inhibitors.<br />

Altogether, these data argue for new roles for the plasma vitamin K-dependent factor,<br />

protein S in regulating endothelial cells proliferation occurring during angiogenesis.<br />

1 Burstyn-Cohen T. et al., J Clin Invest (2009)<br />

2 Stitt T.N. et al., Cell (1995)<br />

3 Burnier L. et al., Blood (2010)<br />

4 O'Donnell K. et al., Am J Pathol (1999)<br />

5 Gallicchio M. et al., Blood (2005)<br />

171


Session 8: Gene expression networks in health <strong>and</strong> disease Poster 6<br />

Tungsten carbide cobalt nanoparticles exert hypoxia-like effects on the gene expression<br />

level in human keratinocytes<br />

Wibke Busch 1 , Dana Kühnel 1 , Kristin Schirmer 2 , Stefan Scholz 1<br />

1 Helmholtz-Centre for Environmental Research Leipzig - UFZ, Department<br />

Bioanalytical Ecotoxicology, Permoserstr. 15, 04318 Leipzig, Germany<br />

2 Eawag, Swiss Federal Institute of Aquatic Science <strong>and</strong> Technology, 8600 Dübendorf,<br />

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

Tungsten carbide (WC) <strong>and</strong> tungsten carbide cobalt (WC-Co) nanoparticles are of<br />

occupational health relevance because of the increasing usage in hard metal industries. Earlier<br />

studies showed an enhanced toxic potential for WC-Co compared to WC or cobalt ions alone.<br />

Therefore, we investigated the impact of these particles on the global gene expression level in<br />

a human keratinocyte cell line (HaCaT) <strong>and</strong> compared the effects to those of cobalt ions<br />

(CoCl2). Changes in gene expression patterns were analysed after 3 hours <strong>and</strong> 3 days of<br />

exposure using whole genome microarrays <strong>and</strong> were confirmed by qPCR. WC nanoparticles<br />

exerted very little effects on transcription patterns. In contrast, WC-Co nanoparticles caused<br />

significant transcriptional changes that were similar to those provoked by CoCl2. However,<br />

the transcriptional response - both in terms of the number of affected genes as well as with<br />

respect to the relative fold changes - appeared to be more pronounced for CoCl2. Gene set<br />

enrichment analyses revealed that the differentially expressed genes were related to hypoxia<br />

response, carbohydrate metabolism <strong>and</strong> endocrine pathways. Furthermore, the identified<br />

genes were targets of several transcription factors whereas a majority of effects was found to<br />

be mediated by the transcription factor HIF1 (hypoxia inducible factor 1) which is known to<br />

be stabilised by cobalt ions. In conclusion, we show that WC nanoparticles caused low<br />

transcriptional responses while WC-Co nanoparticles are able to exert responses similar to<br />

that of free cobalt ions, particularly the induction of hypoxia-like effects via interactions with<br />

HIF1! in human keratinocytes. Since almost no particle specific expression patterns were<br />

found, this study shows that particle uptake is hardly detectable on the gene expression level<br />

<strong>and</strong> that leached ions are of great relevance in nanoparticle toxicity.<br />

!"#$%&'(&)*+&!,-&.'/012$#&34546&55789<br />

172


Session 8: Gene expression networks in health <strong>and</strong> disease Poster 7<br />

Thalassemia mouse model for human !-globin gene IVS-I-6 mutation<br />

Giulia Breveglieri, 1 Alessia Finotti, 2 Irene Mancini, 2 Nicoletta Bianchi, 2 Ilaria<br />

Lampronti, 2 Francesca Salvatori, 1 Giordana Feriotto, 2 Cristina Zuccato, 2 Monica<br />

Borgatti, 2 Gianni Car<strong>and</strong>ina, 3 Claudia Mel<strong>and</strong>ri, 3 Fiorella Altruda, 4 Sharmila<br />

Fagoonee, 4 Maddalena Iannicella, 4 Laura Breda, 5 Stefano Rivella 5 <strong>and</strong> Roberto<br />

Gambari 1,2<br />

1 Laboratory for the Development of Pharmacological <strong>and</strong> Pharmacogenomic Therapy<br />

of Thalassemia, Biotechnology Center, Ferrara University, Italy; e-mail:gam@unife.it;<br />

2 BioPharmaNet, Department of Biochemistry <strong>and</strong> Molecular Biology, Ferrara<br />

University, Italy; 3 Laboratory for Chemical <strong>and</strong> Clinical Analysis <strong>and</strong> Microbiology,<br />

University Hospital, Ferrara, Italy; 4 Molecular Biotechnology Center, University of<br />

Turin, Italy; 5 Division of Hematology-Oncology, Department of Pediatrics, Weill Cornell<br />

Medical College, New York, NY, USA<br />

In !-thalassemias, mutations of the !-globin gene or its regulatory regions cause absence (!°)<br />

or reduced (! + ) synthesis of !-globin chains, associated with a corresponding excess of the<br />

complementary "-globins. Mouse models for the different mutations (>200) of the !-globin<br />

gene causing thalassemia are very important to test in vivo the activity of novel mutationspecific<br />

therapeutic approaches. The IVS-I-6 mutation is the most frequent in the middle-east<br />

region <strong>and</strong> recurrent in Italy <strong>and</strong> Greece. This mutation leads to the activation of three criptic<br />

splicing sites, which generate three aberrantly spliced RNAs. In this study we report the<br />

production <strong>and</strong> characterization of a transgenic mouse line carrying a human !-globin gene<br />

containing the IVS-I-6 thalassemia point mutation. Our TG-!-IVS-I-6 mouse model displays<br />

a tissue specific expression of the transgene (fully overlapping with that of the endogenous<br />

murine !-globin gene) <strong>and</strong> produces normally spliced human !-globin mRNA (giving rise to<br />

!-globin production <strong>and</strong> formation of a human-mouse tetrameric chimeric hemoglobin " m 2! h )<br />

<strong>and</strong> unspliced aberrant !-globin-IVS-I-6 RNA in blood. Therefore, the TG!-IVS-I-6 mouse<br />

might be used as an in vivo model to characterize the effects of antisense ODN targeting the<br />

criptic site responsible for the generation of aberrantly spliced human !-globin RNA<br />

sequences.<br />

Supported by Telethon, UE Ithanet Project, Fondazione Cassa di Risparmio di Padova e<br />

Rovigo, AVLT (Rovigo Association for the Fight Against Thalassemia).<br />

173


Session 8: Gene expression networks in health <strong>and</strong> disease Poster 8<br />

Digoxin <strong>and</strong> ouabain increase the synthesis of cholesterol in human liver cells.<br />

Joanna Kopecka, Ivana Campia, Amalia Bosia <strong>and</strong> Chiara Riganti.<br />

Department of Genetics, Biology <strong>and</strong> Biochemistry, University of Torino <strong>and</strong> Research<br />

Center on Experimental Medicine (CeRMS), Via Santena, 5/bis, 10126, Torino, Italy.<br />

joanna.kopecka@unito.it, ivana.campia@unito.it, amalia.bosia@unito.it,<br />

chiara.riganti@unito.it.<br />

Cardioactive glycosides, like digoxin <strong>and</strong> ouabain inhibit the Na + /K + -ATPase: forcing the<br />

Na + /Ca 2+ exchanger to extrude Na + in exchange with Ca 2+ <strong>and</strong> thus increasing the [Ca 2+ ]i<br />

concentration, they exert a strong inotropic effect on heart. For this reason they are used in the<br />

treatment of congestive heart failure <strong>and</strong> cardiac arrhythmias. Recent findings have shown<br />

that cardioactive glycosides exert pleiotropic effects on cells, independently from the<br />

inhibition of Na + /K + -ATPase: for instance, they affect the cell response to hypoxia, modulate<br />

several signalling pathways involved in cell death <strong>and</strong> proliferation, regulate the transcription<br />

of different genes. Moreover, having a sterol-like chemical structure, cardioactive glycosides<br />

can modulate the metabolism of steroid hormones in vivo, probably taking part into fine feedback<br />

mechanisms. Until now no evidence has been provided about the effects of glycosides<br />

on the synthesis of the steroids hormones precursor, that is cholesterol.<br />

Here we report that digoxin <strong>and</strong> ouabain increase the synthesis of cholesterol in human liver<br />

HepG2 cells, enhancing the activity <strong>and</strong> the expression of the 3-hydroxy-3-methylglutarylcoenzyme<br />

A reductase (HMGCR), the rate-limiting enzyme of sterol biosynthesis. This effect<br />

was mediated by the binding of the sterol regulatory element binding protein-2 (SREBP-2) to<br />

the HMGCR promoter, <strong>and</strong> was lost in cells silenced for SREBP-2 or loaded with increasing<br />

amounts of cholesterol. Digoxin <strong>and</strong> ouabain competed with cholesterol for the binding on the<br />

sterol sensor domain of SREBP-cleavage-activating protein (SCAP), thus appearing as new<br />

critical regulators of cholesterol synthesis in human liver cells.<br />

Our results might have physiopathological significance: for instance it is known that different<br />

levels of cholesterol in plasma membranes may modify the activity of Na + /K + -ATPase <strong>and</strong> its<br />

affinity for inhibitors, thus affecting the therapeutic efficacy of glycosides. Since alterations<br />

of cholesterol levels <strong>and</strong> heart diseases are frequently associated, the potential cross-talk<br />

between digoxin <strong>and</strong> endogenous synthesis of cholesterol should be taken into consideration,<br />

because it may critically modulate the clinical response to cardioactive glycosides.<br />

174


Session 8: Gene expression networks in health <strong>and</strong> disease Poster 9<br />

Towards domino systems biology <strong>and</strong> an integrated ATP-centric model of regulation in<br />

Saccharomyces cerevisiae<br />

Malkhey Verma 1 , Maria Nardelli 1 <strong>and</strong> Hans V Westerhoff 1,2<br />

1 Manchester Interdisciplinary Biocentre, The University of Manchester, UK<br />

2 MCISB, The University of Manchester, UK<br />

Email: malkhey.verma@manchester.ac.uk; hans.westerhoff@manchester.ac.uk<br />

The domino systems biology approach was developed <strong>and</strong> implemented in the resolution of<br />

three paradoxical observations. These concern the response of ATP levels to perturbations in<br />

glycolysis, drug efflux pumping <strong>and</strong> DNA repair in S. cerevisiae. The analysis reinforced that<br />

the energetics of these three aspects of cell function cannot be understood in terms of these<br />

functions alone, but depend on the dynamic integration of the multiple cell functions that<br />

operate in the corresponding conditions. Even for underst<strong>and</strong>ing a functional component<br />

process of the cell, one must therefore implement systems biology. We developed domino<br />

systems biology shown to be an effective way of doing this.<br />

175


Posters are classified by session<br />

<strong>and</strong> then in alphabetical order (PRESENTING AUTHOR)<br />

(Late breaking abstracts are at the end of the <strong>book</strong>)<br />

Session 9: Neurodegenerative Diseases<br />

176


Session 9: Neurodegenerative Diseases Poster 1<br />

Parkin-mediated transcriptional function links p53 to autosomal recessive Parkinson’s<br />

disease.<br />

Cristine A. Costa <strong>and</strong> Frédéric Checler<br />

Institut de Pharmacologie Moléculaire et Cellulaire <strong>and</strong> Institut de NeuroMédecine<br />

Moléculaire, UMR6097 CNRS/UNSA, 660 route des Lucioles, 06560 Valbonne, France,<br />

acosta@ipmc.cnrs.fr<br />

Parkinson’s disease (PD) is the second cause of age-related neurodegenerescence in human<br />

beings. Although its etiology is mainly sporadic, a subset of genetic cases that is characterized<br />

by early onset <strong>and</strong> exacerbated loss of dopaminergic neurons of the substantia nigra pars<br />

compacta have been identified. Familial cases of PD can be associated with a dominant or<br />

recessive inheritance. Parkin is responsible for most of the autosomal recessive cases of PD.<br />

The main function of this protein appeared to be supported by its associated ubiquitin-ligase<br />

enzymatic activity <strong>and</strong> several lines of evidence indicated that impairment of this activity<br />

could contribute to the pathology.<br />

Although never demonstrated, a putative parkin-associated transcription factor activity could<br />

have been suspected from several observations including parkin localization in the nucleus,<br />

control of several genes activated in stress conditions <strong>and</strong> a Ring-IBR-Ring structure that<br />

predicts possible transcription factor properties. Based on these arguments we have<br />

documented a novel function of parkin as a transcription factor capable of transrepressing<br />

p53. We have demonstrated that parkin reduces p53 promoter transactivation, mRNA <strong>and</strong><br />

protein levels. The parkin-associated function was fully prevented by p53 depletion. We have<br />

described a physical interaction between parkin <strong>and</strong> the p53 promoter by both gel shift <strong>and</strong><br />

chromatin immunoprecipitation assays <strong>and</strong> we established that parkin binds to p53 promoter<br />

via its Ring1 domain. Importantly, the influence of parkin on p53 was also observed in PDaffected<br />

brains. Interestingly, parkin-associated repression of p53 promoter transactivation<br />

was prevented by all familial mutations studied in our work, independently of their ability to<br />

abolish or preserve parkin ubiquitin-ligase activity. This data pinpoints the fact that some<br />

functions harbored by parkin may be independent of its canonic ubiquitin-ligase activity.<br />

177


Session 9: Neurodegenerative Diseases Poster 2<br />

The TOR Signalling Pathway Mediates Batten Disease Phenotypes in Dictyostelium<br />

discoideum<br />

Paige K. Smith 1 , Anita Chavan 1 , Bethany S<strong>and</strong>s 1 , Gemma Shanley 1 , Paul R. Fisher 1 ,<br />

Sarah J. Annesley 1<br />

1 Department of Microbiology, La Trobe University, Bundoora, VIC, Australia<br />

Batten Disease is the common name given to a group of lysosomal storage disorders known<br />

as the Neuronal Ceroid Lipofuscinoses (NCLs). Eight forms of the disease have been<br />

identified <strong>and</strong> are characterised by their age of onset <strong>and</strong> an accumulation of a fluorescent<br />

lipofuscin-like pigment. Whilst each is genetically different, symptoms are similar in the<br />

different forms of the disease. We have used a simple eukaryotic organism, Dictyostelium<br />

discoideum to create models of Batten disease. We selected five different genetic forms of<br />

