29.12.2012 Views

Abstracts (poster) - Wissenschaft Online

Abstracts (poster) - Wissenschaft Online

Abstracts (poster) - Wissenschaft Online

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

<strong>Abstracts</strong> (<strong>poster</strong>)<br />

Mark Wossidlo<br />

γH2AX in the mouse zygote – implications of DNA repair in epigenetic reprogramming [<strong>poster</strong>]<br />

Bernd Schmeck, Janina Lorenz, Philippe Dje N'Guessan, Antje Flieger, Vincent van Laak, Norbert Suttorp, Stefan<br />

Hippenstiel<br />

L. pneumophila induce histone modifications in human lung epithelial cells [<strong>poster</strong>]<br />

Luke Dannenberg, Leo Iniguez, Heather Holster, Peggy Farnham, Bing Ren, David Fisher, Gerd Pfeifer, Hui Liu, Jacob<br />

Kitzman, Fatih Ozsolak<br />

A flexible, high-density array platform for genome-wide characterization of epigenetic and<br />

transcriptional regulatory mechanisms involved in cancer [<strong>poster</strong>]<br />

Anna Katharina Sedello, Gabriele Putz, Frank Buchholz<br />

A Polycomb Group Protein Synergizes with Runx1 in Blocking HSC Differentiation. [<strong>poster</strong>]<br />

Jennifer Cropley, Catherine Suter, David Martin<br />

A progressive multigenerational shift in epigenotype with continuous methyl donor<br />

supplementation [<strong>poster</strong>]<br />

Maja Klug, Sven Heinz, Lucia Schwarzfischer, Sabine Pape, Michael Rehli<br />

Active demethylation of promoter CpGs in post-mitotic cells [<strong>poster</strong>]<br />

Tobias Paprotka, Holger Jeske<br />

Analysis of geminiviral cytosine methylation [<strong>poster</strong>]<br />

Christine Paprotka, Mareike Rust, Katharina Kohl, Jörg Leers, Rainer Renkawitz<br />

Analysis of the effect of CTCF and NuRD on chromatin structure [<strong>poster</strong>]<br />

Nadine Obier, Albrecht M. Müller<br />

Analyzing epigenetic modifications in pluripotent stem cells via intranuclear flow cytometry<br />

[<strong>poster</strong>]<br />

Markus Nees, Christian Hammann, Manu Dubin, Jonathan Chubb, Wolfgang Nellen<br />

Argonaute proteins in Dictyostelium discoideum [<strong>poster</strong>]<br />

Osman El-Maarri1 , Tim Becker2 , Thomas Mikeska3 , Judith Junen1 , Syed Saadi Manzoor1 , Amalia Diaz-Lacava2 , Rainer<br />

Schwaab1 , Thomas Wienker2 , Andreas Waha3 , Johannes Oldenburg1 Association analysis between DNA methylation from total blood and polymorphisms in DNA<br />

methyltransferase (DNMT) genes in healthy individuals: A tendency toward higher methylation<br />

levels in males [<strong>poster</strong>]<br />

Rafal Archacki, T. Sarnowski, J. Halibart-Puzio, Daniel Buszewicz, M. Prymakowska-Bosak, M. Kuras, C. Koncz, A.<br />

Jerzmanowski<br />

ATBRM bromodomain-ATPase and ATSWI3C, representing putative subunits of SWI/SNF<br />

chromatin remodeling complexes, control similar developmental functions in Arabidopsis [<strong>poster</strong>]<br />

Christine Champion, Loïc Ponger, Catherine Senamaud-Beaufort, Dominique Guianvarc'h, Ludovic Halby, Anne-Laure<br />

Guieysse-Peugeot, Paola B. Arimondo<br />

Biochemical approaches to study the DNA methylation mechanisms involved in tumorigenesis<br />

[<strong>poster</strong>]<br />

Christian Rohde, Yingying Zhang, Tomasz P. Jurkowski, Heinrich Stamerjohanns, Richard Reinhardt*, Albert Jeltsch<br />

Bisulfite sequencing Data Presentation and Compilation (BDPC) web server – a useful tool for<br />

DNA methylation analysis [<strong>poster</strong>]<br />

Rafal Archacki, T.J. Sarnowski, J. Halibart-Puzio, D. Buszewicz, M. Prymakowska-Bosak, M. Kuras, C. Koncz, A.<br />

Jerzmanowski<br />

BRM bromodomain-ATPase and ATSWI3C, representing putative subunits of SWI/SNF chromatin<br />

remodeling complexes, control similar developmental functions in Arabidopsis [<strong>poster</strong>]


Katerina Krizova, Miloslava Fojtova, Ann Depicker, Ales Kovarik<br />

Callus-induced epiallelism of an invertedly repeated transgene locus influences its transsilencing<br />

abilities [<strong>poster</strong>]<br />

Britta Wallmen, Simon Wöhrle, Andreas Hecht<br />

Cell specific inducibility of Wnt target genes correlates with epigenetic modifications and<br />

differential promoter occupancy by TCF/LEF proteins [<strong>poster</strong>]<br />

Peter Hemmerich, Stefanie Weidtkamp-Peters, Christian Hoischen, Lars Schmiedeberg, Indri Erliandri, Stephan<br />

Diekmann<br />

CENP-I as a new epigentic mark at centromere chromatin [<strong>poster</strong>]<br />

Claudia Gebhard, Elmar Schilling, Lucia Schwarzfischer-Pfeilschifter, Mathias Ehrich, Michael Rehli<br />

Comparative methylation profiling of tumor samples using methyl-CpG-immuno precipitation<br />

(MCIp) and CpG island microarrays [<strong>poster</strong>]<br />

Ruxandra Farcas, Eberhard Schneider, Ulrich Zechner, Achim Tresch, Hans Zischler, Angelika Daser, Thomas Haaf<br />

Comparison of human and non-human primate methylation status of CpG islands in the promoter<br />

region of CCRK [<strong>poster</strong>]<br />

Martin Herold, Dorte Bohla, Marek Bartkuhn, Imke Panzer, Rainer Renkawitz<br />

CTCF, the highly conserved boundary factor of Drosophila and vertebrates [<strong>poster</strong>]<br />

Jürgen Geisel, Heike Schorr, Gunar H. Heine, Marion Bodis, Ulrich Hübner, Jean-Pierre Knapp, Wolfgang Herrmann<br />

Decreased p66Shc promoter methylation in patients and end-stage renal disease [<strong>poster</strong>]<br />

Martina Dadejova, K. Yoong Lim, Roman Matyasek, Andrew Leitch, Ales Kovarik<br />

Developmental activation of silent rRNA genes is associated with increased transcription activity<br />

of rDNA loci in synthetic hybrids of Nicotiana [<strong>poster</strong>]<br />

OLUSOLA DOKUN, WOLFGANG SCHULZ<br />

DNA hypomethylation of SNCG (synuclein-gamma) in cancer: tumor-specific or cell typespecific?<br />

[<strong>poster</strong>]<br />

Joachim Weitzel<br />

DNA methylation impairs activation of haploid expressed genes in male germ cells. [<strong>poster</strong>]<br />

Christina Klaus, Daniela Kremer, Victoria Kolb-Bachofen<br />

DNA methyltransferases and the influence of cytokines and nitric oxide (NO) on DNA methylation<br />

[<strong>poster</strong>]<br />

Stefanie Stepanow, Kathrin Reichwald, Klaus Huse, Matthias Platzer<br />

Do epigenetic effects at MCHR1 contribute to obesity? [<strong>poster</strong>]<br />

Perrine Gaub, Andrea Tedeschi, Antonio Schmandke, Radhika Puttagunta, Tuan Nguyen, Simone Di Giovanni<br />

Enhancement of neuronal acetylation promotes neurite and axon outgrowth [<strong>poster</strong>]<br />

Jana Krejci, Eva Bartova, Andrea Harnicarova, Roman Hajek, Gabriela Galiova, Stanislav Kozubek<br />

Epigenetic changes in multiple myeloma cells [<strong>poster</strong>]<br />

Georgios J. Vlachojannis, Andreas M. Zeiher, Stefanie Dimmeler<br />

Epigenetic control of the eNOS promoter by DNA methylation in vasculogenic progenitor cell<br />

populations [<strong>poster</strong>]<br />

Maria Elena Torres-Padilla<br />

Epigenetic mechanisms in early mouse development [<strong>poster</strong>]<br />

Michael Michalkiewicz, Teresa Michalkiewicz, Kyle MacGillis<br />

Epigenetic mechanisms in hypertension [<strong>poster</strong>]


Silke Götze, Sonja Sievers, Oliver Müller<br />

Epigenetic regulation in the Wnt signalling pathway [<strong>poster</strong>]<br />

Nadia Sellami, Sabine Adam-Klages, Reiner Siebert, Hans-Jürgen Heidebrecht<br />

Epigenetic Regulation of the Cancer Testis Antigen CT45 [<strong>poster</strong>]<br />

Svend Petersen-Mahrt, Wolf Reik, Siim Pauklin, Heather Coker<br />

Epigenetic Reprogramming of 5-meC via DNA Deamination and DNA Repair [<strong>poster</strong>]<br />

Robert Liefke, Daniela Salat, Jörg Wiedenmann, Franz Oswald, Tilman Borggrefe<br />

ETO, but not AML1/ETO, augments RBP-Jk/Sharp-mediated transcriptional repression of Notch<br />

target genes [<strong>poster</strong>]<br />

Yvonne Möller-Steinbach, Cristina Madeira Alessandre, Vivien Exner, Patti Taranto, Claudia Köhler, Lars Hennig<br />

Function of Polycomb group proteins in the transition to flowering in plants [<strong>poster</strong>]<br />

Cordula Tschuch, Angela Schulz, Armin Pscherer, Meinhard Hahn, Peter Lichter, Daniel Mertens<br />

Functional analysis of candidate genes localized in 13q14.3, a region commonly affected in B-CLL<br />

[<strong>poster</strong>]<br />

Devi Thiagarajan, Sanjeev Khosla<br />

Functional characterisation of mDnmt2 [<strong>poster</strong>]<br />

Soyoung Lim, Johannes Schulte, Hans-Ulrich Schildhaus, Uta Flucke, Phillip Kahl, Roland Schüle, Reinhard Büttner,<br />

Jutta Kirfel<br />

Functional role of Lysine-specific histone methylase-1 in carcinogenesis [<strong>poster</strong>]<br />

Andreas Werner, Mark Carlile<br />

Functional short RNAs from naturally occurring sense/antisense transcripts [<strong>poster</strong>]<br />

Sandra Weiss, Ralf Gilsbach, Frederico Barreto, Achim Lother, Lutz Hein<br />

Heart failure and fibrosis induced by overexpression of methyl-CpG- binding protein 2 (MeCP2) in<br />

transgenic mice [<strong>poster</strong>]<br />

Alexandra Moosmann, Coen Campsteijn, Martina Schmid,, Eric M. Thompson<br />

High diversity of developmental stage-specific histone variants in the larvacean, Oikopleura<br />

dioica [<strong>poster</strong>]<br />

Irene Tiemann-Boege, Christina Curtis, Darryl Shibata, Simon Tavaré<br />

High-throughput analysis of methylation patterns to track cell divisions [<strong>poster</strong>]<br />

Tzvetina Brumbarova, Cecile Doyen, Emilie Bonnefoy, Guillermo Orsi, Pierre Couble, Benjamin Loppin<br />

HIRA functions in Drosophila [<strong>poster</strong>]<br />

Michael Haberland, Rusty Montgomery, Eric N. Olson<br />

Histone deacetylases 1 & 2 control adipogenesis [<strong>poster</strong>]<br />

RAFFAELE TEPERINO, MICHELE LONGO, PAOLA MIRRA, PIETRO FORMISANO, FRANCESCO BEGUINOT, PAOLA UNGARO<br />

HNF4 DIRECTS HISTONE METHYLATION TO SILENCE PED/PEA-15 EXPRESSION IN HUMAN<br />

HEPATOCYTES [<strong>poster</strong>]<br />

Eva Bartova, Abdrea Harnicarova, Jana Krejci, Gabriela Galiova, Stanislav Kozubek<br />

Human embryonic stem cells are characterized by distinct patterns of histone modifications in<br />

comparison with cells of feeder layer [<strong>poster</strong>]<br />

Francesco Nicassio1 , Joseph Vissers4 , Nadia Corrado1 , Liliana Areces1,2 , Steven Bergink3 , Jurgen Marteijn3 , Wim<br />

Vermeulen3 , Maarten van Lohuizen4 , Pier Paolo di Fiore1,2 , Elisabetta Citterio4 Human USP3 is a chromatin modifier required for S-phase progression and genome stability<br />

[<strong>poster</strong>]


Andrea Tedeschi, Tuan Nguyen, Radhika Puttagunta, Perrine Gaub, Simone Di Giovanni<br />

Identification of a novel transcription module for axon outgrowth and regeneration [<strong>poster</strong>]<br />

Japke Polman, E. Ronald de Kloet, Nicole Datson<br />

Identification of binding sites of the Glucocorticoid Receptor in the brain [<strong>poster</strong>]<br />

Philipp Rathert, Arunkumar Dhayalan, Xing Zhang, Renata Jurkowska, Raluca Tamas, Yoichi Shinkai, Xiaodong Cheng,<br />

Albert Jeltsch<br />

Identification of new non-histone targets of the human G9a protein methyltransferase using<br />

peptide arrays [<strong>poster</strong>]<br />

Sylvia Erhardt, Craig M. Betts, Barbara G. Mellone, Gary H. Karpen, Aaron F. Straight<br />

Identification of novel regulators of centromeric chromatin by genome-wide RNAi screening<br />

[<strong>poster</strong>]<br />

Bastian Stielow, Alexandra Sapetschnig, Imme Krüger, Michael Boutros, Guntram Suske<br />

Identification of SUMO-dependent chromatin-associated transcriptional repression components<br />

by a genome-wide RNA interference screen [<strong>poster</strong>]<br />

Harriet Wikman, Michaela Kraemling, Dirk Kemming, Klaus Pantel<br />

Identification of Target Genes in Micrometastatic Lung Cancer by Methylation Arrays [<strong>poster</strong>]<br />

Isabelle GUILLERET, Maria-Chiara OSTERHELD, Richard BRAUNSCHWEIG, Véronique GASTINEAU, Suzanne TAILLENS<br />

Imprinting of tumor-suppressor genes in human placenta. [<strong>poster</strong>]<br />

Alexandre Ceccaldi, Dominique Guianvarc'h, Catherine Senamaud-Beaufort, Renata Jurkowska, Daniel Dauzonne,<br />

Albert Jeltsch, Paola B Arimondo<br />

In quest of DNMT inhibitors [<strong>poster</strong>]<br />

Careen Katryniok, Bernd L. Sorg, Dieter Steinhilber<br />

INDUCTION OF HUMAN 5-LIPOXYGENASE GENE EXPRESSION BY THE HISTONE DEACETYLASE<br />

INHIBITOR TRICHOSTATIN A - INVESTIGATIONS ON THE MECHANISM [<strong>poster</strong>]<br />

Daniela Kremer, Wolfgang Schulz, Victoria Kolb-Bachofen<br />

iNOS-generated NO plasy an critical role in DNA-methylation [<strong>poster</strong>]<br />

Nathalie Jurisch, Bjoern Textor, Peter Angel, Marina Schorpp-Kistner<br />

Involvement of JunB in post-translational HDAC6 regulation and chromatin remodelling [<strong>poster</strong>]<br />

Annette Scharf, Karin Meier, Volker Seitz, Alexander Brehm, Axel Imhof<br />

Kinetics of histone modifications during in vitro chromatin assembly [<strong>poster</strong>]<br />

Fabio Mohn, Michael Weber, Michael Rebhan, Tim Roloff, Jens Richter, Michael Stadler, Miriam Bibel, Dirk Schübeler<br />

Lineage-specific Polycomb targets and de novo DNA methylation define restriction and potential<br />

of neuronal progenitors [<strong>poster</strong>]<br />

Maciej Meglicki, Marta Teperek, Ewa Borsuk<br />

Localization of heterochromatin protein 1α during mouse oogenesis and early embryonic<br />

development [<strong>poster</strong>]<br />

Christian Schmidl, Maja Klug, Tina Böld, Petra Hoffmann, Matthias Edinger, Michael Rehli<br />

Locus-wide detection of cell type specific DNA methylation patterns using comparative methyl-<br />

CpG-Immunoprecipitation (MCIp) [<strong>poster</strong>]<br />

Nicole Happel, Stefan Stoldt, Detlef Doenecke<br />

M-phase specific phosphorylation of histone H1.5 at threonine 10 by GSK3 [<strong>poster</strong>]<br />

Stephanie Jungmichel, Christoph Spycher, Manuel Stucki<br />

Mechanism of MDC1 dimerization [<strong>poster</strong>]


Gunter Reuter, Thomas Rudolph, Sandro Lein, Matthias Walther, Heiko Baisch, Sameer Phalke, Christian Apelt, Sandy<br />

Mietsch<br />

Mechanisms of chromatin differentiation during early embryogenesis of Drosophila [<strong>poster</strong>]<br />

Anette Tippelt<br />

Methyl Primer Express® Software and the Influence of Amplicon Characteristics to the success<br />

rate in DNA Sequencing of bis treated gDNA [<strong>poster</strong>]<br />

Sonja Röhrs, Julia Romani, Wilhelm Dirks, Hans G. Drexler, Hilmar Quentmeier<br />

Methylation profiles of tumour suppressor genes in Hodgkin and non-Hodgkin lymphoma cell<br />

lines [<strong>poster</strong>]<br />

Aditi Kanhere, Vingron Martin, Haas Stefan<br />

Methylation status of promoter depends on its CpG content [<strong>poster</strong>]<br />

Szabolcs Sörös, Wolfgang Fischle<br />

Molecular insights into HP1-chromatin interaction [<strong>poster</strong>]<br />

Renata Jurkowska, Da Jia, Sergey Ragozin, Nils Ansbach, Claus Urbanke, Xing Zhang, Richard Reinhardt, Wolfgang<br />

Nellen, Xiaodong Cheng, Albert Jeltsch<br />

Multimerisation of the Dnmt3L-Dnmt3a complex on DNA and its mechanistic implications [<strong>poster</strong>]<br />

Timo Quante, Lars Tögel, Wolfgang Deppert, Genrich V. Tolstonog<br />

Mutp53 as a modulator of global chromatin organisation [<strong>poster</strong>]<br />

Sascha Tierling, Yingying Zhang, Christian Rohde, Nina Pälmke, Julia Arand, Diana Santacruz, Matthias Platzer, Richard<br />

Reinhardt, Albert Jeltsch, Jörn Walter<br />

NAME21: The National Methylome Project of Human Chromosome 21 [<strong>poster</strong>]<br />

Yingying Zhang, Christian Rohde, Sascha Tierling, Heinrich Stamerjohanns, Matthias Platzer, Richard Reinhardt, Jörn<br />

Walter, Albert Jeltsch<br />

NAME21: The National Methylome Project of Human Chromosome 21 [<strong>poster</strong>]<br />

Franck COURT, Marion BANIOL, Hélène HAGEGE, Julie BORGEL, Jacques PIETTE, Guy CATHALA, Thierry FORNE<br />

NEW INSIGHTS INTO THE IMPRINTED MOUSE Igf2/H19 LOCUS BY 3C-qPCR METHOD [<strong>poster</strong>]<br />

Wibke Peters, Thomas Macherey, Mike Duisken, Sophie Willnow, Bernhard Lüscher, Elmar Weinhold<br />

New S-Adenosyl-L-methionine Analogues to Investigate the Methylome [<strong>poster</strong>]<br />

Andrea Felten, Peter Leister, Karl Heinz Scheidtmann<br />

Novel Coactivators of Androgen Receptor: AATF and ZIP Kinase [<strong>poster</strong>]<br />

Andrea Harni•arová, Eva Bártová, Jana Krej•í, Gabriela Galiová, Stanislav Kozubek<br />

Nuclear location of Oct3/4 and c-myc genes in human embryonic stem cells undergoing<br />

differentiation [<strong>poster</strong>]<br />

Niels Boeckel, Masamichi Koyanagi, Masayoshi Iwasaki, Andreas M. Zeiher, Stefanie Dimmeler<br />

Oct3/4 and Klf4 promoter status in multipotent circulating mesangioblasts [<strong>poster</strong>]<br />

Emilia Jarochowska, Pawe• Krawczyk, Anna •ach, Micha• Krzyszto•<br />

Presentation of Students' Society of Genetics and Epigenetics [<strong>poster</strong>]<br />

Daniel Buszewicz, Marta Teperek, Pawel Krawczyk, Emilia Jarechowska, Michal Krzyszton<br />

Presentation of Students' Society of Genetics and Epigenetics [<strong>poster</strong>]<br />

Arunkumar Dhayalan, Tomasz Jurkowski, Heike Laser, Richard Reinhardt, Da Jia, Xiaodong Cheng, Albert Jeltsch<br />

Protein - protein interaction analysis by Absence of Interference approach [<strong>poster</strong>]<br />

Michael Grzendowski, Markus J. Riemenschneider, Marietta Wolter, Uwe Schlegel, Helmut E. Meyer, Guido<br />

Reifenberger, Kai Stühler<br />

Proteome analysis of human glioma with 1p/19q LOH [<strong>poster</strong>]


Rudolf Engelke, Gerhard Mittler<br />

Proteomic analysis of the nuclear matrix in pre-B cells. [<strong>poster</strong>]<br />

Levin Böhlig, Kurt Engeland<br />

Regulation of an intronic microRNA and its host gene by the tumor suppressor p53 [<strong>poster</strong>]<br />

Huan Shu, Lars Hennig<br />

Restructuring of epigenetic landscapes during plant development [<strong>poster</strong>]<br />

Lin XU, Rozenn MENARD, Alexandre BERR, Denise MEYER, Wen-Hui SHEN<br />

Role of Histone Ubiquitination in Arabidopsis Development [<strong>poster</strong>]<br />

Annelen Schemm, Sabine Neumann, Pamela Strissel, Cord-Michael Becker<br />

Role of NRSF/ hREST4 in Neuronalisation of Tumors [<strong>poster</strong>]<br />

Ernst Aichinger, Aleksandra Erilova, Grigory Makarevich, Claudia Köhler<br />

Role of the Mi-2 homolog PICKLE in repression of Polycomb group target genes in Arabidopsis<br />

[<strong>poster</strong>]<br />

Lorenz Kallenbach, Patrick Heun<br />

Role of the SUMO E3 ligase PIAS in chromosome and nuclear organization in Drosophila<br />

melanogaster [<strong>poster</strong>]<br />

Amos Tanay<br />

Selection and mutation in the evolution of CpG islands [<strong>poster</strong>]<br />

Gabriela Galiová, Eva Bártová, Andrea Harni•arová, Jana Krej•í, Stanislav Kozubek<br />

Single-cell c-myc gene expression in human embryonic stem cells and human teratocarcinoma<br />

NTERA cells [<strong>poster</strong>]<br />

Henriette Franz, Steven A. Jacobs, C. David Allis, Sepideh Khorasanizadeh, Wolfgang Fischle<br />

SPECIFICITY OF THE CDY FAMILY OF CHROMODOMAINS FOR METHYLATED ARKS MOTIFS IN<br />

CHROMATIN [<strong>poster</strong>]<br />

Bastian Stielow, Alexandra Sapetschnig, Christina Wink, Guntram Suske<br />

SUMO-modified transcription factors repress transcription by provoking local heterochromatic<br />

gene silencing [<strong>poster</strong>]<br />

Günter Kahl, Carlos Molina Medina, Björn Rotter, Peter Winter, Hideo Matsumura, Ryohei Terauchi<br />

SuperSAGE: A complete genome-wide quantitative expression profiling platform [<strong>poster</strong>]<br />

Peter R. Lange, Andreas Finke, Claus Wasternack<br />

TFL2 as an epigenetic regulator in Arabidopsis development [<strong>poster</strong>]<br />

Stephan Hupfer, Julia Brill, Cord-Michael Becker, Kristina Becker<br />

The entla mouse - a model for human absence epilepsy [<strong>poster</strong>]<br />

Jennifer Gerke, Özgür Bayram, Gerhard H. Braus<br />

The velvet complex coordinates light, fungal development and secondary metabolism in<br />

Aspergillus nidulans [<strong>poster</strong>]<br />

Filip Senigl, Jiri Plachy, Jiri Hejnar<br />

The CpG island core element protects retroviral vectors from transcriptional silencing [<strong>poster</strong>]<br />

Sarantis Chlamydas, Patrick Heun, Ruggiero Caizzi<br />

The Drosophila melanogaster centromeric region: a chromosomal domain in a dynamic state<br />

[<strong>poster</strong>]<br />

Andrea Just, Falk Butter, Esther Lizano, Michelle Trenkmann, Tony Heitkam, Heike Betat, Mario Mörl


The function of two conserved elements in the bacterial Poly(A)Polymerase and CCA-adding<br />

enzyme [<strong>poster</strong>]<br />

Stefan Ehrentraut, Jan Weber, Ann E. Ehrenhofer-Murray<br />

The HDAC Rpd3 functions in boundary formation by removal of Sir2 substrate [<strong>poster</strong>]<br />

Marcus Buschbeck, Iris Uribesalgo, Luciano Di Croce<br />

The histone variant macroH2A regulates key developmental genes [<strong>poster</strong>]<br />

Andreas May, Daniela Weise, Kurt Reifenberg, Thomas Haaf, Ulrich Zechner<br />

The impact of ovarian stimulation on the cellular epigenome in preimplantation mouse embryos<br />

[<strong>poster</strong>]<br />

Andreas Thomae, Dagmar Pich, Jan Brocher, Christian Berens, Robert Hock, Wolfgang Hammerschmidt, Aloys<br />

Schepers<br />

The interaction between ORC and the high mobility group protein HMGA1a creates site-specific<br />

replication origins [<strong>poster</strong>]<br />

Tomasz Jurkowski, Nils Anspach, Lilia Kulishova, Wolfgang Nellen, Albert Jeltsch<br />

The M.EcoRV DNA methyltransferase uses DNA bending for recognition of an expanded EcoDam<br />

recognition site. [<strong>poster</strong>]<br />

Yamuna Gangadharan, Gary Karpen, Patrick Heun<br />

The role of Drosophila SUMO E3 ligase dPIAS in Chromosome and Nuclear Organization [<strong>poster</strong>]<br />

Tuan Nguyen, Mahmoud Youness, Andrea Tedeschi, Andrew Green, Kirsi Forsberg, Simone Di Giovanni<br />

The role of NFAT in axonal outgrowth and regeneration [<strong>poster</strong>]<br />

Christine Vogler, Tanja Waldmann, Lora Braun, Mirek Dundr, Robert Schneider<br />

The tale of a tail - Histone H2A and its C-terminal tail [<strong>poster</strong>]<br />

Myriam Ekici, Mathias Hohl, Gerald Thiel<br />

Transcription of genes encoding synaptic vesicle proteins in human neural stem cells:chromatin<br />

accessability, histone methylation pattern and essential role of REST [<strong>poster</strong>]<br />

Madeleine Meusburger, Mark Helm, Frank Lyko<br />

tRNA targets methylated by the Dnmt2 methyltransferase [<strong>poster</strong>]<br />

Chandan Goswami, Tim Hucho<br />

TRPV4 Biochemically And Functionally Interacts With The Cytoskeleton [<strong>poster</strong>]<br />

Akuma Divine Saningong, Peter Bayer, Jonathan Wolf Mueller<br />

Unravelling the Function of Human DNA-Binding Protein Par14 in the Cellular Nucleus [<strong>poster</strong>]<br />

Agnieszka Sokol, Aleksandra Kwiatowska, Andrzej Jerzmanowski, Marta Prymakowska-Bosak<br />

Up-regulation of stress-inducible genes in tobacco and Arabidopsis cells in response to abiotic<br />

stresses and ABA treatment correlates with dynamic changes in histone H3 and H4 modifcations<br />

[<strong>poster</strong>]


Mark Wossidlo<br />

γH2AX in the mouse zygote – implications of DNA repair in<br />

epigenetic reprogramming<br />

Wossidlo M, Lepikhov K, Paelmke N, Walter J<br />

University of Saarland, Natural Sciences – Technical Faculty III, FR 8.3, Biological<br />

Sciences, Genetics/Epigenetics, Saarbrücken, Germany<br />

In mammals shortly after the fertilization of the oocyte the paternal genome undergoes<br />

dramatic epigenetic changes. The paternal DNA in mouse zygotes is rapidly<br />

demethylated by an apparently active mechanism, while the maternal DNA stays<br />

methylated. It still remains unknown which enzymes are responsible for the paternal<br />

DNA demethylation, same holds true for the mechanisms behind the process. Therefore<br />

we examined whether this paternal demethylation is mediated by a ubiquitous DNA<br />

repair process. To address this question we used the indirect immunofluorescense<br />

approach to detect the presence of DNA repair associated phosphorylated histone H2AX<br />

(γH2AX) in mouse zygotes at different pronuclear stages.<br />

We found out that the common DNA strand break associated marker γH2AX<br />

preferentially appears in the paternal pronucleus at certain pronuclear stages. In present<br />

work we describe the dynamic changes of γH2AX pattern, which is influenced by DNA<br />

polymerase inhibitor aphidicolin and at certain pronuclear stages independent on<br />

replication. This replication independent preferential localization of γH2AX in the paternal<br />

pronucleus in early zygotic stages indicates that active DNA demethylation in zygotes<br />

might be linked to DNA repair.<br />

contact:<br />

Mark Wossidlo<br />

Universität des Saarlandes<br />

Genetik / Epigenetik<br />

m.wossidlo@mx.uni-saarland.de<br />

Universitäts Campus Geb. A2.4<br />

66123 Saarbrücken (Germany)


Bernd Schmeck, Janina Lorenz, Philippe Dje N'Guessan, Antje Flieger, Vincent van<br />

Laak, Norbert Suttorp, Stefan Hippenstiel<br />

L. pneumophila induce histone modifications in human lung<br />

epithelial cells<br />

Legionella pneumophila causes community- and hospital-acquired pneumonia. Their<br />

effect on histone marks is unknown.<br />

L. pneumophila wildtype strain 130b induced time- and dose-dependently expression of<br />

the important chemoattractant IL-8 and global, genome wide histone modifications in<br />

human lungs epithelial A549 cells. We analyzed the promoter of the important<br />

proinflammatory chemokine IL-8 and found that histone H4 was acetylated and H3 was<br />

phosphorylated at Ser-10 and acetylated at Lys-14, followed by recruitment of<br />

transcription factor NF-κB, and RNA polymerase II as well as gene transcription. L.<br />

pneumophila strain 130b-induced IL-8 expression was decreased by histone<br />

acetyltransferase (HAT) inhibitor anacardic acid and enhanced by the histone<br />

deacetylase (HDAC) inhibitor trichostatin A. Accordingly, after L. pneumophila infection<br />

HATs p300 and CBP were time-dependently recruited to the il8 promoter, whereas<br />

HDAC1 and HDAC5 first vanished and later reappeared at the promoter. Interestingly, L.<br />

pneumophila specifically induced expression of HDAC5 but not of other HDACs in lung<br />

epithelial cells. Furthermore, L. pneumophila-induced cytokine release, promoter specific<br />

histone modifications and Pol-II recruitment were reduced in infection with flagellindeletion<br />

mutants.<br />

In summary, histone acetylation seems to be important for the regulation of<br />

proinflammatory gene expression in L. pneumophila infected lung epithelial cells.<br />

contact:<br />

PD Dr. Bernd Schmeck<br />

Charité - Universitätsmedizin Berlin<br />

Medizinische Klinik m.S. Infektiologie und Pneumologie<br />

Bernd.Schmeck@charite.de<br />

Augustenburger Platz 1<br />

13353 Berlin (Germany)


Luke Dannenberg, Leo Iniguez, Heather Holster, Peggy Farnham, Bing Ren, David<br />

Fisher, Gerd Pfeifer, Hui Liu, Jacob Kitzman, Fatih Ozsolak<br />

A flexible, high-density array platform for genome-wide<br />

characterization of epigenetic and transcriptional regulatory<br />

mechanisms involved in cancer<br />

Epigenetic mechanisms, such as DNA methylation and histone modification, and altered<br />

transcription factor binding play major roles in the development of many human<br />

diseases, most notably cancer. High-density and highly flexible DNA microarrays,<br />

derived from a unique combination of photolithography and a digital micromirror device,<br />

are now allowing researchers the opportunity to examine epigenetic events at an<br />

unprecedented scale and resolution. Phenomenon such as de novo DNA methylation of<br />

tumor suppressor gene promoters silences their expression, hence creating a gateway<br />

for uncontrolled cell division. In addition, the binding of many transcription factors<br />

becomes increased in a cancerous cell to promote the expression of genes involved in<br />

initiating carcinogenesis and metastasis. By looking on a genome-wide scale using highdensity<br />

microarrays, a comprehensive picture of DNA methylation patterns, chromatin<br />

structure, and transcription factor binding can be generated to aid in the characterization<br />

of the differences between normal and cancer cells, different cancer types, the same<br />

cancer from two different individuals, and drug treatment studies. Novel platform<br />

developments, specifically the 2.1 million probe (HD2) long-oligonucleotide array, have<br />

expanded the horizon of genome-wide studies for its application in elucidating epigenetic<br />

and transcriptional regulatory mechanisms involved in cancer. Cancer studies using<br />