Batten’s disease, those resulting from mutant alleles at the CLN1, 2, 3, 5 <strong>and</strong> 7 loci. Multiple<br />

Dictyostelium mutant cell lines (strains) were created, in each of which the expression of the<br />

Dictyostelium homologue of one of these proteins was antisense-inhibited. Phenotypic<br />

analysis of the mutant strains revealed a consistent pattern of aberrant phenotypes – impaired<br />

aggregation <strong>and</strong> defective growth accompanied by increased rates of nutrient uptake. We<br />

show the results of Batten disease created through down regulation of the CLN2 protein as<br />

one example. Comparison of the phenotypes produced by Batten disease <strong>and</strong> those predicted<br />

to be caused by defective TOR signalling implicated the TOR signalling pathway as a<br />

mediator of the Batten disease phenotypes. This was confirmed by rapamycin treatment of<br />

wild type cells <strong>and</strong> antisense-inhibition of expression of Rheb, an upstream activator of TOR<br />

signalling, both of which caused the same phenotypes as Batten disease. The results could<br />

reveal possible drug targets for treatment in mammalian systems.<br />

178


Session 9: Neurodegenerative diseases Poster 3<br />

The comparison of the survival of P19-derived neuroprogenitors <strong>and</strong> P19 naive cells<br />

after intracerebellar application in a mice model with <strong>and</strong> without neurodegeneration.<br />

Cedikova M. (1), Pitule P. (1, 2), Houdek Z. (3, 5), Cendelin J. (3), Kulda V. (4),<br />

Kralickova M. (1, 5), Vozeh F. (3), Babuska V. (5), Pachernik J. (6), Zech N. (5, 7), Uher<br />

P. (5).<br />

(1) Department of Histology <strong>and</strong> Embryology, (2) Department of Surgery, (3)<br />

Department of Pathophysiology, (4) Department of Medical Chemistry <strong>and</strong><br />

Biochemistry, Faculty of Medicine, Charles University, Plzen, (5) IVF Centers Prof.<br />

Zech - Pilsen, Plzen,<br />

(6) Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno,<br />

(7) Departments for Obstetrics <strong>and</strong> Gynecology, Unit of Gynecological Endocrinology<br />

<strong>and</strong> Reproductive Medicine, University of Graz, Austria.<br />

Email adress of the first author: mirka.cedikova@seznam.cz<br />

Mouse embryonal carcinoma (EC) cells (P19 line) were studied for their survival <strong>and</strong><br />

developmental potential in the intact cerebellum of B6CBA mice <strong>and</strong> a model of<br />

neurodegeneration, in Lurcher mutant mice. Lurcher (Lc) mutant mice represent a model of<br />

olivocerebellar degeneration.<br />

The P19 cells were labeled with a green fluorescent protein <strong>and</strong> either cultured without<br />

differentiation or differentiated into neuroprogenitors using the retinoic acid. The<br />

intracerebellar application was performed in 73 mice - group A consisted of wt mice that<br />

received neuroprogenitors (n=21), group B of wt mice that received naive P19 cells (n=22),<br />

group C consisted of Lc mice that received neuroprogenitors (n=15) <strong>and</strong> group D of Lc mice<br />

that received naive P19 cells (n=15). The morphology of transplanted cells in the context of<br />

surrounding cerebellar tissue was evaluated after three weeks. The groups were compared for<br />

statistically significant difference using the Fisher’s test.<br />

Transplantations resulted in survival <strong>and</strong> further neurodifferentiation of cells in 13 cases in<br />

group A, in 7 cases in group B, in 3 cases in group C <strong>and</strong> in 3 cases in group D which means<br />

that the survival rates are 62%, 32%, 20% <strong>and</strong> 20%. Statistically, there is a significant<br />

difference between the groups of Wt (group A <strong>and</strong> B) <strong>and</strong> Lc mice (group C <strong>and</strong> D) (P=2,77).<br />

This is a pilot study comparing the fate of transplanted EC cells in the cerebellum with <strong>and</strong><br />

without a presence of neurodegenerative process. Our results confirm that EC cells can further<br />

differentiate in the host tissue. It seems that the differentiated elements do not survive less<br />

than undifferentiated <strong>and</strong> that the neurodegeneration affects the survival of EC derived<br />

elements. We plan to follow the impact of such transplantations on restoration of cerebellar<br />

functions. With further validation, the EC cells could become a valuable model for cell<br />

therapy of neurodegenerative diseases. This work has been supported by grants VZ MSM<br />

021620816, COST B30/2007 OC 152 <strong>and</strong> COST BM0603 No. OC10038, by the grant from<br />

the Czech Science Foundation no. 301/08/0717 <strong>and</strong> by SVV-2010- 260 806.<br />

179


Session 9: Neurodegenerative diseases Poster 4<br />

Alteration of MAP Kinase activity mediates the neurotoxic effects of the prion protein<br />

recombinant fragment hPrP90-231: reversal by minocycline.<br />

Aless<strong>and</strong>ro Corsaro 1 , Valentina Villa 1 , Stefano Thellung 1 , Mario Nizzari 1 , Antonio<br />

Aceto* <strong>and</strong> Tullio Florio 1<br />

1 Laboratory of Pharmacology, Dept. Oncology Biology <strong>and</strong> Genetics, University of<br />

Genova, Genova, Italy. E-mail Tullio.Florio@unige.it<br />

2 Department of Biomedical Sciences, Section of Biochemistry, University G.<br />

D’Annunzio, Chieti, Italy<br />

The recombinant peptide corresponding to the protease-resistant core of the human prion<br />

protein (hPrP90-231) can be converted in a PrP SC -like conformation (Corsaro, Paludi et al.<br />

2006) representing a useful model to study the molecular mechanism of neurotoxicity<br />

occurring during prion diseases. SH-SY5Y human neuroblastoma cells, in the absence of the<br />

neurotrophic factors present in fetal calf serum (FCS), undergo to marked activation of the<br />

apopotic pathway due to the loss of phosphorylation/activation of ERK1/2. Apoptosis is a<br />

tightly regulated process, where molecular cascades, triggered by both pro-survival <strong>and</strong> proapoptotic<br />

stimuli, elicit modifications of MAP kinase activity. In particular, the activation of<br />

ERK1/2 <strong>and</strong> p38 was reported to act in a mutually antagonistic way to induce cell survival or<br />

commitment to apoptosis. Interestingly, SH-SY5Y treatment with hPrP90–231 significantly<br />

affects the ability of FCS-contained neurotrophic factors to activate ERK1/2. The abolishment<br />

of ERK1/2 activity in these cells caused the removal of a tonic inhibition of p38 MAP kinase<br />

that is activated <strong>and</strong>, in turn, causes caspase 3 activation. This observation suggests that<br />

hPrP90-231 causes cell death inhibiting the pro-survival ERK1/2 activity of neurotrophic<br />

factors, sustaining the activity of the pro-apoptotic p38 MAP kinase <strong>and</strong> the subsequent<br />

caspase 3 activation. We also demonstrate that minocycline treatment causes a significant<br />

inhibition of hPrP90-231 induced apoptosis. In our experimental model minocycline prevents<br />

the down-regulation of the ERK1/2 pro-survival pathway induced by hPrP90-231, <strong>and</strong> the<br />

increased ERK1/2 activity keeps p38 silenced, preventing caspase 3 activation. In<br />

conclusion, we demonstrated that neurotoxic hPrP90-231 induces apoptosis unbalancing<br />

ERK1/2 <strong>and</strong> p38 MAP kinase activities <strong>and</strong> minocycline may inhibit this effect restoring<br />

ERK1/2 neurotrophic trasductional signalling.<br />

Reference<br />

Corsaro, A., D. Paludi, et al. (2006). "Conformation dependent pro-apoptotic activity of the<br />

recombinant human prion protein fragment 90-231." Int J Immunopathol Pharmacol 19(2):<br />

339-56.<br />

180


Session 9: Neurodegenerative diseases Poster 5<br />

Implication of the Notch pathway in the adult CNS during neurodegenerative processes<br />

Gauthier Dorban, Paul Felten, Claire Hoenen, Paul Heuschling <strong>and</strong> Eleonora Morga<br />

Life Sciences RU, FSTC, University of Luxembourg, 162a avenue de la Faïencerie, L-<br />

1511 Luxembourg. gauthier.dorban@uni.lu<br />

In the CNS, the Notch pathway is implicated in many aspects of cellular development <strong>and</strong><br />

functions. Recent studies have highlighted the role of Notch pathway in neurodegenerative<br />

diseases. It is now established that Notch plays a critical role in steering an immune response<br />

toward inflammation by regulating expression of various cytokines <strong>and</strong> proinflammatory<br />

compounds. Inappropriate or chronic deployment of the inflammation can become a<br />

destructive force in neuropathogenic processes such as multiple sclerosis (MS) <strong>and</strong><br />

Alzheimer’s disease (AD). The reexpression of Notch pathway components, especially the<br />

lig<strong>and</strong> Jagged1, is induced during neurodegenerative diseases <strong>and</strong> seems to create an<br />

environment that induces neuronal death.<br />

The aim of our study is to underst<strong>and</strong> the implication of the Notch pathway in the adult CNS<br />

during neurodegenerative processes. Using a mouse model, we have, identified Jagged1expressing<br />

cells in the forebrain before <strong>and</strong> after a stab wound. The gliosis was induced in the<br />

motorcortex of B6 mice by a mechanical lesion. A kinetic study has been realized to analyze<br />

the modulation of Jagged1 expression in the lesion until 20 days after the stab wound injury.<br />

Next, we have validated two methods to modulate in vivo Jagged1 expression. In our first<br />

model, cholesterol-labelled siRNA directed against Jagged1 has been injected within the<br />

lesion <strong>and</strong> allowed to spread around the stab wound. In the second model, mice bearing<br />

homozygously floxed alleles for Jagged1 were crossed to transgenic mice expressing a<br />

tamoxifen inducible Cre-recombinase. These in vivo strategies enabled us to study the<br />

induced modulations of the inflammatory reaction in the CNS. Jagged1 inhibition<br />

significantly decreases microglial activation <strong>and</strong> astrogliosis within the lesion.<br />

Our results show that Jagged1 plays an important role in the control of inflammatory reactions<br />

<strong>and</strong> could thus allow development of new therapeutic strategies helping to reduce the gliosis<br />

<strong>and</strong> the inflammatory response in the CNS.<br />

181


Session 9: Neurodegenerative diseases Poster 6<br />

Reduced p53 transcriptional activity in long term hypoxia increases chemoresistance in<br />

Medulloblastoma<br />

Yuen Ngan Fan, Daniel Meley, Barry Pizer, Violaine Sée<br />

Medulloblastoma (MB) is the most common malignant paediatric brain tumour. We have<br />

previously shown the key role of p53 signalling in medulloblastoma sensititvity to<br />

chemotherapeutic drugs (Meley et al, Cell Death <strong>and</strong> Disease 2010). Here, we have explored<br />

how hypoxia affects p53 activity <strong>and</strong> cell sensitivity upon chemotherapeutic treatment. We<br />

have shown that the miRNA miR34a is a p53 target <strong>and</strong> is up-regulated upon treatment with<br />

the chemotherapeutic drug etoposide. We also demonstrated that a miR34a mimic is able to<br />

induce a death program independent on p53 status. This suggests that we can bypass p53dependent<br />

drug resistance by directly targeting miR34a. We have found that MB cells (D283)<br />

become less sensitive to etoposide after long-term hypoxia exposure, while short-term<br />

hypoxia did not alter their sensitivity. We have further demonstrated that the increased drug<br />

resistance was concomitant with the inhibition of etoposide-induced p53 transcriptional<br />

activity <strong>and</strong> hence mir34a transcription. In conclusion, miR34a mimic can constitute a new<br />

molecular target for tumour treatment especially in p53 mutated cells or hypoxic tumour cells.<br />

182


Session 9: Neurodegenerative diseases Poster 7<br />

<strong>Inflammation</strong> induces transitory dedifferentiation of astrocytes<br />

Sebastien Gabel, Tony Heurtaux, Paul Felten, Eleonora Morga, Paul Heuschling <strong>and</strong><br />

Luc Gr<strong>and</strong>barbe<br />

Life Sciences Research Unit, FSTC, University of Luxembourg, 162A, avenue de la<br />

Faïencerie, L-1511 Luxembourg, (e-mail: luc.gr<strong>and</strong>barbe@uni.lu)<br />

Brain inflammation, currently described in some neurodegenerative diseases, has been<br />

recognized as a complex phenomenon that generates negative consequences, like<br />

neurotoxicity. However, several studies suggest that inflammatory signals could also exert a<br />

potentially positive influence on neural cell proliferation, survival, migration <strong>and</strong><br />

differentiation. In the adult CNS, astrocytes with neural stem cell capacity exist in specific<br />

neurogenic niches but adult parenchymal astrocytes are widely considered as terminally<br />

differentiated cells with no neurogenic capacity. This orthodoxy has been challenged by<br />

several observations that have shown that both immature parenchymal <strong>and</strong> reactive astrocytes<br />

demonstrate multipotency. Recently correlative studies have suggested that in injured<br />

conditions astrocytes undergo a process of dedifferentiation, <strong>and</strong> may acquire the potential of<br />

neural stem cells (NSC). However this process of dedifferentiation following injury remains<br />

unclear. Here, we report that during early response of reactive gliosis, inflammation induces a<br />

conversion of mature astrocytes into neural progenitors. After 24 h of TNF treatment most of<br />

the astrocytes lose the expression of GFAP <strong>and</strong> some of them re-express markers of the<br />

stemness state, such as CD133 <strong>and</strong> Musashi-1 (Msi-1). Thus TNF treatment results in the<br />

appearance of cells with neural progenitor phenotype, which are able to proliferate <strong>and</strong><br />

differentiate into neurons or astrocytes. To confirm that inflammatory signaling pathway<br />

activation directs the dedifferentiation of astrocytes in vivo, we realized a cortical lesion by a<br />

stab wound injury in adult mice brains. The in vivo data obtained, suggest that inflammation<br />

induces a decrease of GFAP expression in astrocytes <strong>and</strong> the re-expression of stemness<br />

markers in the area of the lesion after 24 h. The data presented here reveal the plasticity of<br />

astrocytes, which could be a potential target for the therapeutic strategies in injured CNS<br />

regeneration.<br />

183


Session 9: Neurodegenerative diseases Poster 8<br />

Abeta disaggregating proteins counteract Abeta induced neurotoxic responses<br />

Ana Gabriela Henriques, Bárbara Gomes, Raquel Ruivo, Edgar F. da Cruz e Silva* <strong>and</strong><br />

da Odete A. B. Cruz e Silva<br />

Laboratório de Neurociências, Centro de Biologia Celular, Universidade de Aveiro,<br />

3810-193 Aveiro, Portugal.<br />

*Laboratório de Transdução de Sinais, Centro de Biologia Celular, Universidade de<br />

Aveiro, 3810-193 Aveiro, Portugal.<br />

odetecs@ua.pt<br />

Abeta peptide is recognized as a key player in Alzheimer´s disease pathology, being<br />

responsible for the induction of several cellular responses associated with neurotoxicity <strong>and</strong><br />

neurodegeneration. Of note, it was shown that the fibrillar/aggregated species of Abeta are<br />

highly toxic. Thus, several therapeutic strategies aimed at depolymerizing Abeta provide<br />

attractive avenues of intervention. Laminin <strong>and</strong> gelsolin provide such a model, given that they<br />

both prevent Abeta aggregation by forming Abeta-laminin <strong>and</strong> Abeta-gelsolin complexes.<br />

Further, these complexes attenuate the neurotoxic effects of Abeta. Additionally, we have<br />

recently shown that Abeta can lead to intracellular accumulation of the neurotrophic <strong>and</strong><br />

neuroprotector secreted Alzheimer´s amyloid precursor protein fragment (sAPP) <strong>and</strong> preincubation<br />

of Abeta with laminin <strong>and</strong> gelsolin also renders it less potent in this respect. In<br />

conclusion, our data indicates that Abeta-laminin <strong>and</strong> Abeta-gelsolin complexes are less<br />

apoptotic <strong>and</strong> less potent at inducing intracellular sAPP retention than fibrillar Abeta. Hence,<br />

these data validate the potential of both proteins as therapeutic targets to prevent Abetainduced<br />

effects both at the neurotoxic <strong>and</strong> at the APP metabolic levels.<br />

184


Session 9: Neurodegenerative diseases Poster 9<br />

Microglia in astroglial culture activation: an underestimated role<br />

Tony Heurtaux, Sophie Losciuto, Gauthier Dorban, Sebastien Gabel, Luc Gr<strong>and</strong>barbe,<br />

Eleonora Morga, Paul Heuschling<br />

Life Sciences Research Unit, FSTC, University of Luxembourg, 162A, avenue de la<br />

Faïencerie, L-1511 Luxembourg (e-mail: tony.heurtaux@uni.lu)<br />

Development <strong>and</strong> progression of neurodegenerative diseases is associated with a chronic<br />

inflammatory state. <strong>Inflammation</strong> is probably not a primary pathogenic event in most forms of<br />

neurodegenerative diseases, however, there is substantial evidence that inflammatory<br />

responses of microglia, astrocytes <strong>and</strong> other immune cells contribute to disease severity.<br />

In accordance with different ontogenic origins of astrocytes <strong>and</strong> microglial cells, their<br />

physiological roles <strong>and</strong> many of their responses upon activation are also different. The<br />

purpose of this work is to evaluate the consequences of the presence of microglia in the<br />

activation of astroglial cultures. Indeed, the presence <strong>and</strong> role that microglial cells play in<br />

astroglial cultures are often underestimated. Thus, we have compared the impact of proinflammatory<br />

events on different glial cultures. We have stimulated murine microglia, mixed<br />

glial cultures, astrocyte-enriched cultures <strong>and</strong> neurosphere-derived astrocytes with proinflammatory<br />

compounds (lipopolysaccharide LPS <strong>and</strong> interferon-gamma IFNg). Cell<br />

cultures have been characterized by immunocytochemistry. mRNA <strong>and</strong> protein expression<br />

levels have been determined by PCR <strong>and</strong> ELISA assays respectively.<br />

Microglial cultures were strongly activated in presence of LPS <strong>and</strong> IFNg. Compared to<br />

microglia, the same treatments induced a lower activation of mixed glial cultures whereas a<br />

weak activation was observed on astrocyte-enriched cultures <strong>and</strong> neurosphere-derived<br />

astrocytes. In conclusions, we clearly show that microglia-devoided cultures (astrocyteenriched<br />

cultures <strong>and</strong> neurosphere-derived astrocytes) are poorly activated by proinflammatory<br />

agents. These results support the hypothesis that activation of astrocytes during<br />

neuroinflammatory states may be due to prior microglial activation inducing finally a vicious<br />

circle <strong>and</strong> a chronic inflammatory state. We present evidence that the magnitude of the proinflammatory<br />

response is proportional to the percentage of microglial cells present in glial<br />

cultures.<br />

185


Session 9: Neurodegenerative diseases Poster 10<br />

The role of the ESCRT-II complex in Dictyostelium discoideum<br />

Jasmina Ilievska 1 , Paul R Fisher 1 , Naomi E Bishop 1 <strong>and</strong> Sarah J Annesley 1<br />

1 Department of Microbiology, La Trobe University, Bundoora 3086, Victoria, Australia.<br />

Email: p.fisher@latrobe.edu.au<br />

The ESCRT (endosomal sorting complexes required for transport) machinery is composed of<br />

four complexes (0, I, II, III) integral to the degradation of cargo through the endocytic <strong>and</strong><br />

autophagic pathway. Mutations of ESCRT genes are associated with neurodegenerative<br />

diseases such as Alzheimer’s, Parkinson’s <strong>and</strong> Huntington’s. These diseases are characterised<br />

by accumulation of intracellular protein aggregates <strong>and</strong> defective autophagic compartments.<br />

The ESCRT complexes are involved in autophagy however their role in autophagic cell death<br />

(ACD) is not well known. Research of the autophagic pathway is often complicated by the<br />

additional cell death pathway of apoptosis. Both pathways result in the same cell death<br />

morphology making the two processes morphologically indistinguishable. The NIH<br />

recognised model eukaryotic system Dictyostelium discoideum provides a unique way to<br />

study autophagic cell death as it lacks an apoptosis pathway.<br />

The aim of the research is to alter the expression of the ESCRT-II proteins (Vps22, 25 <strong>and</strong> 36)<br />

in Dictyostelium discoideum <strong>and</strong> determine their roles in the autophagic cell death pathway.<br />

Preliminary results suggest that antisense inhibition of Vps22, 25 <strong>and</strong> 36 causes defects in cell<br />

growth, development, endocytosis <strong>and</strong> autophagic cell death. Here we present results for two<br />

of the members of the ESCRT-II complex Vps22 <strong>and</strong> Vps25.<br />

186


Session 9: Neurodegenerative diseases Poster 11<br />

A Possible Role of GSK-3beta-Nrf2 Signaling Pathway for the Prevention <strong>and</strong>/or<br />

Treatment of Alzheimer’s Disease<br />

Chan Lee 1 , Young-Joon Surh 2 , <strong>and</strong> Jung-Hee Jang 1<br />

1 College of Oriental Medicine, Daegu Haany University, Daegu 706-828, 2 College of<br />

Pharmacy, Seoul National University, Seoul 151-742, South Korea<br />

beta-Amyloid peptide (Abeta) is the major component of senile plaques accumulated in the<br />

brains of patients with Alzheimer’s disease (AD) <strong>and</strong> has been reported to cause neuronal cell<br />

death via oxidative stress. Therefore, attention has been focused on identifying redoxsensitive<br />

transcription factors <strong>and</strong> their target genes protecting against Abeta-induced<br />

oxidative cell death. Nrf2 plays a pivotal role in the transcriptional regulation of antioxidant<br />

proteins <strong>and</strong> detoxification enzymes <strong>and</strong> blocks apoptosis caused by a wide array of death<br />

signals. Ectopic expression of Nrf2 rescued cells from Abeta-induced cytotoxicity, apoptosis,<br />

intracellular accumulation of reactive oxygen species <strong>and</strong> oxidative damages. Moreover, Nrf2<br />

overexpression increased the expression of gamma-glutamylcysteine ligase (GCL), a ratelimiting<br />

enzyme in cellular glutathione biosynthesis. Conversely, knockdown of Nrf2 gene<br />

expression with siRNA or dominant negative mutant Nrf2 exacerbated Abeta-induced<br />

oxidative cell death. To further elucidate the upstream regulator for Nrf2 activation, we have<br />

focused on glycogen synthase kinase-3beta (GSK-3beta). Inhibition of Abeta-induced GSK-<br />

3beta activation by pharmacological inhibitors such as LiCl led to nuclear accumulation Nrf2<br />

<strong>and</strong> transcriptional activation of Nrf2 downstream target genes <strong>and</strong> protected against Abetamediated<br />

oxidative cell death. In another experiment, some dietary <strong>and</strong> medicinal<br />

phytochemicals attenuated A!-induced oxidative cell death via suppression of GSK-3beta <strong>and</strong><br />

subsequent activation of Nrf2. Taken together, these findings suggest that GSK-3beta-Nrf2<br />

signaling pathway may act as a survival mediator against AD.<br />

187


Session 9: Neurodegenerative diseases Poster 12<br />

Neuroprotective <strong>and</strong> Memory Enhancing Effects of Cypress-containing Essential Oils on<br />

Scopolamine-induced Amnesia in C57BL/6 Mice<br />

Gil-Yong Lee <strong>and</strong> Chan-Ik Park<br />

Department of Cosmeceutical Science, Daegu Haany University, Gyeongsangbuk-do<br />

712-715, S. Korea (E-mail : cipark@dhu.ac.kr)<br />

This study was performed to investigate the memory enhancing effects of essential oils from<br />

rosemary, thyme red, bergamot, pine needle, lavender <strong>and</strong> cypress against scopolamineinduced<br />

amnesia in C57BL/6 mice. To induce amnesia, scopolamine (1mg/kg) was<br />

intraperitonically injected into C57BL/6 mice 30 min before starting behavior tests. We have<br />

conducted Y-maze, Morris water-maze, passive avoidance <strong>and</strong> fear conditioning tests to<br />

monitor learning <strong>and</strong> memory functions. Essential oils of bergamot, pine needle, lavender <strong>and</strong><br />

cypress effectively reversed scopolamine-induced memory impairment in C57BL/6 mice<br />

which was represented by improvement of spontaneous alterations in Y-maze test, mean<br />

escape time in water-maze test, step-through latency in passive avoidance test <strong>and</strong> freezing<br />

response in fear conditioning tests. Particularly, combination of cypress with other essential<br />

oils (cypress + lavender > cypress + pine needle > cypress + bergamot) exhibited enhanced<br />

memory enhancing effects. However, compared with aforementioned natural essential oils,<br />

synthetic perfume mixtures showed relatively lower activities under the same experimental<br />

condition. These results suggest that cypress-containing essential oils may have therapeutic<br />

potentials <strong>and</strong> be applied for the prevention <strong>and</strong>/or treatment of amnesia-related neurological<br />

disorders.<br />

Keywords : amnesia, scopolamine, learning <strong>and</strong> memory, essential oil, cypress<br />

188


Session 9: Neurodegenerative diseases Poster 13<br />

Development of Natural Resources for the Prevention <strong>and</strong> Treatment of Alzheimer's<br />

Disease via Activating Nrf2-HO-1 Signaling Pathway<br />

Chan Lee <strong>and</strong> Jung-Hee Jang<br />

Department of Pathology, College of Oriental Medicine, Daegu Haany University,<br />

Daegu 706-828, Korea<br />

beta-Amyloid peptide (Abeta), the major component of senile plaques is considered to have a<br />

causal role in the development <strong>and</strong> progression of Alzheimer's disease (AD). Increasing<br />

evidence supports that Abeta-induced cell death is mediated by oxidative stress. Induction of<br />

heme oxygenase-1 (HO-1), the rate-limiting enzyme in heme degradation process, has been<br />

associated with adaptive survival response to oxidative insults. Treatment of SH-SY5Y cells<br />

with Abeta time-dependently increased protein expression of HO-1. Furthermore pretreatment<br />

of these cells with ZnPP, an inhibitor of HO-1 activity, aggravated Abeta-induced apoptotic<br />

cell death. Abeta treatment resulted in a transient activation of upstream redox-sensitive<br />

transcription factor, NF-E2-related factor 2 (Nrf2). Conversely, knockdown of Nrf2 gene<br />

expression abolished Abeta-induced HO-1 expression. In another experiment, we have<br />

searched for phytochemicals which can attenuate Abeta-induced apoptosis <strong>and</strong> memory<br />

impairment via Nrf2-HO-1 signaling pathway. We have investigated the neuroprotective<br />

effect of luteolin, 3´,4´,5,7-tetrahydroxyflavone present in the perilla leaf <strong>and</strong> seed, against<br />

Abeta-induced neuronal cell death <strong>and</strong> cognitive deficit in Sprague-Dawley (SD) rats. Abetacaused<br />

learning <strong>and</strong> memory deficits were effectively improved by oral administration of<br />

luteolin as assessed by several behavior tests such Morris water-maze, passive avoidance, fear<br />

conditioning tests through up-regulation of HO-1 by Nrf2 activation. Under the same<br />

experimental condition, luteolin attenuated Abeta-induced pro-apoptotic signals in the cortex<br />

<strong>and</strong> hippocampus such as activation of JNK <strong>and</strong> increased ratio of Bax to Bcl-2. Taken<br />

together, these findings suggest that up-regulation of HO-1 via activation of Nrf2 may act as a<br />

survival signal <strong>and</strong> have therapeutic potentials for the prevention <strong>and</strong> treatment of AD.<br />