NimbleGen 385K microarrays will be presented and discussed as well as data on the HD2<br />

platform.<br />

contact:<br />

Product Manager Luke Dannenberg<br />

Roche NimbleGen<br />

ldannenberg@nimblegen.com<br />

500 S. Rosa Road<br />

53719 Madison, Wisconsin (United States)


Anna Katharina Sedello, Gabriele Putz, Frank Buchholz<br />

A Polycomb Group Protein Synergizes with Runx1 in Blocking<br />

HSC Differentiation.<br />

Our lab previously showed that Runx1 deletion in adult mice leads to a block in<br />

differentiation but not to leukemia development. We used a loss-of-function approach to<br />

identify proteins cooperating with Runx1 in processes determining whether<br />

hematopoietic stem and progenitor cells undergo differentiation or maintain stem-celllike<br />

properties. By transducing lineage negative hematopoietic cells from Runx1Δ/Δ mice<br />

with a pooled shRNA library we obtained cells with a proliferative advantage in a serial<br />

colony-forming-cell assay. Genomic DNA isolated from these immortalized cells revealed<br />

an shRNA targeting a member of the polycomb group protein family, which is involved in<br />

chromatin remodeling. Runx1Δ/Δ cells transduced with shRNA targeting the polycomb<br />

group protein member form distinct colonies for more than double as long as nontransduced<br />

cells in serial colony-forming-cell assays. We hypothesize that block of<br />

differentiation by Runx1 deletion cooperates with a reactivation of self-renewal programs<br />

by knock down of the polycomb group protein to immortalize hematopoietic stem and<br />

progenitor cells, possibly mimicking leukemia.<br />

Literature<br />

Putz G, Rosner A, Nuesslein I, Schmitz N, Buchholz F. AML1 deletion in adult mice<br />

causes splenomegaly and lymphomas. Oncogene (2006) 25, 929-939.<br />

Bernards R, Brummelkamp TR, Beijersbergen RL. shRNA libraries and their use in cancer<br />

genetics. Nature Methods (2006) 3, 701-706.<br />

contact:<br />

Anna Katharina Sedello<br />

Max-Planck Institute of Molecular Cell Biology and Genetics<br />

asedello@mpi-cbg.de<br />

Pfotenhauerstr. 108<br />

01307 Dresden (Germany)<br />

additional information<br />

Gabriele Putz, currently at Osiris Therapeutics, Inc., 7015 Albert Einstein Dr., Colombia, MD 21046,<br />

USA


Jennifer Cropley, Catherine Suter, David Martin<br />

A progressive multigenerational shift in epigenotype with<br />

continuous methyl donor supplementation<br />

The epigenetic state of a locus can be affected by environmental factors such as diet.<br />

The murine A vy (agouti viable yellow) allele is one such locus: dietary supplementation<br />

of pregnant dams with methyl donors changes the epigenetic state of the locus in the<br />

offspring. At A vy , an IAP retrotransposon is inserted upstream of agouti. When<br />

epigenetically active the IAP usurps transcriptional control, driving ectopic expression of<br />

agouti signalling protein to produce the characteristic obese yellow phenotype. The<br />

epigenetic state of the IAP is unstable in the germline, so that isogenic mice show wide<br />

variation in the somatic epigenetic state of the IAP, with resultant broadly variable<br />

penetrance and expressivity. Supplementation of maternal diet with methyl donors<br />

promotes epigenetic silencing of the IAP, shifting the spectrum of offspring phenotypes<br />

away from obese yellow. We have previously shown that methyl donors can affect the<br />

germline epigenetic state of the A vy IAP. Here we show that continual supplementation<br />

of A vy mice over five generations leads to progressive germline stabilisation of the IAP<br />

epigenotype, so that the silent state becomes more strongly heritable and thus<br />

significantly more prevalent in the population. In unsupplemented populations the IAP is<br />

completely silent in 13% of mice. In a supplemented population, successive breeding of<br />

males carrying a silent IAP increases the prevalence of the silent allele almost three-fold<br />

(to 31%) by the fifth generation. These results suggest that long-term exposure to an<br />

environmental stimulus can effect epigenetic changes throughout a population. Such<br />

mechanisms may contribute to adaptive evolution via stable epigenetic silencing in the<br />

germline.<br />

contact:<br />

Dr Jennifer Cropley<br />

Victor Chang Cardiac Research Institute<br />

j.cropley@victorchang.edu.au<br />

384 Victoria st<br />

2010 Darlinghurst (Australia)<br />

additional information<br />

Dr Catherine Suter: Victor Chang Cardiac Research Institute, Darlinghurst, Australia<br />

Prof David Martin: Childrens Hospital Oakland Research Institute, Oakland, CA, USA


Maja Klug, Sven Heinz, Lucia Schwarzfischer, Sabine Pape, Michael Rehli<br />

Active demethylation of promoter CpGs in post-mitotic cells<br />

Within the last decades, it has become increasingly evident that the epigenetic code,<br />

including chromatin structure, DNA methylation, as well as histone modifications, plays<br />

an important role in regulating gene expression. The dynamics of DNA methylation, in<br />

particular the regulated, active removal of methyl-CpG marks, has remained a mystery,<br />

partly due to the lack of appropriate model systems. The differentiation of human blood<br />

monocytes into macrophages or dendritic cells proceeds without proliferation,<br />

representing an excellent model system for analyzing active demethylation processes in<br />

post-mitotic cells. In earlier studies, we observed the strong up-regulation of the CCL13<br />

gene specifically in monocyte-derived dendritic cells. The transcriptional activation<br />

coincides with the demethylation of three defined CpG residues in the CCL13-promoter<br />

during differentiation of monocytes into dendritic cells, whereas the promoter remains<br />

methylated and silent in monocyte-derived macrophages. Here we present a detailed<br />

time-course analysis of the epigenetic/chromatin status of the CCL13 promoter by<br />

quantitative mRNA expression analysis, chromatin immunoprecipitation, methyl-CpG<br />

immunoprecipitation and MNase-hypersensitivity, and restriction enzyme accessibility<br />

assays. We detected a strong correlation between active DNA demethylation,<br />

nucleosome remodeling, Histone H3 lysine 4 methylation and transcription of the CCL13<br />

gene during dendritic cell differentiation. Our data suggest that active demethylation<br />

proceeds in parallel with chromatin remodeling and gene activation.<br />

contact:<br />

Dipl Maja Klug<br />

Uniklinikum Regensburg<br />

Hämatologie/Onkologie<br />

maja.klug@klinik.uni-regensburg.de<br />

Franz-Josef-Strauss Allee 11<br />

93053 Regensurg (Deutschland)


Tobias Paprotka, Holger Jeske<br />

Analysis of geminiviral cytosine methylation<br />

Geminiviruses are important plant pathogens, causing yield losses in crop plants all over<br />

the world. They consist of a single stranded DNA genome which varies in size from 2.5<br />

to 3.0 kb, depending on the virus and is packed into icosahedral twin shaped particles.<br />

Plant defence mechanisms against geminiviruses, including post transcriptional gene<br />

silencing (PTGS) and transcriptional gene silencing (TGS) have been observed and are<br />

competed by the virus with silencing suppressors. The methylation of partial viral DNA<br />

sequences has been shown, but a complete determination of the cytosine methylation<br />

pattern of a viral genome has not yet been accomplished. A recent advance in<br />

geminivirus discovery and diagnostics was the application of rolling circle amplification<br />

(RCA), using the Φ29 polymerase. To analyse the complete methylation pattern of a<br />

geminivirus genome a method based on bisulphite sequencing combined with RCA was<br />

developed. The unmethylated cytosines are converted conventionally, but the<br />

subsequent PCR step is replaced through amplification with RCA. Preferential<br />

amplification of particular sequences is avoided by the use of random hexamer primers.<br />

The obtained DNA is then digested with restriction enzymes that miss Cs or Gs in their<br />

recognition sequence and randomly cloned into pBluescript. After colony PCR or RCA the<br />

cloned fragments can be sequenced directly using universal primers. A diverse pattern<br />

was observed including methylation of CpG, CpNpG and asymmetrical sites, which<br />

indicates the enforcement by the plants TGS machinery. An increase of cytosine<br />

methylation during the course of infection was also detected. This approach therefore<br />

shows the usefulness of RCA in bisulphite sequencing as a beneficial method to observe<br />

methylation patterns of small circular DNA molecules.<br />

contact:<br />

Dipl.-Biol. Tobias Paprotka<br />

University of Stuttgart<br />

Institute of Biology, Department of Molecular Biology and Virology of Plants<br />

tobias.paprotka@bio.uni-stuttgart.de<br />

Pfaffenwaldring 57<br />

70550 Stuttgart (Germany)


Christine Paprotka, Mareike Rust, Katharina Kohl, Jörg Leers, Rainer Renkawitz<br />

Analysis of the effect of CTCF and NuRD on chromatin<br />

structure<br />

CTCF, a ubiquitously expressed transcription factor, is the only protein in vertebrates<br />

known to mediate enhancer blocking. The mechanism of enhancer blocking is still<br />

unclear, but there are several models postulated. All these models share the idea, that<br />

the modification, remodeling and three-dimensional arrangement of chromatin play a<br />

major role.<br />

The NuRD complex is a multi-subunit protein complex with enzymatic activities involving<br />

chromatin remodeling and histone deacetylation. Targeting of NuRD to methylated CpG<br />

sequences leads to gene repression and is mediated through the methyl-binding-domain<br />

proteins MBD2 and MBD3 [1-4].<br />

Here we present a strategy to analyse the influence of CTCF and distinct NuRD<br />

complexes on chromatin structure in vivo at defined genomic regions. For our studies we<br />

utilize a well established system using the ability of the Lac-repressor to bind to the Lacoperator.<br />

LacO repeat clusters stably integrated into the genome have been generated<br />

[5]. Using this system we analyze CTCF and NuRD recruited to the array via LacI-MBD2/-<br />

CTCF. Using immunofluorescence microscopy we can analyze the recruitment and<br />

assembly of NuRD components. In addition histone modification depending on CTCF or<br />

NuRD can be analysed by chromatin immunoprecipitation as well. This allows to address<br />

the question wether CTCF has an influence on chromatin structure and if spreading of<br />

repressed chromatin occures after NuRD binding.<br />

Literature<br />

[1] Brackertz, M., Gong, Z., Leers, J., Renkawitz, R., 2006, Nucleic Acids Res, Vol.34,<br />

397-406<br />

[2] Gong, Z., Brackertz, M., Renkawitz, R. 2006, Mol. Cell. Biol., Vol. 26, 4519-4528<br />

[3] Zhang et al., 2002,Mol. Cell. Biol., Vol 22, 536-546<br />

[4] Guezennec et al., 2006, Mol. Cell. Biol.,Vol 26, 843-851<br />

[5] T. Jegou, K. Rippe, unpublished data<br />

contact:<br />

Christine Paprotka<br />

Justus-Liebig-Universität Giessen<br />

Institut für Genetik<br />

christine.paprotka@gen.bio.uni-giessen.de<br />

Heinrich-Buff-Ring 58 -62<br />

35392 Giessen (Germany)


Nadine Obier, Albrecht M. Müller<br />

Analyzing epigenetic modifications in pluripotent stem cells<br />

via intranuclear flow cytometry<br />

Pluripotency describes the ability of embryonic stem cells (ES cells) to self-renew and to<br />

give rise to all cell types of the developing embryo including the germline. Our goal is to<br />

define pluripotency by identifying the entry point of cells into an irreversible<br />

differentiation state with restricted differentiation potential.<br />

In this regard we developed a FACS-based protocol which can quantitatively display<br />

levels of different histone modifications in diverse cell types, such as in ES cells and in<br />

their differentiated derivatives. Applying this intranuclear flow cytometric method, we<br />

detected differences in global histone H4 acetylation levels between cells that were<br />

either treated or not treated with the HDAC inhibitor TSA. Further, we observed a<br />

distinct reduction of histone H3 lysine 27 tri-methylation (H3K27me3) levels in ES cells<br />

lacking the protein EED, which – as a critical component of the Polycomb-group<br />

repressor complex 2 - is participating in enzymatic methylation of H3K27. Our<br />

preliminary studies on global levels of H3K4me3 and H3K27me3 in combination with ckit-surface<br />

expression revealed that c-kit high immunophenotypic ES cells are also<br />

highly positive for both H3K4m3 and H3K27me3 “chromatin-immunophenotypes”.<br />

Together, we developed a new method for the analysis of global histone modifications by<br />

intra-nuclear flow cytometry. This method represents a promising tool to simultaneously<br />

study cellular properties, such as cell proliferation, apoptosis, surface marker expression<br />

and intranuclear “chromatin-immunophenotype”, on the single cell level of large<br />

quantities of cells.<br />

contact:<br />

M.Sc. Nadine Obier<br />

Universität Würzburg<br />

Institut für medizinische Strahlenkunde und Zellforschung (MSZ)<br />

nadineobier@web.de<br />

Versbacher Str. 5<br />

97078 Würzburg (Germany)


Markus Nees, Christian Hammann, Manu Dubin, Jonathan Chubb, Wolfgang Nellen<br />

Argonaute proteins in Dictyostelium discoideum<br />

M. Nees, C. Hammann, M. Dubin, J. Chubb*, W. Nellen<br />

Department of Genetics, Universität Kassel, D-34132 Kassel, Germany.<br />

* School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH<br />

The genom of Dictyostelium discoideum contains six genes which resemble Argonaute or<br />

Piwi proteins from other organisms. Argonautes are involved in slicing of mRNAs in the<br />

RNAi pathway and in translational inhibition in the miRNA pathway. Piwi proteins are<br />

expressed specifically in the germline where they associate with another class of<br />

approximately 29-nucleotide-long small RNAs, named piRNAs. The function of piRNAs is<br />

not yet fully understood.<br />

To elucidate the function of Argonaute proteins in Dictyostelium discoideum we present<br />

first data on the cellular localization, the expression pattern during the developmemtal<br />

cycle and the phenotype of knock-out mutants.<br />

contact:<br />

Markus Nees<br />

Universität Kassel<br />

Genetik<br />

neemar@web.de<br />

Heinrich-Plett-Straße 40<br />

34132 Kassel (Germany)


Osman El-Maarri 1 , Tim Becker 2 , Thomas Mikeska 3 , Judith Junen 1 , Syed Saadi<br />

Manzoor 1 , Amalia Diaz-Lacava 2 , Rainer Schwaab 1 , Thomas Wienker 2 , Andreas Waha 3 ,<br />

Johannes Oldenburg 1<br />

Association analysis between DNA methylation from total<br />

blood and polymorphisms in DNA methyltransferase (DNMT)<br />

genes in healthy individuals: A tendency toward higher<br />

methylation levels in males<br />

To examine the contribution of polymorphisms in DNMT genes to methylation variations<br />

we performed a search for polymorphisms in coding regions of all known human DNA<br />

methyltransferases genes (DNMT1, 3A, 3B, 3L and 2=TRDMT1) in 96 normal males and<br />

96 normal females. Global methylation was estimated by studying two repetitive DNA<br />

elements, namely Line-1 and Alu repeats, while single loci were investigated at three<br />

differentially methylated regions: PEG3, NESP55 and H19; two additional single loci were<br />

also studied at Xq28 and 19q13.4. All studied CpGs showed a slightly higher methylation<br />

in males (P


Rafal Archacki, T. Sarnowski, J. Halibart-Puzio, Daniel Buszewicz, M. Prymakowska-<br />

Bosak, M. Kuras, C. Koncz, A. Jerzmanowski<br />

ATBRM bromodomain-ATPase and ATSWI3C, representing<br />

putative subunits of SWI/SNF chromatin remodeling<br />

complexes, control similar developmental functions in<br />

Arabidopsis<br />

Among the factors that serve to modify chromatin structure, SWI/SNF chromatin<br />

remodeling complexes define conserved and well-characterized group. However, no<br />

SWI/SNF complex has been purified and characterized in higher plants so far, yet its<br />

existence is highly probable. Four genes encoding homologues of Swi2/Snf2 ATPase<br />

(BRM, SYD, CHR12 and CHR23) and four encoding homologues of Swi3 subunit<br />

(ATSWI3A, ATSWI3B, ATSWI3C and ATSWI3D), as well as a single SNF5 orthologue<br />

(BSH) have been identified in Arabidopsis. This makes a number of possibilities for<br />

assembly of plant SWI/SNF complexes. In the lack of structural and biochemical data,<br />

homology analyses and interpretation of genetic and in vitro interactions are the best<br />

tools for investigating SWI/SNF complex composition and function.<br />

Here we show a comparative analysis of brm and atswi3c null mutants. Both of them<br />

display similar (but not identical) developmental alterations, including semidwarfism,<br />

leaf curling, inhibition of root elongation, homeotic-like changes in flowers, and defects<br />

in pollen development. These observations, together with the results showing that BRM<br />

and SWI3C interact in yeast two-hybrid assay (Farrona et al., 2004), suggest that BRM<br />

and SWI3C proteins exist in the same SWI/SNF chromatin remodeling complex. Our<br />

analyses of brm atswi3c double mutants further support this hypothesis, as the brm<br />

atswi3c plants display brm phenotype. Nonetheless, certain differences between<br />

phenotypic traits of atswi3c and brm mutants, such as complete sterility of brm and the<br />

occurrence of unfused carpels in brm flowers, indicate that the biological functions of<br />

these two SWI/SNF subunits are not completely overlapping.<br />

contact:<br />

M. Sc. Daniel Buszewicz<br />

Warsaw University<br />

Laboratory of Plant Molecular Biology<br />

dbuszewicz@gmail.com<br />

Pawinskiego 5A/F<br />

02-106 Warsaw (Poland)


Christine Champion, Loïc Ponger, Catherine Senamaud-Beaufort, Dominique<br />

Guianvarc'h, Ludovic Halby, Anne-Laure Guieysse-Peugeot, Paola B. Arimondo<br />

Biochemical approaches to study the DNA methylation<br />

mechanisms involved in tumorigenesis<br />

Cancer cells show a highly disturbed epigenetic landscape, with a global<br />

hypomethylation of the genome that induces abnormal expression of genes (such as<br />

oncogenes) and a local hypermethylation of promotors that silences tumor suppressor<br />

genes (TSG). DNA methylation is catalysed by a family of enzymes called DNA<br />

methyltransferases (DNMTs) and only occurs at position 5 of cytosines in CpG<br />

dinucleotides (in Vertebrates) that are not randomly distributed in the genome but<br />

mainly grouped in CpG islands. Yet the mechanism by which specific de novo<br />

methylation is directed to the TSG promotors remains still unknown. Indeed, it would be<br />

of great therapeutic interest to block this specific hypermethylation of TSG promotors in<br />

order to restore in malignant cells their natural ability to block tumorigenesis.<br />

We have chosen to focus on prostate cancer and two TSG for which inactivation is due to<br />

promotor hypermethylation : RASSF1A and RARβ2.<br />

On one hand, we investigate whether short DNA sequences influence the DNA<br />

methylation pattern. By a bioinformatic analysis we have found 10 DNA motifs that are<br />

overrepresented in hypermethylated promotors in prostate cancer versus non<br />

hypermethylated promotors.<br />

On the other hand, we are developing two affinity chromatography approaches to<br />

identify the protein partners of DNMTs involved in DNA methylation.<br />

contact:<br />

PhD Student Christine Champion<br />

Museum National d'Histoire Naturelle<br />

MNHN USM 503, CNRS UMR5153, INSERM U565<br />

christine.champion@mnhn.fr<br />

43, rue Cuvier<br />

75231 Paris cedex 5 (France)<br />

additional information<br />

Dominique Guianvarc'h : CNRS UMR7613-Université Paris VI


Christian Rohde, Yingying Zhang, Tomasz P. Jurkowski, Heinrich Stamerjohanns,<br />

Richard Reinhardt*, Albert Jeltsch<br />

Bisulfite sequencing Data Presentation and Compilation<br />

(BDPC) web server – a useful tool for DNA methylation<br />

analysis<br />

During bisulfite genomic sequencing projects large amount of data is generated. The<br />

BDPC web interface (http://biochem.jacobs-university.de/BDPC/) automatically analyzes<br />

bisulfite datasets prepared using the BiQ Analyzer (Bock et al. 2005, Bioinformatics 21,<br />

4067-8). BDPC provides the following output: 1) MS-Excel compatible files compiling for<br />

each PCR product i) the average methylation level, the number of clones analyzed, and<br />

the percentage of CG sites analyzed (which is an indicator of data quality), ii) the<br />

methylation level observed at each CG site, and iii) the methylation level of each clone.<br />

2) A methylation overview table compiling the methylation of all amplicons in all tissues.<br />

3) Publication grade figures in PNG format showing the methylation pattern for each PCR<br />

product embedded in an HMTL file summarizing the methylation data, the DNA sequence<br />

and some basic statistics. 4) A summary file compiling the methylation pattern of<br />

different tissues, which is linked to the individual HTML result files, and can be directly<br />

used for presentation of the data in the internet. 5) A condensed file, containing all<br />

primary data in simplified format for further downstream data analysis, and 6) a custom<br />

track file for display of the results in the UCSC genome browser.<br />

contact:<br />

Christian Rohde<br />

Jacobs University Bremen<br />

School of Engineering and Science<br />

c.rohde@jacobs-university.de<br />

Campus Ring 1<br />

28759 Bremen (Germany)<br />

additional information<br />

*Max Planck Institute for Molecular Genetics, Ihnestrasse 63-73, D-14195 Berlin-Dahlem, Germany


Rafal Archacki, T.J. Sarnowski, J. Halibart-Puzio, D. Buszewicz, M. Prymakowska-<br />

Bosak, M. Kuras, C. Koncz, A. Jerzmanowski<br />

BRM bromodomain-ATPase and ATSWI3C, representing<br />

putative subunits of SWI/SNF chromatin remodeling<br />

complexes, control similar developmental functions in<br />

Arabidopsis<br />

Among the factors that serve to modify chromatin structure, SWI/SNF chromatin<br />

remodeling complexes define conserved and well-characterized group. However, no<br />

SWI/SNF complex has been purified and characterized in higher plants so far, yet its<br />

existence is highly probable. Four genes encoding homologues of Swi2/Snf2 ATPase<br />

(BRM, SYD, CHR12 and CHR23) and four encoding homologues of Swi3 subunit<br />

(ATSWI3A, ATSWI3B, ATSWI3C and ATSWI3D), as well as a single Snf5 orthologue<br />

(BSH) have been identified in Arabidopsis (1). This makes a number of possibilities for<br />

assembly of plant SWI/SNF complexes. In the lack of structural and biochemical data,<br />

homology analyses and interpretation of genetic and in vitro interactions are the best<br />

tools for investigating SWI/SNF complex composition and function.<br />

Here we show a comparative analysis of brm and atswi3c null mutants. Both of them<br />

display similar (but not identical) developmental alterations, including semidwarfism,<br />

leaf curling, inhibition of root elongation, homeotic-like changes in flowers, and defects<br />

in pollen development. These observations, together with the results showing that BRM<br />

and SWI3C interact in yeast two-hybrid assay (2), suggest that BRM and SWI3C proteins<br />

exist in the same SWI/SNF chromatin remodeling complex. Our analyses of brm atswi3c<br />

double mutants further support this hypothesis, as the brm atswi3c plants display brm<br />

phenotype. Nonetheless, certain differences between phenotypic traits of atswi3c and<br />

brm mutants, such as complete sterility of brm and the occurrence of unfused carpels in<br />

brm flowers, indicate that the biological functions of these two SWI/SNF subunits are not<br />

completely overlapping.<br />

Literature<br />

(1) Jerzmanowski A. SWI/SNF chromatin remodeling and linker histones in plants.<br />

Biochim Biophys Acta. 2007 May-Jun;1769(5-6):330-45<br />

(2) Farrona S, Hurtado L, Bowman JL, Reyes JC. The Arabidopsis thaliana SNF2 homolog<br />

AtBRM controls shoot development and flowering. Development. 2004 Oct;131(20):4965-<br />

75<br />

contact:<br />

M.Sc Rafal Archacki<br />

University of Warsaw<br />

Laboratory of Plant Molecular Biology<br />

rafa@ibb.waw.pl<br />

Pawinskiego 5A<br />

02-106 Warsaw (Poland)<br />

additional information<br />

Affiliation of T.J. Sarnowski, J. Halibart-Puzio, and D. Buszewicz: Polish Academy of Sciences,<br />

Institute of Biochemistry and Biophysics, Pawinskiego 5A, 02-106 Warsaw, Poland<br />

Second affiliation of M. Prymakowska-Bosak and A. Jerzmanowski: Polish Academy of Sciences,<br />

Institute of Biochemistry and Biophysics, Pawinskiego 5A, 02-106 Warsaw, Poland<br />

Affiliation of M. Kuras: University of Warsaw, Department of Ecotoxicology, Miecznikowa 1, 02-096<br />

Warsaw, Poland<br />

Affiliation of C. Koncz: Max-Planck Institut für Züchtungsforschung, Carl-von-Linné-Weg 10, D-<br />

50829 Köln, Germany


Katerina Krizova, Miloslava Fojtova, Ann Depicker, Ales Kovarik<br />

Callus-induced epiallelism of an invertedly repeated transgene<br />

locus influences its trans-silencing abilities<br />

Using a two component transgene system involving two epiallelic variants of an<br />

invertedly repeated silencing locus (1) we have studied stability of trans-silencing<br />

interactions in tobacco cell culture and regenerated plants. In parental hybrids the<br />

posttranscriptionally but not transcriptionally silenced epiallele of locus 1 trans-silenced<br />

and trans-methylated target locus (2). Expression and methylation of both silenced<br />

(Lo1/Lo2) and non-silenced (Lo1E/Lo2) hybrids were stable over several generations in<br />

plants. However, in early Lo1E/Lo2 callus decreased expression of the nptII reporter<br />

gene was observed while the expression in Lo1/Lo2 remained unchanged. Analysis of<br />

small RNA species and coding region methylation suggested that the nptII genes were<br />

silenced by a PTGS mechanism in both cultures. Expression changes were correlated<br />

with changes in locus 1 promoter methylation status: the PTGS variant in Lo1/Lo2 line<br />

acquired methylation while the TGS epiallele in Lo1E/Lo2 line showed reduced<br />

methylation compared to the parental plant. Bisulfite genomic sequencing of locus 1<br />

revealed molecules with no, intermediate and high level of methylation. These data<br />

indicated that a cell culture process brought two epialleles of the silencer locus 1 to the<br />

same epigenetic ground characterized by high epilallelic diversity. In regenerated plants<br />

about 75% of Lo1E/Lo2 individuals returned to the original non-silenced phenotype while<br />

25% of individuals were silenced. From Lo1/Lo2 callus, 25% of regenerated plants<br />

showed increased expression whereas 75% of individuals remained silenced. The results<br />

demonstrated sensitivity of transgenes containing inverted structures towards epigenetic<br />

changes imposed by cell culture.<br />

contact:<br />

Mgr. Katerina Krizova<br />

Academy of Sciences<br />

Institute of Biophysics v.v.i.<br />

krizova@ibp.cz<br />

Kralovopolska 135<br />

CZ-61265 Brno (Czech Republic)


Britta Wallmen, Simon Wöhrle, Andreas Hecht<br />

Cell specific inducibility of Wnt target genes correlates with<br />

epigenetic modifications and differential promoter occupancy<br />

by TCF/LEF proteins<br />

Transcription factors of the T-cell factor (TCF)/lymphoid enhancer factor (LEF) family are<br />

considered to act in conjunction with corepressors and coactivators as bimodal switches<br />

for the activation or repression, respectively, of Wnt/beta-catenin target genes.<br />

Accordingly, TCF/LEF proteins are thought to remain constantly bound to the promoter<br />

regions of their target genes. However, constant promoter occupancy by TCF/LEF factors<br />

does not readily explain how distinct groups of Wnt target genes can be differentially<br />

regulated in a cell-type specific and developmentally controlled manner. We<br />

systematically compared known target genes with respect to Wnt-responsiveness,<br />

promoter occupancy by TCF/LEF proteins and epigenetic features in different cell lines.<br />

In E14 embryonic stem cells, in the neural cell line C17.2 and in C2C12 myogenic cells<br />

we find that Axin2, Cdx1 and T/Brachyury are differentially expressed and regulated.<br />

Activation of these target genes is predominantly mediated by a subset of TCF/LEF<br />

factors. Analysis of DNA methylation patterns and histone modifications at promoter<br />

regions revealed that Wnt-inducibility correlates with DNA hypomethylation and active<br />

histone marks. In contrast, non-responsive promoters showed hypermethylation and<br />

repressive histone marks. Moreover, Wnt-responsiveness correlates with differential<br />

promoter occupancy by TCF/LEF proteins. Notably, in contrast to current models,<br />

TCF/LEF transcription factors are not present at promoter regions of non-responding<br />

genes. We hypothesize that distinct promoter occupancy by TCF/LEF proteins and<br />

epigenetic control mechanisms form a multi-layered control system to achieve<br />

differential regulation of Wnt target gene expression.<br />

contact:<br />

Britta Wallmen<br />

University of Freiburg<br />

Institute of Molecular Medicine and Cell Research<br />

britta.wallmen@mol-med.uni-freiburg.de<br />

Stefan-Meier-Str. 17<br />

79104 Freiburg (Germany)


Peter Hemmerich, Stefanie Weidtkamp-Peters, Christian Hoischen, Lars Schmiedeberg,<br />

Indri Erliandri, Stephan Diekmann<br />

CENP-I as a new epigentic mark at centromere chromatin<br />

Epigenetic marking of a DNA locus may be realized by posttranslational modifications of<br />

nucleosomal histones or by stable binding of a specific protein at that locus. Centromere<br />

identity is believed to be conveyed by CENP-A, a specialized histone H3 analog that<br />

substitutes canonical H3 within centromeric nucleosomes. CENP-A is constitutively<br />

present at centromeres and required for the association of all other kinetochore proteins.<br />

To test whether these epigenetic properties are unique to CENP-A we have assessed the<br />

exchange rates of inner centromere proteins by quantitative microscopy throughout the<br />

cell cycle in living human cells (1). We demonstrate that, in addition to CENP-A, CENP-I<br />

is also a stable centromere component that does at no time exchange with soluble pools<br />

at centromeres. Loading of CENP-I onto centromeric chromatin occurs co-replicationally,<br />

while CENP-A is loaded in early G1. A subfraction of CENP-H (~20%) also stays stably<br />

bound to centromeres throughout the cell cyle. In contrast, CENP-B, CENP-C, and<br />

hMis12 turn over completely at centromeres with residence times ranging between<br />

seconds to hours. Our data reveal a wide range of cell cycle-specific assembly plasticity<br />

of the centromere during the cell-cycle and identify CENP-I as a potentially additional<br />

epigentic marker at centromeres.<br />

Literature<br />

(1) Hemmerich et al., J. Cell Biol. (2008) in press<br />

contact:<br />

PhD Peter Hemmerich<br />

Leibniz Institute for Age Research<br />

Fritz-Lipmann-Institute<br />

phemmer@fli-leibniz.de<br />

Beutenbergstr. 11<br />

07745 Jena (Germany)


Claudia Gebhard, Elmar Schilling, Lucia Schwarzfischer-Pfeilschifter, Mathias Ehrich,<br />

Michael Rehli<br />

Comparative methylation profiling of tumor samples using<br />

methyl-CpG-immuno precipitation (MCIp) and CpG island<br />

microarrays<br />

Department of Hematology, University Hospital Regensburg, 93042 Regensburg,<br />

Germany &<br />

*Sequenom, Inc., San Diego, CA, 92121, USA<br />

Methylation of CpG islands is associated with transcriptional repression and, in cancer,<br />

leads to the abnormal silencing of tumor suppressor genes. We have previously<br />

developed a genome wide methylation profiling assay based on a recombinant, antibodylike<br />

MBD-Fc fusion protein that allows the detection of CpG methylation independent of<br />

chemical DNA modification using bisulfite or methylation-sensitive restriction. Here, we<br />

present an in depth comparison of comparative methylation data obtained with an<br />

optimized MCIp/hybridization procedure and quantitative methylation data obtained by<br />

base-specific cleavage of bisulfite amplification products and Matrix-Assisted Laser<br />

Desorption/Ionization Time-Of-Flight Mass Spectrometry (MALDI-TOF MS). MCIp<br />

methylation profiles of appr. 27 000 CpG islands were obtained from two myeloid<br />

leukemia cell lines using different hybridization conditions and various amounts of<br />

starting material (1-4 µg of genomic DNA). A set of 140 genes (covered by 1300<br />

different amplicons) that were selected based on the array results were analyzed by<br />

MALDI-TOF MS. The comparison of both techniques shows an excellent correlation<br />

between bisulfite and MCIp data sets. Our comprehensive validation study shows that<br />

robust methylation profiles can be obtained with as little as 1 µg of genomic DNA and<br />

demonstrates the high sensitivity and reproducibility of the MCIp approach.<br />

contact:<br />

Elmar Schilling<br />

Uniklinik Regensburg<br />

Hämatologie und Onkologie<br />

elmar.schilling@klinik.uni-regensburg.de<br />

Franz-Josef-Strauss Allee 11<br />

93042 Regensburg (Germany)


Ruxandra Farcas, Eberhard Schneider, Ulrich Zechner, Achim Tresch, Hans Zischler,<br />

Angelika Daser, Thomas Haaf<br />

Comparison of human and non-human primate methylation<br />

status of CpG islands in the promoter region of CCRK<br />

Little is known about how the human brain differs from that of our closest relatives,<br />

although it is known that humans and primates share a high extent of DNA sequence<br />

homology. One explanation are species differences in regulation of gene expression.<br />

Here we focused our attention on differences in promoter DNA methylation in human<br />

and non-human primate brains. For comparative methylation analysis, we performed<br />

bisulphite sequencing of DNA from frontal cortex of 11 humans, one chimpanzee, two<br />

baboons, and one rhesus monkey. Species-specific methylation patterns were found for<br />

the cell-cycle related kinase (CCRK) gene that activates CDK2 and is indispensable for<br />

cell growth. CCRK has an intermediate CpG promoter with tendency to high-CpG<br />

promoter. In the analyzed CpG island, we could distinguish three different regions, two<br />

whose methylation status is conserved and one with differences in the methylation<br />

status between the analyzed species. The first region, an ALU-Sg repeat, was almost<br />

completely methylated in all human and primate samples. The second region, a block of<br />