Keywords: beta-amyloid, glycogen synthase kinase-3beta, NF-E2-related factor-2, oxidative<br />

stress, phytochemicals<br />

189


Session 9: Neurodegenerative diseases Poster 14<br />

REGULATION OF INFLAMMATORY PHENOTYPES OF MICROGLIA AND<br />

MICROGLIA-ASTROCYTE COMMUNICATION BY LIVER X RECEPTORS.<br />

Sophie Losciuto, Xavier C. Hever, Tony Heurtaux, Eleonora Morga <strong>and</strong> Paul<br />

Heuschling<br />

Life Sciences Research Unit, FSTC, University of Luxembourg, 162A, avenue de la<br />

Faïencerie, L-1511 Luxembourg (e-mail: xavier.hever@uni.lu)<br />

Alzheimer’s Disease is a neurodegenerative disorder characterized by the accumulation <strong>and</strong><br />

aggregation of Amyloid beta (Ab) peptide. In this pathology, microglia are frequently<br />

activated by a local inflammation, in part caused by the presence of Ab. Pro-inflammatory<br />

compounds secreted by activated microglia attract <strong>and</strong> activate astrocytes. These astrocytes<br />

also produce pro-inflammatory molecules leading to an inflammatory vicious circle in the<br />

brain accelerating neuronal death. It is known that activated microglia can differentiate<br />

towards a continuous spectrum of phenotypes of which the two extreme states are called M1<br />

for the pro-inflammatory state <strong>and</strong> M2 for the anti-inflammatory state. The Liver X Receptors<br />

(LXRs) are lig<strong>and</strong>-activated nuclear receptors known to regulate inflammatory responses by<br />

repressing pro-inflammatory gene expressions in activated microglia.<br />

In this study, Ab was used to induce the M1 pro-inflammatory state of microglia. We first<br />

showed that activated LXR was able to decrease the M1 state of activated microglia but was<br />

not able to induce the M2 anti-inflammatory state. These results suggest that LXR activation<br />

induces in microglia an intermediary state between M1 <strong>and</strong> M2. Since activated LXR reduces<br />

microglial activation, we analysed whether this effect could have an impact on the cellular<br />

communication between microglia <strong>and</strong> astrocytes. We showed that LXR activation has no<br />

direct effect on astrocyte activation. In co-culture experiments, preliminary observations<br />

showed that microglia, treated with an LXR agonist, were able to down-regulate astrocytic<br />

activation. Activated LXR appears to be indirectly able to modulate the phenotype of<br />

astrocytes through its action on microglia.<br />

This work emphasizes the role of activated LXR in cellular communication between<br />

microglia <strong>and</strong> astrocytes. Thus, LXR activation could reduce brain inflammation <strong>and</strong> may<br />

protect from neuronal death.<br />

190


Session 9: Neurodegenerative diseases Poster 15<br />

Cellular Antioxidant Adaptive Survival Response to 6-Hydroxydopamine-induced<br />

Nitrosative Cell Death in C6 Glioma Cells<br />

Chan Lee 1 , Gyu Hwan Park 2 , <strong>and</strong> Jung-Hee Jang 1*<br />

1 College of Oriental Medicine, Daegu Haany University, Daegu 706-828, South Korea,<br />

2 Department of Neurology, Columbia University, New York, NY 10032, USA<br />

Parkinson’s disease (PD) is a progressive neurodegenerative movement disorder characterized<br />

by selective loss of dopaminergic neurons in the substantia nigra. 6-Hydorxydopamine (6-<br />

OHDA) is a catecholaminergic neurotoxin widely used to produce experimental models of PD<br />

<strong>and</strong> has been reported to cause oxidative <strong>and</strong>/or nitrosative stress. In this study, we have<br />

investigated 6-OHDA-induced nitrosative cell death <strong>and</strong> its self-defense mechanism in C6<br />

glioma cells. Treatment of C6 cells with 6-OHDA increased expression of inducible nitric<br />

oxide synthase (iNOS) <strong>and</strong> subsequent production of nitric oxide (NO). Furthermore 6-<br />

OHDA treatment led to peroxynitrite generation <strong>and</strong> nitrotyrosine formation. 6-OHDAinduced<br />

nitrosative stress ultimately caused apoptotic cell death as determined by decreased<br />

Bcl-2/Bax ratio, activation of c-Jun N-termianl kinase (JNK), <strong>and</strong> cleavage of caspase-3 <strong>and</strong><br />

poly(ADP-ribose)polymerase (PARP), which were attenuated by peroxynitrite decomposition<br />

catalyst, FeTPPS. In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in<br />

an increased expression of HO-1 <strong>and</strong> 6-OHDA-induced cytotoxicity was effectively<br />

suppressed by the HO-1 inducer SnCl2, supporting the cytoprotective role of HO-1. To<br />

elucidate the molecular mechanism underlying 6-OHDA-mediated HO-1 induction, we have<br />

examined the possible involvement of NF-E2-related factor 2 (Nrf2), which plays an<br />

important role in the transcriptional regulation of phase II detoxification <strong>and</strong> antioxidant<br />

enzymes. 6-OHDA treatment increased nuclear translocation <strong>and</strong> transcriptional activity of<br />

Nrf2, which seemed to be mediated by activation of Akt/ protein kinase B. Taken together<br />

these findings suggest that HO-1 up-regulation via Nrf2 activation may mediate cellular<br />

adaptive survival response to 6-OHDA-induced nitrosative cell death in C6 glioma cells.<br />

191


Session 9: Neurodegenerative diseases Poster 16<br />

Influence of free fatty acids on mitochondrial function in human brain cell line exposed<br />

to amyloid-beta.<br />

Agnieszka !liwa, Joanna Góralska, Barbara Zapa"a, Anna Gruca, Urszula Czech, Anna<br />

Knapp, Magdalena Awsiuk, Aldona Dembi#ska-Kie$<br />

Department of Clinical Biochemistry, Collegium Medicum, Jagiellonian University,<br />

Kopernika 15a, 31-501 Krakow, Pol<strong>and</strong><br />

Background:<br />

The cellular effects of nonestrified fatty acids (FFA) are complex <strong>and</strong> related in between to<br />

their role as substrates for energy production, as ion channel modulators, mitochondrial<br />

uncoupling agents <strong>and</strong> modulators of genes expression.<br />

The nutrient-induced changes in cellular metabolism is the recent target in the preventive<br />

medicine.<br />

Aim: The aim of the study was to investigate the effect of dietary FFA on mitochondrial<br />

function of the neuronal origin cells.<br />

Methods: LN-18 cells (glioblastoma) were preincubated with FFA: PA, OA, AA, EPA <strong>and</strong><br />

TTA (30µM) for 24h <strong>and</strong> exposed to !-amyloid (25 µM) for the last 18 hours. Measurement<br />

of mitochondrial membrane potential ("#) was performed by using flow cytometry <strong>and</strong> BD<br />

Bioimager 855 microscopy.Mitochondrial metabolic activity was monitored by measurements<br />

of the mitochondrial oxygen consumption rates (OROBOROS ® Oxygraph-2k) <strong>and</strong> ATP<br />

generation (ATP Lite Parkin Elmer).<br />

Results:<br />

In LN-18 cells PA significantly decreased mitochondrial membrane potential, mitochondrial<br />

respiration <strong>and</strong> ATP level. ATP generation was also diminished by OA, <strong>and</strong> the effect was<br />

equal to effect of !-amyloid alone. Preincubation of LN-18 cells with EPA prevent the<br />

negative effect of !-amyloid on mitochondrial respiration, though did not prevent the decrease<br />

in oxidative phosphorylation capacity.<br />

Conclusion:<br />

Our data suggest negative effect of PA on mitochondrial functions of the CNS-origin cells.<br />

Potentially neuroprotective action of EPA may be related to preventing !-amyloid induced<br />

mitochondrial dysfunction.<br />

Acknowledgments: this study was supported by Polish-Norwegian grant no. PNRF-104-AI-<br />

1/07, K/ZDS/000935 as well as the EU COST Action FA0602.<br />

192


Session 9: Neurodegenerative diseases Poster 17<br />

Study of the role of the Parkinson’s disease gene, Park2, in the development of gliomas.<br />

Julien Viotti, Eric Duplan, Frédéric Checler <strong>and</strong> Cristine Alves da Costa<br />

Institut de Pharmacologie Moléculaire et Cellulaire <strong>and</strong> Institut de NeuroMédecine<br />

Moléculaire, UMR6097 CNRS/UNSA, 660 route des Lucioles, 06560 Valbonne, France,<br />

viotti@ipmc.cnrs.fr<br />

Cancer is a huge age-related problem of public health that is mainly characterized by cell<br />

death control impairment. In contrast, Parkinson’s disease (PD) is a neurodegenerative<br />

disorder that is due to a massive loss of dopaminergic neurons of the substantia nigra by<br />

exacerbated apoptosis. Interestingly, several epidemiological data evidenced a negative<br />

correlation between PD <strong>and</strong> cancer. This negative correlation raises the question of whether<br />

despite phenotypically distinct, these two diseases could share common protein effectors. In<br />

this context, it is worth noting that most of familial PD-associated proteins are implicated in<br />

cell cycle control <strong>and</strong> often abnormally expressed in tumors. We have just ascribed a new<br />

function to one of them, parkin, as a transcriptional factor capable of trans-repressing p53. Of<br />

most interest several works pinpoint the role of parkin as a potential tumor suppressor protein,<br />

the mutations of which have been associated to glioblastoma generation. The main goal of the<br />

present work was to explore this newly identified transcription factor function of parkin in the<br />

etiology of human brain tumors. Our first results show that the overexpression of parkin can<br />

increase cell viability, decrease both the number of pre-apoptotic nuclei <strong>and</strong> caspase-3 activity<br />

<strong>and</strong> modulate cell cycle kinetics. In addition, we have observed a modulation of both parkin<br />

protein <strong>and</strong> mRNA levels according to the grade of gliomas. Our data suggest that the<br />

putative utilization of parkin as a brain tumor biomarker could be used in replacement of the<br />

subjective histological analysis performed in routine to date <strong>and</strong> perhaps allow an earlier <strong>and</strong><br />

bona fide diagnosis of this devastating <strong>and</strong> incurable type of cerebral cancer.<br />

193


Notes<br />

194


Notes<br />

195


Notes<br />

196


List of participants<br />

(In alphabetical order)<br />

(Updated January 13 th , 2011)<br />

197


Miss Siti Aminah Abdul Rahim<br />

norlux neuro oncology laboratory<br />

crp santé<br />

84 val fleuri<br />

L-1526 luxembourg<br />

LUX<br />

Email: sitiaminah.abdulrahim@crpsante.lu<br />

Mr. Soheil Akbari<br />

molecular biology <strong>and</strong> cancer research lab<br />

Dokuz Eylul University school of<br />

medicine<br />

inciralti /balcova<br />

35350 Izmir<br />

TUR<br />

Email: s.akbari84@gmail.com<br />

Mr. Rolf Altenburger<br />

Dept Bioanalytical Ecotoxicology<br />

UFZ Helmholtz Centre for Environmental<br />

Research<br />

Permoserstr. 15<br />

4318 Leipzig<br />

DEU<br />

Email: rolf.altenburger@ufz.de<br />

Dr. Salomon Amar<br />

The Center for Anti-Inflammatory<br />

Therapeutics<br />

Boston University<br />

650 Albany St., X-343<br />

MA Boston<br />

USA<br />

Email: samar@bu.edu<br />

Pr. Frank Antonicelli<br />

Dermatology<br />

URCA<br />

51 Rue Cognacq-jay<br />

51420 Reims<br />

FRA<br />

Email: frank.antonicelli@univ-reims.fr<br />

Dr. Aicha Ba<br />

Bioorganische Chemie-School of<br />

Pharmacy<br />

Saarl<strong>and</strong> University<br />

Universitaet Camous<br />

66123 Saarbruecken<br />

DEU<br />

Email: la.ba-bernardi@mx.unisaarl<strong>and</strong>.de<br />

Mr. Khalil Abouelaradat<br />

Radiobiology Unit<br />

SCK-CEN<br />

Boeretang 200<br />

B-2400 Mol<br />

BEL<br />

Email: kabouela@sckcen.be<br />

Miss Hélène Albert<br />

LIMBP<br />

Université Paul Verlaine Metz<br />

Rue du Général Delestraint<br />

57070 METZ<br />

FRA<br />

Email: albert2@univ-metz.fr<br />

Dr. Cristine Alves Da Costa<br />

IPMC<br />

CNRS<br />

660 ROUTE DES LUCIOLES<br />

6560 VALBONNE<br />

FRA<br />

Email: acosta@ipmc.cnrs.fr<br />

Mr. Alex<strong>and</strong>er Anderson<br />

Integrated Mathematical Oncology<br />

H. Lee Moffitt Cancer Center<br />

12901 Magnolia Drive<br />

33612 Tampa<br />

USA<br />

Email: alex<strong>and</strong>er.<strong>and</strong>erson@moffitt.org<br />

Pr. Safiye Nese Atabey<br />

Department of Medical Biology <strong>and</strong><br />

Genetics<br />

DEU Medical School<br />

Mithatpasa caddesi, Temel Bilimler<br />

Binasi, Kat 3<br />

35340 izmir<br />

TUR<br />

Email: nese.atabey@deu.edu.tr<br />

Mrs. Jaehee Bae<br />

Pharmacology<br />

Konyang University<br />

685, Gasuwondong, Seogu<br />

302-718 Daejeon<br />

KOR<br />

Email: jaeku@skku.edu<br />

198<br />

Mr. Adebayo Adewunmi Ebenezer<br />

UNIVERSITY COLLEGE HOSPITAL<br />

NO 27 AYORINDE STREET EKOTEDO,<br />

OPP SHOP 142 ADAMASINGBA<br />

SHOPPING COMPLEX<br />

2234 IBADAN<br />

NGA<br />

MASTERLARYYCHO@YAHOO.COM<br />

Miss Claire Allan<br />

Microbial Cell Biology<br />

La Trobe University<br />

Kingsbury Drive<br />

3086 Bundoora<br />

AUS<br />

Email: c_y_a_001@hotmail.com<br />

Dr. Sarah Annesley<br />

Micorbial Cell Biology<br />

La Trobe University<br />

Plenty Rd<br />

3086 Bundoora<br />

AUS<br />

Email: S.Annesley@latrobe.edu.au<br />

Pr. Marina Aunapuu<br />

Histology<br />

University of Tartu<br />

Ravila 19<br />

50411 Tartu<br />

EST<br />

Email: marina.aunapuu@ut.ee<br />

Pr. Denyse Bagrel<br />

LIMBP<br />

Université Paul Verlaine-Metz<br />

Rue du Général Delestraint<br />

57070 Metz<br />

FRA<br />

Email: bagrel@univ-metz.fr


Mrs. Emilie Bana<br />

Laboratoire d'Ingénierie Moléculaire et<br />

Biochimie Pharmacologique<br />

Université Paul Verlaine - Metz<br />

Avenue du Général Delestraint<br />

57070 METZ<br />

FRA<br />

Email: emilie.bana@free.fr<br />

Dr. Henry Bayele<br />

Structural <strong>and</strong> Molecular Biology<br />

University College London<br />

Gower Street<br />

WC1E 6BT London<br />

GBR<br />

Email: h.bayele@medsch.ucl.ac.uk<br />

Miss Cristina Belgiovine<br />

IGM-CNR<br />

Via Abbiategrasso 207<br />

27100 Pavia<br />

ITA<br />

Email: belgiovine@igm.cnr.it<br />

Dr. Rafi Benotmane<br />

Radiobiology Unit<br />

SCK-CEN<br />

Boeretang 200<br />

B-2400 Mol<br />

BEL<br />

Email: abenotma@sckcen.be<br />

Pr. Afig Berdeli<br />

MOLECULAR MEDICINE<br />

LABORATORY<br />

EGE UNIVERSITY FACULTY OF<br />

MEDICINE CHILD HOSPITAL<br />

BORNOVA<br />

35100 IZMIR<br />

TUR<br />

Email: afigberdeli@yahoo.com<br />

Miss Kim BoAe<br />

Formulation Lab. of New Cosmeceuticals<br />

Daegu Haany University<br />

290, Yugok-dong<br />

712-715 Gyeongsan-si, Gyeongsangbukdo<br />

KOR<br />

Email: gajette@hanmail.net<br />

Miss Burcin Baran<br />

molecular biology <strong>and</strong> cancer research lab<br />

dokuz eylul school of medicine<br />

Inciralti/Balcova<br />

35350 Izmir<br />

TUR<br />

Email: burcinbrn@yahoo.com<br />

Dr. Juergen Becker<br />

PromoCell GmbH<br />

PromoCell GmbH<br />

Sickingenstrasse 63/65<br />

69126 Heidelberg<br />

DEU<br />

Email: info@promocell.com<br />

Miss Houda Benboubker<br />

LIMBP<br />

Campus Bridoux<br />

57000 Metz<br />

FRA<br />

Email: houda@benboubker.com<br />

Mrs. Lynda Benyoub<br />

AUREUS PHARMA<br />

174 Quai de Jemmapes<br />

75010 PARIS<br />

FRA<br />

Email: sabine.clement@aureuspharma.com<br />

Mrs. Geert Bienvenue<br />

GE Healthcare<br />

koterveldstraat 2<br />

1831 Diegem<br />

BLZ<br />

Email: geert.bienvenue@ge.com<br />

Mr. William Booth<br />

AMSBIO<br />

184 Milton Park<br />

OX14 4SE Abingdon<br />

GBR<br />

Email: stephanieg@amsbio.com<br />

199<br />

Dr. Daniela Basso<br />

Department of Laboratory Medicine<br />

University Hospital of Padova<br />

Via Giustiniani 2<br />

35128 Padova<br />

ITA<br />

Email: daniela.basso@sanita.padova.it<br />

Mrs. Adeline Beillerot<br />

LIMBP<br />

UPV-Metz<br />

rue du général Delestraint<br />

57070 Metz<br />

FRA<br />

Email: beillero@univ-metz.fr<br />

Dr. Aline Bennasroune<br />

Laboratoire d'Ingénierie Moléculaire et<br />

Biochimie Pharmacologique<br />

Rue du general Delestraint<br />

57070 Metz<br />

FRA<br />

Email: aline.bennasroune@univ-metz.fr<br />

Mr. Omar Benzakour<br />

Institut de Physiologie et Biologie<br />

Cellulaires<br />

Université de Poitiers<br />

1, rue Georges Bonnet<br />

86022 Poitiers<br />

FRA<br />

Email: omar.benzakour@univ-poitiers.fr<br />

Dr. Romain Blasius<br />

prophac<br />

Prophac<br />

5 rangwee<br />

2412 luxembourg<br />

LUX<br />

Email: romain.blasius@prophac.lu<br />

Dr. Monica Borgatti<br />

BIOCHEMISTRY AND MOLECULAR<br />

BIOLOGY DEPARTMENT<br />

FERRARA UNIVERSITY<br />

VIA FOSSATO DI MORTARA 74<br />

44121 FERRARA<br />

ITA<br />

Email: brgmnc@unife.it


Dr. Pieter Borger<br />

Departement Biomedizin Pulmonology<br />

University Hospital Basel<br />

Hebelstrasse 20<br />

4031 Basel<br />

CHE<br />

Email: pieter.borger@unibas.ch<br />

Dr. Angela Brieger<br />

Biomedical Research Laboratory<br />

JWG University Frankfurt a.M.<br />

Theodor-Stern-Kai 7<br />

60590 Frankfurt<br />

DEU<br />

Email: a.brieger@em.uni-frankfurt.de<br />

Pr. Jean-Luc Bueb<br />

Life Sciences Research Unit<br />

Université du Luxembourg<br />

162a, avenue de la Faïencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: jean-luc.bueb@uni.lu<br />

Dr. Mario Capecchi<br />

Department of Human Genetics<br />

University of Utah <strong>and</strong> The Howard<br />

Hughes Medical Institute<br />

15 North 2030 East #5440<br />

84112 Salt LakeCity<br />

USA<br />

Email: mario.capecchi@genetics.utah.edu<br />

Pr. Park ChanIk<br />

Formulation Lab. of New Cosmeceuticals<br />

Daegu Haany University<br />

290, Yugok-dong<br />

712-715 Gyeongsan-si, Gyeongsangbukdo<br />

KOR<br />

Email: gajette@hanmail.net<br />

Dr. Elena Chernolovskaya<br />

Laboratory of Nucleic Acids<br />

Biochemistry<br />

Institute of Chemical Biology <strong>and</strong><br />

Fundamental Medicine SB RAS<br />

Lavrentiev ave., 8<br />

630090 Novosibirsk<br />

RUS<br />

Email: elena_ch@niboch.nsc.ru<br />

Mr. Michael Bots<br />

Lab for Exp. Oncology <strong>and</strong> Radiobiology<br />

Meibergdreef 9<br />

1105 AZ Amsterdam<br />

NLD<br />

Email: m.bots@amc.uva.nl<br />

Dr. Juergen Brieger<br />

Molecular Tumor Biology<br />

University Medical Center of the<br />

Johannes Gutenberg University Mainz<br />

Department of Otolaryngology,<br />

55131 Mainz<br />

DEU<br />

Email: juergen.brieger@unimedizinmainz.de<br />

Mrs. Wibke Busch<br />

Department Bioanalytical Ecotoxicology<br />

Helmholtz-Centre for Environmental<br />

Research - UFZ<br />

Permoser Str. 15<br />

4318 Leipzig<br />

DEU<br />

Email: wibke.busch@ufz.de<br />

Dr. Miroslava Cedikova<br />

Department of Histology <strong>and</strong> Embryology<br />

Karlovarska 48<br />

301 00 Plzen<br />

CZE<br />

Email: mirka.cedikova@seznam.cz<br />

Dr. Beatrice Charreau<br />

INSERM UMR643<br />

30 bd Jean Monnet<br />

44093 Nantes<br />

FRA<br />

Email: Beatrice.Charreau@univ-nantes.fr<br />

Dr. Andy Chevigné<br />

Retrovirology<br />

CRP-Santé<br />

Val Fleuri<br />

L-1526 Luxembourg<br />

LUX<br />

Email: <strong>and</strong>y.chevigne@crp-sante.lu<br />

200<br />

Mr. Koen Breyne<br />

Biochemistry - Fac. Veterinary Sciences<br />

Ghent University<br />

Salisburylaan 133<br />

9820 Merelbeke (Ghent)<br />

BEL<br />

Email: koen.breyne@ugent.be<br />

Pr. Pedro Buc Calderon<br />

PMNT 7369<br />

université catholique de Louvain<br />

73, avenue E. Mounier<br />

1200 Bruxelles<br />

BEL<br />

Email: pedro.buccalderon@uclouvain.be<br />

Pr. Vittorio Calabrese<br />

Clinical Biochemistry <strong>and</strong> clinical<br />

molecular Biology<br />

University of Catania<br />

Viale Andrea Doria 6<br />

95125 CATANIA<br />

ITA<br />

Email: calabres@unict.it<br />

Dr Claudia Cerella<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: claudia.cerella@lbmcc.lu<br />

Dr Sebastien Chateauvieux<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: sebastien.chateauvieux@lbmcc.lu<br />

Miss Claude Condé<br />

labotory of fundamental virology <strong>and</strong><br />

immunology<br />

Ulg<br />

1, avenue de ll'hôpital Bat B34<br />

4000 Liège<br />

BEL<br />

Email: claude_conde@hotmail.com


Dr. Aless<strong>and</strong>ro Corsaro<br />

Laboratory of Pharmacology, Dept.<br />

Oncology Biology <strong>and</strong> Genetics,<br />

University of Genova<br />

Viale benedetto XV<br />

16132 Genova<br />

ITA<br />

Email: ale.corsaro@unige.it<br />

Miss Brigitte Czepukojc<br />

Bioorganische Chemie-School of<br />

Pharmacy<br />

Saarl<strong>and</strong> University<br />

Universitaet Campous<br />

66123 Saarbruecken<br />

DEU<br />

Email: b.czepukojc@mx.uni-saarl<strong>and</strong>.de<br />

Mrs. Saskia De Meyer<br />

LSCA<br />

Agilent Technologies<br />

Pegasus Park - De Kleetlaan 5 Bus 9<br />

1831 Diegem<br />

BEL<br />

Email: godelieve_minet@agilent.com<br />

Mrs. Debora Dell'Era Dosch<br />

Europe<br />

Enzo Life Sciences<br />

Industriestrasse 17<br />

CH-4415 Lausen<br />

CHE<br />

Email: agrainger@enzolifesciences.com<br />

Dr. Dieter Demon<br />

Biochemistry - Fac. Veterinary Sciences<br />

Ghent University<br />

Salisburylaan 133<br />

9820 Merelbeke (Ghent)<br />

BEL<br />

Email: dieter.demon@ugent.be<br />

Mr. Luciano Di Croce<br />

Epigenetic events in Cancer<br />

CRG/ICREA<br />

Dr. Aiguader 88<br />

8003 Barcelona<br />

ESP<br />

Email: luciano.dicroce@crg.es<br />

Pr. Pascale Cossart<br />

Interactions Bactéries Cellules<br />

Institut Pasteur<br />

25 rue du Docteur Roux<br />

75015 Paris<br />

FRA<br />

Email: pcossart@pasteur.fr<br />

Mr. Incarnato Danny<br />

NEUROSCIENCE DEPARTMENT<br />

SIENA BIOTECH SPA<br />

STRADA DEL PETRICCIO E<br />

BELRIGUARDO 35<br />

53100 SIENA<br />

ITA<br />

Email: dincarnato@sienabiotech.it<br />

Mr. Nicolas Dejeans<br />

PMNT 7369<br />

université catholique de Louvain<br />

73, avenue E. Mounier<br />

1200 Bruxelles<br />

BEL<br />

Email: nicolas.dejeans@uclouvain.be<br />

Pr. Aldona Dembinska-Kiec<br />

Department of Clinical Biochemistry<br />

Jagiellonian University<br />

Kopernika 15 a<br />

31-501 Cracow<br />

POL<br />

Email: mbkiec@cyf-kr.edu.pl<br />

Mr. Harald Desch<br />

Leica Microsystems<br />

3, rue des Joncs<br />

L-1818 Howald<br />

LUX<br />

Email: steve.engel@lecuit.lu<br />

Dr. Maria Di Girolamo<br />

G-Protein-Mediated Signalling<br />

Laboratory<br />

Consorzio Mario Negri Sud<br />

Via Nazionale 8/A<br />

66030 S. Maria Imbaro<br />

ITA<br />

Email: mdigirolamo@negrisud.it<br />

201<br />

Miss Isabelle Coupienne<br />

Virology <strong>and</strong> Immunology<br />

1, avenue de l'Hopital<br />

4000 Liege<br />

BEL<br />

Email: icoupienne@ulg.ac.be<br />

Mrs. Odete de Cruz e Silvia<br />

Laboratorio Neurociencias<br />

Centro Biologia Celular, Universidade de<br />

Aveiro<br />

Campus Santiago<br />

3810-193 Aveiro<br />

PRT<br />

Email: aghenriques@ua.pt<br />

Pr. Antonio del Sol<br />

Computational Biology Unti<br />

Luxembourg Center for Systems<br />

Biomedicine-University of Luxembourg<br />

162a, avenue de la faiencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: antonio.delsol@uni.lu<br />

Mrs. Kristel Demeyere<br />

Biochemistry-Fac veterinary sciences<br />

Ghent University<br />

Salisburylaan 133<br />

9820 MERELBEKE (GHENT)<br />

BEL<br />

Email: Kristel.demeyere@ugent.be<br />

Mr. Brian DeWitt<br />

Biorefinery<br />

IBBL<br />

6, rue Nicolas Barblé<br />

1210 Luxembourg<br />

LUX<br />

Email: brian.dewitt@ibbl.lu<br />

Pr. Cinzia Di Pietro<br />

biology <strong>and</strong> genetics<br />

university of catania<br />

via s. sofia 87<br />

95124 catania<br />

ITA<br />

Email: dipietro@unict.it


Dr Marc Diederich<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: marc.diederich@lbmcc.lu<br />