6 CpGs at the end of the ALU-Sg repeat, was mostly unmethylated in the 11 humans<br />

and rhesus monkey, but highly methylated in chimpanzee and the two baboons. The<br />

third region, corresponding to the end of the CpG island, was completely unmethylated<br />

in all human and primate samples. We conclude that methylation status of the second<br />

region varies between human and rhesus monkey on the one hand and chimpanzee and<br />

baboon on the other hand. In future investigations, we will focus on relating our findings<br />

to gene expression data of CCRK in primate brain.<br />

contact:<br />

Ruxandra Farcas<br />

Johannes Gutenberg University<br />

Institute for Human Genetics<br />

farcas@humgen.klinik.uni-mainz.de<br />

Langenbeckstraße 1<br />

55101 Mainz (Germany)


Martin Herold, Dorte Bohla, Marek Bartkuhn, Imke Panzer, Rainer Renkawitz<br />

CTCF, the highly conserved boundary factor of Drosophila and<br />

vertebrates<br />

Insulator sequences guide the function of distantly located enhancer elements to the<br />

appropriate target genes by blocking inappropriate interactions. In Drosophila dCTCF is<br />

the only insulator binding protein known to be conserved in vertebrates. We found that<br />

the structurally related factors dCTCF and Su(Hw) have distinct binding targets, whereas<br />

the Su(Hw) interacting factor CP190 largely overlaps with dCTCF binding sites. Analysis<br />

of the bithorax complex revealed that six of the borders between the parasegment<br />

specific regulatory domains are bound by dCTCF and CP190 in vivo and some of them<br />

act as insulators (1,2). We have shown that the function of one boundary, Fab-8, is<br />

dependent on binding of dCTCF, as mutations of the dCTCF target sites abolish Fab-8<br />

insulator function (1,3).<br />

Since dCTCF is critical for Fab-8 enhancer blocking in Drosophila S2 cells, we used cells<br />

with integrated reporter constructs. These contain variants of the Fab-8 insulator<br />

(Ciavatta et al., 2007), which allow us to investigate the interaction between dCTCF and<br />

CP190 by RNAi experiments and studies of other dCTCF interaction partners, which we<br />

identified by Flag-tag IPs.<br />

As it is known, that CTCF mediates the interchromosomal colocalization between<br />

Igf2/H19 and Wsb1/Nf1 (Ling et al., 2007) we wanted to investigate the influence of<br />

dCTCF on the 3D conformation of the chromatin. For this purpose we use 3C-assay to<br />

analyse the interaction between dCTCF target sites in the presence and absence of<br />

dCTCF.<br />

Literature<br />

1. Mohan, M., Bartkuhn, M., Herold, M., Philippen, A., Heinl, N., Bardenhagen, I., Leers,<br />

J., White, R. A., Renkawitz-Pohl, R., Saumweber, H., and Renkawitz, R. (2007) Embo J<br />

26(19)<br />

2. Holohan, E. E., Kwong, C., Adryan, B., Bartkuhn, M., Herold, M., Renkawitz, R.,<br />

Russell, S., and White, R. (2007) PloS Genet 3(7)<br />

3. Moon, H. et al, (2005) EMBO Rep 6(2)<br />

contact:<br />

Dipl. Biol. Martin Herold<br />

Justus-Liebig-Universität Giessen<br />

Institut für Genetik<br />

martin.herold@bio.uni-giessen.de<br />

Heinrich-Buff-Ring 58<br />

35392 Giessen (Germany)


Jürgen Geisel, Heike Schorr, Gunar H. Heine, Marion Bodis, Ulrich Hübner, Jean-Pierre<br />

Knapp, Wolfgang Herrmann<br />

Decreased p66Shc promoter methylation in patients and endstage<br />

renal disease<br />

p66Shc is a stress response protein and partially regulated by epigenetic modifications.<br />

Mice lacking p66Shc have reduced atherosclerosis and a prolonged life time. The aim of<br />

the present study is to compare promoter methylation of the p66Shc gene between<br />

healthy controls and patients with end-stage renal disease (ESRD). There are two<br />

reasons for studying patients with ESRD. First, patients with ESRD have a disturbed<br />

homocysteine metabolism and second an increased risk for cardiovascular disease.<br />

In our study we measured fasting levels of homocysteine, S-adenosylmethionine (SAM),<br />

S-adenosylhomocysteine (SAH) and 8-isoprostane in 22 patients and in 26 healthy, age-<br />

and sex-matched controls. The methylation of the p66Shc promoter and Line-1, as<br />

marker of whole genome methylation was quantified in peripheral blood mononuclear<br />

cells.<br />

In comparison to the control group homocysteine, SAM, SAH, 8-isoprostane and whole<br />

genome methylation were significantly elevated in ESRD patients, while the p66Shc<br />

promoter methylation was significantly reduced. A significant correlation was found<br />

between SAH and p66Shc promoter methylation in the patient group. This observation<br />

underlines the role of SAH as a potent inhibitor of methyltransferases. Using backward<br />

regression analysis, we demonstrated that 8-isoprostane has a significant influence on<br />

p66Shc promoter methylation. In the control group and in patients with ESRD increasing<br />

8-isoprostane levels were linked to an elevated promoter methylation.<br />

Under physiological conditions, based on the results of the control group, the p66Shc<br />

expression is more silenced through epigenetic modifications. The atheroclerotic risk is<br />

dramatically increased in ESRD patients; therefore our experimental results of<br />

methylation are in accordance with the clinical situation.<br />

Literature<br />

1)Migliaccio E, Giorgio M, Pelicci PG. Apoptosis and aging: Role of p66Shc redox protein.<br />

Antioxid. Redox Signal. 2006; 8:600-8.<br />

2)Geisel J, Schorr H, Heine GH, Bodis M, Hübner U, Knapp JP, Herrmann W. Decreased<br />

p66Shc promoter methylation in patients and end-stage renal disease. Clin. Chem. Lab.<br />

Med. 2007;45:1764-70<br />

contact:<br />

Prof. Dr. Jürgen Geisel<br />

University of Saarland<br />

Department of Clinical Chemistry<br />

kchjgei@uniklinikum-saarland.de<br />

Kirrbergerstr<br />

66421 Homburg (Germany)


Martina Dadejova, K. Yoong Lim, Roman Matyasek, Andrew Leitch, Ales Kovarik<br />

Developmental activation of silent rRNA genes is associated<br />

with increased transcription activity of rDNA loci in synthetic<br />

hybrids of Nicotiana<br />

The formation of allopolyploid plant is often associated with homogenization and<br />

expression changes of repeated sequences including tandem arrays of units coding for<br />

18-5.8-26S nuclear ribosomal DNA (rDNA). We have studied inheritance and expression<br />

of parental rDNA (nucleolar dominance) in Nicotiana rustica (2n = 4x = 48), which is a<br />

natural 10 000 years-dated allotetraploid between the diploid species N. paniculata (2n<br />

= 2x = 24, maternal P- genome donor) and N. undulata (2n = 2x = 24, paternal U-<br />

genome donor). We also studied synthetic F1 diploid (1n = 2x = 24) and allotetraploid<br />

(2n = 4x = 48) hybrids derived from respective progenitor species. Natural N. rustica<br />

has approx. three times higher number of U-derived genes than expected from gene<br />

additivity due to interlocus homogenization process, in synthetic hybrids structure and<br />

relative amount of P- and U-type units was not altered (Dadejova et al., 2007).<br />

Expression of rDNA (internal transcribed spacers ITS1 and ITS2) loci was examined by<br />

RT-CAPS method. Transcription analysis revealed silencing of P-type units in leaves of<br />

both N. rustica and synthetic hybrids. In roots, calli and floral organs of the synthetic<br />

hybrids the silent P-units were derepressed. Quantitative PCR showed several fold higher<br />

levels of primary ETS-18S-5.8S-26S transcripts in root tips, floral organs and calli than<br />

in leaves.<br />

Several conclusions can be drawn from this study: (i) developmental switches of<br />

nucleolar dominance occur in synthetic Nicotiana hybrids and polyploids with balanced<br />

numbers of parental rRNA genes (ii) uniparental silencing is broken in tissues with<br />

increased transcription of rDNA (iii) no such switches occur in natural N. rustica with<br />

partially homogenized parental units.<br />

Literature<br />

Dadejová M, Lim KY, Sou•ková-Skalická K, Matyášek R, Grandbastien MA, Leitch AR,<br />

Kovarik A. 2007. Transcription activity of rRNA genes correlates with a tendency towards<br />

intergenomic homogenization in Nicotiana allotetraploids. New Phytologist 174: 658-668.<br />

contact:<br />

Mgr. Martina Dadejova<br />

Academy of Sciences<br />

Institute of Biophysics v.v.i.<br />

dadejova@ibp.cz<br />

Kralovopolska 135<br />

CZ-61265 Brno (Czech Republic)


OLUSOLA DOKUN, WOLFGANG SCHULZ<br />

DNA hypomethylation of SNCG (synuclein-gamma) in cancer:<br />

tumor-specific or cell type- specific?<br />

SNCG is one of few single-copy genes reported to be activated by DNA hypomethylation<br />

in human cancers. Accordingly, a microarray comparison of cultured urothelial carcinoma<br />

(UC) and normal cells from the same patient indicated a 5-fold upregulation. Of 13 UC<br />

cell lines, 6 showed overexpression, but 7 very low levels vs. normal cells. Similarly, in<br />

indivdiual carcinoma tissues, both increased and strongly diminished SNCG expressions<br />

were found. Treatment with the DNA methyltransferase inhibitor 5-aza-2-deoxycytidine<br />

induced expression in non-expressing cell lines. Bisulfite sequencing revealed dense<br />

methylation of the SNCG promoter in UC lines without expression, whereas normal<br />

urothelial cells and expressing UC lines lacked methylation. Investigation of a larger<br />

range of cells showed partial methylation of SNCG in fibroblasts, blood leukocytes and<br />

ureteral connective tissue. In fibroblasts low expression was found. The promoter was<br />

unmethylated in normal prostate epithelial cells, but densely methylated in prostate<br />

cancer cell lines. These data demonstrate cell type-specific methylation of the SNCG<br />

promoter, i.e. low methylation in epithelia and partial methylation in connective tissue<br />

and blood. In carcinomas, downregulation of SNCG appears associated with<br />

hypermethylation, whereas upregulation occurs in cells with an already unmethylated<br />

promoter and not by hypomethylation. We propose SNCG as a new example of a gene<br />

subject to tissue-specific methylation.<br />

Literature<br />

Hoffmann MJ, Schulz WA. 2005. Causes and consequences of DNA Hypomethylation in<br />

human cancer. Biochem cell bio. 83:296-321.<br />

•Liu H, Liu W, Wu Y, Zhou Y, Xue R, Luo C, Wang L, Zhao W, Jiang JD, Liu J. 2005. Loss<br />

of epigenetic control of synuclein-gamma gene as a molecular indicator of metastasis in<br />

a wide range of human cancers. Cancer Res 65:7635-7643.<br />

contact:<br />

MR OLUSOLA DOKUN<br />

HEINRICH HEINE UNIVERSITY<br />

DEPT. OF UROLOGY<br />

olusolayakub.dokun@uni-duesseldorf.de<br />

UNIVERSITATSTR. 1<br />

40225 DUESSELDORF (GERMANY)


Joachim Weitzel<br />

DNA methylation impairs activation of haploid expressed<br />

genes in male germ cells.<br />

Spermatogenesis is a complex developmental process starting from a pluripotent<br />

spermatogonial stem cell finally leading to highly specialized spermatozoa. The<br />

developmental program of spermatogenesis is regulated by several testis-specific<br />

transcription factors, e.g. the cAMP response element modulator tau (CREMtau) or the<br />

germ cell nuclear factor (GCNF). CREMtau, a testis-specific transcriptional activator, is<br />

an alternative splice product of the CREM gene belonging to a family of proteins, which<br />

are regulated by cAMP and bind CRE sites. The consensus sequence of the CRE site (5’-<br />

TGACGTCA-3’) contains a central CpG which is a target of DNA methylation. Here we<br />

show that methylation of the consensus CRE site prevents the CREM protein for binding<br />

in electrophoretic mobility shift assays. Furthermore, CREM is unable to activate a<br />

methylated promoter-reporter construct in cell experiments. In silico analysis identified<br />

variants of the CRE site within several haploid expressed gene. One of these modified<br />

CRE sites within the mGPDH gene promoter lost the critical CpG motif; however,<br />

generated an additional binding site for the nuclear receptor GCNF. GCNF is a<br />

transcriptional repressor and belongs to the nuclear receptor superfamily of ligandactivated<br />

transcription factors; however, a ligand for GCNF is unknown. Transcriptional<br />

activity of CREM leads to an activation of a promoter-reporter construct in cell culture<br />

whereas additional activation of GCNF leads to an alleviation of gene transcription. The<br />

activation of CREM and GCNF correlates with modifications of the chromatin structure<br />

(e.g. histone acetylation rates), suggesting a balanced activation of CREM and GCNF for<br />

appropriate gene expression in haploid male germ cells. Since several haploid-expressed<br />

target genes contain CREM and GCNF sites and these sites are sensitive to epigenetic<br />

modifications this regulation schema might be a general regulation mechanism for<br />

several post-meiotically expressed genes implicated in sperm motility and male fertility.<br />

Literature<br />

Rajkovi•,M., Middendorff,R., Wetzel,M.G., Frkovi•,D., Damerow,S., Seitz,H.J. and<br />

Weitzel,J.M. (2004). Germ cell nuclear factor relieves cAMP-response-element modulator<br />

tau-mediated activation of the testis-specific promoter of human mitochondrial glycerol-<br />

3-phosphate dehydrogenase.<br />

Journal of Biological Chemistry 279, 52493-52499.<br />

contact:<br />

Dr Joachim Weitzel<br />

FBN Dummerstorf<br />

Department of Reproductive Biology<br />

weitzel@fbn-dummerstorf.de<br />

Wilhelm-Stahl-Allee 2<br />

18196 Dummerstorf (Germany)


Christina Klaus, Daniela Kremer, Victoria Kolb-Bachofen<br />

DNA methyltransferases and the influence of cytokines and<br />

nitric oxide (NO) on DNA methylation<br />

Chronic inflammation has been recognized to play a role in the regulation of DNA<br />

methylation, but so far no molecular mechanisms are known.<br />

During the inflammation reaction proinflammatory cytokines lead to the expression of<br />

the inducible NO synthase (iNOS) which then generates high output NO synthesis.<br />

In a parallel contribution we describe that NO causes DNA remethylation (see “iNOS –<br />

generated NO plays a critical role in DNA-methylation” by Kremer D., Schulz W. and Kolb-<br />

Bachofen V.) but it is not known in which way NO influences DNA methylation.<br />

We here searched for modulation of Dnmt expression by cytokines and/or by NO.<br />

Expression of Dnmt 1, 3a, 3b and Gadd 45• at the mRNA level as well as the protein<br />

level were maintained by real-time PCR and by Western blotting experiments.<br />

The cell line A549iNOS, stably transfected with the iNOS promoter, was treated with 5aza-2´-deoxycytidine<br />

(Aza) and Trichostatin A (TSA) for three days.<br />

The cells were exposed to the cytokines IL-1•, IFN-• and TNF-• for two days with<br />

continued presence of Aza/TSA.<br />

In some experiments the NOS inhibitor N5-(1-Iminoethyl)-L-ornithine (L-NIO) was also<br />

added and/or the chemical NO donor DetaNO at nontoxic concentrations or alternatively<br />

Deta as control.<br />

In a third experiment the cells were treated with DetaNO or Deta only.<br />

Real-time PCR at 24 hours and 48 hours after treatment with Aza/TSA results in<br />

significant increases of all four mRNA-species. Neither cytokine addition nor NOS<br />

inhibition altered this response, with the exception of Gadd 45• mRNA, which was<br />

significantly reduced by these additions. All four mRNA species were completely<br />

suppressed by the additional presence of the NO donor.<br />

Protein levels of Dnmt 1, Dnmt 3a and Gadd 45• were determined 48 hours after<br />

treatment with the different agents and analyzed in Western blots.<br />

We see no significant alterations in protein levels by the various treatments, except that<br />

the additional presence of the NO donor increases protein levels of Dnmt 1, 3b and Gadd<br />

45•.<br />

In summary, these results demonstrate that indeed Dnmt´s are modulated in their<br />

expression levels by inhibitor treatment and by NO.<br />

Further experiments will have to show, whether an impact or enzyme activity is also<br />

seen.<br />

contact:<br />

Christina Klaus<br />

Heinrich-Heine-Universität Düsseldorf<br />

Arbeitsgruppe Immunbiologie<br />

christina.klaus@uni-duesseldorf.de<br />

Universitätsstrasse 1<br />

40225 Düsseldorf (Germany)


Stefanie Stepanow, Kathrin Reichwald, Klaus Huse, Matthias Platzer<br />

Do epigenetic effects at MCHR1 contribute to obesity?<br />

Obesity is a major cause of morbidity & mortality in western community. It is associated<br />

with an increased risk of type 2 diabetes, heart disease, cancer etc. Melaninconcentrating<br />

hormone receptor 1 (MCHR1) plays a significant role in regulation of<br />

energy balance, food intake and body weight in humans and rodents. Wermter et al.<br />

(2005) detect an association and transmission disequilibrium with obesity for two MCHR1<br />

SNPs (rs133072, rs133073) in obese German children and adolescents. However, this<br />

finding could not be confirmed in four independent study samples from Germany,<br />

France, Denmark & USA. Accordingly, results of two other groups were contradictory.<br />

Gibson et al. (2004) did not find association of these SNPs with obesity in a cohort of<br />

British Caucasians aged 40-65 years. In contrast, Bell et al. (2005) reported a weak<br />

association for rs133072 and obesity in a French Caucasian study group comprising<br />

morbidly obese children and adults. These findings raise the question of age-dependent<br />

mechanisms and/or epigenetic effects.<br />

SNPs rs133072 and rs133073 are in tight linkage and form two haplotypes in which one<br />

allele of either SNP represents a methylation site. We hypothesize that epigenetic factors<br />

contribute to the contradictory results reported for the association of MCHR1 SNPs and<br />

obesity. We have analyzed both SNPs and 16 CpGs in their vicinity with respect to a<br />

haplotype specific DNA methylation in human individuals of different age and will present<br />

initial results.<br />

Literature<br />

Wermter & Reichwald et al. 2005: Mutation analysis of the MCHR1 gene in human<br />

obesity.Eur J Endocrin 152(6): 851-862.<br />

Gibson et al. 2004: Melanin-Concentrating Hormone Receptor Mutations and Human<br />

Obesity: Functional Analysis. Obesity 12: 743-749.<br />

Bell et al. 2005: Association of Melanin-Concentrating Hormone Receptor 1 5'<br />

Polymorphism With Early-Onset Extreme Obesity. Diabetes 54: 3049-3055.<br />

contact:<br />

Stefanie Stepanow<br />

Leibniz institute for age research<br />

Fritz-Lipmann institute<br />

stepanow@fli-leibniz.de<br />

Beutenbergstr. 11<br />

07745 Jena (Germany)


Perrine Gaub, Andrea Tedeschi, Antonio Schmandke, Radhika Puttagunta, Tuan<br />

Nguyen, Simone Di Giovanni<br />

Enhancement of neuronal acetylation promotes neurite and<br />

axon outgrowth<br />

Spontaneous axon regeneration following injury in the central nervous system is<br />

extremely limited due to the presence of an inhibitory environment, mediated by<br />

disrupted myelin and extracellular matrix proteins, and to a deficiency of the intrinsic proaxon<br />

outgrowth gene expression program.<br />

Here, we have employed a novel strategy to switch the intrinsic neuronal genetic<br />

program from a “non permissive” to a “permissive” pattern for neurite/axon outgrowth<br />

by increasing acetylation by facilitating the histone acetyltransferases (HAT) activity. In<br />

fact, HAT regulate the remodelling of chromatin and of the dynamic changes in the<br />

nucleosomal packaging, by transferring acetyl groups to core histone proteins and<br />

transcription factors. These modifications promote chromatin relaxation, accessibility to<br />

chromosomal DNA, and ultimately facilitate transcription.<br />

Previously, increased neuronal acetylation has been shown to protect from apoptosis and<br />

to promote differentiation..<br />

Here, we show that the enhancement of endogenous acetylation in neurons, by using<br />

specific deacetylase inhibitory drugs, leads to increased neurite/axon outgrowth.<br />

Specifically, we demonstrate that increased acetylation enhances axon outgrowth and<br />

reduced growth cone collapse in primary neurons on both permissive (poly-D-lysine) and<br />

non-permissive (myelin, CSPG) substrates. In addition, we show that these effects are<br />

largely dependent upon the HATs CBP/p300 and P/CAF and the acetylation of the<br />

transcription factor p53.<br />

Finally, in vivo experiments in models of axon injury further address the relevance of<br />

acetylation on axon regeneration.<br />

These findings may contribute to developing new strategies for axonal outgrowth and<br />

regeneration after injury.<br />

contact:<br />

MD, PhD Simone Di Giovanni<br />

University of Tuebingen<br />

HIH<br />

simone.digiovanni@medizin.uni-tuebingen.de<br />

otfried mueller strasse 27<br />

72076 Tuebingen (Germany)


Jana Krejci, Eva Bartova, Andrea Harnicarova, Roman Hajek, Gabriela Galiova,<br />

Stanislav Kozubek<br />

Epigenetic changes in multiple myeloma cells<br />

Chromosomal rearrangements can be frequently observed in many cancer cells. Multiple<br />

myeloma represents one such example. Karyotypic abnormalities of MM correlated with<br />

the stage of this disease and drug response. Here, we focused on two MM cell lines;<br />

CD138- ARH-77 and CD138+ MOLP-8 cells which represent interesting models for their<br />

analogy with the bone marrow environment of multiple myeloma patients. CD138- cells<br />

are considered as a physiological component while CD138+ cells are considered as a<br />

malignant cell population. ChIP-PCR analyses of epigenetic profiles such as H3K9<br />

acetylation and H3K9 di-methylation at promoters and coding regions of the c-myc and<br />

CCND1 genes showed specific epigenetic patterns of these histone modifications induced<br />

by cytostatics such as bortezomib, dexamethason, melphalan and after gamma<br />

irradiation. Analyzing H3K9 acetylation at selected promoter regions through the use of<br />

ChIP on chip methodology, we observed interesting differences between the control and<br />

melphalan treated ARH-77 cells. Our preliminary data showed that melphalan causes<br />

changes in H3K9 acetylation at certain region of human chromosomes.<br />

These experiments were supported by grant LC06027 and by other grants<br />

AVOZ50040507 and AVOZ50040702.<br />

contact:<br />

PhD Jana Krejci<br />

Institute of Biophysics AS CR, v.v.i.<br />

krejci@ibp.cz<br />

Kralovopolska 135<br />

61265 Brno (Czech Rebublic)


Georgios J. Vlachojannis, Andreas M. Zeiher, Stefanie Dimmeler<br />

Epigenetic control of the eNOS promoter by DNA methylation<br />

in vasculogenic progenitor cell populations<br />

DNA methylation has been shown to play an essential role in both the transcriptional<br />

regulation and endothelial cell-specific expression of the human endothelial nitric oxide<br />

synthase (eNOS) gene. Further, recent data emphasizes an important role of eNOS in<br />

stem cell biology in particular with regard to mobilization and vasculoprotective<br />

properties. We assessed the hypothesis that stem and vasculogenic progenitor cells will<br />

exhibit different DNA methylation patterns of the eNOS promoter region dependent on<br />

their vascular fate. Genomic DNA from either cultivated cells i.e. endothelial progenitor<br />

cells (EPCs), mesangioblasts, CD34+, HUVECs and microvascular endothelial cells<br />

(MVECs) or sorted stem cell subpopulations (CD34 / KDR) were subjected to sodium<br />

bisulfite treatment. The final PCR products were subcloned and sequenced (5-10 clones).<br />

Whereas the eNOS proximal promoter was either devoid or very lightly methylated in the<br />

human endothelial cell types including HUVECs and MVECs, the promoter was heavily<br />

methylated in the examined progenitor and stem cell types namely CD34+ and<br />

mesangioblasts. Surprisingly, EPCs and CD34+/KDR+ subpopulations exhibit a profound<br />

methylation of the eNOS promoter. Finally, preliminary in vivo data suggest that the<br />

eNOS promoter methylation of in vivo implanted Matrigel-EPCs partly dissolves after 14<br />

days. In conclusion, we have demonstrated that the examined progenitor and stem cells -<br />

although having prospects to a vascular fate - are in contrast to endothelial cell types<br />

heavily methylated in the promoter region of the eNOS gene suggesting epigenetic<br />

silencing at this level of maturation. However, preliminary data show an in vivo effect on<br />

methylation patterns of the eNOS promoter in EPCs.<br />

contact:<br />

Dr. med. Georgios Vlachojannis<br />

J.W.Goethe-University Frankfurt<br />

Molecular Cardiology<br />

georgios.vlachojannis@kgu.de<br />

Theodor-Stern-Kai 7<br />

60590 Frankfurt am Main (Germany)


Maria Elena Torres-Padilla<br />

Epigenetic mechanisms in early mouse development<br />

The early preimplantation mouse embryo is a unique system where it is possible to<br />

explore the foundations of totipotency and differentiation. Following fertilisation, a single<br />

cell, the zygote, will give rise to all tissues of the organism. The first signs of<br />

differentiation in the embryo are evident at the blastocyst stage with the formation of<br />

the trophectoderm, a differentiated tissue that envelopes the inner cell mass. Although<br />

the cells of the preimplantation embryo are totipotent, as the embryo develops some<br />

differences appear to develop between them which are, at least partially, related to the<br />

epigenetic information of each of these cells. We have previously shown that specific<br />

histone marks might predispose cells in the embryo towards the inner cell mass<br />

compartment.We are currently investigating the mechanisms that underlie chromatin<br />

plasticity in the early embryo and the processes that regulate this first cell fate decision.<br />

We are interested in identifying new players involved in the establishment of the<br />

chromatin structure in the preimplantation mouse embryo. We will present our new<br />

results on such players, including chromatin modifiying enzymes and their impact in<br />

embryonic development.<br />

contact:<br />

Dr Maria Elena Torres-Padilla<br />

IGBMC<br />

metp@igbmc.u-strasbg.fr<br />

1 Rue Laurent Fries<br />

67404 Illkirch (France)


Michael Michalkiewicz, Teresa Michalkiewicz, Kyle MacGillis<br />

Epigenetic mechanisms in hypertension<br />

Epigenetic variations may underlie hypertension and in fact, better than variation in DNA<br />

sequence, may explain the late onset and quantitative nature of this disease. Genomewide<br />

DNA methylation patterns in hypertensive Dahl S (SS) and normotensive Brown<br />

Norway (BN) rats were assessed in a high throughput manner by methylated DNA<br />

immunoprecipitation combined with a competitive hybridization on a microarray<br />

(NimbleGene) covering 22K promoters. More than 2K promoters were hypermethylated<br />

in the SS kidney. The heart methylomes were less different. Gene encoding enzymes<br />

and involved in inflammation were the mostly methylated in the hypertensive kidney<br />

whereas the genes involved in signal transmission were the least affected. We then<br />

zoomed in on the known hypertension QTL areas and further narrowed down<br />

hypermethylated targets. We then selected a few candidate genes for a detailed<br />

nucleootide specific quantitative methylation assay using pyrosequencing of bisulfite<br />

converted DNA. Pyrosequencing revealed hypermethylation of the renin promoter (but<br />

not a distal CpG island) in the hypertensive kidney, consistent with the reduced renin<br />

expression in this strain. Thus, an epigenetic mechanism may underlie some forms of<br />

hypertension. However, since the kidney is both the culprit and the victim of<br />

hypertension, the cause-effect relationship between the epigenome and hypertension<br />

has to be established. Support: NIH HL-082798 and AHW MCW.<br />

contact:<br />

Assoc Professor Michael Michalkiewicz<br />

Medical College of Wisconsin<br />

Department of Physiology<br />

mmichalk@mcw.edu<br />

8701 Watertown Plank Road<br />

53226 Milwaukee (USA)


Silke Götze, Sonja Sievers, Oliver Müller<br />

Epigenetic regulation in the Wnt signalling pathway<br />

Epigenetic mechanisms are involved in controlling gene expression. The two main<br />

mechanisms of epigenetic gene regulation are DNA methylation and histone<br />

modification, which affect each other. Histone modifications like acetylation, methylation<br />

and phosphorylation are known to be highly dynamic and are likely to be modulated by<br />

signalling pathways. In contrast very little is known about dynamic changes of DNA<br />

methylation, specifically if they occur and how they might be regulated. Therefore we<br />

want to investigate, which epigenetic changes occur due to Wnt signalling and what<br />

impact these changes have on expression of Wnt target genes.<br />

For this approach we use DMH (differential methylation hybridisation) on a CpG island<br />

microarray to identify changes in the DNA methylation. In a further step we will also<br />

perform a ChIP (chromatin immunoprecipitation) on chip analysis to detect changes in<br />

the histone H3 acetylation.<br />

In a first step we evaluated a proper Wnt model system with a Wnt inactive and a Wnt<br />

active state. We used western blotting and RNA expression analysis to find a suitable<br />

model system. DMH with this model system led to a number of candidate genes, which<br />

showed differential DNA methylation on the microarray. We performed bisulfite<br />

sequencing of some candidate genes but we could not verify the microarray results. The<br />

high number of false positive results was likely caused by incomplete restriction during<br />

DMH. Thus we established internal controls to monitor the restriction during repetition of<br />

the DMH experiment.<br />

contact:<br />

M.Sc. Silke Götze<br />

MPI für molekulare Physiologie<br />

Abteilung Strukturbiologie<br />

Silke.Goetze@mpi-dortmund.mpg.de<br />

Otto-Hahn-Strasse 11<br />

44227 Dortmund (Deutschland)


Nadia Sellami, Sabine Adam-Klages, Reiner Siebert, Hans-Jürgen Heidebrecht<br />

Epigenetic Regulation of the Cancer Testis Antigen CT45<br />

The cancer testis (CT) antigen CT45 is encoded by a family of six highly similar genes<br />

that are clustered in tandem on Xq26.3. (1) The CT45-protein is found in the nucleus of<br />

male germ cells, germ-cell-derived tumors and Hodgkin lymphoma.<br />

The functions of the various CT antigens are highly diverse and little is known about<br />

their regulation. For a set of CT antigens with a CpG-rich promoter, DNA methylation has<br />

been shown to be the primary silencing mechanism. (2) The possibility of epigenetic<br />

regulation of CT45 is indicated by experiments showing that CT45 is inducible in nonexpressing<br />

cells after treatment with the demethylating agent 5-aza-2’-deoxycytidine.<br />

(3)<br />

Our experiments focus on the the methylation profile of the CT45 promoter in CT45expressing<br />

and non-expressing cells to find out whether this CT-X antigen is also<br />

regulated by DNA methylation. Initial experiments show that a small region that contains<br />

3 CpGs in the CT45 transcription start region is differentially methylated. In those, DNA<br />

of CT45-expressing cells from testis or L428 Hodgkin lymphoma cells was about 35 %<br />

methylated whereas DNA of non-expressing cells derived from tonsils, HeLa cancer cells<br />

or cervix was approximately 85 % methylated. After treatment of HeLa cells with 5-aza-<br />

2’-deoxycytidine, around 20 % of the cells expressed CT45. DNA of those cells enriched<br />

by FACS was also less methylated than the CT45-non-expressing cells. We propose that<br />

DNA methylation is a regulatory mechanism for CT45.<br />

Literature<br />

(1) Chen YT et al. Proc Natl Acad Sci U S A 2005;102:7940-5.<br />

(2) De Smet C et al. Mol Cell Biol 1999;19:7327-35.<br />

(3) Heidebrecht HJ et al. Clin Cancer Res 2006;12(16):4804-11.<br />

contact:<br />

Nadia Sellami<br />

Christian-Albrechts-University, Kiel<br />

Institute of Pathology<br />

nsellami@path.uni-kiel.de<br />

Michaelisstr. 11<br />

24105 Kiel (Germany)<br />

additional information<br />

co-author affiliations:<br />

H.J. Heidebrecht: Institute of Pathology<br />

S. Adam-Klages: Institute of Immunology<br />

R. Siebert: Institute of Human Genetics


Svend Petersen-Mahrt, Wolf Reik, Siim Pauklin, Heather Coker<br />

Epigenetic Reprogramming of 5-meC via DNA Deamination and<br />

DNA Repair<br />

Activation-induced cytidine deaminase (AID) coordinates B-cell class switch<br />

recombination and somatic hypermutation, to diversify the expressed immunoglobulin<br />

repertoire of a healthy immune system. Activity of AID at the immunoglobulin locus has<br />

been confirmed by the presence of uracil, the product of AID mediated cytosine<br />

deamination. Other forms of cytosine have not previously been thought to be significant<br />

substrates, however we have demonstrated that in vitro and in E. coli 5’-methyl cytosine<br />

(5’me-C), an important element within epigenetic regulation, is also deaminated by AID.<br />

Such activity results in conversion of 5’me-C to thymine with subsequent repair of the<br />

T:G mismatch not reinstating the methyl cytosine, thus potentially altering the gene’s<br />

epigenetics status. Furthermore, AID is co-expressed with a surrounding cluster of<br />

pluripotency genes during early development, linking expression to function. The<br />

analysis has now been extended to in vivo mouse transgenics. We are able to show that<br />

AID, if targeted to a methylated locus can actively (depending on the catalytic activity of<br />

AID) initiate epigenetic reprogramming. Our current work focuses on the molecular<br />

mechanism on how AID and other DNA deaminases can initiate and process the lesions<br />

at 5’me-C sites in DNA. By comparing and contrasting the different cell milieus for<br />

immunoglobulin diversification and epigenetic reprogramming, as well as identify AID<br />

interacting proteins, our current model states, that AID can alter the balance of the DNA<br />

repair pathways and lead to mutagenic or non-mutagenic repair depending on the status<br />

of the cell.<br />

contact:<br />

Dr Svend Petersen-Mahrt<br />

Cancer Research UK<br />

DNA Editing - Clare Hall Laboratories<br />

skpm@cancer.org.uk<br />

Balnche Lane<br />

EN6 3LD South Mimms (UK)


Robert Liefke, Daniela Salat, Jörg Wiedenmann, Franz Oswald, Tilman Borggrefe<br />

ETO, but not AML1/ETO, augments RBP-Jk/Sharp-mediated<br />

transcriptional repression of Notch target genes<br />

Notch is a transmembrane receptor that determines<br />

cell fates and pattern formation in all animal species.<br />

After ligand binding, proteolytic cleavage steps occur<br />

and the intracellular part of Notch (Notch-IC)<br />

translocates to the nucleus, where it targets the DNA<br />

binding protein RBP-Jκ/CBF1 (reviewed in 1). In the<br />

absence of Notch, RBP-Jκ represses Notch target<br />

genes through recruitment of a corepressor complex.<br />

We and others have identified SHARP, CtIP and CtBP<br />

as a component of this complex (3, 4, 5). Here, we<br />

show that co-repressor ETO, as well as the<br />

leukemogenic fusion protein AML1/ETO, directly<br />

interact with SHARP, that ETO is part of the<br />

endogenous RBP-Jκ-containing co-repressor<br />

complex and that it is found at the Notch target gene<br />

promoters in ChIP experiments. In functional assays<br />

co-repressor ETO, but not leukemogenic fusion<br />

protein AML1/ETO, augments SHARP-mediated<br />

repression. Furthermore, overexpression of<br />

AML1/ETO activates endogenous Notch target genes<br />

whereas knock-down of AML1/ETO leads to<br />

downregulation of Notch target genes. Therefore, we<br />

propose that AML1/ETO can disturb the normal,<br />

repressive function of ETO at Notch target genes.<br />

This activating (derepressing) effect of AML1/ETO<br />

may contribute to its oncogenic potential in myeloid<br />

leukemia.<br />

Literature<br />

1) Bray SJ.. (2006) Nat Rev Mol Cell Biol 7:678-89.<br />

2) Licht JD (2001) Oncogene 20:566079<br />

3) Oswald, F., et al. (2002) Embo J 21:5417-26.<br />

4) Kuroda, K. et al. (2003) Immunity 18:301-12.<br />

5) Oswald, F. et al. (2005) Mol Cell Biol 25:10379-90<br />

contact:<br />

Robert Liefke<br />

Max-Planck Institute of Immunobiology<br />

liefke@immunbio.mpg.de<br />

Stübeweg 51<br />

79108 Freiburg (Germany)


Yvonne Möller-Steinbach, Cristina Madeira Alessandre, Vivien Exner, Patti Taranto,<br />

Claudia Köhler, Lars Hennig<br />

Function of Polycomb group proteins in the transition to<br />

flowering in plants<br />

Polycomb group (PcG) proteins function as silencers of gene transcription by establishing<br />

repressive chromatin environments at specific target loci. In animals, PcG proteins form<br />

protein complexes, such as Polycomb repressive complex PRC1 and PRC2. Plants also<br />

contain PcG proteins, but probably have only PRC2- and no PRC1-homologs. One well<br />

studied function of PcG proteins in plants is to control the decision to flower, which is<br />

based on the integration of several environmental signals. One such key signal is<br />

vernalization, the exposure to prolonged periods of cold, such as typically experienced<br />

during winters. A vernalization requirement enables plants to winter in vegetative phase<br />

before initiating reproduction with approaching spring. The molecular basis of<br />

vernalization involves an epigenetic cellular memory of the duration of cold. Prolonged,<br />

but not short exposures to cold inhibit transcription of FLC, a potent repressor of<br />

flowering. The inactive state of FLC is maintained even if plants are returned to ambient<br />

temperature, and this maintenance requires the VRN2, a homolog of the PRC2 subunit<br />

Su(Z)12. Here, we will present results concerning the composition and function of the<br />

plant PRC2-like VRN2 complex.<br />

contact:<br />

Dipl Biochem Yvonne Möller-Steinbach<br />

ETH Zuerich<br />

Plant Sciences<br />

syvonne@ethz.ch<br />

Universitätstr.2<br />

8092 Zürich (Schweiz)


Cordula Tschuch, Angela Schulz, Armin Pscherer, Meinhard Hahn, Peter Lichter, Daniel<br />

Mertens<br />

Functional analysis of candidate genes localized in 13q14.3, a<br />

region commonly affected in B-CLL<br />

In B-cell Chronic Lymphocytic Leukemia (B-CLL), loss of region 13q14.3 is the most<br />

frequent genomic imbalance. Several characteristics of this region point to an epigenetic<br />

pathomechanism: 1) candidate genes lack point mutations, 2) these genes are<br />

downregulated in tumors, 3) the presence of large non-coding RNA genes is reminiscent<br />

of imprinted regions where only one gene copy is active. Recently, we could show that<br />

the two copies of the critical region are replicating asynchronously suggesting differential<br />

chromatin packaging. Furthermore, a number of genes in the region are monoallelically<br />

expressed in healthy probands. This led us to the functional characterization of genes in<br />

the critical region which will allow identification of their role in the molecular<br />

pathomechanism.<br />

Candidate genes were overexpressed in a hematopoetic cell line and RNAi technology<br />

was used for the knock down of candidate genes. After modulation of expression, RNA<br />

was isolated at different time points to identify effects in downstream target genes via<br />

expression profiling.<br />

After overexpression and knock down of candidate genes, we could identify target genes<br />

involved in pathways for which a tumor suppressor function has been described. Thus,<br />

NOTCH signaling is involved in regulation of cell cycle, apoptosis and survival.<br />

Furthermore, we showed that candidate genes are involved in NF•B signaling. This<br />

pathway has the potential to promote survival of B-cells and plays a major role in B-cell<br />

development.<br />

Our analysis shed light on the function and the pathways involved in the<br />

pathomechanism localized in 13q14.3. The essential role in early B-cell development,<br />

make them highly interesting candidate pathways for elucidation of the pathomechanism<br />

in B-CLL.<br />

contact:<br />

Dr. Cordula Tschuch<br />

German Cancer Research Center<br />

c.tschuch@dkfz.de<br />

Im Neuenheimer Feld 580<br />

69120 Heidelberg (Germany)


Devi Thiagarajan, Sanjeev Khosla<br />

Functional characterisation of mDnmt2<br />

Inspite of having all the signature motifs for a DNA methyl transferase, Dnmt2 seems to<br />

have a weak DNA methyl transferase activity. It has also been shown to have a tRNA<br />

methyltransferase activity whose significance is not clear. The purpose of this study was<br />

to probe the function of the mouse Dnmt2 protein through its interacting partners. We<br />

have identified a few interacting partners of Dnmt2 through Immunoprecipitation and<br />

GST pull down assays. Most of the interacting partners were found to be involved in RNA<br />

metabolism. These interactions have been confirmed through mammalian two hybrid<br />

and reverse Co-Immunoprecipitation experiments. The significance of Dnmt2 interacting<br />

partners, in light of our experiments examining the subcellular localization of Dnmt2 and<br />

its various deletion mutations would be presented in correlation to Dnmt2 function.<br />

contact:<br />

Senior research fellow Devi Thiagarajan<br />

Centre for DNA Fingerprinting and Diagnostics<br />

Centre for DNA Fingerprinting and Diagnostics<br />

devi@cdfd.org.in<br />

ECIL Road, Nacharam<br />

500076 Hyderabad (India)


Soyoung Lim, Johannes Schulte, Hans-Ulrich Schildhaus, Uta Flucke, Phillip Kahl,<br />

Roland Schüle, Reinhard Büttner, Jutta Kirfel<br />

Functional role of Lysine-specific histone methylase-1 in<br />

carcinogenesis<br />

Mutations in the molecular pathways that regulate cell proliferation, differentiation, and<br />

cell death all contribute to cancer formation. Enzymes that covalently modify histones<br />

affect these pathways by controlling the dynamic remodelling of the chromatin structure.<br />

While acetylation of histone is generally related to transcriptional activation, methylation<br />

at the lysine residue either activates or represses gene transcription depending on the<br />

sites of methylation or the number of methyl groups. LSD1 (Lysine-Specific Histone<br />

Demethylase), which is the first discovered lysine-specific histone demethylase, can<br />

function as both a histone diMe H3-K4 and a diMe H3-K9 demethylase. Since the<br />

catalytic domain of LSD1 has sequence homology to monoaminoxidase (MAO) and uses<br />

the same demethylating mechanism, the possible inhibitory effect of MAO inhibitors<br />

(MAOIs) on LSD1 was suggested.<br />

Recently, it has been shown that changes in global levels of individual histone<br />

modification are associated with cancer and that these changes are predictive of clinical<br />

outcome. We showed that LSD1 is highly expressed in different types of cancer,<br />

suggesting the relevance of LSD1 with tumour malignancy in various tumour types. The<br />

potential use of LSD1 as prognostic or even diagnostic marker in different types of<br />

cancer will be evaluated. In addition, the inhibitory effect of clinically used MAOIs will be<br />

tested in vitro and in vivo and discussed.<br />

contact:<br />

Soyoung Lim<br />

Bonn Medical School<br />

Institute of Pathology<br />

limsoyoung@gmx.de<br />

Sigmund-Freud Str. 25<br />

53127 Bonn (Germany)


Andreas Werner, Mark Carlile<br />

Functional short RNAs from naturally occurring<br />

sense/antisense transcripts<br />

Overlapping sense/antisense RNAs transcribed in opposite directions from the same<br />

gene are common in vertebrates but the phenomenon is poorly understood.<br />

We investigated how sense/antisense RNAs of a conserved phosphate transporter gene<br />

(Slc34a) are processed and hypothesize how this may influence gene expression.<br />

Sense/antisense transcripts are transiently co-expressed during zebrafish embryonic<br />

development. We monitored sense/antisense transcript processing using Xenopus<br />

oocytes as an expression system. In the cytoplasm RNAs were stable in whatever<br />

combination expressed. In the nucleus RNA duplexes of >30 bp were degraded into<br />

short RNAs of about 23 bases within 4 hours. The small RNAs triggered degradation of a<br />

reporter RNA. We could also detect small RNAs in zebrafish embryos at stages that show<br />

co-expression of sense/antisense transcripts. Remarkably, both strands of the small RNA<br />

are detectable and the prevalence of sense- or antisense- short RNAs is developmentally<br />

regulated.<br />

Comparable results were obtained with sense/antisense transcripts from the homologous<br />

locus in mouse. Strand selection was non-random with short RNAs complementary to the<br />

antisense transcript detectable in tissues that express the sense encoded Na/phosphate<br />

cotransporter.<br />

We present the first evidence in vertebrates that sense/antisense transcript pairs feed<br />

into an RNA interference related process. The regulated prevalence of sense- or<br />

antisense oriented short RNAs represents a strand specific molecular switch. A model for<br />

the biological role of antisense transcription is presented: We predict that antisense<br />

transcripts provide the tools to perform a dry run of the transcriptome and play an<br />

essential role in genomic quality control of higher eukaryotes.<br />

contact:<br />

Dr Andreas Werner<br />

Newcastle University<br />

Cell and Molecular Biosciences<br />

andreas.werner@ncl.ac.uk<br />

Framlington Place<br />

NE2 4HH Newcastle (United Kingdom)


Sandra Weiss, Ralf Gilsbach, Frederico Barreto, Achim Lother, Lutz Hein<br />

Heart failure and fibrosis induced by overexpression of methyl-<br />

CpG- binding protein 2 (MeCP2) in transgenic mice<br />

Regulation of gene expression by histone acetylation/deacetylation plays a basic role in<br />

the development and progression of cardiac hypertrophy and failure. Genes can be<br />

silenced by histone deacetylation and binding of methyl-CpG-binding proteins to<br />

methylated promoter areas. In the present study, we analyzed expression of MeCP2 in<br />

the heart during the development of heart failure and investigated its cardiac function in<br />

a transgenic mouse model.<br />

Expression of MeCP2 was determined in wild-type (WT) and α 2ABC -adrenoceptordeficient<br />

(α 2ABC KO) mice after induction of cardiac hypertrophy by transverse aortic<br />

constriction (TAC). In WT mice, TAC increased ventricle/body weight ratio by 154±12%<br />

after 8 weeks. In WT TAC mice, cardiac MeCP2 mRNA levels were decreased by 92±5%<br />

as compared to the sham group. Chronic elevation of circulating catecholamines in<br />

α 2ABC KO mice caused a similar decrease in cardiac MeCP2 mRNA levels to 27±15 % of<br />

WT controls. To test if MeCP2 expression affects cardiac myocyte growth, rat neonatal<br />

cardiac myocytes were cotransfected with GFP and MeCP2 or empty vector. Cardiac<br />

myocyte areas increased from 467±26 µm 2 at baseline to 637±44 µm 2 after<br />

phenylephrine treatment and to 755±40 µm 2 after phenylephrine plus MeCP2<br />

transfection. Beyond transgenic mice expressing MeCP2 under control of the α-MHC gene<br />

promoter were generated. Unexpectedly, all MeCP2 transgenic founder mice were lost at<br />

6-8 weeks of age due to a severe cardiomyopathy.<br />

These results reveal that expression of MeCP2 is regulated during heart failure and<br />

cardiac-specific expression in transgenic mice induces severe postnatal cardiomyopathy.<br />

Thus, MeCP2 may play an important role in the control of gene expression during the<br />

development of heart failure.<br />

contact:<br />

Sandra Weiss<br />

Universität Freiburg<br />

Inst. für Experimentelle und Klinische Pharmakologie<br />

sandra.weiss@pharmakol.uni-freiburg.de<br />

Albertstr. 25<br />

79104 Freiburg (Germany)


Alexandra Moosmann, Coen Campsteijn, Martina Schmid,, Eric M. Thompson<br />

High diversity of developmental stage-specific histone<br />

variants in the larvacean, Oikopleura dioica<br />

Histone variants alter the composition of individual nucleosomes and expand the PTM<br />

repertoire. With the exception of histone H4, several different variants of each histone<br />

subtype have been reported for most organisms. Oikopleura dioica is an abundant panglobal<br />

zooplankton with interesting life history features concerning histone gene<br />

organization and regulation, as well as the spatial and temporal use of histone variants.<br />

The life cycle of this urochordate is characterized by a developmental switch between<br />

mitotic and endocycling cells. We are using Oikopleura dioica as a model to study histone<br />

variant expression and their post-translational modifications in mitotic, endocycling and<br />

meiotic nuclei. The Oikopleura histone complement comprises 47 genes encoding 36<br />

different histone isoforms, a number exceeding the library known in mammals and other<br />

organisms. Most mammalian variants are also present in the Oikopleura histone<br />

complement. Performing quantitative RT-PCR we have assessed the expression profile<br />

for all histone genes throughout the Oikopleura development. Histone gene expression in<br />

Oikopleura is coregulated in clusters and several variants are exclusively expressed at<br />

very distinct developmental stages. We also find 14 variants being male specific. Further<br />

we have begun to determine the nuclear localization of the H3 variant family members<br />

with EGFP fusion constructs to investigate their possible functions in Oikopleura.<br />

contact:<br />

Alexandra Moosmann<br />

University of Bergen<br />

Sars International Centre for Marine Molecular Biology<br />

alex.moosmann@gmail.com<br />

Thormøhlensgt. 55<br />

5008 Bergen (Norway)


Irene Tiemann-Boege, Christina Curtis, Darryl Shibata, Simon Tavaré<br />

High-throughput analysis of methylation patterns to track cell<br />

divisions<br />

Errors in epigenetic DNA methylation at non-functional or neutral CpG islands can be<br />

used as a “molecular clock” to track cell replication (1). Methylation errors occur on the<br />

newly replicated DNA strand during the reestablishment of the parental methylation<br />

patterns. Like nucleotide substitutions, methylation of neutral CpG sites is heritable and<br />

cumulative, but occurs at a much higher frequency; thus, the methylation state of 8-12<br />

CpGs in a neutral CpG island can be used as a binary code of information to follow cell<br />

replication ‘a <strong>poster</strong>iori’ in somatic tissues (2-5). A series of studies have already<br />

recorded methylation patterns at 3 neutral CpG islands and modelled the replication<br />

history in different tissues of the human body (2-4). Methylation patterns in these<br />

studies were assessed by sequencing bacterial clones derived from bisulfite treated<br />

genomic DNA. In order to derive a more complete picture of the replication history based<br />

on many more cells and individuals, we developed a high-throughput assay using a new<br />

technology similar to the next generation sequencing platforms. Our assay amplifies<br />

individual molecules on magnetic beads in microscopic aqueous compartments of an oilbuffer<br />

emulsion, resulting in each bead being covered by many identical PCR copies (6,<br />

7). The methylation status of each original molecule can then be assessed by fluorescent<br />

probes under a microscope. With this technology we are capable of interrogating<br />

~400,000 CpG strings in a single experiment. In addition, a tagging strategy allows<br />

analyzing simultaneously many individuals. We have tested the reliability of this<br />

technology on known mixtures of CpG strings. With this technology it should be possible<br />

to capture with great detail the diversity of methylation patterns observed in replicating<br />

tissues.<br />

Literature<br />

1. D. Shibata, S. Tavare, Cell Cycle 5, 610 (Mar, 2006).<br />

2. J. Y. Kim, K. D. Siegmund, S. Tavare, D. Shibata, BMC Med 3, 10 (2005).<br />

3. J. Y. Kim, S. Tavare, D. Shibata, Proc Natl Acad Sci U S A 102, 17739 (Dec 6, 2005).<br />

4. J. Y. Kim, S. Tavare, D. Shibata, BMC Biol 4, 2 (2006).<br />

5. P. Nicolas, K. M. Kim, D. Shibata, S. Tavare, PLoS Comput Biol 3, e28 (Mar 2, 2007).<br />

6. J. Shendure et al., Science 309, 1728 (Sep 9, 2005).<br />

7. M. Li, F. Diehl, D. Dressman, B. Vogelstein, K. W. Kinzler, Nat Methods 3, 95 (Feb,<br />

2006).<br />

contact:<br />

Research Asscociate Irene Tiemann-Boege<br />

University of Cambridge<br />

Cancer Research Cambridge Research Institute<br />

itiemann@gmail.com<br />

Robinson Way<br />

CB2 0RE Cambridge (UK)


Tzvetina Brumbarova, Cecile Doyen, Emilie Bonnefoy, Guillermo Orsi, Pierre Couble,<br />

Benjamin Loppin<br />

HIRA functions in Drosophila<br />

The histone chaperone HIRA is a conserved chromatin assembly factor that is specifically<br />

involved in the assembly of nucleosomes containing the H3 histone variant H3.3. HIRA is<br />

involved in the replication-independent (RI) deposition of core histones, in contrast to<br />

the CAF-1 complex which is responsible for the DNA replication coupled (RC) nucleosome<br />

assembly. Previous work in our group has shown that HIRA has a critical role for the<br />

formation of the male pronucleus during fertilization in Drosophila by allowing the<br />

paternal DNA to recover a nucleosomal chromatin structure and to replicate in<br />

coordination with the maternal DNA [1]. Thus, assembly of paternal chromatin<br />

represents a peculiar case of RI assembly at the scale of a whole nucleus.<br />

Although HIRA is only absolutely required for male pronucleus formation in Drosophila,<br />

analysis of a series of recently generated mutant alleles suggests that this histone<br />

chaperone plays other functions in somatic cells. Data supporting these possible new<br />

roles of HIRA in Drosophila will be presented and discussed.<br />

Literature<br />

[1] Bonnefoy E. et al. (2007) PLoS Genetics 3(10): 1991-2006<br />

contact:<br />

PhD Tzvetina Brumbarova<br />

University of Lyon<br />

tzvetina.brumbarova@cgmc.univ-lyon1.fr<br />

43 Bd du 11 Novembre 1918<br />

69622 Villeurbanne (France)


Michael Haberland, Rusty Montgomery, Eric N. Olson<br />

Histone deacetylases 1 & 2 control adipogenesis<br />

Adipogenesis is a tightly orchestrated process in which mesenchymal precursor cells<br />

differentiate into mature fat cells. This process is under the control of a well established<br />

cascade of transcription factors including the C/EBP family, SREBP and PPARgamma. In<br />

vitro studies have shown that these adipogenic core transcription factors interact with<br />

histone deacetylases, a conserved family of chromatin modifying enzymes that usually<br />

act as transcriptional repressors. Thus, in the classical model of adipocyte differentiation,<br />

HDACs are thought to be inhibitors of the adipogenic program by directly repressing the<br />

transcriptional activity of adipogenic transcription factors.<br />

We used genetic and pharmacological models to test the influence of HDACs on<br />

adipogenesis. Treatment of pre-adipocytes (3T3-L1) and mouse embryonic fibroblasts<br />

(MEFs) with diverse HDAC inhibitors (TSA, SAHA, Scriptaid) lead to a robust block of<br />

adipocyte differentiation in vitro. Time course analyses indicated that distinct phases in<br />

the differentiation program are sensitive to HDAC inhibition. In order to elucidate targets<br />

that are responsible for the observed phenotype, we used MEFs with conditionally<br />

targeted alleles for HDAC1, HDAC2 and HDAC 8. Efficient deletion of the floxed alleles<br />

was obtained by using either a self-deleting lentiviral CRE or a Tamoxifen-inducible CRE.<br />

We found that deletion of any single HDAC did not lead to a block in adipogenesis.<br />

However deletion of both HDAC1 and HDAC2 completely blocked adipocyte<br />

differentiation without being detrimental to cell survival. A detailed molecular analysis<br />

indicated that this phenotype was due to defective chromatin remodelling during the<br />

differentiation process as well as to perturbed post-translational modifications of<br />

adipogenic transcription factors. Deletion of HDAC1 and HDAC2 in vivo using aP2-CRE<br />

HDAC1/2 double-conditional mice indicated that histone deacetylases are required for<br />

homeostasis of adipose tissue in animals.<br />

contact:<br />

Dr. Michael Haberland<br />

UT Southwestern Medical School<br />

Department of Molecular Biology<br />

michael.haberland@utsouthwestern.edu<br />

5323 Harry Hines BLVD<br />

75390 Dallas, TX (USA)


RAFFAELE TEPERINO, MICHELE LONGO, PAOLA MIRRA, PIETRO FORMISANO,<br />

FRANCESCO BEGUINOT, PAOLA UNGARO<br />

HNF4 DIRECTS HISTONE METHYLATION TO SILENCE PED/PEA-<br />

15 EXPRESSION IN HUMAN HEPATOCYTES<br />

Ped/Pea-15 is a gene commonly overexpressed in tissues from type 2 diabetic<br />

individuals and healthy subjects at high risk of developing diabetes (such as first degree<br />

relatives). Indeed, its overexpression alone is able to determine both insulin-resistance<br />

and beta-cell dysfunction in a transgenic animal model. HNF4alpha is a liver-enriched<br />

nuclear receptor that controls the expression of a broad programme of metabolic genes<br />

and thereby controls energy homeostasis in vivo. Recent evidences indicate that<br />

HNF4alpha inhibits Ped/Pea-15 expression in liver by binding its responsive element on<br />

Ped/Pea-15 promoter. However, the mechanisms by which HNF4alpha exerts its action<br />

remain not fully understood. Here we show that HNF4alpha is essential to direct histone<br />

methylation and maintain Ped/Pea-15 gene repression in human hepatocytes.<br />

HNF4alpha expression promotes the assembly of histone deacetylase (HDAC), as well as<br />

histone methyl transferase (HMT) complexes on the Ped/Pea-15 promoter and leads to<br />

the di-methylation of Lysine9 on histone H3 as determined by chromatin<br />

immunoprecipitation assays. HNF4alpha•interacts with and recruits SMRT (Silencing<br />

Mediator of Retinoic Acid and Thyroid Hormone Receptor) corepressor to Ped/Pea-15<br />

promoter inducing the compaction of the associated chromatin, as assessed by ReChIP<br />

assays. These results suggest that HNF4alpha serves as a scaffold for both HDAC and<br />

HMT activities to inhibit Ped/Pea-15 transcription, thus representing a new potential<br />

molecular tool to target Ped/Pea-15 expression.<br />

contact:<br />

Undergraduate RAFFAELE TEPERINO<br />

UNIVERSITY OF NAPLES<br />

FEDERICO II<br />

raftep@alice.it<br />

VIA PANSINI, 5<br />

80137 NAPLES (ITALY)


Eva Bartova, Abdrea Harnicarova, Jana Krejci, Gabriela Galiova, Stanislav Kozubek<br />

Human embryonic stem cells are characterized by distinct<br />

patterns of histone modifications in comparison with cells of<br />

feeder layer<br />

Higher-order chromatin structure in parallel with epigenetic modification of histones<br />

plays an important role in the regulation of nuclear processes. Here, nuclear patterns of<br />

H3K4me2, H3K4me3, H3K9 acetylation, H3K9me1, H3K9me2, H3K9me3, H3K27me2,<br />

H3K27me3 H3K79me1 and DNA methylation were studied in pluripotent human<br />

embryonic stem cells (hESCs). These interphase profiles were compared with mouse<br />

embryonic fibroblasts (MEFs) that were used as a feeder layer for hESC cultivation.<br />

Different nuclear patterns between hESCs and MEFs were found for H3K4me3,<br />

H3K9me3, H3K27me3 and DNA methylation. Observed differences in the nuclear<br />

arrangement were probably influenced by the presence of large clusters of<br />

heterochromatin, called chromocentres in MEFs, which are densely H3K4me3, H3K9me3,<br />

and DNA methylated. H3K27me3 pronouncedly accumulated at mouse inactive<br />

chromosome X, while both female chromosomes X in pluripotent hESCs did not contain<br />

such a high level of H3K27 tri-methylation. Changes in the epigenetic interphase profiles<br />

were accompanied by relatively stable levels of the proteins studied, which were<br />

determined by western blots. Our preliminary data imply that histone epigenetic patterns<br />

are cell-type specific and undergo changes more frequently in comparison with their<br />

protein levels.<br />

This work was supported by the Grant Agency of the Czech Republic, grant Nos:<br />

204/06/0978, and by other grants: AVOZ50040507 and AVOZ50040702. We thank Prof.<br />

Douglas Melton (HHMI/Harvard University) for providing us with hESCs.<br />

contact:<br />

PhD Eva Bartova<br />

Institute of Biophysics Academy of Sciences of the Czech Republic<br />

bartova@ibp.cz<br />

Kralovopolska 135<br />

61265 Brno (Czech Republic)


Francesco Nicassio 1 , Joseph Vissers 4 , Nadia Corrado 1 , Liliana Areces 1,2 , Steven<br />

Bergink 3 , Jurgen Marteijn 3 , Wim Vermeulen 3 , Maarten van Lohuizen 4 , Pier Paolo di<br />

Fiore 1,2 , Elisabetta Citterio 4<br />

Human USP3 is a chromatin modifier required for S-phase<br />

progression and genome stability<br />

Protein ubiquitination is critical for several aspects of the DNA damage response. Our<br />

studies are focused on the molecular mechanisms and the enzymes involved in the<br />

ubiquitination of histone proteins and how deregulation of this process influences<br />

genome stability and tumorigenesis. Although histones are the most abundant monoubiquitin<br />

conjugates in mammalian cells, the role of mono-ubiquitinated H2A (uH2A) and<br />

H2B (uH2B) remain poorly understood. In particular, little is known about mammalian<br />

deubiquitinating enzymes (DUBs) that catalyze the removal of ubiquitin from<br />

uH2A/uH2B. Through a biochemical approach aimed at the isolation of histone-specific<br />

ubiquitin ligases and DUBs, we identified a ubiquitin-specific protease, USP3, as a<br />

deubiquitinating enzyme for uH2A and uH2B. Biochemical analysis and FRAP-based<br />

dynamics studies showed that USP3 dynamically associates with chromatin, and<br />

deubiquitinates H2A/H2B in vivo. The ZnF-UBP domain of USP3 mediates its interaction<br />

with uH2A. Functional ablation of USP3 by RNAi leads to delay of S-phase progression,<br />

and to accumulation of DNA breaks, with ensuing activation of an ATR/ATM- regulated<br />

DNA damage checkpoint response. In addition, we present evidence supporting the<br />

involvement of USP3 and uH2A in the response to ionizing radiation. Our studies identify<br />

USP3 as a novel regulator of H2A and H2B ubiquitination, highlight its role in preventing<br />

replication stress and suggest its involvement in the response to DNA double strand<br />

breaks. Together our results implicate USP3 as a novel chromatin modifier in the<br />

maintenance of genome integrity.<br />

contact:<br />

MSc Joep Vissers<br />

Netherlands Cancer Institute<br />

j.vissers@nki.nl<br />

Plesmanlaan 121<br />

1066CX Amsterdam (Netherlands)<br />

additional information<br />

1 IFOM, Istituto FIRC di Oncologia Molecolare, 2 Istituto Europeo di Oncologia, Milan, Italy; 3 Erasmus<br />

MC, Rotterdam, and 4 The Netherlands Cancer Institute, Amsterdam, The Netherlands.


Andrea Tedeschi, Tuan Nguyen, Radhika Puttagunta, Perrine Gaub, Simone Di<br />

Giovanni<br />

Identification of a novel transcription module for axon<br />

outgrowth and regeneration<br />

Transcription plays an important role during neurite and axon outgrowth and<br />

regeneration.<br />

However, to date, no transcriptional complexes have been shown to control axon<br />

outgrowth and regeneration by regulating axon growth genes. Here, we investigate the<br />

role of the tumor suppressor p53 and the histone acetyltransferases CBP/p300 and<br />

P/CAF, which promote chromatin relaxation and promoter accessibility, on axon<br />

outgrowth and regeneration.<br />

We show that specifically p53 and CBP/p300 form a transcriptional complex that<br />

regulates the growth associated protein 43 (GAP-43), a key factor for axon outgrowth<br />

and regeneration, by occupying its promoter. Acetylated p53 at K372-3-82 promotes<br />

neurite outgrowth, and induces GAP-43 expression by binding specific elements on the<br />

neuronal GAP-43 promoter in a chromatin environment via CBP/p300 signaling.<br />

Importantly, in an axon regeneration model, CBP and p53 K372-3-82 are induced in<br />

axotomized facial motor neurons, where p53 K372-3-82 occupancy of GAP-43 promoter<br />

is enhanced as shown by in vivo chromatin immunoprecipitation. Finally, by comparing<br />

wt and p53 null mice, we demonstrate that the p53/GAP-43 transcriptional module is<br />

specifically switched on during axon regeneration in vivo.<br />

These data contribute to the understanding of gene regulation in axon outgrowth and<br />

may suggest new molecular targets for axon regeneration.<br />

contact:<br />

Simone Di Giovanni<br />

Univeristy of Tuebingen<br />

Hertie Institute for Clinical Brain Research<br />

simone.digiovanni@medizin.uni-tuebingen.de<br />

otfried mueller strasse 27<br />

76072 Tuebingen (Germany)


Japke Polman, E. Ronald de Kloet, Nicole Datson<br />

Identification of binding sites of the Glucocorticoid Receptor in<br />

the brain<br />

The Glucocorticoid Receptor (GR) is activated by the stress hormone cortisol, after which<br />

the hormone-receptor complex migrates to the nucleus where it functions as a<br />

transcription factor modulating the transcription of various target genes. Using Serial<br />

Analysis of Gene Expression (SAGE) and Affymetrix GeneChips our group has extensively<br />

characterised large-scale expression profiles of GR-regulated genes in vivo in rat<br />

hippocampus, ex vivo in rat hippocampal slices and in neuronally differentiated PC12<br />

cells (1-3). This has given clear insight into the genome-wide effects of the GR and the<br />

various cell functions it is involved in. However, it is not clear which of these genes are<br />

primary GR-targets mediated by direct GR binding to glucocorticoid response elements<br />

and which are downstream targets influenced by GR-induced pathways. The focus of the<br />

current project is to identify the primary gene-targets of GR using Serial Analysis of<br />

Chromatin Occupancy (SACO) in PC12 cells and in vivo in rat hippocampus. SACO<br />

combines Chromatin ImmunoPrecipitation (ChIP) and SAGE and enables large scale<br />

analysis of binding sites of the GR throughout the genome in an unbiased manner (4). In<br />

a later stage we will investigate in animal models of early life stress, known to induce<br />

long-lasting changes in stress-responsiveness, how the capability of the GR to bind to its<br />

primary targets is affected via epigenetic mechanisms.<br />

Literature<br />

1. Datson,N.A. et al. Eur.J. Neurosci. 14, 675-689 (2001).<br />

2. Morsink,M.C. et al. Journal of Neurochemistry 99, 1282-1298 (2006).<br />

3. Morsink,M.C. et al. Journal of Neuroendocrinology 18, 239-252 (2006).<br />

4. Impey,S. et al. Cell 119, 1041-1054 (2004).<br />

contact:<br />

MSc Japke Polman<br />

Division of Medical Pharmacology<br />

Leiden/Amsterdam Center for Drug Research, Leiden University Medical Centre<br />

jpolman@lacdr.leidenuniv.nl<br />

P.O. Box 9503<br />

2300 RA Leiden (the Netherlands)


Philipp Rathert, Arunkumar Dhayalan, Xing Zhang, Renata Jurkowska, Raluca Tamas,<br />

Yoichi Shinkai, Xiaodong Cheng, Albert Jeltsch<br />

Identification of new non-histone targets of the human G9a<br />

protein methyltransferase using peptide arrays<br />

Histone methylation is essential for gene regulation and chromatin dynamics. We<br />

employed methylation of peptide SPOT arrays comprising up to 420 different substrates<br />

per array to study the sequence specificity of the G9a histone methyltransferase, a<br />

major euchromatin-associated histone H3 lysine 9 methyltransferase. The enzyme<br />

mostly recognized the RK dipeptide sequence for lysine methylation. The activity of G9a<br />

is strongly inhibited at substrates methylated at H3R8, suggesting that methylation of<br />