Dr. Catherine Dostert<br />

Laboratoire de Neurobiologie<br />

Université du Luxembourg<br />

162A, avenue de la faïencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: catherine.dostert@uni.lu<br />

Dr. Joanna Dulinska-Litewka<br />

Chair of Medical Biochemistry<br />

Jagiellonian University Medical College<br />

Kopernika 7<br />

31-034 Krakow<br />

POL<br />

Email: mblitewk@cyf-kr.edu.pl<br />

Mr. Steve Engel<br />

Lecuit Opto-Technical / Leica<br />

Microsystems<br />

3, rue des Joncs<br />

L-1818 Howald<br />

LUX<br />

Email: steve.engel@lecuit.lu<br />

Miss Yuen Ngan Fan<br />

university of Liverpool<br />

Crown street<br />

L69 7ZB Liverpool<br />

GBR<br />

Email: carolfan@liverpool.ac.uk<br />

Pr. Dean Felsher<br />

Felsher Laboratory<br />

Stanford University<br />

269 Campus Drive<br />

94305-5151 Stanford<br />

USA<br />

Email: dfelsher@stanford.edu<br />

Dr. Sascha Dietrich<br />

Adaptive Immunregulation (G331)<br />

Im Neuenheimer Feld 580<br />

69120 He<br />

Im Neuenheimer Feld 410<br />

69120 Heidelberg<br />

DEU<br />

Email: sascha.dietrich@med.uniheidelberg.de<br />

Mr. Peng Du<br />

Bioorganische Chemie-School of<br />

Pharmacy<br />

Saarl<strong>and</strong> University<br />

Universitaet Campous<br />

66123 Saarbruecken<br />

DEU<br />

Email: du-peng@live.com<br />

Dr. Eric Duplan<br />

IPMC<br />

CNRS<br />

660 Route des lucioles<br />

6560 Valbonne<br />

FRA<br />

Email: duplan@ipmc.cnrs.fr<br />

Dr. Esra Erdal<br />

Department of Medical Biology <strong>and</strong><br />

Genetics<br />

DEU Medical School<br />

Mithatpasa caddesi, Temel Bilimler<br />

Binasi, Kat 3<br />

34350 izmir<br />

TUR<br />

Email: esra.erdal@deu.edu.tr<br />

Dr. Margarida Fardilha<br />

Signal Transduction<br />

Campus de Santiago<br />

3810-193 Aveiro<br />

PRT<br />

Email: mfardilha@ua.pt<br />

Mr. Paul Felten<br />

Laboratoire de Neurobiologie<br />

Université du Luxembourg<br />

162A, avenue de la faïencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: paul.felten@uni.lu<br />

202<br />

Dr. Gauthier Dorban<br />

Laboratoire de Neurobiologie<br />

Université du Luxembourg<br />

162A, avenue de la faïencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: gauthier.dorban@uni.lu<br />

Mrs. Elodie Dubus<br />

AUREUS PHARMA<br />

174 Quai de Jemmapes<br />

75010 PARIS<br />

FRA<br />

Email: elodie.dubus@aureus-pharma.com<br />

Mrs. Bessem Cecilia Elango<br />

Laboratory Techniciant<br />

Institude Of Biomedical Research<br />

Rose Garden Muea-Buea po box 485<br />

south west region Cameroon<br />

237 Buea<br />

CMR<br />

Email: kimbifuen@yahoo.co.uk<br />

Dr. Zsolt Fabian<br />

Regenerative Medicine Institute<br />

University Road<br />

Co. Galway Galway<br />

IRL<br />

Email: zsolt.fabian@nuigalway.ie<br />

Pr. Dean Felsher<br />

Felsher laboratory<br />

Stanford<br />

269 Campus Dr<br />

94305-5151 Stanford<br />

USA<br />

Email: dfelsher@stanford.edu<br />

Mrs. Sanjanie Fern<strong>and</strong>o<br />

PRF lab, Department of Microbiology<br />

Latrobe University<br />

Kingsbury Drive<br />

VIC3086 Bundoora<br />

AUS<br />

Email:<br />

sgfern<strong>and</strong>o@students.latrobe.edu.au


Pr. Paul Fisher<br />

Microbial Cell Biology<br />

Kingsbury Drive<br />

VIC 3086 Melbourne<br />

AUS<br />

Email: P.Fisher@latrobe.edu.au<br />

Mr. Sylvain Fraineau<br />

Institut de Physiologie et Biologie<br />

Cellulaires<br />

Université de Poitiers<br />

1 rue Georges Bonnet<br />

86022 Poitiers<br />

FRA<br />

Email: sylvain.fraineau@univ-poitiers.fr<br />

Mr Francois Gaascht<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: francois.gaascht@lbmcc.lu<br />

Dr. Michael Gale<br />

STRIDE Center<br />

University of Washington<br />

Box 357650<br />

98195-7650 Seattle<br />

USA<br />

Email: mgale@uw.edu<br />

Dr. Dorota Gil<br />

Chair of Medical Biochemistry<br />

Jagiellonian University Medical College<br />

Kopernika 7<br />

31-034 Krakow<br />

POL<br />

Email: dgil@poczta.fm<br />

Dr. Bernhard Goetz<br />

Prophac<br />

5 rangwee<br />

2412 luxembourg<br />

LUX<br />

Email: bernhard.goetz@prophac.lu<br />

Dr Cristina Florean<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: cristina.florean@lbmcc.lu<br />

Mrs Elodie Frenger<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: elodie.frenger@lbmcc.lu<br />

Miss Karoline Gäbler<br />

Signal Transduction Laboratory<br />

162a, avenue de la Faincerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: karoline.gaebler@uni.lu<br />

Pr. Roberto Gambari<br />

BIOCHEMISTRY AND MOLECULAR<br />

BIOLOGY DEPARTMENT<br />

FERRARA UNIVERSITY<br />

VIA FOSSATO DI MORTARA 74<br />

44121 FERRARA<br />

ITA<br />

Email: gam@unife.it<br />

Mrs. Agnes Giot<br />

GE Healthcare<br />

koterveldstraat 2<br />

1831 Diegem<br />

BEL<br />

Email: agnes.giot@ge.com<br />

Dr Stefania Gonfloni<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: stefania.gonfloni@lbmcc.lu<br />

203<br />

Dr. Paola Fogar<br />

Department of Laboratory Medicine<br />

University-Hospital of Padova<br />

Via Giustiniani 2<br />

35128 Padova<br />

MRT<br />

Email: paola.fogar@unipd.it<br />

Dr. Nicolai Fricker<br />

Department of Immunogenetics, DKFZ<br />

Im Neuenheimer Feld 267<br />

69120 Heidelberg<br />

DEU<br />

Email: n.fricker@dkfz.de<br />

Dr Anthoula Gaigneaux<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: anthoula.gaigneaux@lbmcc.lu<br />

Dr. Alex Gamma<br />

Zurich University Psychiatric Hospital<br />

Lenggstrasse 31<br />

8032 Zurich<br />

CHE<br />

Email: agamma@ethz.ch<br />

Mr. Christophe Glorieux<br />

PMNT 7369<br />

université catholique de Louvain<br />

73, avenue E. Mounier<br />

1200 Bruxelles<br />

BEL<br />

Email: christophe.glorieux@uclouvain.be<br />

Dr. Stefania Gonfloni<br />

Molecular Genetics<br />

University of Rome 'Tor Vergata<br />

via della Ricerca Scientifica<br />

133 Rome<br />

ITA<br />

Email: Stefania.Gonfloni@uniroma2.it


Dr. Marie-Lise Gougeon<br />

Antiviral Immunity, Biotherapy <strong>and</strong><br />

Vaccine Unit<br />

INSTITUT PASTEUR<br />

25-28 rue du Dr. Roux<br />

75015 Paris<br />

FRA<br />

Email: mlgougeo@pasteur.fr<br />

Dr. Luc Gr<strong>and</strong>barbe<br />

Laboratoire de Neurobiologie<br />

Université du Luxembourg<br />

162A, avenue de la faïencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: luc.gr<strong>and</strong>barbe@uni.lu<br />

Mr. Anne Grosse-Wilde<br />

Ozinsky Lab<br />

Institute for Systems Biology<br />

1441 N 34th St<br />

98103 Seattle<br />

USA<br />

Email:<br />

AGWILDE@SYSTEMSBIOLOGY.ORG<br />

Dr. Yvette Habraken<br />

Virology <strong>and</strong> Immunology<br />

University of Liège<br />

Allée de l'Hopital 1, Bât B34<br />

4000 Liège<br />

BEL<br />

Email: yvette.habraken@ulg.ac.be<br />

Miss Luciana Harumi Osaki<br />

Gastrointestinal Epithelium Biology<br />

Av. Prof. Lineu Prestes<br />

5508900 Sao Paulo<br />

BRA<br />

Email: luciana.osaki@usp.br<br />

Mrs Liliane Hermes<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: liliane.hermes@lbmcc.lu<br />

Miss Aysim Gozukizil<br />

Molecular biology research labratory<br />

Dokuz Eylul University<br />

inciralti<br />

35330 izmir<br />

TUR<br />

Email: a.gkizil@hotmail.com<br />

Dr Cindy Gr<strong>and</strong>jenette<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: cindy.gr<strong>and</strong>jenette@lbmcc.lu<br />

Miss Geraldine Guerin-Peyrou<br />

Technical Support<br />

Polyplus-transfection<br />

Bioparc, Bd S. Brant<br />

67401 Illkirch<br />

FRA<br />

Email: ggp@polyplus-transfection.com<br />

Mrs Sheherazade Hajjouli<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: sheherazade.hajjouli@lbmcc.lu<br />

Mrs Estelle Henry<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: estelle.henry@lbmcc.lu<br />

Dr. Tony Heurtaux<br />

Laboratoire de Neurobiologie<br />

Université du Luxembourg<br />

162A, avenue de la faïencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: tony.heurtaux@uni.lu<br />

204<br />

Miss Astrid Grainger<br />

Benelux<br />

Enzo Life Sciences<br />

Melkerijweg 3<br />

BE-2240 Z<strong>and</strong>hoven<br />

BEL<br />

Email: agrainger@enzolifesciences.com<br />

Mrs Christina Grigorakaki<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: christina.grigorakaki@lbmcc.lu<br />

Dr. Claude Haan<br />

Signal Transduction Laboratory<br />

University of Luxembourg<br />

162A avenue de la faiencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: claude.haan@uni.lu<br />

Pr. Pirkko Härkonen<br />

Institute of Biomedicine<br />

University of Turku<br />

Kiinamyllynkatu 10<br />

20720 Turku<br />

FIN<br />

Email: harkonen@utu.fi<br />

Miss Ivana Hermanova<br />

CLIP Charles University in Prague<br />

V Uvalu 84<br />

15006 Prague<br />

CZE<br />

Email: ivana.hermanova@lfmotol.cuni.cz<br />

Dr. Xavier Hever<br />

Laboratoire de Neurobiologie<br />

Université du Luxembourg<br />

162A, avenue de la faïencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: xavier.hever@uni.lu


Mr. Kasper Hoebe<br />

Cincinnati Children's Hospital<br />

3333 Burnet Avenue<br />

45229 Cincinnati<br />

USA<br />

Email: kasper.hoebe@cchmc.org<br />

Dr. Andrew Hufton<br />

Molecular Systems Biology<br />

Meyerhofstr. 1<br />

69117 Heidelberg<br />

DEU<br />

Email: <strong>and</strong>rew.hufton@embo.org<br />

Pr. Claus Jacob<br />

Bioorganische Chemie-School of<br />

Pharmacy<br />

Saarl<strong>and</strong> University<br />

Universitaet Camous<br />

66123 Saarbruecken<br />

DEU<br />

Email: c.jacob@mx.uni-saarl<strong>and</strong>.de<br />

Mr. Patryk Janus<br />

Center for Translational Research <strong>and</strong><br />

Molecular Biology of Cancer<br />

Wybrzeze Armii Krajowej 15<br />

44-101 Gliwice<br />

POL<br />

Email: patrykjanus@gmail.com<br />

Mr. Jang-Yeon Jeong<br />

Immunobiology<br />

Jeju National University<br />

66 jejudaehakno<br />

690756 Jeju<br />

KOR<br />

Email: ckdgml3735@hanmail.net<br />

Pr. Jang Jung-Hee<br />

College of Oriental Medicine<br />

Daegu Haany University<br />

165 Sang-dong, Suseong-gu<br />

706-828 Daegu<br />

KOR<br />

Email: pamy202@paran.com<br />

Miss Claire Hoenen<br />

Laboratoire de Neurobiologie<br />

Université du Luxembourg<br />

162A, avenue de la faïencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: claire.hoenen@uni.lu<br />

Miss Jasmina Ilievska<br />

Microbial Cell Biology<br />

La Trobe University<br />

Plenty Rd<br />

3086 Bundoora<br />

AUS<br />

Email: jilievska@students.latrobe.edu.au<br />

Dr Monika Jain<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: monika.jain@lbmcc.lu<br />

Mr. R.G.P.T. Jayasooriya<br />

Immunobiology<br />

Jeju National University<br />

66 jejudaehakno<br />

690756 Jeju<br />

KOR<br />

Email: www.jayasamanthas@gmail.com<br />

Mrs. Elisabeth John<br />

Life Sciences Research Unit,<br />

FSTCUniveristy of Luxembourg<br />

Avenue de la Faiencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: elisabeth.john@uni.lu<br />