H3R8 may control deposition of H3K9 methylation. Using the specificity profile derived<br />

for G9a, we identified several non-histone protein targets and showed G9a mediated<br />

methylation of five non-histone proteins as well as automethylation of G9a. The new<br />

targets are nuclear proteins known to participate in epigenetic signaling. We<br />

demonstrated potential downstream signaling pathways for methylation of non-histone<br />

proteins, because methylated peptides interacted with HP1ß in a methylation dependent<br />

manner in vitro.<br />

contact:<br />

Ph.D Philipp Rathert<br />

Jacobs University<br />

Biochemistry<br />

p.rathert@jacobs-university.de<br />

Campus Ring 1<br />

28759 Bremen (Germany)


Sylvia Erhardt, Craig M. Betts, Barbara G. Mellone, Gary H. Karpen, Aaron F. Straight<br />

Identification of novel regulators of centromeric chromatin by<br />

genome-wide RNAi screening<br />

The centromere is a specialized chromosomal site that is the structural and functional<br />

foundation for mitotic kinetochore formation. Kinetochores attach chromosomes to the<br />

mitotic spindle, monitor proper attachment through the mitotic checkpoint, and couple<br />

spindle and motor protein forces to move chromosomes in mitosis. How the site of<br />

centromere formation is specified (centromere identity) and propagated through multiple<br />

cell divisions remain unclear. Centromere formation is epigenetically regulated in<br />

metazoans, and the best candidate for an epigenetic mark that specifies centromere<br />

identity is the CENP-A family of centromere-specific histone H3 variants. CENP-A is<br />

essential for kinetochore assembly, chromosome attachment to the spindle, and<br />

chromosome segregation in all eukaryotes. Nearly all kinetochore proteins require CENP-<br />

A for their assembly, and CENP-A mislocalization to non-centromeric regions produces<br />

functional ectopic kinetochores. We identified factors required for centromere formation<br />

using genome-wide RNAi screening for defects in CENP-A (CID) localization in<br />

Drosophila. We found that centromere assembly is coupled to the cell cycle by Cyclin A<br />

and by the APC regulator RCA1/Emi1 through regulation of the CDH1/Fizzy-related<br />

activator of the Anaphase Promoting Complex (APC). We also identified a novel<br />

centromere protein CLD2 and the known Centromere Protein-C (CENP-C) as essential for<br />

the assembly of centromeric chromatin. CID and the factors we have identified are<br />

mutually dependent for centromere localization, and disruption of any one of these<br />

factors results in a loss of centromere function. Our findings identified essential<br />

components of the epigenetic machinery that ensure proper specification and<br />

propagation of the centromere, and suggest a mechanism for coordinating centromere<br />

duplication with cell division.<br />

contact:<br />

PhD Sylvia Erhardt<br />

Universität Heidelberg<br />

ZMBH<br />

s.erhardt@zmbh.uni-heidelberg.de<br />

Im Neuenheimer Feld 282<br />

69120 Heidelberg (Germany)


Bastian Stielow, Alexandra Sapetschnig, Imme Krüger, Michael Boutros, Guntram<br />

Suske<br />

Identification of SUMO-dependent chromatin-associated<br />

transcriptional repression components by a genome-wide RNA<br />

interference screen<br />

SUMO modification of many transcription factors is linked to transcriptional repression.<br />

The molecular mechanisms by which SUMO attachment represses transcription are<br />

largely unknown. Using the transcription factor Sp3 as a paradigm we have performed a<br />

genome-wide RNA interference screen in Drosophila melanogaster cells for components<br />

regulating and mediating SUMO-dependent transcriptional repression. Analysis of<br />

>21,000 double-stranded RNAs (dsRNAs) identified 120 genes whose dsRNA-mediated<br />

knockdowns impaired SUMO-dependent transcriptional repression. Several of these<br />

genes encode chromatin-associated proteins including the ATP-dependent chromatin<br />

remodeler dMi-2, the D. melanogaster ortholog of the C. elegans protein MEP-1 and the<br />

polycomb protein dSfmbt. Knockdown of these proteins did not impair SUMO conjugation<br />

demonstrating that they act downstream of SUMO attachment. Biochemical analyses<br />

revealed that dMEP-1, dMi-2 and dSfmbt interact with each other, bind to SUMO and are<br />

recruited to promoters in a SUMOylation-dependent manner. Our results suggest that<br />

dMEP-1, dMi-2 and dSfmbt are part of a common repression complex established by<br />

DNA-bound SUMO-modified transcription factors.<br />

This work was supported by a grant of the Deutsche Forschungsgemeinschaft to G.S.<br />

Literature<br />

Sapetschnig, A., Rischitor, G., Braun, H., Doll, A., Schergaut, M., Melchior, F. and Suske,<br />

G. (2002). Transcription factor Sp3 is silenced through SUMO modification by PIAS1.<br />

EMBO J. 21, 5206-5215.<br />

Stielow, B., Sapetschnig, A., Krüger, I., Kunert, N., Brehm, A., Boutros, M., and Suske,<br />

G. (2008). Identification of SUMO-dependent chromatin-associated transcriptional<br />

repression components by a genome-wide RNA interference screen. Mol. Cell, in press.<br />

contact:<br />

Prof. Dr. Guntram Suske<br />

Philipps-Universität Marburg<br />

Institut für Molekularbiologie und Tumorforschung (IMT)<br />

Suske@imt.uni-marburg.de<br />

Emil-Mannkopff-Str. 2<br />

D-35032 Marburg (Germany)


Harriet Wikman, Michaela Kraemling, Dirk Kemming, Klaus Pantel<br />

Identification of Target Genes in Micrometastatic Lung Cancer<br />

by Methylation Arrays<br />

We have recently identified specific molecular patterns associated with the presence of<br />

disseminated tumor cells (DTCs) in the bone marrow (BM) in patients with primary early<br />

stage lung cancer. We performed a combined expression and copy number (CGH)<br />

profiling of primary lung tumors, and detected five chromosomal regions differentiating<br />

BM-negative from BM-positive patients. Heterozygotic loss of chromosome 4q12-q32 in<br />

BM-positive patients was the most prominent finding. The same loss was also found to<br />

be common in brain metastases from lung cancer patients. The 4q region spanned over<br />

107.1 Mbp and contained 73 differentially expressed genes. In order to narrow down the<br />

potential target gene in this region, a methylation array-screening was performed. 3<br />

lung cancer cell lines showing loss of 4q were treated with 5-aza-2' deoxycytidine, a<br />

demethylating agent. By comparing the mRNA expression of treated and non-treated cell<br />

line one can identify genes silenced through methylation. As a control for cellular stress<br />

a normal bronchial epithelial cell line was used. Treatments were done in triplicate and<br />

RNA was pooled before the competitive hybridization on Agilent 4x44K arrays. All<br />

together 620 genes were found 2-fold up-regulated in at least two of the cancer cell lines<br />

but not in the control cell line. Of these genes 12 are located to the 4q12-q32. 5 genes<br />

were excluded as no expression could be detected in normal lung tissue or no differential<br />

expression could be found between normal and tumor tissue. We are currently mapping<br />

the methylation sites of the 7 remaining potential target genes and will perform MSI-PCR<br />

on primary lung tumors in order to verify the findings and map down the possible target<br />

gene on 4q responsible for the early micrometastatic spread of lung cancer.<br />

contact:<br />

Dr Harriet Wikman<br />

University Medical Center Hamburg-Eppendorf<br />

Intst. Tumor Biology<br />

h.wikman@uke.uni-hamburg.de<br />

Martinistrasse 52<br />

20246 Hamburg (Germany)


Isabelle GUILLERET, Maria-Chiara OSTERHELD, Richard BRAUNSCHWEIG, Véronique<br />

GASTINEAU, Suzanne TAILLENS<br />

Imprinting of tumor-suppressor genes in human placenta.<br />

Transcriptional deregulation in cancer has been shown to be associated with epigenetic<br />

alterations, in particular in tumor-suppressor-gene (TSG) promoters. In contrast, DNA<br />

methylation in TSG is absent in normal differentiated cells. Nevertheless, we previously<br />

showed that the promoter of the tumor-suppressor gene APC was methylated on one<br />

allele only, in normal gastric cells (1). Recently, RASSF1A has been shown to be<br />

imprinted in normal human placenta (2). To clarify putative TSG methylation in normal<br />

tissues, 23 placenta at the first trimester, on both decidua and villi, and 4 normal nongestational<br />

endometrium were screened for DNA methylation by methylation-sensitive<br />

single-strand conformation analysis (MS-SSCA) and sequencing after bisulfite<br />

modification, on a panel of 12 genes known to be implicated in carcinogenesis. In all<br />

placental villi, 4 promoter TSG genes - APC, SFRP2, RASSF1A and WIF1 - were<br />

hypermethylated, whereas all decidua and normal endometrium did not show any<br />

methylation. Allele-specific methylation analysis revealed that this methylation was<br />

monoallelic. Furthermore, the comparison with maternal DNA indicated that APC and<br />

WIF1 were methylated on the maternal allele, whereas SFRP2 was found methylated on<br />

the paternal allele. Imprinting status of these 4 genes is conserved during pregnancy.<br />

Sequence analysis of WIF1 mRNA revealed that only the unmethylated paternal allele<br />

was transcribed. These results indicate that TSG imprinting is pre-existent in normal<br />

human placenta and should not be confused with carcinogenesis or pathology-induced<br />

methylation.<br />

Literature<br />

1. Clément G, Bosman FT, Fontolliet C, Benhattar J. Monoallelic methylation of the APC<br />

promoter is altered in normal gastric mucosa associated with neoplastic lesions. Cancer<br />

Res. 2004, 64(19):6867-73.<br />

2. Chiu RW, Chim SS, Wong IH, Wong CS, Lee WS, To KF, Tong JH, Yuen RK, Shum AS,<br />

Chan JK, Chan LY, Yuen JW, Tong YK, Weier JF, Ferlatte C, Leung TN, Lau TK, Lo KW, Lo<br />

YM. Hypermethylation of RASSF1A in human and rhesus placentas. Am J Pathol. 2007,<br />

170(3):941-50.<br />

contact:<br />

Dr. Isabelle GUILLERET<br />

University of Lausanne - CHUV<br />

University Institute of Pathology - CHUV<br />

isabelle.guilleret@chuv.ch<br />

Rue du Bugnon 25<br />

1011 Lausanne (Switzerland)


Alexandre Ceccaldi, Dominique Guianvarc'h, Catherine Senamaud-Beaufort, Renata<br />

Jurkowska, Daniel Dauzonne, Albert Jeltsch, Paola B Arimondo<br />

In quest of DNMT inhibitors<br />

not submitted<br />

contact:<br />

Dr Paola B Arimondo<br />

CNRS<br />

UMR5153 INSERM U565 MNHN USM503<br />

arimondo@mnhn.fr<br />

43 rue Cuvier<br />

75005 Paris (France)<br />

additional information<br />

Alexandre Ceccaldi 1, Dominique Guianvarc'h 2, Catherine Senamaud-Beaufort 1, Renata Jurkowska<br />

3, Daniel Dauzonne 4, Albert Jeltsch 3, Paola B. Arimondo 1<br />

1UMR5153 CNRS-MNHN USM503; INSERM U565, 43 rue Cuvier 75005 Paris France; email:<br />

arimondo@mnhn.fr<br />

2UMR7613 CNRS-UPMC, Université Pierre et Marie Curie, boîte 182, 4, place Jussieu, 75005 Paris,<br />

France<br />

3Jacobs University Bremen, Campus Ring 1, 28759 Bremen, Germany<br />

4UMR 176 CNRS Institut Curie, 26 rue d'Ulm, 75005 Paris, France


Careen Katryniok, Bernd L. Sorg, Dieter Steinhilber<br />

INDUCTION OF HUMAN 5-LIPOXYGENASE GENE EXPRESSION<br />

BY THE HISTONE DEACETYLASE INHIBITOR TRICHOSTATIN A -<br />

INVESTIGATIONS ON THE MECHANISM<br />

5-lipoxygenase (5-LO) is the key enzyme in the formation of leukotrienes which are<br />

important inflammatory mediators. The 5-LO promoter lacks a TATA or CCAT box, but<br />

posseses a unique GC-rich sequence (1). We have shown previously that epigenetic<br />

mechanisms play a role in the transcription of 5-lipoxygenase. The 5-LO transcription is<br />

silenced by DNA methylation (2,3) and activated by treatment with histone desacetylase<br />

inhibitior trichostatin A (TsA) (4).<br />

In this study, we continued the investigations concerning the activation of the 5-LO<br />

promoter by TsA. We explored the time dependency of the effect of TsA by rt-PCR and<br />

achieved the maximal effect 8-16 hours after incubation. Subsequently we analysed<br />

changes in the histone modifications of three different cell lines after TsA incubation, two<br />

of which are transcriptionally active (HL-60 and MM6 cells), whereas the third cell line is<br />

methylated and silenced (U937). We could discover differences between this cell lines in<br />

the acetylation pattern of the core histone proteins H3 and H4 using the chromatin<br />

immunoprecipitation assay. In TsA treated HL-60 and, surprisingly in U937 cells, we<br />

could detect an increase of acetylated Histon H3 and H4 with a maximum after 8-16<br />

hours. Interestingly, no change in histone acetylation status is seen in the cell line MM6.<br />

It looks as if TsA does not act via the classical mechanism, which is increase of<br />

acetylation state of histone proteins, in MM6 cells.<br />

Literature<br />

1. Hoshiko, S., Rådmark, O., and Samuelsson, B. (1990) Proceedings of the National<br />

Academy of Sciences of the United States of America 87, 9073-9077<br />

2. Uhl, J., Klan, N., Rose, M., Entian, K. D., Werz, O., and Steinhilber, D. (2002) Journal<br />

of Biological Chemistry 277, 4374-4379<br />

3. Uhl, J., Klan, N., Rose, M., Entian, K. D., Werz, O., and Steinhilber, D. (2003)<br />

Advances in Experimental Medicine & Biology 525, 169-172<br />

4. Klan, N., Seuter, S., Schnur, N., Jung, M., and Steinhilber, D. (2003) Biological<br />

Chemistry 384, 777-785<br />

contact:<br />

Careen Katryniok<br />

University of Frankfurt<br />

Institute of Pharmaceutical Chemistry<br />

Katryniok@pharmchem.uni-frankfurt.de<br />

Max-von-Laue-Str. 9<br />

60438 Frankfurt (Germany)


Daniela Kremer, Wolfgang Schulz, Victoria Kolb-Bachofen<br />

iNOS-generated NO plasy an critical role in DNA-methylation<br />

Methylation profiles are subject to alterations during cellular life cycles and hypo- as well<br />

as hypermethylation are hallmarks of cancer development. Increasing evidence points to<br />

chronic inflammation as a key player in the regulation of DNA methylation. Chronic<br />

inflammation is characterized by a formation of proinflammatory cytokines, which leads<br />

to the expression of the inducible nitric oxide synthase (NOS2) and high-output nitric<br />

oxide (NO) synthesis. The regulation of the NOS2 in human cells is highly complex and<br />

so far only one publication suggests that it is also involved in epigenetic control.<br />

We used the cell line A549iNOS (human lung-epithelial cell line), stably transfected with<br />

the NOS2 promoter in front of luciferase. The cells were treated with 2-aza-5’deoxycytidine<br />

(Aza-CdR) and trichostatin A (TSA) and activated with cytokines (IL-1•,<br />

TNF-•, IFN-•).<br />

Treatment with Aza-CdR/TSA and simultaneous cytokine activation (48h) leads to<br />

moderate but significant increases in NOS2 expression (2, 5 • above cytokines only).<br />

Addition of NIO, an inhibitor of NO formation, results in a dramatic increase in promoter<br />

activity (6 times), in mRNA level (27 times) and also in enzyme activity.<br />

If instead an NO-Donor was added, NOS2 expression was completely shut down, despite<br />

the continued Aza-CdR/TSA treatment.<br />

We then have used an ELISA-based assay to assess whole genome methylation. The<br />

whole genome methylation status of A549iNOS (100%) is decreased to 25% by the Aza-<br />

CdR/TSA treatment, is completely reversed during the cytokine activation period, but<br />

stays low in the presence of NIO and is fully reverted in the additional presence of a<br />

chemical NO-donor.<br />

In summary, we here find a feedback-mechanism, where de-methylation of the iNOS<br />

promoter leads to an increase of its expression following increased production of NO,<br />

which in turn causes re-methylation which then stops or limits iNOS expression.<br />

These processes may play a significant role in chronic situations with aberrant iNOS<br />

activity as occurs for instance during substrate restriction e.g. in psoriasis and asthma or<br />

BH4 deficiency e.g. in megaloblastic anemia.<br />

contact:<br />

Daniela Kremer<br />

Heinrich-Heine University Düsseldorf<br />

Research Group Immunobiology<br />

daniela.kremer@uni-duesseldorf.de<br />

Universitätsstrasse 1<br />

40225 Düsseldorf (Germany)


Nathalie Jurisch, Bjoern Textor, Peter Angel, Marina Schorpp-Kistner<br />

Involvement of JunB in post-translational HDAC6 regulation<br />

and chromatin remodelling<br />

The AP-1 transcription factor, consisting of homo- or heterodimers between the<br />

members of the Jun and Fos protein families, is implicated in a number of biological<br />

processes such as cellular differentiation, cell cycle progression, apoptosis and<br />

tumorigenesis. Within the AP-1 family, JunB has an exceptional position since it has<br />

been early demonstrated to function as a repressor on a variety of AP-1 target genes.<br />

Moreover, studies on conventional and conditional junB knock-out mice have revealed<br />

both positive and negative functions for JunB in influencing the control of cell<br />

proliferation, apoptosis and differentiation. Since microarray analysis revealed that JunB<br />

can repress many different targets genes and only activate a few ones at the same time<br />

in the same cell, one could speculate whether JunB exerts its specific regulatory function<br />

via chromatin remodelling.<br />

In order to investigate the putative role of JunB in epigenetics, we first screened the<br />

expression of histones deacetylases (HDACs) as well as the expression of some<br />

scaffolding proteins targeting these enzymes to the DNA. JunB deficient primary and<br />

immortalised mouse embryonic fibroblasts showed a decreased level of the histone<br />

deacetylase HDAC6. Surprisingly, this effect was not due to transcriptional regulation by<br />

JunB but rather due to protein instability in absence of JunB. Current studies address<br />

first the consequences of the decreased HDAC6 expression on gene de-repression and<br />

second the molecular mechanism responsible for HDAC6 instability in the JunB-deficient<br />

fibroblasts.<br />

The final goal of these studies will be to determine the genetic network driven by JunB<br />

and HDAC6 that controls cell identity and function in normal development but also in<br />

disease such as inflammation-associated disorders and cancer.<br />

contact:<br />

Nathalie Jurisch<br />

German Cancer Research Centre<br />

n.jurisch@dkfz.de<br />

Im Neuenheimer Feld 280<br />

69120 Heidelberg (Germany)


Annette Scharf, Karin Meier, Volker Seitz, Alexander Brehm, Axel Imhof<br />

Kinetics of histone modifications during in vitro chromatin<br />

assembly<br />

Histone modifications play a crucial role in the establishment and maintenance of gene<br />

expression patterns. In order to faithfully maintain a particular set of modifications,<br />

mechanisms must exist that allow the modification of the newly deposited histones<br />

according to the pre-existing pattern. In order to analyse the changes of histone<br />

modifications during chromatin assembly and maturation, we used an in vitro chromatin<br />

assembly system from Drosophila embryo extracts. In accordance with what has been<br />

observed in vivo, we find a deacetylation of the initially diacetylated isoform of histone<br />

H4, which is dependent on chromatin assembly. Immediately after deposition of the<br />

histones onto DNA H4 is monomethylated at K20, which is required for an efficient<br />

deacetylation of the H4 molecule. K20 methylation dependent l(3)MBT association with<br />

chromatin and the identification of a l(3)MBT-dRPD3 complex suggest that a deacetylase<br />

is specifically recruited to the monomethylated substrate through interaction with<br />

l(3)MBT. Moreover, the monomethylation of K20 also facilitates the release of members<br />

of the chromatin assembly factor CAF1 from the newly assembled chromatin. Our data<br />

suggest that an ordered appearance and disappearance of histone modification marks<br />

occurs at newly assembled chromatin and regulates the maturation of chromatin.<br />

contact:<br />

Annette Scharf<br />

Ludwig Maximilians Universität<br />

Adolf Butenandt Institut<br />

annette.scharf@med.uni-muenchen.de<br />

Schillerstrasse 44<br />

80336 München (Germany)


Fabio Mohn, Michael Weber, Michael Rebhan, Tim Roloff, Jens Richter, Michael Stadler,<br />

Miriam Bibel, Dirk Schübeler<br />

Lineage-specific Polycomb targets and de novo DNA<br />

methylation define restriction and potential of neuronal<br />

progenitors<br />

Reduction of cellular potency during development is thought to employ epigenetic<br />

restriction. We have used a murine neuronal differentiation system that progresses from<br />

embryonic stem cells to lineage-committed progenitors and further to postmitotic,<br />

terminally differentiated neurons to identify genes targeted by two repressive epigenetic<br />

pathways: DNA methylation and Polycomb-mediated methylation of histone H3<br />

(H3K27me3).<br />

We show that CpG-rich promoters are unmethylated in stem cells, yet several hundred<br />

become DNA methylated in lineage-committed progenitor cells with no further<br />

methylation during terminal differentiation. Targeted promoters control pluripotency and<br />

germline-specific genes, suggesting a role for DNA methylation in stabilizing loss of<br />

pluripotency.<br />

Conversely, we detect acquisition and loss of H3K27me3 at novel targets at both<br />

progenitor and terminal state. Surprisingly, neuron-specific genes that are poised to be<br />

activated upon further terminal differentiation, become Polycomb targets in progenitor<br />

cells. Moreover, the presence of H3K27me3 in stem cells primes for differentiationcoupled<br />

DNA methylation, suggesting context-dependent crosstalk between Polycomb<br />

and DNA methylation.<br />

Our analysis illustrates that de novo DNA methylation and dynamic switches in Polycomb<br />

targets cooperate to establish epigenetic states specific for restricting pluripotency and<br />

defining the developmental potential of progenitor cells.<br />

contact:<br />

Fabio Mohn<br />

Friedrich Miescher Institute for Biomedical Research<br />

fabio.mohn@fmi.ch<br />

Maulbeerstrasse 66<br />

4058 Basel (Switzerland)<br />

additional information<br />

Address affiliation of Miriam Bibel and Jens Richter:<br />

Novartis Institutes for Biomedical Research, Neurodegeneration Department, 4002 Basel,<br />

Switzerland


Maciej Meglicki, Marta Teperek, Ewa Borsuk<br />

Localization of heterochromatin protein 1α during mouse<br />

oogenesis and early embryonic development<br />

Proper organization of pericentric heterochromatin is vital for proper chromosome<br />

segregation and for the maintenance of genome stability in eukaryotic cells. The<br />

structure of pericentric heterochromatin depends mainly on presence of certain histone<br />

variants, non-histone proteins and specific pattern of epigenetic modifications of core<br />

histones. One of the non-histone proteins, which is thought to be indispensable for the<br />

formation and maintenance of heterochromatin, is HP1 (heterochromatin protein 1). Two<br />

isoforms of HP1, HP1α and HP1β, have been found at pericentric regions of somatic cells<br />

at interphase. In these cells, HP1β and majority of HP1α dissociate into the cytoplasm<br />

before prometaphase and re-associate with pericentric regions at the end of mitosis.<br />

Localization of HP1α in mouse oocytes and early embryos was unknown.<br />

Here we show localization of HP1α protein during oogenesis and in cleaving mouse<br />

embryos. HP1α was not detected in primordial oocytes. It appeared in pericentric<br />

heterochromatin at the beginning of the growth phase. Subsequently, it dissociated from<br />

these regions in fully-grown oocytes during cessation of transcription and was absent in<br />

maturing and ovulated oocytes. It was not observed in zygotes, but reappeared in 2-cell<br />

embryos. At this stage and in 4- and 8-cell embryos, the association of HP1α with<br />

pericentric heterochromatin was restricted to defined stages of the interphase (S/G2).<br />

Literature<br />

Dormann et al., 2006. Dynamic Regulation of Effector Protein Binding to Histone<br />

Modifications. Cell Cycle 5: 2842-51<br />

Garagna et al., 2004. Three-dimensional localization and dynamics of centromeres in<br />

mouse oocytes during folliculogenesis. J Mol Histol 35: 631-638<br />

Martin et al., 2006. Genome restructuring in mouse embryos during reprogramming and<br />

early development. Dev Biol 292: 317-332<br />

van der Heijden et al., 2005. Asymmetry in histone H3 variants and lysine methylation<br />

between paternal and maternal chromatin of the early mouse zygote. Mech Dev<br />

122:1008-1022<br />

contact:<br />

M.Sc Maciej Meglicki<br />

University of Warsaw<br />

Zoology<br />

mmeglicki@biol.uw.edu.pl<br />

ul. Przy Agorze 28<br />

01-930 Warsaw (Poland)


Christian Schmidl, Maja Klug, Tina Böld, Petra Hoffmann, Matthias Edinger, Michael<br />

Rehli<br />

Locus-wide detection of cell type specific DNA methylation<br />

patterns using comparative methyl-CpG-Immunoprecipitation<br />

(MCIp)<br />

DNA-methylation is a vital epigenetic mark. It participates in establishing and<br />

maintaining chromatin structures and regulates gene transcription during mammalian<br />

development and cellular differentiation. The extent, function and regulation of tissue- or<br />

cell type-specific DNA-methylation, however, is largely unknown. Here we present a<br />

locus-wide DNA-methylation analysis of CD4+CD25+ regulatory T-cells (Treg) and<br />

conventional CD4+CD25- T-cells (Tconv). Comparative methylation profiling was<br />

performed by fractionation of genomic DNA in hyper- and hypomethylated subsets using<br />

methyl-CpG-immuno precipitation (MCIp) followed by microarray hybridisation and<br />

combined analysis. In total, 69 genomic regions (124 genes and covering 12 Mb of the<br />

human genome) that were selected based on differential gene expression in both cell<br />

types, were analysed for differentially methylated areas. Microarray results were<br />

independently validated using qPCR and/or bisulfite sequencing. In total, we detected<br />

approximately 130 locations that were specifically demethylated in one or the other cell<br />

type (appr. 100 in Treg, 30 in Tconv). Such areas are detected in almost 2/3rds of the<br />

analysed regions, particularly in cell-type specific genes like FOXP3, IL2RA and IKZF2 in<br />

Tregs or CD40LG and IFNG in Tconv. Interestingly, the majority of cell-type specifically<br />

demethylated areas overlap with evolutionary conserved sequences suggesting<br />

regulatory functions for these areas. Our pilot study demonstrates the feasibility of our<br />

approach for exploring genome-wide methylation differences in normal cells and<br />

provides a first comprehensive, locus-wide analysis of cell type-specific methylation<br />

patterns in regulatory T-cells and conventional CD4+ T-cells.<br />

contact:<br />

Christian Schmidl<br />

Uniklinikum Regensburg<br />

Hämatologie/Onkologie<br />

christian.schmidl@klinik.uni-regensburg.de<br />

Franz-Josef-Strauss Allee 11<br />

93053 Regensurg (Deutschland)


Nicole Happel, Stefan Stoldt, Detlef Doenecke<br />

M-phase specific phosphorylation of histone H1.5 at threonine<br />

10 by GSK3<br />

H1 histones are progressively phosphorylated during the cell cycle. The number of<br />

phosphorylated sites increases from zero to three in late S-phase up to five or six in late<br />

G2 and M-phase. It is assumed that this phosphorylation modulates chromatin<br />

condensation and decondesation but until now its specific role remains unclear. Recently<br />

it was shown by mass spectrometry that the subtype H1.5 becomes<br />

pentaphosphorylated during mitosis, with phosphorylated threonine 10 (T10) being the<br />

last phosphorylation site to occur.<br />

We have generated an antiserum specific for H1.5 phosphorylated at T10.<br />

Immunofluorescence labelling of HeLa cells with this antiserum revealed that the<br />

phosphorylation at this site appears in prophase and disappears in telophase, and that<br />

this hyperphosphorylated form of H1.5 is chromatin bound in metaphase, when the<br />

chromatin condensation is maximal. In search of the kinase responsible for the<br />

phosphorylation at this site, we found that T10 can not be phosphorylated by<br />

CDK1/CyclinB and CDK5/p35, respectively, but by GSK3 in vitro. Furthermore, addition<br />

of specific GSK3 inhibitors led to a reduction of the phosphorylation at this site both in<br />

vivo and in vitro.<br />

contact:<br />

Dr. Nicole Happel<br />

University of Göttingen<br />

Institute for Biochemistry and Molecular Cell Biology<br />

nhappel@gwdg.de<br />

Humboldtallee 23<br />

37073 Göttingen (Germany)


Stephanie Jungmichel, Christoph Spycher, Manuel Stucki<br />

Mechanism of MDC1 dimerization<br />

In response to DNA double strand breaks (DSB), numerous proteins engage in DNA<br />

repair and DNA-damage checkpoint signaling pathways. In mammalian cells extensive<br />

phosphorylation of the histone variant H2AX by members of the PIKK family is the key<br />

regulatory processes of IR-induced nuclear foci formation [1]. This modification is<br />

followed by accumulation of DNA damage response (DDR) factors in microscopically<br />

discernible nuclear foci with an emerging role of mediator proteins, that may serve as<br />

landing platforms for the efficient recruitment of other DDR factors to sites of DSBs.<br />

MDC1 (mediator of DNA damage checkpoint 1) directly binds phosphorylated H2AX via<br />

its C-terminal tandem BRCT-domain thereby acting as a bridging factor between<br />

damaged chromosomes and other DDR proteins [2].<br />

Besides BRCT domains, MDC1 also features a FHA domain at its N-terminus. The precise<br />

role of this phosphopeptide-interacting FHA domain however has not been understood so<br />

far. We recently identified a novel phospho-specific binding partner of the MDC1 FHA<br />

domain. Surprisingly, this binding partner is MDC1 itself. We showed that a conserved<br />

threonine residue close to the N-terminus of MDC1 becomes phosphorylated by ATM in<br />

vitro and in vivo. When phosphorylated, the MDC1 N-terminus interacts in trans with the<br />

FHA domain of another MDC1 molecule. Thus, we hypothesize that the MDC1 FHA<br />

domain functions to promote homodimerization of MDC1 which may help to regulate the<br />

cellular response to DSBs.<br />

Literature<br />

[1] Rogakou, E.P., et al., DNA double-stranded breaks induce histone H2AX<br />

phosphorylation on serine 139. J Biol Chem, 1998. 273(10): p. 5858-68.<br />

[2] Stucki, M., et al., MDC1 directly binds phosphorylated histone H2AX to regulate<br />

cellular responses to DNA double-strand breaks. Cell, 2005. 123(7): p. 1213-26.<br />

contact:<br />

Dipl. Biochemikerin Stephanie Jungmichel<br />

University of Zuerich<br />

Institute of Veterinary Biochemistry and Molecular Biology<br />

jung@vetbio.uzh.ch<br />

Winterthurerstrasse 190<br />

8057 Zuerich (Switzerland)


Gunter Reuter, Thomas Rudolph, Sandro Lein, Matthias Walther, Heiko Baisch, Sameer<br />

Phalke, Christian Apelt, Sandy Mietsch<br />

Mechanisms of chromatin differentiation during early<br />

embryogenesis of Drosophila<br />

Cleavage chromatin is transcriptional inactive and indexed by specific histone<br />

modification marks. A chromatin complex containing the demethylase SU(VAR)3-3, the<br />

DmWHSC1 methyltransferase and the SU(VAR)2-1 protein, a novel key factor of<br />

chromatin regulation, controls transcriptional quiescence in cleavage and primordial<br />

germ line nuclei. Differentiation of euchromatin and heterochromatin is controlled after<br />

cleavage when syncytial blastoderm shows an apico-basal polarity by two chromatin<br />

complexes containing either the SU(VAR)3-3 or the LID demethylase. Mutant analysis<br />

revealed coordinated function of both complexes in defining the balance between eu-<br />

and heterochromatin. Heterochromatic gene silencing in PEV becomes established<br />

already in cycle 10-12 embryos and is afterwards stably maintained. An alternative DNA<br />

methylation dependent pathway controls retrotransposon silencing. Genetic and<br />

molecular analysis revealed that in early embryogenesis silencing of retrotransposons is<br />

initiated by Dnmt2 dependent DNA methylation of LTR sequences. Later in<br />

embryogenesis no DNA methylation is found and silencing is maintained by SUV4-20<br />

dependent H4K20 trimethylation. Enzyme complexes, which define heterochromatin in<br />

somatic cells, also function in control of transcriptional quiescence in germ line cells.<br />

Molecular analysis of new Su(var) genes furthermore allowed identification of the<br />

maternal signals which initiate differentiation of euchromatin and heterochromatin.<br />

contact:<br />

Prof. Dr. Gunter Reuter<br />

Martin Luther University Halle-Wittenberg<br />

Biology/Genetics<br />

reuter@genetik.uni-halle.de<br />

Weinbergweg 10<br />

06120 Halle/Saale (D)


Anette Tippelt<br />

Methyl Primer Express® Software and the Influence of<br />

Amplicon Characteristics to the success rate in DNA<br />

Sequencing of bis treated gDNA<br />

A well known method to study methylation patterns is to treat gDNA with sodium<br />

bisulfite to distinguish methylated cytosines (5mC) from unmethylated onse. During this<br />

treatment Cytosines (C) are deaminated to uracils (U) and replaced by thymine (T) in<br />

subsequent PCR amplifications. 5mC however remains unchanged. The base composition<br />

undergoes a dramatic change during this process. Originally complementary DNA<br />

strands have no complementary counterpart after this chemical interaction. This must be<br />

taken into consideration during primer and amplicon design for MSP or BSP reactions.<br />

A new PC based software called “Methyl Primer Express®” significantly facilitates the<br />

primer design process. It highlights CpGs islands and converts gDNA into a bisulfite<br />

treated sequence. Custom primer design settings for BSP or MSP PCR are applicable. In<br />

addition the base composition of a final amplicon is presented to reveal homopolymer<br />

stretches. This information is crucial for subsequent sequencing.<br />

The software is available free from the web: https://www.appliedbiosystems.com<br />