Dr. Stefanie Kaempf<br />

Medical Manager<br />

NonWoTecc Medical GmbH<br />

Nattermannallee 1<br />

50829 Cologne<br />

DEU<br />

Email: s.kaempf@nonwotecc.de<br />

205<br />

Mr. Jean Marie Hoornaert<br />

EUROGENTEC<br />

5 rue du Bois Saint Jean<br />

4102 Seraing<br />

BEL<br />

Email: j.m.hoornaert@eurogentec.com<br />

Dr. Ute Inegbenebor<br />

Medical <strong>and</strong> Research Laboratory<br />

Ambrose Alli University<br />

Benin Auchi Way<br />

23401 Ekpoma<br />

NGA<br />

Email: druteinegbenebor@yahoo.com<br />

Mr. Yeon-Jeong Jang<br />

Immunobiology<br />

Jeju National University<br />

66 jejudaehakno<br />

690756 Jeju<br />

KOR<br />

Email: ckdgml3735@hanmail.net<br />

Dr. Yongtark Jeon<br />

Seoul National University Bundang<br />

Hospital<br />

300 Gumidong<br />

463-707 Sungnam<br />

KOR<br />

Email: asidof1@snu.ac.kr<br />

Dr. Hye Jin Jung<br />

Department of Biotechnology<br />

Yonsei University<br />

262 Seongsanno<br />

120-749 Seoul<br />

KOR<br />

Email: hjjung96@gmail.com<br />

Mr. Thorben Kaetzel<br />

lsru<br />

Université du Luxembourg<br />

162a, avenue de la Faïencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: thorben.kaetzel@uni.lu


Mr. Surasee Kamlua<br />

Centre for Research <strong>and</strong> Development of<br />

Medical Diagnostic Laboratories<br />

Khon Kaen University<br />

Mitrapap<br />

40002 Khon Kaen<br />

THA<br />

Email: kamikaze_me@hotmail.com<br />

Mr. Chang-Hee Kang<br />

Immunobiology<br />

Jeju National University<br />

66 jejudaehakno<br />

690756 Jeju<br />

KOR<br />

Email: ckdgml3735@hanmail.net<br />

Dr. Michael Katze<br />

STRIDE Center<br />

University of Washington<br />

Box 358070<br />

WA Seattle<br />

USA<br />

Email: honey@uw.edu<br />

Dr Mareike Kelkel<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: mareike.kelkel@lbmcc.lu<br />

Pr. Yong Beom Kim<br />

Department of OB&GYN, Seoul National<br />

University Bundang Hospital<br />

166 Gumi-ro, Bundang-gu<br />

463-707 SEONGNAM<br />

KOR<br />

Email: ybkimlh@snubh.org<br />

Dr. Hee Seung Kim<br />

Seoul National University<br />

Unavailable<br />

110-744 Seoul<br />

KOR<br />

Email: bboddi0311@snu.ac.kr<br />

Mrs. Emine K<strong>and</strong>emis<br />

molecular biology <strong>and</strong> cancer research lab<br />

dokuz eylul school of medicine<br />

Inciralti/Balcova<br />

35350 Izmir<br />

TUR<br />

Email: eminecelik@gmail.com<br />

Mr Tommy Karius<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: tommy.karius@lbmcc.lu<br />

Dr. Alex<strong>and</strong>er Kel<br />

Chief Scientific Officer<br />

geneXplain GmbH<br />

Am Exer 10b<br />

D-38302 Wolfenbuettel<br />

DEU<br />

Email: alex<strong>and</strong>er.kel@genexplain.com<br />

Mr. Khairan Khairan<br />

Bioorganische Chemie-School of<br />

Pharmacy<br />

Saarl<strong>and</strong> University<br />

Universitaet Campous<br />

66123 Saarbruecken<br />

DEU<br />

Email: khannazia-yusuf@yahoo.com<br />

Mr. Gi-Young Kim<br />

Immunobiology<br />

Jeju National University<br />

66 jejudaehakno<br />

690756 Jeju<br />

KOR<br />

Email: immunkim@jejunu.ac.kr<br />

Dr. Olga Kofanova<br />

Biorefinery<br />

IBBL<br />

6, rue Nicolas Barblé<br />

1210 Luxembourg<br />

LUX<br />

Email: olga.kofanova@ibbl.lu<br />

206<br />

Pr. Jaeku Kang<br />

Department of Pharmacology<br />

College of Medicine, Konyang University<br />

685, Gasuwon-dong, Seo-gu,<br />

302-718 Daejeon<br />

KOR<br />

Email: jaeku@konyang.ac.kr<br />

Dr. Vladimir Katanaev<br />

Katanaev<br />

University of Konstanz<br />

Universitätsstrasse 10, Box 643<br />

78457 Konstanz<br />

DEU<br />

Email: vladimir.katanaev@unikonstanz.de<br />

Dr. Olga Kel-Margoulis<br />

Applied Life Science Informatics<br />

geneXplain GmbH<br />

Am Exer 10b<br />

D-38302 Wolfenbuettel<br />

DEU<br />

Email: olga.kelmargoulis@genexplain.com<br />

Miss Kiyoon Kim<br />

Department of Biochemistry <strong>and</strong><br />

Molecular Biology, Rm 419<br />

School of Medicine, Kyung Hee<br />

University<br />

1 Hoegidong, Dongdaemungu<br />

130-701 Seoul<br />

KOR<br />

Email: navy07i@naver.com<br />

Dr. Hyun-Duck Kim<br />

Preventive <strong>and</strong> social dentistry<br />

Seoul Ntional University School of<br />

Dentistry<br />

28 Yeonkeon-Dong Jongno-Ku<br />

110-749 Seoul<br />

KOR<br />

Email: hyundkim@snu.ac.kr<br />

Pr. Andrew Koff<br />

Cell Cycle Regulation<br />

Memorial Sloan-Kettering Cancer Center<br />

1275 York Avenue<br />

10021 New York, New York<br />

USA<br />

Email: koffa@mskcc.org


Mr. Hendrik Koopmans<br />

Cell Biology<br />

Westburg BV<br />

PO Box 214<br />

3830 AE Leusden<br />

NLD<br />

Email: F.v<strong>and</strong>erLoop@westburg.eu<br />

Miss Karolina Kramarzova<br />

CLIP, Charles University in Prague<br />

V Uvalu 84<br />

15006 Prague<br />

CZE<br />

Email: karolina.kramarzova@fnmotol.cz<br />

Miss Imge Kunter<br />

Molecular biology <strong>and</strong> cancer research<br />

lab<br />

Dokuz Eylul University School of<br />

Medicine<br />

inciralti/ balcova<br />

35350 Izmir<br />

TUR<br />

Email: imge_kunter@yahoo.com<br />

Mr. An<strong>and</strong> Lachmansingh<br />

Sales<br />

Bio-Connect B.V.<br />

Begonialaan 3a<br />

NL-6851 TE Huissen<br />

NLD<br />

Email: peters@bio-connect.nl<br />

Mr. Chae Hyeong Lee<br />

Dongguk University Ilsan Hospital<br />

Siksa-dong 814<br />

410-773 Goyang<br />

KOR<br />

Email: gynelee@paran.com<br />

Mrs. Elisabeth Letellier<br />

Signal Transduction Laboratory<br />

University of Luxembourg<br />

162 A avenue de la Faïencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: elisabeth.letellier@uni.lu<br />

Mr. Joanna Kopecka<br />

Department of Genetics, Biology <strong>and</strong><br />

Biochemistry, University of Torino<br />

Via Santena 5/bis<br />

10126 Torino<br />

ITA<br />

Email: joanna.kopecka@unito.it<br />

Dr. Stephanie Kreis<br />

Signal Transduction Laboratory<br />

University fo Luxembourg<br />

162A Ave de la Faiencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: stephanie.kreis@uni.lu<br />

Miss Hoi Tung Kwan<br />

NGAN HEXTAN<br />

DEPARTMENT OF O & G, LKS<br />

FACULTY OF MEDICINE, THE<br />

UNIVERSITY OF HONG KONG<br />

21 SASSON ROAD<br />

0 HONG KONG<br />

HKG<br />

Email: virg1234@hotmail.com<br />

Dr. Inna Lavrik<br />

DKFZ/Bioquant<br />

Im Neuenheimer Feld 280<br />

69120 Heidelberg<br />

DEU<br />

Email: i.lavrik@dkfz.de<br />

Miss Helene Leger<br />

IHES<br />

35 route de chartres<br />

91440 Bures-sur-Yvette<br />

FRA<br />

Email: leger@ihes.fr<br />

Mrs. Nan Li<br />

Institute of Immunology<br />

Second Military Medical University<br />

800 Xiangyin Road<br />

200433 Shanghai<br />

CHN<br />

Email: linan@immunol.org<br />

207<br />

Mrs. Peyda Korhan<br />

Molecular Biology <strong>and</strong> Cancer Research<br />

Lab<br />

Dokuz Eylul University School of<br />

Medicine<br />

Inciralti, Balcova<br />

35350 Izmir<br />

TCA<br />

Email: korhanpeyda@hotmail.com<br />

Pr. Guido Kroemer<br />

INSERM U848<br />

UniversitÈ Paris DÈscartes<br />

15 rue del l\'Ecole de MÈdecine<br />

75006 Paris<br />

FRA<br />

Email: kroemer@orange.fr<br />

Pr. Ho Jeong Kwon<br />

Department of Biotechnology<br />

Yonsei University<br />

262 Seongsanno<br />

120-749 Seoul<br />

KOR<br />

Email: kwonhj@yonsei.ac.kr<br />

Miss Aurore Lecat<br />

Virology <strong>and</strong> Immunology (GIGA-R)<br />

avuenue de l'hopital<br />

4000 Liège<br />

BEL<br />

Email: aurore.lecat@ulg.ac.be<br />

Mrs Noemie Legr<strong>and</strong><br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: noemie.legr<strong>and</strong>@lbmcc.lu<br />

Miss Maria Liivr<strong>and</strong><br />

Systems Biology, LSRU, FSTC<br />

University of Luxembourg<br />

162a, avenue de la Faiencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: marialiivr<strong>and</strong>@gmail.com


Mrs. Chen Lin<br />

AG Katanaev<br />

Universitaet Konstanz<br />

Universitaetsstrasse 10<br />

78464 Konstanz<br />

DEU<br />

Email: Chen.Lin@uni-konstanz.de<br />

Mrs. Evgeniya Logashenko<br />

Laboratory of biochemistry of nucleic<br />

acids<br />

8, Lavrentev ave<br />

630090 Novosibirsk<br />

RUS<br />

Email: evg_log@niboch.nsc.ru<br />

Mr. Ev<strong>and</strong>ro Luis de Oliveira Niero<br />

Cell <strong>and</strong> Molecular Biology<br />

Av. Prof. Lineu Prestes<br />

5508900 Sao Paulo<br />

BRA<br />

Email: eloniero@usp.br<br />

Dr. Thomas Luft<br />

Adaptive Immune Regulation<br />

University of Heidelberg<br />

Im Neuenheimer Feld 410<br />

69120 Heidelberg<br />

DEU<br />

Email: t.luft@dkfz.de<br />

Dr. Eva Matalova<br />

Department of Physiology<br />

University of Veterinary <strong>and</strong><br />

Pharmaceutical Sciences<br />

Palackeho 1-3<br />

612 42 Brno<br />

CZE<br />

Email: matalova@iach.cz<br />

Pr. Gerry Melino<br />

Dip Experimental Medicine<br />

University Rome Tor Vergata<br />

via Montpellier 1<br />

133 Rome<br />

ITA<br />

Email: melino@uniroma2.it<br />

Mrs. Qiuyan Liu<br />

Institute of Immunology<br />

Second Military Medical University<br />

800 Xiangyin Road<br />

200433 Shanghai<br />

CHN<br />

Email: lqy1969@yahoo.com.cn<br />

Miss Axelle Loriaux<br />

Promega Benelux<br />

Schipholweg 1<br />

2316XB LEIDEN<br />

NLD<br />

Email: axelle.loriaux@promega.com<br />

Dr. Francesca Luchetti<br />

Cytometry<br />

University<br />

Via Ca' le Suore 2<br />

61029 Urbino<br />

ITA<br />

Email: francesca.luchetti@uniurb.it<br />

Mrs Fabienne Mack<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: fabienne.mack@lbmcc.lu<br />

Dr. Conny Mathay<br />

Biorefinery<br />

IBBL<br />

6, rue Nicolas Barblé<br />

1210 Luxembourg<br />

LUX<br />

Email: conny.mathay@ibbl.lu<br />

208<br />

Miss Julia Lochead<br />

PromoCell GmbH<br />

PromoCell GmbH<br />

Sickingenstrasse 63/65<br />

69126 Heidelberg<br />

DEU<br />

Email: info@promocell.com<br />

Miss Sophie Losciuto<br />

Laboratoire de Neurobiologie<br />

Université du Luxembourg<br />

162A, avenue de la faïencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: sophie.losciuto@uni.lu<br />

Miss Anne-Marie Lüchtenborg<br />

AG Katanaev<br />

Universität Konstanz<br />

Universitätsstrasse 10<br />

78457 Konstanz<br />

DEU<br />

Email: anne-marie.luechtenborg@unikonstanz.de<br />

Dr. Irena Manov<br />

Pediatric Research <strong>and</strong> Electron<br />

Microscopy Unit<br />

Technion-Israel Institute of Technology<br />

Efron 1<br />

31096 Haifa<br />

ISR<br />

Email: irmanov@tx.technion.ac.il<br />

Dr. Mark Mattson<br />

Laboratory of Neurosciences<br />

National Institute on Aging<br />

251 Bayview Boulevard - BRC 5C214<br />

21224 Baltimore<br />

USA<br />

Email: mattsonm@grc.nia.nih.gov<br />

Miss Mohd Daud Melor<br />

INSTITUTE OF MEDICAL<br />

MOLECULAR BIOTECHNOLOGY<br />

UITM SUNGAI BULOH CAMPUS<br />

JALAN HOSPITAL<br />

47000 SUNGAI BULOH<br />

MYS<br />

Email: impaxmelor@gmail.com


Mr. Romuald Menth<br />

Technical Support<br />

Polyplus-transfection<br />

Bioparc, Bd S. Brant<br />

67401 Illkirch<br />

FRA<br />

Email: rmenth@polyplustransfection.com<br />

Dr. Nicola Miglino<br />

Departement Biomedizin Pulmonology<br />

University Hospital Basel<br />

Hebelstrasse 20<br />

4031 Basel<br />

CHE<br />

Email: nicola.miglino@unibas.ch<br />

Dr Franck Morceau<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: franck.morceau@lbmcc.lu<br />