In this <strong>poster</strong> we present tools and tips for robust primer and amplicon design for a BSP<br />

PCR based approach and illustrate examples for DNA sequencing.<br />

contact:<br />

Dr. Anette Tippelt<br />

Applied Biosystems<br />

anette.tippelt@eur.appliedbiosystems .com<br />

Scheefstrasse 64<br />

72074 Tübingen (Germany)


Sonja Röhrs, Julia Romani, Wilhelm Dirks, Hans G. Drexler, Hilmar Quentmeier<br />

Methylation profiles of tumour suppressor genes in Hodgkin<br />

and non-Hodgkin lymphoma cell lines<br />

Epigenetic inactivation of tumour suppressor genes (TSGs) by promoter CpG island<br />

methylation is frequently observed in neoplasia. It is generally agreed, that CpG island<br />

hypermethylation profiles are specific for different tumour types. Therefore, the<br />

methylation patterns of TSGs might prove useful in cancer diagnosis and indicative of<br />

cellular drug responses. Here, we set out to elucidate whether the methylation profile of<br />

TSGs would allow the classification of diverse lymphoma entities. We analysed the<br />

methylation status of 24 different TSGs in combination with copy number changes.<br />

Thirty-nine lymphoma cell lines were tested, representing Hodgkin lymphoma plus five<br />

distinct subtypes of non-Hodgkin lymphoma. For this approach we used a methylationspecific<br />

MLPA (Multiplex Ligation-dependent Probe Amplification) assay. Using<br />

quantitative real time PCR, we confirmed transcriptional silencing of hypermethylated<br />

TSGs, discovered by MLPA. We identified a group of TSGs, generally methylated or<br />

deleted in all analysed lymphoma cell lines (e.g. CDH13, IGSF4, RARbeta). We also<br />

found several TSGs, that were preferentially methylated in specific lymphoma subtypes<br />

(e.g. CD44, CHFR, RASSF1A). Our results support methylation studies (e.g. for RARbeta<br />

and RASSF1A) performed on primary lymphoma patient material 1,2 , confirming the<br />

applicability of cell lines as model systems. This screening method, thus revealed<br />

potential novel epigenetic markers for Hodgkin and non-Hodgkin lymphoma.<br />

Literature<br />

1 Murray P. G., Qiu G. H., Fu L. et al. (2004) Frequent epigenetic inactivation of the<br />

RASSF1A tumor suppressor gene in Hodgkin´s lymphoma. Oncogene 23 (6): 1326-31.<br />

2 Shi H., Guo J., Duff D. J., Rahmatpanah F. et al. (2007) Discovery of novel epigenetic<br />

markers in non-Hodgkin´s lymphoma. Carcinogenesis 28, 1: 60-70.<br />

contact:<br />

Dr. Sonja Röhrs<br />

DSMZ - Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH<br />

Department of human and animal cell lines<br />

sro07@dsmz.de<br />

Inhoffenstr. 7B<br />

38124 Braunschweig (Germany)


Aditi Kanhere, Vingron Martin, Haas Stefan<br />

Methylation status of promoter depends on its CpG content<br />

Methylation of cytosine residue in CpG dinucleotides is a frequent epigenetic modification<br />

which influences gene expression. Around 70% of CpG sites in human genome are<br />

methylated. The human genome is generally depleted of CpG dinucleotides except for<br />

short stretches of DNA known as CpG islands. In contrast to bulk DNA, the CpG islands<br />

are generally methylation-free. On the other hand, methylation of CpG island is<br />

important in X-chromosomal inactivation, gene imprinting and cancer related abberant<br />

gene expression. Although non-CpG island associated CpG methylation plays an<br />

important role in tissue-specific gene regulation, CpG island methylation is also reported<br />

in testis specific gene expression. Hence, analysis of methylation vis-a-vis CpG<br />

distribution and tissue-specific gene expression is very important.<br />

In this study we utilized methylation profiles of chromosomes 6, 21 and 22 collected on<br />

different tissues (Eckhardt et al. 2006 Nature Genetics 1359-60). We correlated the<br />

methylation profile with observed gene expression. We also calculated the Normalized<br />

CpG frequency in the corresponding promoters.<br />

We observe that gene expression is inversely correlated with methylation density of a<br />

promoter. While promoters with high CpG content are generally non-methylated, those<br />

with low CpG content show higher levels of methylation. There is higher variance in<br />

methylation status of low CpG promoters in different tissues as compared to methylation<br />

status of high CpG promoters. Additionally we also find higher variance in expression of<br />

low CpG promoters as compared to high CpG promoters.<br />

Our results support previous observations that high CpG promoters which are associated<br />

with house-keeping genes are generally unmethylated. The high variance in methylation<br />

status as well as gene expression in low CpG promoter indicate possible involvment in<br />

tissue-specific gene expression. CpG density of promoter is thus an important factor in<br />

regulation of gene expression.<br />

Literature<br />

Eckhardt F, Lewin J, Cortese R, Rakyan VK, Attwood J,et al.(2006)DNA methylation<br />

profiling of human chromosomes 6, 20 and 22. Nat Genet.38,1378-85.<br />

Saxonov S, Berg P, Brutlag DL.(2006) A genome-wide analysis of CpG dinucleotides in<br />

the human genome distinguishes two distinct classes of promoters.<br />

Proc Natl Acad Sci U S A. 103,1412-7.<br />

contact:<br />

Dr Aditi Kanhere<br />

Max Planck Institute for Molecular Genetics<br />

kanhere@molgen.mpg.de<br />

Ihnestr 63-73<br />

14195 Berlin (Germany)


Szabolcs Sörös, Wolfgang Fischle<br />

Molecular insights into HP1-chromatin interaction<br />

Heterochromatin protein 1 (HP1) was initially identified as a non-histone component of<br />

chromatin that predominantly localizes to pericentromers [1] . In vivo colocalization of HP1<br />

with H3K9me3 in heterochromatic regions is dependent on both, the chromo-domain and<br />

a functional chromo-shadow-domain [2] . Systematic examination of Drosophila mutants<br />

and various in vivo experiments implicate HP1 in formation and propagation of a<br />

heterochromatic environment defined by tight chromatin structures and transcriptional<br />

silencing [3, 4] . However, on a molecular level, very little is known about how HP1<br />

mediates heterochomatinization.<br />

Here, we present a well-defined in vitro oligonucleosome system that allows precise<br />

characterization of both, the nature of HP1 association with chromatin and the<br />

consequences of HP1 recruitment for chromatin structure. Our data show that binding of<br />

HP1 to recombinant oligonucleosomes is predominantly determined by the H3K9<br />

methylation status. Very alike to the in vivo situation, the association is mediated by<br />

both, the HP1 chromo-domain and the chromo-shadow-domain. Interestingly, binding is<br />

enhanced under ionic conditions that lead to a condensed 30 nm-like oligonucleosome<br />

structure. Using biochemical and biophysical approaches like dynamic light scattering,<br />

and atomic force microscopy we examined the impact of HP1 on chromatin structure.<br />

Our results suggest that HP1 mediates a dramatic change in conformation and<br />

association of oligonucleosomal arrays. This effect is strongly dependent on the<br />

methylation status of H3K9, the HP1 chromo-domain, and its chromo-shadow-domain.<br />

Our findings manifest HP1 as a key effector in establishing higher chromatin<br />

condensation states in heterochromatic regions.<br />

Literature<br />

1. James, T.C. and S.C. Elgin, Mol Cell Biol, 1986. 6: p. 3862-3872.<br />

2. Cheutin, T., A.J. McNairn, T. Jenuwein, D.M. Gilbert, P.B. Singh, and T. Misteli,<br />

Science, 2003. 299(5607): p. 721-5.<br />

3. Cryderman, D.E., M.H. Cuaycong, S.C. Elgin, and L.L. Wallrath, Chromosoma, 1998.<br />

107(5): p. 277-85.<br />

4. Brink, M.C., Y. van der Velden, W. de Leeuw, J. Mateos-Langerak, A.S. Belmont, R.<br />

van Driel, and P.J. Verschure, Histochem Cell Biol, 2006. 125(1-2): p. 53-61.<br />

contact:<br />

Szabolcs Sörös<br />

Max Planck Institute for Biophysical Chemistry<br />

ssoeroe@gwdg.de<br />

Am Fassberg 11<br />

37077 Göttingen (Germany)


Renata Jurkowska, Da Jia, Sergey Ragozin, Nils Ansbach, Claus Urbanke, Xing Zhang,<br />

Richard Reinhardt, Wolfgang Nellen, Xiaodong Cheng, Albert Jeltsch<br />

Multimerisation of the Dnmt3L-Dnmt3a complex on DNA and<br />

its mechanistic implications<br />

DNA methyltransferase 3a (Dnmt3a) and its regulatory factor, DNA methyltransferase 3like<br />

protein (Dnmt3L), are both required for de novo DNA methylation of imprinted<br />

genes in mammalian germ cells, though the basis for imprinting-associated methylation<br />

is not yet clear. X-ray structure analysis shows that the C-terminal domain of Dnmt3L<br />

interacts with the catalytic domain of Dnmt3a. The Dnmt3a-C/Dnmt3L-C complex further<br />

dimerizes through Dnmt3a-3a interaction, forming a tetrameric complex with two active<br />

sites. Substitution of key noncatalytic residues in the Dnmt3a-3L interface or Dnmt3a-3a<br />

interface eliminated enzymatic activity of Dnmt3a and reduced its cofactor and DNA<br />

binding ability, indicating the requirement of the intact interfaces for the function of the<br />

enzyme. Biochemical data show that Dnmt3a-C binds strongly to DNA and forms an<br />

oligomeric, nucleoprotein filament in a very cooperative reaction. We further visualised<br />

the Dnmt3a (and Dnmt3a/Dnmt3L) – DNA filaments using scanning force microscopy<br />

and demonstrated that the multimerization of Dnmt3a is required for its localization to<br />

pericetromeric heterochromatin.<br />

Molecular modeling of a DNA-Dnmt3a dimer suggested that the two active sites are<br />

separated by approximately one DNA helical turn. A periodicity in the activity of Dnmt3a<br />

on long DNA revealed a correlation of methylated CpG sites at distances of 8-10 base<br />

pairs, suggesting that oligomerization leads Dnmt3a to methylate in a periodic pattern. A<br />

similar periodicity of 9.5 base pairs is observed for the frequency of CpG sites in the<br />

differentally-methylated regions of 12 maternally-imprinted mouse genes. These results<br />

suggest a basis for the recognition of differentially-methylated regions in imprinted<br />

genes, involving detection of both nucleosome modification and CpG spacing.<br />

Literature<br />

Jia D*, Jurkowska RZ*, Zhang X, Jeltsch A, Cheng X. A. Structure of Dnmt3a bound to<br />

Dnmt3L suggests a model for de novo DNA methylation. (2007) Nature 449(7159):248-<br />

51.<br />

Jurkowska RZ, Ragozin S, Ansbach N, Urbanke C, Jia D, Reinhard R, Nellen W, Xiaodong<br />

C, Jeltsch A Formation of nucleoprotein filaments by mammalian DNA methyltransferase<br />

Dnmt3a and its complex with regulator Dnmt3L. (2008) Manuscript in preparation.<br />

contact:<br />

Renata Jurkowska<br />

Jacobs University Bremen<br />

Department of Biochemistry<br />

r.jurkowska@jacobs-university.de<br />

Campus Ring 1<br />

28759 Bremen (Germany)<br />

additional information<br />

Renata Jurkowska, Sergey Ragozin and Albet Jeltsch: Biochemistry Lab, School of Engineering and<br />

Science, Jacobs University Bremen, Campus Ring 1, 28759 Bremen, Germany<br />

Nils Ansbach and Wolfgang Nellen: Abt. Genetik, CINSaT, Universität Kassel, Heinrich-Plett-Str. 40,<br />

34132 Kassel, Germany<br />

Claus Urbanke: Medizinische Hochschule, Abteilung Strukturanalyse OE 8830, Carl Neuberg Str. 1,<br />

30625 Hannover, Germany<br />

Da Jia, Xing Zhang and Xiaodong Cheng: Department of Biochemistry, Emory University School of<br />

Medicine, 1510 Clifton Road, Atlanta, GA 30033, USA<br />

Richard Reinhard: Max Planck Institute for Molecular Genetics, Ihnestrasse 63-73, D-14195 Berlin-<br />

Dahlem, Germany


Timo Quante, Lars Tögel, Wolfgang Deppert, Genrich V. Tolstonog<br />

Mutp53 as a modulator of global chromatin organisation<br />

Missense mutations in the TP53 gene are frequent genetic alterations in human tumor<br />

tissue and cell lines. In contrast to its wild-type counterpart, the mutant p53 (mutp53)<br />

protein is long-lived and has lost the transcriptional activity towards pro-apoptotic and<br />

growth arrest genes, but retained the propensity for targeting to chromatin. Expression<br />

of mutp53 is advantageous for tumor cells, however the molecular mechanism of<br />

mutp53 action is still not known. We used tumor cell lines expressing endogenous and<br />

inducible mutp53 proteins as models to study the role of mutp53 in transcriptional<br />

regulation. Mutp53 has lost sequence specificity in DNA binding activity but retained the<br />

property to recognize DNA secondary structures, and based on our ChIP-Seq data is<br />

prone to interact preferentially with repetitive sequences possessing conformational<br />

flexibility. Therefore we assumed that mutp53 operates on the level of global chromatin<br />

organization rather than on modulating the expression of individual genes. In support,<br />

we found that differentially regulated genes frequently map to the same chromosomal<br />

locations or even are organized as physically-linked gene clusters. This implies that<br />

mutp53 might be a factor involved in regulating the recruitment of gene promoters to<br />

immobile transcription factories containing active RNA polymerases. To test this<br />

hypothesis, physical DNA contacts in several genomic loci encompassing co-regulated<br />

gene clusters were profiled using the chromosome conformation capture (3C) method.<br />

Preliminary results strongly support the proposed function of mutp53 in modulating the<br />

gene expression program at a higher level.<br />

contact:<br />

Timo Quante<br />

Heinrich-Pette-Institute of Experimental Virology and Immunology, University of Hamburg<br />

Department of Tumor Virology<br />

timo.quante@hpi.uni-hamburg.de<br />

Martinistr. 52<br />

20251 Hamburg (Germany)


Sascha Tierling, Yingying Zhang, Christian Rohde, Nina Pälmke, Julia Arand, Diana<br />

Santacruz, Matthias Platzer, Richard Reinhardt, Albert Jeltsch, Jörn Walter<br />

NAME21: The National Methylome Project of Human<br />

Chromosome 21<br />

DNA methylation is essential for embryonic development, immunological memory and<br />

adult brain function. Despite of the complex biological importance of DNA methylation<br />

for the development and pathophysiology of human, genome wide maps of DNAmethylation<br />

distribution are still lacking. NAME 21 will contribute to fill this gap by<br />

providing such a comprehensive map for chromosome 21. We analyzed all gene<br />

promoters in two primary cell types (peripheral blood and primary fibroblasts) and two in<br />

vitro cell lines (HEK293 and HEPG2) by bisulfite sequencing and found most amplicons to<br />

be consistently unmethylated. Adressing inter-species methylation variation we found 10<br />

out of 28 amplicons to be different (25-50%) between human and chimpanzee.<br />

Focussing on structural promoter features, we found nucleosome positioning not to be<br />

correlated to DNA methylation. However, Z-DNA-like structures in CpG islands might<br />

represent an important structural marker for the prediction of DNA methylation. To<br />

identify intergenic regulatory elements that are potentially involved in transcription and<br />

genome integrity we started to expand our work into evolutionary conserved CpG-rich<br />

sections in transcript-poor and transition (from transcript-rich to transcript-poor) regions<br />

and show first preliminary results. NAME21 is linked to the Human Epigenome<br />

Consortium (HEP) and to the NoE “The EPIGENOME”.<br />

Literature<br />

Bock, C., Paulsen, M., Tierling, S. et al. (2006) PLoS Genet. 2, e26.<br />

Jeltsch, A., Walter, J., Reinhardt, R. & Platzer, M. (2006) Cancer Res. 66, 7378.<br />

Goyal, R., Reinhardt, R. & Jeltsch, A. (2006) Nucl. Acids Res. 34, 1182-8.<br />

Bock, C., Reither, S., Mikeska, T. et al. (2005) Bioinformatics 21, 4067-8.<br />

contact:<br />

Dr. Sascha Tierling<br />

Universität des Saarlandes<br />

Genetik/Epigenetik<br />

s.tierling@mx.uni-saarland.de<br />

Campus Geb. A24<br />

66123 Saarbrücken (Deutschland)<br />

additional information<br />

Address Authors 4,5,6,10 same as first author<br />

Address Authors 2,3,9:<br />

School of Engineering and Science, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen<br />

Address Author 7:<br />

Fritz-Lipmann-Institut, Beutenbergstrasse 11, 07745 Jena<br />

Address Author 8:<br />

Max-Planck-Institut für Molekulare Genetik, Ihnestrasse 63-73, 14195 Berlin


Yingying Zhang, Christian Rohde, Sascha Tierling, Heinrich Stamerjohanns, Matthias<br />

Platzer, Richard Reinhardt, Jörn Walter, Albert Jeltsch<br />

NAME21: The National Methylome Project of Human<br />

Chromosome 21<br />

DNA methylation is an essential epigenetic event involved in development, gene<br />

regulation, imprinting and preserving genome integrity. In the National Methylome<br />

Project for chromosome 21 (NAME21), we analyzed the DNA methylation pattern of 291<br />

amplicons for 190 gene promoters in leukocytes, HEK293, HepG2 and fibroblast cells.<br />

The comparison of the methylation profiles revealed that a fraction of genes is differently<br />

methylated among the tissues. A strong correlation of high methylation in gene<br />

promoter with low gene expression and high expression with low methylation was<br />

demonstrated in HEK293 and leukocytes. The results also indicated that there is a strong<br />

correlation between the absence of DNA methylation and the presence of histone H3 K4<br />

trimethylation and H3 K9/K14 acetylation up to distance of 1000 bps. The analysis of<br />

gene ontology using genes with methylated or unmethylated promoters revealed<br />

overrepresentation of genes in distinct ontology categories. These results documented<br />

an important effect of DNA methylation on both cellular physiology and morphology. A<br />

bioinformatics analysis of the differently methylated promoter regions revealed an<br />

overrepresentation of several transcription factor binding sites. Additionally, several<br />

regions with allele specific methylation pattern were identified.<br />

contact:<br />

PhD student Yingying Zhang<br />

Jacobs University Bremen<br />

School of Engineering and Science<br />

y.zhang@jacobs-university.de<br />

Campus Ring 1<br />

28759 Bremen (Germany)<br />

additional information<br />

Corresponding Author:<br />

a.jeltsch@jacobs-university.de<br />

Address:<br />

Jörn Walter and Sascha Tierling<br />

Institut für Genetik, FB Biowissenschaften, Universität des Saarlandes, Postfach 151150, D-66041<br />

Saarbrücken, Germany<br />

Matthias Platzer<br />

Fritz-Lipmann-Institut, Beutenbergstrasse 11, D-07745 Jena, Germany<br />

Richard Reinhardt<br />

Max Planck Institute for Molecular Genetics, Ihnestrasse 63-73, D-14195 Berlin-Dahlem, Germany


Franck COURT, Marion BANIOL, Hélène HAGEGE, Julie BORGEL, Jacques PIETTE, Guy<br />

CATHALA, Thierry FORNE<br />

NEW INSIGHTS INTO THE IMPRINTED MOUSE Igf2/H19<br />

LOCUS BY 3C-qPCR METHOD<br />

In mammals, higher-order chromatin architecture plays a crucial role in controlling gene<br />

expression notably through long range interactions between regulatory elements.<br />

However, until recently, the in vivo chromatin organization at a scale of 10 to 500 kb<br />

remained poorly known. Since a few years, Chromosome Conformation Capture assays<br />

(3C assays) give us access to this level of genomic organisation (1). Our laboratory<br />

recently developed a new sensitive 3C approach based on real-time quantitative PCR:<br />

the 3C-qPCR method (2). We used this method to investigate long-distance interactions<br />

at the imprinted mouse Igf2/H19 locus. The Insulin-like growth factor 2 (Igf2) gene is<br />

exclusively expressed from the paternal allele and encodes a growth factor required for<br />

normal embryonic development. The H19 gene is maternally expressed and produces an<br />

untranslated RNA of unknown function. Imprinting of both genes is depending on an<br />

Imprinting-Control Region (ICR) which is differentially methylated and controls allelespecific<br />

expression.<br />

Using the 3C-qPCR method and available mutant strains (H19Δ13 & ΔDMR2), we are<br />

able to investigate allele-specific interactions and to obtain functional insights on gene<br />

expression and higher-order chromatin architecture at this locus. These investigations<br />

led to the discovery of a novel paternal intergenic H19/Igf2 transcript (Pihit).<br />

Experiments are in progress to further characterize and determine the potential function<br />

of this transcript.<br />

Literature<br />

(1) Dekker J et al. 2002. Capturing Chromosome Conformation. Science 295, 1306-11.<br />

(2) Hagege H et al. 2007. Quantitative analysis of chromosome conformation capture<br />

assays (3C-qPCR). Nat Protoc. 2, 1722-33<br />

contact:<br />

phd student Franck COURT<br />

UMR 5535 CNRS-UMII<br />

Institut de Génétique Moléculaire de Montpellier<br />

franck.court@igmm.cnrs.fr<br />

1919 route de mende<br />

34293 Montpellier cedex 5 (france)


Wibke Peters, Thomas Macherey, Mike Duisken, Sophie Willnow, Bernhard Lüscher,<br />

Elmar Weinhold<br />

New S-Adenosyl-L-methionine Analogues to Investigate the<br />

Methylome<br />

DNA and protein methyltransferases (MTases) play crucial roles in setting epigenetic<br />

marks on DNA and histone proteins. They catalyse the transfer of the activated methyl<br />

group from the ubiquitous cofactor S-adenosyl-L-methionine (AdoMet or SAM) to specific<br />

residues within their substrates. Since the methyl group is not an attractive reporter<br />

group, we have synthesised two new classes of AdoMet analogues which serve as<br />

efficient cofactors for sequence-specific DNA functionalisation and labelling.<br />

In the first class of cofactor analogues the amino acid side chain of AdoMet is replaced<br />

with an aziridinyl residue and reporter groups are attached to the adenine ring.[1,2]<br />

DNA MTase-catalysed nucleophilic aziridine ring opening leads to sequence-specific<br />

coupling of the whole aziridine cofactor with DNA and hence to covalent DNA labelling. In<br />

the second class of cofactor analogues the methyl group of AdoMet is replaced with<br />

extended carbon chains carrying an unsaturated bond in beta-position to the sulfonium<br />

center.[3,4] The unsaturared bond is essential and leads to efficient transfer of the<br />

extended side chain to DNA.<br />

We have extended this work to protein MTases and find that the latter AdoMet analogues<br />

also function as cofactors for histone MTases. These analogues are used to transfer<br />

bioorthogonal chemical groups for modification by click chemistry and provide a new<br />

chemical tool to label proteins and identify targets of protein MTases.<br />

Literature<br />

[1] Pljevaljcic, F. Schmidt, E. Weinhold, Sequence-specific Methyltransferase-Induced<br />

Labeling of DNA (SMILing DNA), ChemBioChem 2004, 5, 265–269.<br />

[2] Pljevaljcic, F. Schmidt, A. J. Scheidig, R. Lurz, E. Weinhold, Quantitative labeling of<br />

long plasmid DNA with nanometer precision, ChemBioChem 2007, 8, 1516–1519.<br />

[3] C. Dalhoff, G. Lukinavicius, S. Klimasauskas, E. Weinhold, Direct transfer of<br />

extended groups from synthetic cofactors by DNA methyltransferases, Nat. Chem. Biol.<br />

2006, 2, 31–32.<br />

[4] G. Lukinavicius, V. Lapiene, Z. Stasevskij, C. Dalhoff, E. Weinhold, S. Klimasauskas,<br />

Targeted labeling of DNA by methyltransferase-directed Transfer of Activated Groups<br />

(mTAG), J. Am. Chem. Soc. 2007, 129, 2758–2759.<br />

contact:<br />

Professor Elmar Weinhold<br />

RWTH Aachen University<br />

Institute of Organic Chemistry<br />

elmar.weinhold@oc.rwth-aachen.de<br />

Landoltweg 1<br />

52056 Aachen (Germany)<br />

additional information<br />

Mike Duisken, LECO Instrumente GmbH, 41199 Mönchengladbach, Germany; Bernhard Lüscher,<br />

Institute of Biochemistry, Division of Biochemistry and Molecular Biology, RWTH Aachen University<br />

Hospital, 52074 Aachen, Germany.


Andrea Felten, Peter Leister, Karl Heinz Scheidtmann<br />

Novel Coactivators of Androgen Receptor: AATF and ZIP<br />

Kinase<br />

The androgen receptor (AR) is a ligand-dependent transcription factor involved in<br />

differentiation, proliferation and homeostasis. The transcriptional activity of AR is<br />

mediated by interaction with multiple coactivators, which serve in chromatin modification<br />

or remodeling, or provide a link between specific and general transcription factors. We<br />

recently identified AATF (Apoptosis antagonizing transcription factor) and ZIP kinase<br />

(Zipper interacting protein kinase) as novel coactivators of the AR. Following hormone<br />

treatment, AR, AATF, and ZIP kinase formed physical complexes and associated with the<br />

promoter and enhancer of the prostate-specific antigen gene, as revealed by chromatin<br />

immunoprecipitation. AATF seems to serve as adaptor to facilitate or stabilize the<br />

interaction between AR and ZIP kinase while ZIP kinase exerts its function as a kinase.<br />

Significantly, depletion of ZIP kinase by siRNA reduced AR-mediated transactivation.<br />

However, neither AR nor AATF are phosphorylated by ZIP kinase in vitro suggesting that<br />

it phosphorylates other coactivators or chromatin proteins, particularly histones. Indeed,<br />

H3 is phosphorylated at Thr11 in the vicinity of the PSA enhancer. However, a role of ZIP<br />

kinase in this modification has to be established. Other candidate substrates of ZIP<br />

kinase are members of the p160 family, SRC1 and/or SRC3 which are heavily<br />

phosphorylated by different kinases and which form complexes with ZIP kinase. These<br />

issues are presently under investigation.<br />

Supported by DFG<br />

Literature<br />

Leister et al. (2003). Apoptosis Antagonizing Transcription Factor AATF is a novel coactivator<br />

of nuclear hormone receptors. Signal Transduction 3, 17-25.<br />

Leister et al. (2007). ZIP kinase plays a crucial role in androgen receptor-mediated<br />

transcription. Oncogene, 1–9<br />

contact:<br />

Prof Karl Heinz Scheidtmann<br />

University of Bonn<br />

Genetics<br />

kh.scheidtmann@uni-bonn.de<br />

Roemerstr. 164<br />

53177 Bonn (Germany)


Andrea Harni•arová, Eva Bártová, Jana Krej•í, Gabriela Galiová, Stanislav Kozubek<br />

Nuclear location of Oct3/4 and c-myc genes in human<br />

embryonic stem cells undergoing differentiation<br />

Human embryonic stem cells (hES) are pluripotent cell lines derived from the inner cell<br />

mass of the blastocyst. The maintenance of the undifferentiated state in vivo is<br />

dependent on hESC self-renewal ability that is influenced by specific cultivation<br />

conditions. Here, we have studied hES cells differentiation induced by all-trans retinoic<br />

acid (ATRA) which is associated with an increased level of chromatin condensation when<br />

compared with the pluripotent stage. Nuclear patterns of the Oct3/4 (6p21.33) gene,<br />

responsible for hESC pluripotency and the c-myc (8q24.21) gene that controls cell cycle<br />

progression were investigated. In non-differentiated hES cell nuclei, the Oct3/4 were<br />

located on greatly extended chromatin loops, outside their respective chromosome<br />

territories. However, this phenomenon was not observed for the Oct3/4 gene in<br />

differentiated hESCs and not for the c-myc gene in the cell types studied. Analysing the<br />

nuclear radial distances of the Oct3/4 and c-myc genes, we observed that Oct3/4 was<br />

positioned more centrally, at ~65% of nuclear radius then c-myc located at ~71% of<br />

nuclear radius. Neither high level of Oct3/4 expression, associated with Oct3/4 location<br />

on extended chromatin loops, nor differentiation processes influenced the nuclear radial<br />

positioning of this gene. Similarly, differentiation of hESCs using ATRA did not cause cmyc<br />

nuclear radial rearrangement.<br />

This work was supported by the Grant Agency of the Czech Republic, grant Nos:<br />

204/06/0978, and by other grants: AVOZ50040507 and AVOZ50040702. We thank Prof.<br />

Douglas Melton (HHMI/Harvard University) for providing us with hESCs.<br />

contact:<br />

Mgr. Andrea Harni•arová<br />

Institute of Biophysics ASCR, v.v.i.<br />

Institute of Biophysics ASCR, v.v.i.<br />

harand@ibp.cz<br />

Královopolská 135<br />

61265 Brno (Czech Republic)


Niels Boeckel, Masamichi Koyanagi, Masayoshi Iwasaki, Andreas M. Zeiher, Stefanie<br />

Dimmeler<br />

Oct3/4 and Klf4 promoter status in multipotent circulating<br />

mesangioblasts<br />

Histone modification plays an important role in regulation of gene transcription. Active<br />

and inactive promoters are characterized by different histone modifications.<br />

Transcriptional repression is followed e.g. by a pronounced increase in histone H3<br />

methylation on Lysine 9, while transcriptional active promoters are characterized by<br />

acetylation at histone H3 lysine 9 and lysine 14. Previous studies demonstrated<br />

pronounced increase of Histone 3 lysine 9 trimetyhlation and decrease of histone H3<br />

acetylation of the Oct3/4 Promoter during ES cell differentiation. Recently, we identified<br />

clonally expandable, telomerase-expressing stem cells, which can be isolated from<br />

peripheral blood of children undergoing cardiac surgery. The marker profile of the<br />

clonally expanded cells is distinct from endothelial progenitor cells, hematopoietic or<br />

mesenchymal stem cells but resembles multipotent embryonic mesoangioblasts (MAB),<br />

which are multipotent progenitors of mesodermal tissue originally isolated from the<br />

embryonic dorsal aorta and characterised by expression of mesenchymal and endothelial<br />

markers. Indeed, circulating MABs are multipotent and differentiate into endothelial<br />

cells, smooth muscle cells, and cardiomyocytes in vitro and in vivo, and improve<br />

functional recovery after hind limb ischemia and acute myocardial infarction model.<br />

Circulating MAB show telomerase activity and express 3 out of 4 genes previously shown<br />

to induce pluripotency namely Oct3/4, KLF4 and c-myc while Sox2 was not expressed.<br />

Oct3/4 expression was confirmed by RT-PCR and immunostaining. We also performed<br />

chromatin immunoprecipitation to analyze the histone modification status of Oct3/4 and<br />

KLF4 promoters in MAB. We found that histone H3 lysine 9 and 14 were acetylated,<br />

whereas H3 lysine 9 was not trimethylated on both promoters. Taken together, these<br />

findings demonstrate the active status of Oct3/4 and KLF4 promoter in circulating MAB,<br />

which was associated with expression of both genes. Demonstrating active status of<br />

stem cell markers support the evidence of multipotency of adult stem cell.<br />

contact:<br />

Niels Boeckel<br />

University of Frankfurt<br />

Dept. of Internal Medicine III Molecular Cardiology<br />

NielsBoeckel@gmx.de<br />

Theodor-Stern-Kai 7<br />

60590 Frankfurt (Germany)


Emilia Jarochowska, Pawe• Krawczyk, Anna •ach, Micha• Krzyszto•<br />

Presentation of Students' Society of Genetics and Epigenetics<br />

Students’ Society of Genetics and Epigenetics was founded in 2002 under the auspices of<br />

Department of Plant Molecular Biology, University of Warsaw. It gathers BSc, MSc and<br />

PhD students interested in recent developments in genetics and epigenetics. The Society<br />

has completed several projects, such as purification of thermostable DNA polymerase<br />

and analysis of Arabidopsis thaliana mutants in miR161 gene. Currently it is starting a<br />

research project on maize olignucleotide microarray stripping. The Society’s activity<br />

consists also of weekly seminars and scientific conferences allowing students to practice<br />

presentation and discussion on one’s own and others’ research.<br />

contact:<br />

B.Sc. Anna •ach<br />

Warsaw University<br />

Laboratory of Plant Molecular Biology<br />

ankalach@yahoo.co.uk<br />

Pawinskiego 5A/F<br />

PL02-106 Warsaw (Poland)


Daniel Buszewicz, Marta Teperek, Pawel Krawczyk, Emilia Jarechowska, Michal<br />

Krzyszton<br />

Presentation of Students' Society of Genetics and Epigenetics<br />

Students’ Society of Genetics and Epigenetics was founded in 2002 under the auspices of<br />

Department of Plant Molecular Biology, University of Warsaw. It gathers BSc, MSc and<br />

PhD students interested in recent developments in genetics and epigenetics. The Society<br />

has completed several projects, such as purification of thermostable DNA polymerase<br />

and analysis of Arabidopsis thaliana mutants in miR161 gene. Currently it is starting a<br />

research project on maize olignucleotide microarray stripping. The Society’s activity<br />

consists also of weekly seminars and scientific conferences allowing students to practice<br />

presentation and discussion on one’s own and others’ research.<br />

contact:<br />

B. Sc. Michal Krzyszton<br />

Warsaw University<br />

Laboratory of Plant Molecular Biology<br />

mkrzyszton@gmail.com<br />

Pawinskiego 5A/F<br />

PL02-106 Warsaw (Poland)


Arunkumar Dhayalan, Tomasz Jurkowski, Heike Laser, Richard Reinhardt, Da Jia,<br />

Xiaodong Cheng, Albert Jeltsch<br />

Protein - protein interaction analysis by Absence of<br />

Interference approach<br />

Protein-protein interactions are critical to most biological processes and locating proteinprotein<br />

interfaces on protein structures is an important task in Molecular Biology. We<br />

developed a directed evolution method to determine surface residues involved in proteinprotein<br />

interaction of established pairs of interaction proteins. One of the proteins is<br />

subjected to high-level randomization by error-prone PCR. The resulting library is<br />

selected by yeast two-hybrid system for interacting clones, which are isolated and<br />

sequenced. The interaction region can be identified by absence of mutations. It is<br />

displayed on the surface of the structure (or a structural model) of the randomized<br />

protein using a WEB interface (http://biochem.jacobs-university.de/prima/). To explore<br />

the method, we mapped the interface of the catalytic domain of the DNA<br />

methyltransferase Dnmt3a with its regulatory factor Dnmt3L. Dnmt3a was randomized<br />

with high mutational load. 76 interacting clones were isolated and sequenced and 648<br />

mutations were identified. The mutational pattern allowed to identify a unique<br />

interaction region on the surface of Dnmt3a which comprises about 500-600 •². The<br />

results were confirmed by site directed mutagenesis and structural analysis. The<br />

‘Absence of Interference’ method for mapping protein interaction will allow highthroughput<br />

mapping of protein interaction sites suitable for functional studies and<br />

protein docking.<br />

Literature<br />

Dhayalan, A. et al., Mapping of Protein–Protein Interaction Sites by the ‘Absence of<br />

Interference’ Approach, J. Mol. Biol. (2008), doi:10.1016/j.jmb.2007.12.032<br />

contact:<br />

Mr. Arunkumar Dhayalan<br />

School of Engineering and Science<br />

Jacobs University Bremen<br />

a.dhayalan@jacobs-university.de<br />

Research 2, Campus ring1<br />

28759 Bremen (Germany)


Michael Grzendowski, Markus J. Riemenschneider, Marietta Wolter, Uwe Schlegel,<br />

Helmut E. Meyer, Guido Reifenberger, Kai Stühler<br />

Proteome analysis of human glioma with 1p/19q LOH<br />

Gliomas, the most common primary brain tumors, are histologically classified on the<br />

basis of morphological and immunohistochemical features as defined in the World Health<br />

Organization (WHO) classification of tumors of the nervous system. In addition to the<br />

histological assessment, certain genetic factors, such as allelic losses on chromosome<br />

arms 1p and 19q, are able to provide clinically useful information that may help to<br />

stratify gliomas into prognostically distinct subgroups. In particular, recent randomized<br />

trials have strongly associated 1p/19q-deletion with response to radio- and<br />

chemotherapy as well as longer survival in patients with anaplastic oligodendrogliomas<br />

and anaplastic oligoastrocytomas (Cairncross et al., J. Clin. Oncol. 24, 2707-14, 2006;<br />

van den Bent et al., J. Clin. Oncol. 24, 2715-22, 2006).<br />

To identify proteins that are differentially expressed between gliomas with and without<br />

1p/19q-deletion, we performed a proteomic analysis on oligoastrocytomas using<br />

differential gel electrophoresis (DIGE) followed by MALDI-TOF/TOF mass spectrometry.<br />

Thereby, we identified 46 differentially expressed proteins (• ≥ 1.8, p ≤ 0.05). From<br />

these candidate proteins we selected the first promising proteins for further analysis and<br />

confirmed the differential expression of four candidate proteins by using Western-blot<br />

analysis and immunohistochemistry. Subsequent epigenetic analysis revealed promoter<br />

hypermethylation as the major cause for low mRNA and protein expression levels of<br />

these four candidates in oligodendroglial tumors with 1p/19q loss. Further clinical<br />

validation of these differentially expressed proteins on larger patient cohorts is ongoing<br />

to assess their utility as potential biomarkers for classification and prognostic<br />

assessment. This overall approach provides a powerful means to identify proteome wide<br />

alterations associated with allelic status on 1p/19q and thus may help in identifying<br />

novel, highly relevant prognostic glioma markers.<br />

contact:<br />

Michael Grzendowski<br />

Ruhr-Universität Bochum<br />

Medizinisches Proteom-Center<br />

michael.grzendowski@rub.de<br />

Universitätsstr.150<br />

44801 Bochum (Germany)


Rudolf Engelke, Gerhard Mittler<br />

Proteomic analysis of the nuclear matrix in pre-B cells.<br />

The nuclear matrix (NM) is an operationally defined structure that was introduced by<br />

Berezney and Coffey in 1974 to denote a residual nuclear scaffold devoid of bulk<br />

chromatin obtained by sequential salt extractions, detergent and DNase I treatment (1).<br />

Further analysis suggested that this subnuclear structure is a ribonucleoprotein network<br />

comprising the interchromatin space and the nuclear lamina and might therefore play an<br />

important role in compartmentalization of the nucleus as well as the three dimensional<br />

organization of the genome. Consistent with this, nuclear matrix proteins have been<br />

implicated in almost all aspects of nucleic acid metabolism including DNA replication and<br />

repair, transcription as well as RNA processing (2, 3).<br />

In this work we have improved the classical biochemical nuclear matrix preparation<br />

methods utilizing 250 mM ammonium sulfate, 2 M NaCl or 25 mM lithium-3,5,diiodosalicylate<br />

(LIS) as nuclear extraction reagent and analyzed the NM proteome by<br />

nanoLC tandem mass spectrometry leading to the identification of more than 950 NM<br />

protein candidates. Our NM preparation maintains the morphological integrity of the<br />

nucleus as assessed by phase contrast and immunofluorescence microscopy. Cherry-NLS<br />

and DNA DAPI staining served as purification control demonstrating the removal of<br />

chromatin and nucleoplasm. Implications of a quantitative SILAC-based comparison of<br />

the NM versus the nuclear proteome in order to define bona fide NM constituents are<br />

discussed.<br />

Literature<br />

(1) Berezney, R. and D. S. Coffey (1974). "Identification of a nuclear protein matrix."<br />

Biochem Biophys Res Commun 60(4): 1410-7.<br />

(2) Pederson, T. (1998). "Thinking about a nuclear matrix." J Mol Biol 277(2): 147-59.<br />

(3) Zaidi, S. K., et al. (2005). "The dynamic organization of gene-regulatory machinery<br />

in nuclear microenvironments." EMBO Rep 6(2): 128-33.<br />

contact:<br />

Rudolf Engelke<br />

MPI of Immunobiology<br />

engelke@immunbio.mpg.de<br />

Stübeweg 51<br />

79108 Freiburg (Germany)


Levin Böhlig, Kurt Engeland<br />

Regulation of an intronic microRNA and its host gene by the<br />

tumor suppressor p53<br />

The tumor suppressor p53 is a central integrator of different stress signals in the cell.<br />

p53 is a transcription factor and acts by transcriptional activation or repression of<br />

numerous target genes. In addition to protein coding genes, also noncoding transcripts<br />

of the miRNA-34 family have been shown to contribute to p53 function. Thus, microRNAs<br />

appear to have essential functions in mediating cell cycle arrest and apoptosis following<br />

p53 induction. We discovered the p53-dependent expression of the pantothenate kinase<br />

1 gene (PANK1) and its intronic miR-107. We found PANK1 mRNA and protein<br />

upregulated after induction of p53 in a tet-off-regulated DLD-1-derived cell system.<br />

Furthermore, also endogenous levels of p53 induce PANK1 expression whereas no<br />

induction is observed in cells functionally negative for p53. The induction of PANK1<br />

appears to be a result of direct transcriptional activation by p53 because mRNA induction<br />

is also observed after inhibition of translation by cycloheximide. Interestingly, mature<br />

miR-107 is also upregulated after p53 expression which indicates a coregulation of<br />

miRNA and its host gene. By analyzing miRNA target genes we intent to show which<br />

cellular processes are modulated through miR-107 function. In summary, this is the first<br />

example of a target gene being coregulated by p53 together with its intronic microRNA.<br />

contact:<br />

Dipl.-Biochem. Levin Böhlig<br />

Universität Leipzig<br />

Universitätsfrauenklinik<br />

levin.boehlig@medizin.uni-leipzig.de<br />

Semmelweisstraße 14<br />

04103 Leipzig (Germany)


Huan Shu, Lars Hennig<br />

Restructuring of epigenetic landscapes during plant<br />

development<br />

Chromatin is subject to a diverse array of posttranslational modifications (e.g.<br />

methylation, acetylation, ubiquitinylation, ADP-ribosylation and phosphorylation) that<br />

largely impinge on histone amino termini. Distinct histone amino-terminal modifications<br />

and their combinations can synergistically or antagonistically create interaction surfaces<br />

for chromatin-associated proteins, which in turn dictate dynamic transitions between<br />

transcriptionally active or transcriptionally silent chromatin states. Little is known how<br />

genome-wide epigenetic profiles (“epigenetic landscapes”) change during development.<br />

The plant Arabidopsis is a great model to study such changes, because development is<br />

well-characterized, the compact genome is well annotated and because many mutants<br />

deficient in chromatin dynamics are available. One of the most important and beststudied<br />

developmental decisions in plants is the decision when to flower and engage in<br />

reproduction. Here, we will present our approaches and initial results of a project that<br />

probes the changing epigenetic profiles during the transition to flowering of Arabidopsis<br />

by ChIP-chip.<br />

contact:<br />

PhD student Huan Shu<br />

ETH<br />

Institute of Plant Sciences<br />

hshu@ethz.ch<br />

Universitaetstrasse 2<br />

8032 Zuerich (Switzerland)


Lin XU, Rozenn MENARD, Alexandre BERR, Denise MEYER, Wen-Hui SHEN<br />

Role of Histone Ubiquitination in Arabidopsis Development<br />

Histone ubiquitination is a critical epigenetic mark conserved from yeast to human. H2B<br />

mono-ubiquitination is involved in transcriptional activation whereas H2A monoubiquitination<br />

is involved in gene silencing. Recently RING-domain proteins HUB1 and<br />

HUB2, which show highest homologies with the yeast BRE1, were reported to act on H2B<br />

ubiquitination and to be involved in regulation of G2-to-M transition of the cell cycle and<br />

of seed germination (Fleury et al., Plant Cell. 2007 Feb;19(2):417-32; Liu et al., Plant<br />

Cell. 2007 Feb;19(2):433-44). We identified AtUBC1 and AtUBC2 to encode proteins<br />

showing highest homologies with the yeast Rad6. Rad6 is involved, together with BRE1,<br />

in mono-ubiquitination of H2B in yeast. While the single mutant ubc1-1 or ubc2-1 has a<br />

normal flowering phenotype, the double mutant ubc1-1 ubc2-1 showed an earlyflowering<br />

phenotype. We found that hub1 and hub2 mutants also show an earlyflowering<br />

phenotype, suggesting that AtUBC1/2 and HUB1/2 function together in<br />

regulating flowering time through H2B ubiquitination. We also identified two novel genes<br />

of the RING family, LOC1 and LOC2, as being required for the maintenance of the<br />

restriction boundary of the shoot apical meristem (SAM) activity. We suggest that LOC1<br />

and LOC2 repress transcription through a Polycomb-like pathway.<br />

contact:<br />

Dr. Lin XU<br />

UNIVERSITE LOUIS PASTEUR<br />

Institut de Biologie Moléculaire des Plantes du CNRS<br />

lin.xu@ibmp-ulp.u-strasbg.fr<br />

12 rue du Général ZIMMER<br />

67084 STRASBOURG (France)


Annelen Schemm, Sabine Neumann, Pamela Strissel, Cord-Michael Becker<br />

Role of NRSF/ hREST4 in Neuronalisation of Tumors<br />

The neuron-restrictive silencer factor (NRSF) represses transcription of neuronal genes<br />

in non-neuronal tissues by binding to the neuron-restrictive silencer DNA element<br />

(NRSE).<br />

The expression of neuronal cell markers in malignant tumors, e.g. Small Cell Lung<br />

Cancer (SCLC), frequently correlates with paraneoplastic disorders but also with a higher<br />

invasivness and a poorer prognosis (1,2,3). In a variety of cell lines, NRSF and other<br />

neuronal gene expression patterns were determined using RT- PCR. For some cell lines<br />

absence of the NRSF transcript correlated with a presence of neuronal gene expression,<br />

while its presence correlated with neuronal gene silencing.<br />

In some cell lines, however, neuronal transcription occurred despite the presence of an<br />

NRSF transcript. Interestingly the NRSF splice variant, hREST4, was detectable in those<br />

cell lines. hREST4 is known to have nuclear localisation , supporting a role in<br />

transcriptional regulation. To test for a possible activating function of hREST4, luciferase<br />

assays were carried out using cell lines with and without hREST4/ NRSF transcripts,<br />

upon transfection with plasmids containing the luciferase gene under control of the NRSE<br />

of GlyR alpha1. However, no clear hREST4 function could be determined using that multi-<br />

cell line approach in the presense of full length NRSF. But there was a perfect correlation<br />

of hREST4 and GlyR alpha1 transcript levels when hREST4 expression levels were<br />

enhanced via drug treatment within one cell line, supporting our hypothesis.<br />

The mechanism of NRSF loss and the possible transcriptional activity of hREST4 may<br />

contribute to the constitutive activation of neuronal genes leading to deregulated growth<br />

and altered invasion behavior.<br />

Literature<br />

(1) Gurrola-Diaz, C., Lacroix, J., Dihlmann, S., Becker, C.-M., und von Knebel Doeberitz,<br />

M. (2003): Reduced expression of the neuron restrictive silencer factor permits<br />

transcription of glycine receptor α1 subunit in small-cell lung cancer cells. Oncogene 22,<br />

5636-5645.<br />

(2) S.B. Neumann, R. Seitz, A. Gorzella, A. Heister, M. von Knebel-Döberitz and C.-M.<br />

Becker (2004): Relaxation of glycine receptor and onconeural gene transcription in<br />

NRSFdeficient small cell lung cancer cell lines, Brain Res. Mol. Brain Res. 120, 173-181.<br />

(3) Butler, M. H., Hayashi, A., Ohkoshi, N., Villmann, C., Becker, C.-M., Feng, G., de<br />

Camilli, P., und Solimena, M. (2000) Autoimmunity against gephyrin in stiff man<br />

syndrome. Neuron 26, 307–312.<br />

contact:<br />

Annelen Schemm<br />

Universität Erlangen- Nürnberg<br />

Institut für Biochemie<br />

annelen.schemm@biochem.uni-erlangen.de<br />

Fahrsstrasse 17<br />

91054 Erlangen (Deutschland)


Ernst Aichinger, Aleksandra Erilova, Grigory Makarevich, Claudia Köhler<br />

Role of the Mi-2 homolog PICKLE in repression of Polycomb<br />

group target genes in Arabidopsis<br />

Mi-2 subunits are part of the NuRD (Nucleosome remodeling and deacetylase) complex<br />

and are widely conserved within the animal and plant kingdom. Current models propose<br />

varying enzymatic functions in transcriptional repression as well as in derepression.<br />

PICKLE (PKL) is homologous to Mi-2 alpha subunits and is involved in the specification of<br />

different developmental fates. PKL restricts pluripotency by stable repression of<br />

embryonic traits after germination. Moreover, PKL is required throughout the plant life<br />

cycle and lack of PKL function causes activation of the Polycomb group (PcG) target gene<br />

PHERES1. PHERES1 expression is restricted to seed development and is actively<br />

repressed during vegetative development by the action of different Polycomb group<br />

complexes. Therefore, we addressed the question whether activation of Polycomb group<br />

target genes in pkl mutants is caused by a mechanistic interaction of PKL and Polycomb<br />

group complexes.<br />

contact:<br />

Ernst Aichinger<br />

ETH Zürich<br />

Institute of Plant Sciences<br />

aernst@ethz.ch<br />

Universitätsstrasse 2<br />

8092 Zurich (Switzerland)


Lorenz Kallenbach, Patrick Heun<br />

Role of the SUMO E3 ligase PIAS in chromosome and nuclear<br />

organization in Drosophila melanogaster<br />

PIAS proteins were first identified as components of the JAK/STAT signal transduction<br />

pathway. In Drosophila, the only member of the PIAS family, Su(var)2-10, was found to<br />

be a suppressor of position effect variegation (PEV) (Reuter and Wolff, 1981). It is the<br />

only member of the PIAS-protein family in flies. Further studies of dPIAS suggested<br />

diverse roles in chromosome function and nuclear organization (Hari et al., 2001). This<br />

is consistent with the demonstration that PIAS proteins encode an E3 SUMO Ligase,<br />

which is part of the SUMO conjugating system (Heun, 2007). We found that in<br />

Drosophila there are at least 12 PIAS isoforms that result from alternative splicing.<br />

These isoforms share a central core, but differ in their N- and C-termini. Interestingly,<br />

they show different patterns of expression, are present in distinct complexes, and<br />

appear to have different target proteins.<br />

The aim of this project is to determine how dPIAS mediated SUMOylation regulates<br />

heterochromatin formation and nuclear organization. We are currently trying to identify<br />

the dPIAS isoforms involved in PEV and their corresponding target proteins. Then we will<br />

determine the effect of SUMOylation on the function of the target proteins.<br />

Literature<br />

Reuter, G. and I. Wolff, Isolation of dominant suppressor mutations for position effect<br />

variegation in Drosophila melanogaster. Mol Gen Genet, 1981.<br />

Hari, K.L., K.R. Cook, and G.H. Karpen, The Drosophila Su(var)2-10 locus regulates<br />

chromosome structure and function and encodes a member of the PIAS protein family.<br />

Genes Dev, 2001. 15(11): p. 1334-48.<br />

Heun, P., SUMOrganization of the nucleus. Curr Opin Cell Biol, 2007. 19: p. 1-6.<br />

contact:<br />

Lorenz Kallenbach<br />

Max-Planck-Institute of Immunobiology<br />

kallenbach@immunbio.mpg.de<br />

Stuebeweg 51<br />

79106 Freiburg (Germany)


Amos Tanay<br />

Selection and mutation in the evolution of CpG islands<br />

DNA methylation is a key epigenetic modification with a documented role in X<br />

inactivation, imprinting and additional aspects of normal and aberrant development of<br />

mammalian cells. Methylation is affecting CpG dinucleotides across the entire genomes<br />

and in a highly non random fashion that include islands of hypomethylation and neutral<br />

CpG content over an overall background of hypermethylation and low CpG content. We<br />

are developing a comprehensive evolutionary model to describe how the distributions of<br />

CpG dinucleotides evolved to form this intriguing structure. Out model characterize<br />

explicitly the evolutionary forces, selective and other, that are working to form islands. It<br />

improves accuracy by integrating data on primate genomes with high throughput<br />

epigenetic profiles. We identify a large class of CpG islands that are formed due to germline<br />

hypo-methylation and low rates of CpG deamination but not due to selection. We<br />

can associate such low rates of deamination with the activity of at least two protein<br />

complexes (RNA Polymerase and the Polycomb repressor complex 2). The model can<br />

also identify CpG islands that are affected by selection, including known differentially<br />

methylated regions near imprinted genes. The evolutionary approach thereby provides<br />

new and unbiased insight into some of the fundamental questions on the function of the<br />

DNA methylation system during normal development.<br />

Literature<br />

Tanay A, O'Donnell AH, Damelin M, and Bestor T. PNAS 104(13):5521-6 (2007).<br />

contact:<br />

Dr. Amos Tanay<br />

The Weizmann Institute<br />

Department of Computer Science and Applied Math<br />

amos.tanay@weizmann.ac.il<br />

PO BOX 26<br />

76100 Rehovot (Israel)


Gabriela Galiová, Eva Bártová, Andrea Harni•arová, Jana Krej•í, Stanislav Kozubek<br />

Single-cell c-myc gene expression in human embryonic stem<br />

cells and human teratocarcinoma NTERA cells<br />

The c-myc gene was found to be responsible for the self-renewal ability of mouse<br />

embryonic stem cells (mESCs), but in human embryonic stem cells (hESCs) it triggers<br />

apoptosis and induces differentiation (Sumi et al., 2007). Here, nuclear locations of the cmyc<br />

gene and complexes involving c-myc transcripts (c-mycT) have been investigated.<br />

The c-myc gene and its' transcripts were positioned non-randomly within the interphase<br />

nucleus, additionally, c myc RNA signals associated and/or accumulated at nucleoli.<br />

Using oligo-probes, designed to exon II and exon III of the c-myc gene (Levsky et al.,<br />

2002), single c-mycT was preferentially observed in human embryonic stem cells<br />

(hESCs). Conversely, human embryonal teratocarcinoma NTERA cells were characterized<br />

by the presence of multiple c myc RNA signals located in both the nucleoli and<br />

nucleoplasm. In all cell types studied, c mycT, when accumulated at nucleoli, occupied<br />

periphery of this organelle, not that region associated with cultivation surface. Specific<br />

nuclear and nucleolar positioning of the c-mycT probably reflects the kinetics in the cmyc<br />

RNA metabolism.<br />

This work was supported by the Grant Agency of the Czech Republic, grant Nos:<br />

204/06/0978, and by other grants: AVOZ50040507 and AVOZ50040702. We thank Prof.<br />

Douglas Melton (HHMI/Harvard University) for providing us with hESCs.<br />

Literature<br />

1. Sumi T., Tsuneyoshi N., Nakatsuji N., Suemori H. (2007) Apoptosis and differentiation<br />

of human embryonic stem cells induced by sustained activation of c-Myc. Oncogene<br />

26(38):5564-5576.<br />

2. Levsky J.M., Shenoy S.M., Pezo R.C., Singer R.H. (2002) Single-cell gene expression<br />

profiling. Science 297(5582):836-840.<br />

contact:<br />

Mgr. Gabriela Galiová<br />

Institute of Biophysics ASCR, v.v.i.<br />

Institute of Biophysics ASCR, v.v.i.<br />

galiova@ibp.cz<br />

Královopolská 135<br />

61265 Brno (Czech Republic)


Henriette Franz, Steven A. Jacobs, C. David Allis, Sepideh Khorasanizadeh, Wolfgang<br />

Fischle<br />

SPECIFICITY OF THE CDY FAMILY OF CHROMODOMAINS FOR<br />

METHYLATED ARKS MOTIFS IN CHROMATIN<br />

Previous studies have shown two related chromodomain modules in the HP1 and<br />

Polycomb proteins exhibit discriminatory binding interactions with the related<br />

methyllysine marks (containing the ARKS motif) on the histone H3 tail. Recently, the<br />

presence of methylated ARKS motifs have been identified in other chromatin factors<br />

(e.g., linker histone H1b and lysine methylase G9a). These are thought to function as<br />

peripheral docking sites for the HP1 chromodomain. In vertebrates, HP1-like<br />

chromdomains also occur in the chromodomain Y chromosome (CDY) family of proteins<br />

adjacent to a putative catalytic motif. Multiple CDY genes are encoded on the human Y<br />

chromosome and are thought to establish histone H4 acetylation during spermiogenesis.<br />

Two autosomal homologs, CDYL and CDYL2, may associate with multi-subunit<br />

transcription co-repressor complexes in diverse tissues. To improve our understanding of<br />

the function of CDY, CDYL and CDYL2 proteins, we set out to measure the specificity of<br />

their chromodomain modules for potential sites in chromatin. Surprising differences<br />

between these proteins suggest participation in distinct epigenetic pathways. The CDY<br />

chromodomain exhibits discriminatory binding, whereas the CDYL2 chromodomain<br />

exhibits promiscuity and equal binding to a number of ARKS motifs. Subtle amino acid<br />

changes in the CDYL chromodomain prohibit any docking interaction with the ARKS<br />

motifs. In support of the binding studies, in vivo localization studies show CDYL is<br />

targeted to different areas in the cell nucleus as compared to CDYL2. Thus, our<br />

quantitative comparison of CDY family chromodomains highlights how subtle sequence<br />

differences can generate substantial diversity in epigenetic regulations in highly<br />

differentiated species.<br />

contact:<br />

Henriette Franz<br />

Max Planck Institute for Biophysical Chemistry<br />

Chromatin Biochemistry<br />

hfranz@gwdg.de<br />

Am Fassberg 11<br />

37077 Goettingen (Germany)


Bastian Stielow, Alexandra Sapetschnig, Christina Wink, Guntram Suske<br />

SUMO-modified transcription factors repress transcription by<br />

provoking local heterochromatic gene silencing<br />

Following the mechanistic clues provided by our identification of SUMO-dependent<br />

repression components in insect cells (see abstract / <strong>poster</strong> presentation by G. Suske)<br />

we sought to analyze in detail the chromatin changes established by SUMO-modified<br />

transcription factors. We generated a mammalian cell line with a stably integrated<br />

chromatinized Gal4-driven reporter gene that allowed for the readout of the gene<br />

silencing events provoked by SUMOylated transcription factors. Upon transfection,<br />

SUMOylation-competent transcription factors (Gal4 fusions of wildtype Sp3 or<br />

steroidogenic factor 1) repressed transcription whereas the corresponding SUMOylationdeficient<br />

mutants activated transcription of the integrated reporter gene. Chromatinimmunoprecipitations<br />

demonstrate that the promoter-bound SUMO-modified<br />

transcription factors led to the establishment of local repressive chromatin with features<br />

of compacted heterochromatin. SUMO-dependent heterochromatin formation includes<br />

recruitment of the chromatin remodeler Mi-2, the MBT-domain proteins L3MBTL1 and<br />

L3MBTL2, HP1, and the histone methyltransferases SETDB1 and SUV4-20H accompanied<br />

by the establishment of repressive histone modifications such as H3K9 and H4K20<br />

trimethylation. Our results indicate that SUMOylation plays a crucial role in regulating<br />

gene expression by initiating chromatin structure changes that renders DNA inaccessible<br />

to the transcription machinery.<br />

This work was supported by a grant of the DFG to G.S.<br />

Literature<br />

Stielow, B., Sapetschnig, A., Krüger, I., Kunert, N., Brehm, A., Boutros, M. and Suske,<br />

G. (2008). Identification of SUMO-dependent chromatin-associated transcriptional<br />

repression componetnts by a genome-wide RNA interference screen. Mol. Cell, in press.<br />

contact:<br />

Bastian Stielow<br />

Philipps-Universität Marburg<br />

Institut für Molekularbiologie und Tumorforschung<br />

stielow@imt.uni-marburg.de<br />

Emil-Mannkopff-Str. 2<br />

35037 Marburg (Germany)


Günter Kahl, Carlos Molina Medina, Björn Rotter, Peter Winter, Hideo Matsumura,<br />

Ryohei Terauchi<br />

SuperSAGE: A complete genome-wide quantitative expression<br />

profiling platform<br />

SuperSAGE combined with novel sequencing technologies (e.g. 454 pyrosequencing)<br />

allows an ultra-deep analysis of any eukaryotic transcriptome on a scale not yet<br />

possible. An analysis identifies novel genes, alternatively spliced messenger isoforms,<br />

sense and anti-sense transcript pairs, and quantitate each transcript.<br />

SuperSAGE identifies transcripts by a 26 bp “tag”, originating from a unique position of<br />

the cDNA reverse-transcribed from the messenger RNA. Such tags uniquely identify all<br />

transcripts and their isoforms, and distinguish between members of large gene families.<br />

SuperSAGE as “open architecture platform” has advantages over microarrays: (1)the<br />

error-prone transcript quantification on microarrays is avoided by precise counting of<br />

tags, (2) detection of even very rare transcripts, impossible for microarrays, is<br />

commonplace,(3)SuperSAGE identifies new transcripts, and (4)avoids false positives, a<br />

common error arising from cross- hybridization on microarrays.<br />

The 26 bp tags also unambiguously identify transcripts originating from different,<br />

intimately interacting organisms (e.g. a pathogen or parasite and their hosts). For the<br />

first time in the Life Sciences, SuperSAGE permits to analyze the transcriptomes of such<br />

interacting organisms in their natural environment (i.e. without the traditional physical<br />

separation of both), and therefore portrays a realistic picture of the interaction. All<br />

these, not exhaustively listed features, make SuperSAGE the elite technology for a<br />

molecular analysis of eukaryotic parasite/host interactions and for an in-depth<br />

transcriptome analysis with and without epigenetic constraints.<br />

Literature<br />

Matsumura, H., Nasir, K.H.B., Yoshida K., Ito, A., Kahl, G., Krüger D.H., Terauchi, R.<br />

(2006). SuperSAGE-array: The direct use of 26-base-pair transcript tags in<br />

oligonucleotide arrays. Nature Methods 3: 469-474 (2006).<br />

Matsumura H., Reuter M., Krüger D.H., Winter P., Kahl G., Terauchi R. (2007).<br />

SuperSAGE. In: Methods in Molecular Biology 387:” Serial Analysis of Gene<br />

Expression: Digital Gene Expression Profiling”. Ed. K. L. Nielsen, Humana Press Inc.,<br />

Totowa, NJ, USA.<br />

contact:<br />

Professor Günter Kahl<br />

Frankfurt University<br />

Biocenter<br />

kahl@em.uni-frankfurt.de<br />

Max-von-Laue-Strasse 9<br />

60438 Frankfurt am Main (Germany)


Peter R. Lange, Andreas Finke, Claus Wasternack<br />

TFL2 as an epigenetic regulator in Arabidopsis development<br />

A stable repression of gene expression in connection with the inheritance of an<br />

established transcription pattern from cell to cell is an important aspect of eukaryotic<br />

development. Specific combinations of DNA and histone modifications cause local<br />

alterations of the chromatin structure and determine the balance between<br />

heterochromatin and euchromatin. To initiate gene silencing for example the polycomb<br />

repressor complex 2 (PRC2) transfers methyl-groups to specific histone domains. In<br />

animal species this is translated to a stable repression of the corresponding chromatin<br />

domain by the polycomb repressor complex 1 (PRC1). However, in plants PRC1 is<br />

neither structurally nor mechanistically conserved. One possible component of PRC2<br />

dependent maintenance of gene silencing is TERMINAL FLOWER 2 (TFL2). TFL2 was<br />

found to associate with H3K27 tri-methylated euchromatic genes (Turck, 2007). The loss<br />

of TFL2 in Arabidopsis thaliana leads to homeotic alterations in the architecture of<br />

inflorescences and flowers. Constitutive TFL2 over-expression lines generated during our<br />

work also show distinct alterations in inflorescences and the branching pattern.<br />

Molecular analyses mainly focused either on the mechanisms of epigenetic regulation or<br />

on individual target genes. In our work we used the comparison of the tfl2, 35S::TFL2<br />

and wild-type genome-wide transcriptomes to identify specific target genes of TFL2. We<br />

aim to uncover and to functionally characterize target genes of TFL2 involved in plant<br />

development.<br />

Our analysis of the tfl2 mutant transcriptome revealed 199 and 335 significantly up- or<br />

down-regulated genes, respectively. After vigorous selection from the pool of deregulated<br />

genes we are in the process of screening 60 insertion mutant lines for<br />

alterations in the plant architecture. A genetic and molecular characterisation of the<br />

corresponding genes giving rise to developmental phenotypes will be carried out to<br />

define their role in the determination of A. thaliana development.<br />

Literature<br />

Turck F., et al. (2007) PLoS Genet 3(6) e86. doi:10.1371/journal.pgen.0030086<br />

contact:<br />

Dr. Peter Lange<br />

Leibniz-Institut für Pflanzenbiochemie<br />

plange@ipb-halle.de<br />

Weinberg 3<br />

06120 Halle/Saale (Germany)


Stephan Hupfer, Julia Brill, Cord-Michael Becker, Kristina Becker<br />

The entla mouse - a model for human absence epilepsy<br />

Starting from motor abnormalities and a phenotype remniscent of petit mal epilepsy, we<br />

were able to identify the gene defect of the recessive mouse mutant entla by genomewide<br />

linkange analysis (1). This mouse suffers from absence epilepsy, with additional<br />

ataxia as well as paroxysmal dyskinesia evident by postnatal week 4. The entla genome<br />

harbours a mutation within the Cacna2d2 gene, coding for the accessory calcium<br />

channel subunit α2δ2. Comparable Calcium channel defects can be found in the mutants<br />

ducky, stargazer and tottering. These mutants however are often fatal and thus not well<br />

suited for more thorough pathophysiological studies on the course of the disease. Closer<br />

examination of the entla α2δ2 defect in recombinant systems showed changes in<br />

processing as well as in targeting to the plasma membrane. Electrophysiologically, a<br />

reduced Barium current density (pA/pF) and a shift of the channel’s inactivation curve<br />

were evident (1).<br />

A complex phenotype like that of the entla mouse is unlikely to be explained by an<br />

altered function of the product of the mutated gene alone. Rather, secondary and<br />

tertiary changes in the expression and/or function of other gene products may be<br />

partially responsible for pathological alterations in neuronal network function. In the<br />

brain of the entla mouse, the mutation of the calcium channel subunit gene Cacna2d2<br />

leads to a pronounced shift in the expression of subunits of several ligand-gated ion<br />

channels that might be involved in the epileptic pathogenesis in this mouse model.<br />

Literature<br />

(1) Brill, J., Klocke, R., Paul, D., Boison, D., Gouder, N., Klugbauer, N., Hofmann, F.,<br />

Becker, C.-M., Becker, K. (2004) Entla: A novel epileptic and ataxic Cacna2d2 mutant of<br />

the mouse. J. Biol. Chem. 279, 7322-7330.<br />

contact:<br />

Stephan Hupfer<br />

University Erlangen-Nuremberg<br />

Institute of Biochemistry<br />

stephan.hupfer@biochem.uni-erlangen.de<br />

Fahrstr. 17<br />

91054 Erlangen (Germany)<br />

additional information<br />

SH acknowledges support by the IZKF Erlangen<br />

JB is now at the Department of Neurology and Neurological Sciences, Stanford University School of<br />