Dr. Günter Müller<br />

R & D Diabetes Division<br />

Sanofi-Aventis Deutschl<strong>and</strong> GmbH<br />

Industrial Park Hochst Bldg. H821<br />

65926 Frankfurt am Main<br />

DEU<br />

Email: guenter.mueller@sanofiaventis.com<br />

Mr. Olaf Nijst<br />

Unit Vaccinology<br />

RIVM<br />

Anthonie van Leeuwenhoeklaan 9<br />

3721 MA Bilthoven<br />

NLD<br />

Email: Olaf.Nijst@rivm.nl<br />

Mrs Barbora Orlikova<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: barbora.orlikova@lbmcc.lu<br />

Dr. Bernard Metz<br />

Unit Vaccinology<br />

RIVM<br />

A. van Leeuwenhoeklaan 9<br />

3721 MA Bilthoven<br />

NLD<br />

Email: Bernard.Metz@rivm.nl<br />

Dr. Nadezda Mironova<br />

Institute of Chemical Biology <strong>and</strong><br />

Fundamental Medicine SB RAS<br />

Lavrentiev ave. 8<br />

630090 Novosibirsk<br />

RUS<br />

Email: mironova@niboch.nsc.ru<br />

Dr. Eleonora Morga<br />

Laboratoire de Neurobiologie<br />

Université du Luxembourg<br />

162A, avenue de la faïencerie<br />

L-1511 Luxembourg<br />

LUX<br />

Email: eleonora.morga@uni.lu<br />

Dr. Sinem Nalbantoglu<br />

MOLECULAR MEDICINE<br />

LABORATORY<br />

EGE UNIVERSITY FACULTY OF<br />

MEDICINE CHILD HOSPITAL<br />

BORNOVA<br />

35100 IZMIR<br />

TUR<br />

Email: nalbantoglusinem@gmail.com<br />

Mrs Karoline Noworyta<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: karoline.noworyta@lbmcc.lu<br />

Pr. Chris Overall<br />

Centre for Blood Research<br />

University of British Columbia<br />

4.401 Life Sciences Institute, 2350 Health<br />

Sciences Mall<br />

V6T 1Z3 Vancouver<br />

CAN<br />

Email: chris.overall@ubc.ca<br />

209<br />

Pr. Evelyne Meyer<br />

Biochemistry-Fac veterinary sciences<br />

Ghent University<br />

Salisburylaan 133<br />

9820 MERELBEKE (GHENT)<br />

BEL<br />

Email: evelyne.meyer@ugent.be<br />

Miss Brenda Molenberghs<br />

Exhibitor<br />

Cell Signaling Technology Europe<br />

Plesmanlaan 1d<br />

2333 BZ Leiden<br />

NLD<br />

Email: b.molenberghs@bioke.com<br />

Mr Florian Muller<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: florian.muller@lbmcc.lu<br />

Dr. Maria Nardelli<br />

Hans Westerhoff's Research Group,<br />

University of Manchester (UK)<br />

University of Manchester<br />

131 Princess Street<br />

M1 7DN Manchester<br />

GBR<br />

Email: maria.nardelli@manchester.ac.uk<br />

Mrs Marie-Anne Olinger<br />

LBMCC<br />

Hôpital Kirchberg<br />

9, rue Edward Steichen<br />

L-2540 Luxembourg<br />

LUX<br />

Email: marie_anne.olinger@lbmcc.lu<br />

Miss Evin Ozen<br />

Molecular Biology <strong>and</strong> Cancer Research<br />

Lab<br />

Dokuz Eylül Universty Medicine School<br />

Institute of Health Science<br />

Inciralti<br />

35350 Izmir<br />

TUR<br />

Email: evinozen@hotmail.com


Mr. Stefano Papa<br />

Cytometry<br />

University<br />

Via Ca' le Suore 2<br />

61029 Urbino<br />

ITA<br />

Email: stefano.papa@uniurb.it<br />

Mrs. Stéphanie Philippot<br />

LIMBP<br />

Université Paul Verlaine - Metz<br />

Campus Bridoux rue Général Delestraint<br />

57070 METZ<br />

FRA<br />

Email: stephanie.philippot@univ-metz.fr<br />

Mr. Pavel Pitule<br />

Department of Histology <strong>and</strong> Embryology<br />

Karlovarska 48<br />

301 00 Plzen<br />

CZE<br />

Email: pitulep@seznam.cz<br />

Dr. Bali Pulendran<br />

Emory VaccineCenter at Yerkes<br />

Emory University<br />

954 Gatewood Road<br />

30329 Atlanta<br />

USA<br />

Email: pbackma@emory.edu<br />

Dr. Chiara Riganti<br />

Biochemistry Unit, Department of<br />

Genetics, Biology <strong>and</strong> Biochemistry<br />

University of Turin<br />

via Santena 5/bis<br />

10126 Turin<br />

ITA<br />

Email: chiara.riganti@unito.it<br />

!"## $%&%'% ()*)+%&<br />

,"-.&./"%0%+01223'.&./"%<br />

45%'3%06%0&7$.8"+)&<br />

9::: ;";<br />

Email:<br />

?%&%'%@#)*)+%&A#+36%'+@3&/@)B@*%<br />

Dr. Sarah Payne<br />

Cell Technologies<br />

GE Healthcare<br />

Amersham Place<br />

HP7 9NA Little Chalfont<br />

GBR<br />

Email: sarah.payne@ge.com<br />

Dr. Bob Phillips<br />

IBBL<br />

IBBL<br />

6, rue Nicolas Barblé<br />

1210 Luxembourg<br />

LUX<br />

Email: marie-paule.hoffmann@ibbl.lu<br />

Dr. Yulia Pollak<br />

Pediatric Research <strong>and</strong> Electron<br />

Microscopy Unit<br />

Technion-Israel Institute of Technology<br />

Efron 1<br />

31096 Haifa<br />

ISR<br />

Email: mdyulia@tx.technion.ac.il<br />

Mr. Roel Quintens<br />

Radiobiology Unit<br />

SCK-CEN<br />

Boeretang 200<br />

B-2400 Mol<br />

BEL<br />

Email: roel.quintens@sckcen.be<br />

Miss Micaela Rocco<br />

Biochemistry, Department of Life<br />

Sciences<br />

Via Vivaldi 43<br />

81100 Caserta<br />

ITA<br />

Email: micaela.rocco@unina2.it<br />

C-@ 4'') ()&5)+"<br />

D%'+-%0E.-0=".F)'.01'+%-)B+".'#G0<br />

H'"5%-#"+I0D.&&%/%0C3*&"'<br />

=%&E"%&6<br />

9 C3*&"'<br />

1J;<br />

Email: )'')@#)&5)+"AB*'"@3B6@"%<br />

210<br />

Pr. Josef Penninger<br />

imba<br />

dr. bohrgasse 3<br />

1030 vienna<br />

AUT<br />

Email: josef.penninger@imba.oeaw.ac.at<br />

Dr. Jacques Piette<br />

Virology <strong>and</strong> Immunology, GIGA-R<br />

University of Liège<br />

Allée de l'Hopital 1, Bât B34<br />

4000 Liège<br />

BEL<br />

Email: jpiette@ulg.ac.be<br />

Pr. Alex<strong>and</strong>er Pukhalsky<br />

Department of Cystic Fibrosis, Research<br />

Centre for Medical Genetics<br />

1 Moskvorechie Street<br />

115478 Moscow<br />

RUS<br />

Email: osugariver@yahoo.com<br />

Miss Valentine Rech De Laval<br />

IBCP<br />

CNRS<br />

7 passage du Vercors<br />

69367 Lyon Cedex 07<br />

FRA<br />

Email: valentine.rech-de-laval@ibcp.fr<br />

Pr. Bernhard Ryffel<br />

Immunology<br />

CNRS<br />

3B ure de la Ferollerie<br />

45071 Orleans<br />

FRA<br />

Email: bryffel@cnrs-orleans.fr<br />

!"## F)6) ()2)-"<br />

J)6".*".&./I0H'"+<br />

(DKLD>F<br />

=.%-%+)'/0M::<br />

=LM9:: !.&<br />

=>;<br />

Email: ')6)@#)2)-"A#BNB%'@*%


!"# $%&'()% *%&+,&,-.+,/<br />

0.,),12/%&3/1(&(+.-4<br />

'&.5("4.+2/,6/-%+%&.%<br />

5.%/4#/4,6.%/78<br />

9:;?@<br />

Email: A%&'()%4%&+,&,-.+,B1A%.)#-,A<br />

J"# ?W,A%4 *%'+("<br />

*24+(A4/Z.,),12<br />

O&.5("4.+2/,6/F'G(A0,'"1<br />

;N]$EHDI<br />

L.%/@00.%+(1"%44,/A/D('(&W(.A("/M()3/=;X<br />

N9;


!" #$"%& '()(&*+,-%<br />

./!##<br />

012%34&56%"78)*"9<br />

:;5"+4">5'3*%78*?<br />

.@ABCD .


!"# $%"&% '&()*+&"*,%<br />

!-.%"/+-0/1*21&0/-"0%31+-4&5&0-166<br />

73&0&(8+1$8//-")%89<br />

:-349/"#;<<br />

=>?@A '"&-"<br />

!BC<br />

Email: /&()*+&"*,%D+8//-")%89#4-<br />

$"9 K00- '"-583<br />

PV$GG<br />

WX.&/%317&"5)L-"M<br />

@Y1"8-1B4Z%"41Q/-&5)-0<br />

PF?=>A P8S-+L*8"M<br />

PCU<br />

Email: %00-#/"-583D3L+55#38<br />

!"# 'c,-/*+&"% 'c%0*,%<br />

P6$VO<br />

$-/c1C0&,-"9&/[<br />

"8-1481ER0R"%31!-3-9/"%&0/<br />

=NANA $-/c<br />

:IK<br />

Email: /c%0*,%D80&,F+-/c#2"<br />

$"9# K00- J%-"0-Z&e5(<br />

V&*Q5&-05-<br />

EIB6HBI1V6aFaHB<br />

=A1K,-08-1I*&1K3L-"/16<br />

;NfA g-++-3<br />

VBP<br />

Email: %00-#,%-"0-Z&e5(DML*#5*+<br />

O"# Q,-/3%0% J%9&3&-,%<br />

')-*"-/&5%31M-0-/&59<br />

609/&/8/-1V&*5)-+&5%31O)[9&591IKQ<br />

>17*9[M&019/"#<br />

;;@^^> $*95*Z<br />

ICQ<br />

Email: 9,%9&3&-,%D.*3[+-"#5).)#"%9#"8<br />

$"# `83&-0 J-""%S<br />

O$H'1N^


!"## $"%&%' (")*"+"%<br />

,%-).&")./.*0<br />

1,23#4+-%<br />

,5%6(4/6$/%5)"678<br />

93:;%?@.5)*<br />

9AB<br />

Email: &")*"+"%CD"%&%'EF)G3#4+-%C/5<br />

!)C I"/%%+ VO"-%<br />

VO"-%<br />

1[YZ<br />

:P;69"--/%6\/@4+06S-)%%-<br />

7PT] Y%R6K)5+#R"F^Q6YZ<br />

AS\<br />

Email: %"/%%+GRO"-%E*?4"/CF.?<br />

H)C ,.@%)-. V5)-O<br />

94@.)4-.)06.D62O4)?4F./.*0Q6H%G-C6<br />

W+F./.*06K"./.*064+J6X%+%-"F#Q6<br />

A+"&%)#"-06.D6X%+.&4<br />

("4/%6@%+%J%--.6B(<br />

:=:]< X%+.&4<br />

[U\<br />

Email: ).@%)-.CR5)-OE5+"*%C"-<br />

2)C !4)"+4 `%+^.&4<br />

94@.)4-.)06.D6Y5F/%"F6\F"J#6<br />

K".FO%?"#-)0<br />

[+#-"-5-%6.D61O%?"F4/6K"./.*064+J6<br />

$5+J4?%+-4/6!%J"F"+%<br />

94&)%+-"%&64&%CQ67<br />

=]TTPT Y.&.#"@")#^<br />

,AS<br />

Email: ?4)'%+E+"@.FOC+#FC)5<br />

H)C \+J)%" `"+.&0%&<br />

K"."+D.)?4-"F#6H%G4)-?%+-<br />

[+#-"-5-615)"%<br />


Notes<br />

218


Notes<br />

219


Notes<br />

220


Berthold Detection Systems<br />

new! ELISA Workstation<br />

new! Sirius L Tube Luminometer<br />

new! Zoom HT Microplate Washer<br />

�����������������<br />

������������������<br />

��������<br />

������������������<br />

�<br />

�<br />

�����������������������������������<br />

����������<br />

���������������������������<br />

Berthold Detection Systems GmbH<br />

Bleichstraße 56–68<br />

75173 Pforzheim<br />

Phone: +49 (0)7231. 9206-0<br />

www.berthold-ds.com<br />

Luminometers<br />

Choose From the Widest Range of Luminometers<br />

for Research <strong>and</strong> Diagnostics<br />

· Single tube luminometers with up to 2 injectors<br />

· Microplate luminometers with up to 4 injectors<br />

· State-of-the-art PC software for Windows 7<br />

ELISA Workstation<br />

The Walk-away Solution for Immunoassays in Microplate Format<br />

· Combines 5 functions: washer, shaker, dispenser, incubator <strong>and</strong> reader<br />

· Walk-away system: microplate in, results out!<br />

· Ideal for low to medium size labs<br />

· Compact size: only 30 cm wide!<br />

Assay Automation<br />

A Range of Assay Processors for Reliable Start-to-finish Automation<br />

· Scalable stacker magazines<br />

· High processing speed <strong>and</strong> excellent accuracy<br />

· Walk-away functionality<br />

Microplate Washing<br />

Robust, Rapid <strong>and</strong> Easy-to-use Instruments for Automated Plate Washing<br />

· Intuitive protocol-based operation<br />

· Flexible add-on dispensers<br />

· From fast ELISA processing to gentle cell washing<br />

· Integrated stacker option<br />

Microplate Coating<br />

by<br />

The Economical Solution for Your Plate Coating Needs<br />

· Ideal for medium throughput (< 800 plates per day)<br />

· High precision <strong>and</strong> reproducibility<br />

· Robust <strong>and</strong> reliable<br />

by<br />

by


We thank our sponsors:

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

Saved successfully!

Ooh no, something went wrong!