Medicine, Stanford, California 94305, USA


Jennifer Gerke, Özgür Bayram, Gerhard H. Braus<br />

The velvet complex coordinates light, fungal development and<br />

secondary metabolism in Aspergillus nidulans<br />

Differentiation and secondary metabolism are correlated processes in fungi and respond<br />

to light. In Aspergillus nidulans, light inhibits sexual reproduction as well as secondary<br />

metabolism. We identified the heterotrimeric velvet complex as link between lightresponding<br />

developmental regulation and control of secondary metabolism. VeA, which<br />

is primarily expressed in the dark, physically interacts with VelB that is expressed during<br />

sexual development. VeA bridges VelB to the nuclear master regulator of secondary<br />

metabolism LaeA. Deletion of either velB or veA results in defects in both sexual fruiting<br />

body formation and production of secondary metabolites.<br />

contact:<br />

Jennifer Gerke<br />

Georg-August-University Göttingen<br />

Institute of Microbiology and Genetics<br />

jgerke@gwdg.de<br />

Grisebachstr. 8<br />

37077 Göttingen (Germany)


Filip Senigl, Jiri Plachy, Jiri Hejnar<br />

The CpG island core element protects retroviral vectors from<br />

transcriptional silencing<br />

Unmethylated CpG islands are known to maintain adjacent promoters transcriptionally<br />

active. In the adenosine phosphoribosyltransferase gene-adjacent CpG island, the<br />

protection from transcriptional silencing can be attributed to the short CpG-rich core<br />

element containing Sp1 binding sites. We report here the insertion of this CpG island<br />

core element, IE, into the long terminal repeat of a retroviral vector derived from Rous<br />

sarcoma virus, which normally suffers from progressive transcriptional silencing in<br />

mammalian cells. IE insertion into specific position between enhancer and promoter<br />

sequences led to efficient protection of the integrated vector from silencing and gradual<br />

CpG methylation in rodent and human cells. Individual cell clones with IE-modified<br />

reporter vectors display high level of reporter expression for a sustained period and<br />

without substantial variegation in the cell culture. The presence of Sp1 binding sites is<br />

necessary for the protective effect of IE. Modified vectors with IE insertions are also<br />

refractory to the transcriptional silencing during integration as evidenced by colonyforming<br />

assay. We suggest that our strategy of anti-silencing protection by CpG island<br />

core element may prove generally useful in retroviral vectors.<br />

contact:<br />

Jiri Hejnar<br />

Institute of Molecular Genetics<br />

hejnar@img.cas.cz<br />

Videnska 1083<br />

CZ-14220 Prague 6 (Czechia)


Sarantis Chlamydas, Patrick Heun, Ruggiero Caizzi<br />

The Drosophila melanogaster centromeric region: a<br />

chromosomal domain in a dynamic state<br />

Centromeres are the elements of chromosomes, which assemble the proteinaceous<br />

kinetocore, maintain sister chromatid cohesion, regulate chromosome attachment to the<br />

spindle and direct chromosome movement during cell division. Although centromeric<br />

DNA has diverged rapidly during evolution, kinetocore function and organization are<br />

conserved in eukaryotes. The fundamental unit of the centromere locus is the<br />

centromeric nucleosome. This nucleosome contains a centromere specific histone variant<br />

CenH3 (Cid in Drosophila), replacing canonical H3(Henikoff, S.2000). Even though<br />

centromeres are often embedded in “silent” chromatin, their spectrum of histone<br />

modifications is distinct from the flanking heterochromatin. (Sullivan,B.A.2004).<br />

In this study, we have characterized a 2R-chromosomal inversion in the transgenic<br />

Drosophila melanogaster In2Rhplacw strain. The inversion displaces the pericentromeric<br />

heterochromatic block of 2R, which includes Rsp and Bari1 satellite loci and places a<br />

white+ gene reporter near the centric region, under the influence of position effect<br />

variegation (PEV)(Reuter, G 1992). Using experiments of IF-FISH on extended chromatin<br />

fibers, we studied the centromeric epigenetic “landscape”. We determined that CenH3<br />

staining can spreads over adjacent, previously euchromatic, regions. We propose that<br />

Drosophila centromeric regions normally exist in a dynamic state in which a regional<br />

boundary, probably defined by satellite DNA, separates CEN chromatin from flanking<br />

heterochromatin.<br />

Literature<br />

Henikoff, S., Ahmad, K., Platero, J. S. and van Steensel, B. (2000). Heterochromatic<br />

deposition of centromeric histone H3-like proteins Proc. Natl. Acad. Sci. 97: 716-721.<br />

Reuter, G. and Spierei, P. 1992. Position effect variegation and chromatin proteins.<br />

Bioessays 14: 605:612.<br />

Sullivan B.A. and Karpen,G.H. 2004. Centromeric chromatin exhibits a histone<br />

modification pattern that is distinct from both euchromatin and heterochromatin. Nat<br />

Struct Mol Biol.11:1076-83<br />

contact:<br />

Ph.D Sarantis Chlamydas<br />

Max-Planck-Institute of Immunobiology<br />

chlamydas@immunbio.mpg.de<br />

Stubeweg 51<br />

79106 Freiburg (Germany)


Andrea Just, Falk Butter, Esther Lizano, Michelle Trenkmann, Tony Heitkam, Heike<br />

Betat, Mario Mörl<br />

The function of two conserved elements in the bacterial<br />

Poly(A)Polymerase and CCA-adding enzyme<br />

Bacterial Poly(A)polymerases (PAP) and CCA-adding enzymes are both members of the<br />

polymerase β superfamily. Although catalyzing different reactions (PAP adds poly(A) tails<br />

to RNA 3’-ends and CCA-adding enzyme synthesizes the CCA triplet at the tRNA 3’-end),<br />

they show a high sequence similarity. Here, two conserved structural elements of both<br />

enzymes from E. coli were investigated.<br />

The first element – the highly conserved amino acid templating region (EDxxR-motif) –<br />

selects in the CCA-adding enzyme the nucleotide to be incorporated. In PAP, however,<br />

the function of this element is unclear. Therefore, individual amino acid exchanges were<br />

introduced and the resulting proteins tested for activity. Our results indicate that the<br />

EDxxR motif of PAP is essential for ATP specificity of its nucleotide binding pocket,<br />

whereas the CCA-adding enzyme can tolerate mutations in this motif and has a “backup”<br />

mechanism that enables the mutant enzyme to synthesize the CCA triplet.<br />

In the second analyzed region – a flexible loop in the catalytic head domain of both<br />

enzymes – amino acid replacements as well as reciprocal exchanges and deletions were<br />

introduced and tested. The resulting enzyme variants show that this flexible region is an<br />

essential element for the terminal A-addition catalyzed by the CCA-adding enzyme,<br />

whereas it is dispensable in PAP.<br />

Our results indicate how these regions in the enzymes contribute to an effective and<br />

accurate catalysis. Furthermore, it seems that PAP, probably having a structure highly<br />

similar to the CCA-adding enzyme, is fixed in a conformational state that restricts its<br />

nucleotide specificity towards ATP.<br />

contact:<br />

Andrea Just<br />

Universität Leipzig Fakultät für Biowissenschaften<br />

Biochemie<br />

justa@uni-leipzig.de<br />

Brüderstr. 34<br />

04103 Leipzig (Deutschland)


Stefan Ehrentraut, Jan Weber, Ann E. Ehrenhofer-Murray<br />

The HDAC Rpd3 functions in boundary formation by removal of<br />

Sir2 substrate<br />

In Saccharomyces cerevisiae, spreading of the telomeric SIR heterochromatin is<br />

prevented by the activity of boundary elements. So far, boundaries have been<br />

associated with chromatin opening activities, like histone acetyltransferases (HATs) or<br />

histone methyltransferases. Here, we show that the opposite enzymatic activity, the<br />

histone deacetylase (HDAC) Rpd3, was necessary to prevent the encroachment of<br />

heterochromatin into euchromatin at telomeres in S. cerevisiae.<br />

We found by ChIP analysis that in the absence of Rpd3, the SIR complexes were<br />

mislocalized to more centromere-proximal regions. Quantitative RT-PCR showed that SIR<br />

proteins repressed subtelomeric genes in rpd3Δ cells, suggesting a role for Rpd3 in the<br />

restriction of telomeric heterochromatin. When combined with the absence of a known<br />

boundary factor, the HAT SAS-I, rpd3Δ caused inappropriate SIR spreading that was<br />

lethal to yeast cells. Significantly, the lethality of sas2Δ rpd3Δ was suppressed by sir<br />

deletions, suggesting parallel functions for the two enzymes in restricting SIR proteins to<br />

heterochromatin. Furthermore, Rpd3 was capable of creating a boundary when targeted<br />

to the telomere, demonstrating boundary function for Rpd3. Our experiments suggest<br />

that histone deacetylation through Rpd3 deprives the HDAC Sir2 of the ability to<br />

generate the metabolite O-acetyl-ADP-ribose during its NAD+-dependent deacetylation,<br />

which then prevents SIR propagation along the chromatin fiber.<br />

contact:<br />

Stefan Ehrentraut<br />

Universität Duisburg-Essen<br />

Abteilung für Genetik<br />

stefan.ehrentraut@uni-due.de<br />

Universitätsstr. 5<br />

45117 Essen (germany)


Marcus Buschbeck, Iris Uribesalgo, Luciano Di Croce<br />

The histone variant macroH2A regulates key developmental<br />

genes<br />

The most extensive epigenetic modification is the complete exchange of a canonical<br />

histones for a variant protein. Among all known histone variants, the so-termed<br />

macroH2A differs most from its canonical counterpart and is at the same time the least<br />

understood. In addition to a homologous histone domain, macroH2A possesses a large Cterminal<br />

domain of unknown function – the macro domain. Incorporated in nucleosomes,<br />

the macro domain which is about twice the size of the histone domain itself, protrudes<br />

out of the compact structure of the nucleosome. To approach the function of macroH2A<br />

we used a dual approach focusing on the identification of interacting proteins as well as<br />

of target genes. Using a cell culture model for stem cells, we first analyzed chromatin<br />

immunoprecipitations of macroH2A with DNA microarrays covering the promoter regions<br />

of 17.000 genes. Among the identified target genes were many genes known to be<br />

involved in the regulation of development and cell fate decisions. We are currently<br />

testing whether depletion of macroH2A does affect critical steps in development.<br />

contact:<br />

Dr Marcus Buschbeck<br />

Center for Genomic Regulation<br />

PRBB<br />

marcus.busschbeck@crg.es<br />

Dr Aiguader 88<br />

08003 Barcelona (Spain)


Andreas May, Daniela Weise, Kurt Reifenberg, Thomas Haaf, Ulrich Zechner<br />

The impact of ovarian stimulation on the cellular epigenome in<br />

preimplantation mouse embryos<br />

Ovarian stimulation seems to impair genome-wide methylation reprogramming,<br />

implantation and fetal development in mice and to increase the risk for imprinting<br />

disorders in humans. To reveal the impact of ovarian stimulation on imprinted gene<br />

methylation, we analyzed differentially methylated regions of H19 and Snrpn by<br />

conventional bisulphite sequencing as well as bisulphite pyrosequencing in mouse 4-cell,<br />

8-cell, and morula stage embryos derived from superovulated and non-superovulated<br />

matings of C57BL/6J females with either C57BL/6J or Mus musculus castaneus (CAST/Ei)<br />

males. In preimplantation embryos from C57BL/6J inbred matings, a significant loss of<br />

methylation of H19 and Snrpn was found after superovulation. In contrast, our H19<br />

methylation analysis of preimplantation embryos from superovulated and nonsuperovulated<br />

intersubspecific (C57BL/6J x CAST/Ei) matings, which allowed<br />

discrimination of parental alleles by a SNP, revealed no dramatic effect of ovarian<br />

stimulation, but very similar methylation levels and expected methylation patterns with<br />

the paternal and maternal allele predominantly methylated and unmethylated,<br />

respectively. However, a significant percentage of both superovulated and<br />

nonsuperovulated intersubspecific morula stage embryos displayed aberrant methylation<br />

on the maternal H19 allele. The observed discrepancy in methylation levels between<br />

superovulated embryos from inbred and intersubspecific matings may be due to the<br />

action of complex modifiers acting in the intersubspecific genetic background. On the<br />

other hand, in inbred embryos one can not distinguish between parental alleles and,<br />

therefore, not exclude a PCR amplification bias due to the very small number of analyzed<br />

cells.<br />

contact:<br />

Andreas May<br />

Klinikum der Johannes Gutenberg Universität Mainz<br />

Institut für Humangenetik<br />

may@humgen.klinik.uni-mainz.de<br />

Langenbeckstraße 1<br />

55101 Mainz (Deutschland)


Andreas Thomae, Dagmar Pich, Jan Brocher, Christian Berens, Robert Hock, Wolfgang<br />

Hammerschmidt, Aloys Schepers<br />

The interaction between ORC and the high mobility group<br />

protein HMGA1a creates site-specific replication origins<br />

In all eukaryotic cells, origins of DNA replication are characterized by the binding of the<br />

origin recognition complex (ORC). How ORC is positioned to sites where replication<br />

initiates is unknown, because metazoan ORC binds DNA without apparent sequence<br />

specificity. Thus, additional factors might be involved in ORC positioning. Our<br />

experiments indicate that a family member of the high-mobility group proteins,<br />

HMGA1a, can specifically target ORC to DNA. Coimmunoprecipitations and imaging<br />

studies demonstrate that HMGA1a interacts with different ORC subunits in vitro and in<br />

vivo. This interaction occurs mainly in AT-rich heterochromatic regions to which HMGA1a<br />

localizes. Fusion proteins of HMGA1a and the DNA-binding domain of the viral factor<br />

EBNA1 or the prokaryotic tetracycline repressor, TetR, can recruit ORC to cognate<br />

operator sites forming functional origins of DNA replication. When HMGA1a is targeted to<br />

plasmid DNA, the prereplicative complex is assembled during G1 and the amount of ORC<br />

correlates with the local concentration of HMGA1a. Nascent-strand abundance assays<br />

demonstrate that DNA replication initiates at or near HMGA1a-rich sites. Our<br />

experiments indicate that chromatin proteins can target ORC to DNA, suggesting they<br />

might specify origins of DNA replication in metazoan cells.<br />

contact:<br />

Dr Aloys Schepers<br />

Helmholtz Zentrum München<br />

Department of Gene Vectors<br />

schepers@helmholtz-muenchen.de<br />

Marchioninistrasse25<br />

81377 München (Germany)


Tomasz Jurkowski, Nils Anspach, Lilia Kulishova, Wolfgang Nellen, Albert Jeltsch<br />

The M.EcoRV DNA methyltransferase uses DNA bending for<br />

recognition of an expanded EcoDam recognition site.<br />

The M.EcoRV DNA methyltransferases recognizes GATATC sites. It is related to EcoDam,<br />

which methylates GATC sites. The DNA binding domain of M.EcoRV is similar to that of<br />

EcoDam suggesting a similar mechanism of DNA recognition. We show that K11 of<br />

M.EcoRV is involved in recognition of Gua1 and R128 contacts the Gua in base pair six.<br />

These residues correspond to K9 and R124 in EcoDam, which recognize the Gua residues<br />

in both strands of the Dam recognition sequence, indicating that M.EcoRV and EcoDam<br />

make similar contacts to outermost base pair of their recognition sequences. In contrast<br />

to EcoDam M.EcoRV considerably bends the DNA (59±4°) suggesting indirect readout of<br />

the AT rich inner sequence. DNA bending could be a general mechanism for recognition<br />

of expanded target sites during evolution. R128A is inefficient in DNA bending and<br />

binding whereas K11A shows relaxed sequence specificity. These results suggest that<br />

the Gua6-R128 contact forms early and leads to a pause in the linear diffusion of the<br />

enzyme along the DNA. After DNA bending, the K11- Gua1 contact forms.<br />

Literature<br />

Horton, J.R., Liebert, K., Hattman, S., Jeltsch, A. & Cheng, X. (2005) Transition from<br />

nonspecific to specific DNA interactions along the substrate-recognition pathway of dam<br />

methyltransferase. Cell, 121, 349-361.<br />

Beck, C., Cranz, S., Solmaz, M., Roth, M. & Jeltsch, A. (2001) How does a DNA<br />

interacting enzyme change its specificity during molecular evolution? A sitedirected<br />

mutagenesis study at the DNA binding site of the DNA-(Adenine-N6)-methyltransferase<br />

EcoRV. Biochemistry, 40, 10956-10965.<br />

contact:<br />

Tomasz Jurkowski<br />

Jacobs University Bremen<br />

Biochemistry<br />

t.jurkowski@jacobs-university.de<br />

Campus Ring 1<br />

28759 Bremen (Germany)


Yamuna Gangadharan, Gary Karpen, Patrick Heun<br />

The role of Drosophila SUMO E3 ligase dPIAS in Chromosome<br />

and Nuclear Organization<br />

PIAS (Protein Inhibitors of Activated STAT) is a protein family capable of activating and<br />

repressing transcription depending on their interacting proteins. PIAS protein family<br />

members are conserved from yeast to mammals. In Drosophila, dPIAS also called as<br />

Su(Var) 2-101, is encoded by a single gene locus and can also act as a SUMO E3 ligase<br />

for SUMO conjugation. Mutations in Drosophila dPIAS display hypocondensed<br />

chromosomes, disorganized interphase chromosomes, melanotic tumors, and affect<br />

telomere-telomere and telomere-nuclear envelope interactions2. Twelve splice variant<br />

isoforms of dPIAS have been predicted. The protein isoforms of dPIAS are localized in<br />

different patterns in peripheral and internal regions of the nucleus when transfected into<br />

Drosophila Schneider S2 cells. The ectopic expression of dPIAS isoforms also effects the<br />

localization of some potential targets, including the GAGA transcription factor.<br />

Preliminary gel filtration data suggests that different isoforms forms distinct high<br />

molecular weight complexes. The goal of my project is to determine the interacting<br />

partners of dPIAS isoforms<br />

Literature<br />

1) Reuter G. and I. Wolff, “Isolation of dominant suppressor mutations for position-effect<br />

variegation in Drosophila melanogaster” Mol Gen Genet 182, 516–519 (1981).<br />

2) Kumar L. Hari., et al. “The Drosophila Su(Var) 2- 10 locus regulates chromosome<br />

structure and function and encodes a member of the PIAS protein family”. Genes &<br />

Development . 15, 1334 – 1348 (2001).<br />

contact:<br />

Ph.D student Yamuna Gangadharan<br />

Max-Planck-Institute of Immunbiology<br />

gangadharan@immunbio.mpg.de<br />

Stübeweg 51<br />

79108 Freiburg (D)


Tuan Nguyen, Mahmoud Youness, Andrea Tedeschi, Andrew Green, Kirsi Forsberg,<br />

Simone Di Giovanni<br />

The role of NFAT in axonal outgrowth and regeneration<br />

Axons of the adult mammalian central nervous system (CNS) do not regenerate<br />

following injury due to the presence of an inhibitory environment and the lack of intrinsic<br />

pro-growth capacity. However, transcription mediated gene expression, essential for<br />

neurite and axon outgrowth during development, is often recapitulated following axonal<br />

injury in the peripheral nervous system, where sprouting and regeneration do occur.<br />

The nuclear factor for activated T cells (NFAT) is a family of transcription factors that<br />

plays a role in axon growth and guidance during neuronal development, however direct<br />

transcriptional targets for NFAT in developing and regenerating neurons have not been<br />

identified. We hypothesize that developmental genes regulated by NFAT might also play<br />

a role in axon sprouting and regeneration in the adult CNS.<br />

In search for potential NFAT transcriptional targets, we performed an in silico promoter<br />

analysis, which revealed several putative NFAT binding sites within the promoter region<br />

of an important pro-axon outgrowth and pro-regeneration gene, the growth associated<br />

protein-43 (GAP-43).<br />

Chromatin immunoprecipitation and transfection experiments showed that neuronal<br />

enriched NFAT3 occupies the GAP-43 promoter and drives its expression in PC12 cells<br />

and cultured primary neurons. In addition, NFAT3 is found in the nucleus and occupies<br />

the GAP-43 promoter in the cortex during a developmental window where neurons are<br />

actively extending axons and establishing connections (from E16 to P1). Next, we<br />

observed that overexpression of NFAT3 promotes both GAP-43 expression and<br />

neurite/axon outgrowth. Taken together, our data unveil a previously unknown NFAT3<br />

dependent molecular pathway for neurite and axon outgrowth.<br />

contact:<br />

PhD Tuan Nguyen<br />

Universität Tübingen<br />

Hertie Institut<br />

tuan.nguyen@medizin.uni-tuebingen.de<br />

Otfried-Müllerstr. 27<br />

72076 Tübingen (Deutschland)


Christine Vogler, Tanja Waldmann, Lora Braun, Mirek Dundr, Robert Schneider<br />

The tale of a tail - Histone H2A and its C-terminal tail<br />

In the eukaryotic nucleus the DNA is organized in the form of chromatin. The basic unit<br />

of chromatin is the nucleosome consisting of 146 bp of DNA wrapped around an octamer<br />

of the four core histones H2A, H2B, H3 and H4. The histone proteins are not only<br />

important for the compaction of chromatin but also play an important role in the<br />

regulation of DNA-dependent processes such as transcription and repair.<br />

Posttranslational modifications of the flexible tails of the histones are one way to achieve<br />

this regulation. H2A is the only core histone that not only has an N-terminal tail but<br />

additionally contains a flexible C-terminal tail. This tail is thought to be located at the<br />

entry and exit site of the nucleosomal DNA. Almost nothing is known about the role of<br />

this tail in chromatin structure and function nor about proteins interacting with it. We<br />

were able to show that this tail is important for nucleosome stability in vitro and in vivo<br />

and that its deletion significantly increases nucleosome mobility. Furthermore, we found<br />

that expression of C-terminally truncated H2A influences cell proliferation and cell cycle<br />

progression in vivo. This is the first demonstration of a biological function for the H2A Cterminus.<br />

contact:<br />

Christine Vogler<br />

MPI for Immunobiology<br />

Vogler@immunbio.mpg.de<br />

Stuebeweg 51<br />

79108 Freiburg (Germany)


Myriam Ekici, Mathias Hohl, Gerald Thiel<br />

Transcription of genes encoding synaptic vesicle proteins in<br />

human neural stem cells:chromatin accessability, histone<br />

methylation pattern and essential role of REST<br />

Human HNSC.100 neural stem cells differentiate into glia cells when the mitogens bFGF<br />

and EGF are removed from the culture medium. This differentiation was blocked by<br />

activation of a Raf1-estrogen receptor fusion protein, indicating that downregulation of<br />

the ERK signaling pathway is required for gliogenesis. Incubation of the cells with<br />

retinoic acid in the absence of mitogens induced neuronal differentiation that was<br />

accompanied by an upregulation of neuronal gene expression, and a downregulation of<br />

GFAP and nestin expression. Chromatin immunoprecipitation experiments revealed that<br />

retinoic acid treatment changed the histone code of neuronal genes, encoding synapsin<br />

I, synaptophysin and synaptotagmins II, IV and VII, from methylation of lysine residue 9<br />

to methylation of lysine residue 4 of histone 3. Furthermore, binding of histone<br />

deacetylase-1 as well as binding of the transcriptional repressor REST was reduced or<br />

abolished following retinoic acid treatment, indicating that transcriptional repression of<br />

neuronal genes via histone deacetylation was impaired. Accordingly, treatment of neural<br />

stem cells with the histone deacteylase inhibitor trichostatin A induced expression of<br />

synapsin I, synaptophysin, synaptobrevin 2 and synaptotagmins II, IV and VII.<br />

Moreover, expression of a mutant of REST that contained a transcriptional activation<br />

domain enhanced expression of synapsin I, synaptophysin, synaptobrevin 2 and<br />

synaptotagmins II, IV and VII in undifferentiated neural stem cells. These data indicate<br />

that these neuronal genes were accessible for the REST mutant in human neural stem<br />

cells, although the genes were xembedded into a chromatin environment characterized<br />

by the the epigenetic marker for silenced genes, H3metK9.<br />

contact:<br />

myriam ekici<br />

university of saarland medical center<br />

department of medical biochemistry and molecular biology<br />

myriamekici@web.de<br />

gebäude 44<br />

66424 homburg (germany)


Madeleine Meusburger, Mark Helm, Frank Lyko<br />

tRNA targets methylated by the Dnmt2 methyltransferase<br />

Dnmt2 is the most widely conserved member of the family of DNA methyltransferases.<br />

The protein contains ten catalytic motifs that are characteristic for DNA methylating<br />

enzymes. However, the molecular and biological functions of Dnmt2 have remained<br />

enigmatic. It has been shown that the DNA methylating activity is very low in vitro,<br />

which contrasts with its readily detectable methylation of cytosine 38 of tRNA_Asp. In<br />

order to further characterize the activity of Dnmt2 we have used Drosophila Dnmt2 for<br />

the identification of additional RNA targets.<br />

In contrast to published results, we are able to observe in vitro methylating activity of<br />

immunopurified Drosophila Dnmt2 enzyme and in vitro transcribed, unmodified<br />

tRNA_Asp was recognized as a substrate. Furthermore, we identified Drosophila<br />

tRNA_Val as a novel substrate for Dnmt2. Isolation of tRNA_Val out of wildtype and<br />

mutant fly embryos shows an m5C-spot in 2-dimensional thin layer chromatography in<br />

the wildtype tRNA_Val, which is absent in the mutant background. Specific cutting at<br />

position C38 and quantification of the methylation signal was achieved by the application<br />

of the sensitive DNAzyme technique. Our results demonstrate that tRNA methylation by<br />

Dnmt2 is not limited to tRNA_Asp.<br />

contact:<br />

Dipl.-Biol. Madeleine Meusburger<br />

German Cancer Research Center<br />

German Cancer Research Center<br />

M.Meusburger@dkfz-heidelberg.de<br />

Im Neuenheimer Feld 580<br />

69120 Heidelberg (Germany)<br />

additional information<br />

1_Division of Epigenetics, German Cancer Research Center, Im Neuenheimer Feld 580, 69120<br />

Heidelberg, Germany<br />

2_Department of Chemistry, Institute for Pharmacy and Molecular Biotechnology, University of<br />

Heidelberg, Im Neuenheimer Feld 364; 69120 Heidelberg, Germany


Chandan Goswami, Tim Hucho<br />

TRPV4 Biochemically And Functionally Interacts With The<br />

Cytoskeleton<br />

TRPV4 and the cytoskeleton have been reported to influence the development of<br />

mechanical hyperalgesia. If and how these molecules interact is unknown. We now<br />

describe TRPV4 to form a Ca2+-sensitive complex with components of the tubulin-, actin-<br />

and neurofilament-cytoskeleton and the nociceptive signaling molecules PKCe and<br />

CamKII. The C-terminus of TRPV4 is sufficient for complex formation. The interaction<br />

with soluble and filamentous tubulin and actin is direct. Actin and tubulin bind with high<br />

affinity and compete for binding. The presence of TRPV4 strongly increases the amount<br />

of microtubules formed even in presence of microtubule depolymerising agents.<br />

Accordingly, expression of TRPV4 induces striking morphological changes such as<br />

formation of lamellipodia, filopodia, as well as neurite-like structures. These changes are<br />

not restricted to neuron-derived cells but also occur in transfected HaCat, ChoKI, and<br />

HeLa cells. TRPV4 co-localizes with actin and tubulin both in fixed as well as live cells<br />

and stabilizes microtubules at membranous regions. Activation of TRPV4 induces rapid<br />

depolymerization of microtubules. This is accompanied by elongation of filopodial<br />

structures and by a transition of lammelipodial to filopodial structures. Therefore, cell<br />

boundaries and growth cone structures rapidly retract. Accordingly, rat primary sensory<br />

neurons do not extend neurites if cultured in the presence of low doses of TRPV4<br />

agonists. This phenotype is restricted to a subset of nociceptive neurons, which binds to<br />

isolectin B4.<br />

contact:<br />

Dr. rer. nat. Tim Hucho<br />

Max Planck Gesellschaft<br />

Max Planck Institute of molecular Genetics<br />

hucho@molgen.mpg.de<br />

Ihnestrasse 73<br />

14195 Berlin (Germany)


Akuma Divine Saningong, Peter Bayer, Jonathan Wolf Mueller<br />

Unravelling the Function of Human DNA-Binding Protein Par14<br />

in the Cellular Nucleus<br />

The parvulins make up a class of peptidyl prolyl cis/trans isomerases (PPIases, EC<br />

5.2.1.8). PPIases assist in protein folding by accelerating cis/trans isomerisation of<br />

peptide bonds preceding a proline residue within a polypeptide chain. The sequences of<br />

the parvulins (Par) are conserved in both prokaryotes and eukaryotes (Mueller and<br />

Bayer, 2007). On the basis of sequence similarity of Par14 to the well known mitotic<br />

regulator Pin1 and Par14’s enrichment in the cellular nucleus with a dsDNA binding<br />

ability, it has been posited to play a role in cell cycle regulation and chromatin<br />

remodelling (Surmacz et al, 2002; Sekerina et al, 2000). The function of Par14 is still<br />

elusive. In consequence hereof, the theme of this work is devoted to unravel its<br />

function.<br />

Using siRNA technology concomitant with cell culture technique, the expression of the<br />

Par14 was down regulated. Knock down of Par14 was partially ascertained at the<br />

proteomic level. Scrutiny of transfected cells employing fluorescent microscopy revealed<br />

no visible morphological changes. Alternative methods to investigate cell phenotype and<br />

viability are underway.<br />

In an independent approach, three different biochemical sub-nuclear fractionation<br />

methods were applied to study localisation of the endogenous Par14 protein within the<br />

cellular nucleus. With these procedures, it was validated that Par14 was associated with<br />

both the nuclear matrix and chromatin with a higher affinity to the latter.<br />

Proteins which are chromatin and/or nuclear matrix bound have been shown to be<br />

involved in a variety of DNA-dependent activities such as transcription, replication,<br />

recombination, DNA-repair and genomic stability. Par14’s involvement in these<br />

processes is currently being investigated. Taken together, these experiments are<br />

expected to shed more light on the nuclear function of the highly conserved metazoan<br />

protein Par14.<br />

Literature<br />

Mueller JW and Bayer P. 2007. Small Family with Key Contacts: Par14 and Par17<br />

Parvulin Proteins, Relatives of Pin1, Now Emerge in Biomedical Research. Perspectives in<br />

Medicinal Chemistry 2007:1 11–20. Review.<br />

Surmacz TA, Bayer E, Rahfeld JU, Fischer G, Bayer P. 2002. The N-terminal basic<br />

domain of human parvulin hPar14 is responsible for the entry to the nucleus and highaffinity<br />

DNA-binding. J Mol Biol.321(2):235-47.<br />

Sekerina E, Rahfeld JU, Müller J, Fanghänel J, Rascher C, Fischer G, Bayer P. 2000. NMR<br />

solution structure of hPar14 reveals similarity to the peptidyl prolyl cis/trans isomerase<br />

domain of the mitotic regulator hPin1 but indicates a different functionality of the<br />

protein. J Mol Biol. 301(4):1003-17.<br />

contact:<br />

MSc (TUM) Akuma Divine Saningong<br />

University of Duisburg-Essen<br />

ZMB - Biochemie<br />

akuma.saningong@uni-due.de<br />

Universitätstraße 1-5<br />

45117 Essen (Germany)<br />

additional information<br />

1 Structural and Medicinal Biochemistry, University of Duisburg-Essen<br />

2 Division of Molecular Structure, The National Institute of Medical Research (MRC), The Ridgeway,<br />

London NW7 1AA, UK.


Agnieszka Sokol, Aleksandra Kwiatowska, Andrzej Jerzmanowski, Marta<br />

Prymakowska-Bosak<br />

Up-regulation of stress-inducible genes in tobacco and<br />

Arabidopsis cells in response to abiotic stresses and ABA<br />

treatment correlates with dynamic changes in histone H3 and<br />

H4 modifcations<br />

Animal cells react to mitogenic or stress stimuli by rapid up-regulation of immediateearl(IE)<br />

genes and a parallel increase in characteristic modifications of core histones:<br />

chromatin changes, collectively termed the nucleosomal response. With regard to plants<br />

little is known about the accompanying changes at the chromatin level. We have used<br />

tobacco BY-2 and Arabidopsis T87 cell lines to study the nucleosomal response of plant<br />

cells to high salinity, cold and exogenous abscisic acid (ABA). When in quiescent stage,<br />

both tobacco and Arabidopsis cells show the typical nucleosomal response to high<br />

salinity and cold stress, manifested by rapid transient up-regulation of histone H3 Ser-10<br />

phosphorylation, immediately followed by transient up-regulation of H3<br />

phosphoacetylation and histone H4 acetylation. For each of the studied stresses the<br />

observed nucleosomal response was strictly correlated with the induction of stress-type<br />

specifc genes. The dynamics of histone modifications in BY-2 cells in response to<br />

exogenous ABA exhibited a more complex pattern than that evoked by the two abiotic<br />

stresses, probably due to superposition of the primary and secondary effects of ABA. A<br />

rapid increase in H3 Ser-10 phosphorylation was also observed in whole leaves<br />

subjected to high salinity; however, the rate of change in this modification was much<br />

slower than in cultured cells. Together, these results indicate that the quiescent BY-2<br />

and T87 cell lines show a typical nucleosomal response to abiotic stresses and ABA<br />

treatment and may represent suitable models for the study of chromatin-mediated<br />

mechanisms of stress tolerance in plants.<br />

Literature<br />

Axelos M, Curie C, Bardet C, Lescure B (1992) A protocol for transient expression in<br />

Arabidopsis thaliana protoplasts isolated from cell suspension cultures. Plant Physiol<br />

Biochem 30:123–128<br />

Xiong L, Schumaker KS, Zhu J-K (2002) Cell signalling during cold, drought, and salt<br />

stress.<br />

Plant Cell 14:S165–S183<br />

Mahadevan LC, Willis AC, Barratt MJ (1991) Rapid histone H3 phosphorylation in<br />

response to<br />

growth factors, phorbol esters, okadaic acid and protein synthesis inhibitors. Cell<br />

65:775–783<br />

contact:<br />

Ph. D. Marta Prymakowska-Bosak<br />

Warsaw University<br />

Laboratory of Plant Molecular Biology<br />

marta@ibb.waw.pl<br />

Pawinskiego 5A<br />

02-106 Warsaw (Poland)

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

Saved successfully!

Ooh no, something went wrong!