InstItute of GenetIcs
Transcription
InstItute of GenetIcs
2o10 Institute of Genetics & Molecular & Cellular Biology contents Foreword 2 Academics 4 Development & Stem Cells 6 Cancer & Functional Genomics 34 Integrated Structural Biology 62 Translational Medicine & Neurogenetics 80 ICS 100 Platforms 102 Core facilities 108 Facts & Figures 110 Staff scientists 112 Prizes & Distinctions 116 Publications until June 2010 117 Publications 2009 120 Where we are 126 Olivier Pourquié, IGBMC Director Foreword The IGBMC is one of the leading centers in biological research in Europe. It was founded in 1994 by Pierre Chambon, one of the world’s leading figures in biomedical sciences. With its 768 staff the Institute represents one of the largest French biomedical research units associating INSERM, CNRS and the University of Strasbourg. The IGBMC is located close to Strasbourg, a cosmopolitan city hosting the European parliament on the frontier between France and Germany. The IGBMC is a highly international center with 48 different nationalities represented among its personnel and all activities in the Institute are carried out in English. The 43 independent research teams work on a broad array of topics, ranging from the analysis of protein structure to human genetics. The research themes are organised into the following four transversal Programmes. The Integrated Structural Biology Programme consists of teams studying the structure of proteins using a variety of approaches ranging from diffraction analysis and computer modeling to state of the art electron microscopy. The Programme Functional Genomics and Cancer is centered on the analysis of the regulation of gene expression and epigenetics. Several groups in this Programme are also interested in the deregulation of these mechanisms that often lead to cancer. The Programme Development and Stem Cells brings together research groups interested in the formation of the embryo and in stem cell biology in a variety of model systems. Within this Programme, we also see an interface emerging with the physical sciences, associating physicists and cell biologists in the study of complex processes such as morphogenesis. The Programme Translational Medicine and Neurogenetics links all the research teams that are directly interested in biological questions with immediate clinical relevance, such as human genetic diseases. One of IGBMC’s major assets are its excellent core facilities and its outstanding technological platforms, which provide accessible state of the art technologies to the institute’s scientists. One of these platforms, the Mouse Clinical Institute (ICS), is rather unusual in that it is a large structure employing more than 100 people exclusively dedicated to 43 Groups mouse technology. Its activities 106 Staff scientists range from transgenic mouse models production to complex 121 Postdoctoral fellows phenotyping strategies inclu118 PhD students ding behavioral and metabolic 37 Master students analyses. In the following pages 293 Engineers & you will find an overview of Technicians IGBMC’s broad range of re 93 Administrators & search activities and technology General services support, highlighting our stimulating environment which aims to foster excellent training and cross-discipline collaborations. < With the ambition of creating a European centre of excellence in medical research, the IGBMC was founded by Pierre Chambon and installed on its present site in 1994 with the assistance of INSERM, CNRS, the University of Strasbourg and the American laboratories of Bristol Myers-Squibb (BMS). Between 2002 and 2009, following on from Pierre Chambon, the Direction of the IGBMC was assured by Dino Moras and Jean-Louis Mandel, two scientists of international reputation. In October 2009, Olivier Pourquié, a pioneer in research on muscles and skeletal development, elected to return to France to lead the IGBMC, following several years spent in the United States at the Howard Hughes Medical Institute. His principal objective is to confirm and strengthen the status of the IGBMC on the national and international scientific stage. 3 academics academics ● Students & Post-docs Board With its recently launched International PhD Programme, the IGBMC successfully attracts and recruits outstanding students from around the world via a call for applications. In this framework, we provide dedicated scientific and socio-professional training to our students, in accordance with the Graduate School. The training benefits notably from the excellent scientific environment and is tailored to the needs of the students. The courses typically cover all areas of modern biology and state-of-the art technologies offered at the Institute. The training of our students is further enriched by a great diversity of additional activities, of which a flavor is given here: Since October 2004, the Students and Post-Docs Board (SPB) of the IGBMC has been working actively to promote and enhance the scientific and social exchange between the students and post-docs of the Institute, the scientific programmes, the core facilities as well as the platforms. It also links IGBMC’s students/ post-docs with the Direction, and the Life Sciences Graduate School of the University of Strasbourg. After six years of continual growth and improvement, the SPB has become an important partner working in a close relationship with the IGBMC administration. • Special Seminar Series: Eminent international scientists are invited to give a lecture as part of the IGBMC Special Seminar Series. ● Training at IGBMC As one of the leading European centers for biomedical research, the IGBMC provides an outstanding research and training environment. With 239 students and post-docs from over 40 countries, we offer a very lively and dynamic setting, fostering interactions at the IGBMC and beyond. The IGBMC accepts about 37 Master Students and 25 PhD students annually, who are exposed to the latest technological developments and are trained under the supervision of IGBMC group leaders across the Institute’s broad range of research fields. The students are generally enrolled at the Life Sciences Graduate School of the University of Strasbourg, which oversees the thesis progress and delivers the PhD certificate. Several IGBMC researchers teach at the University, including the medical school, thereby promoting links between the University and IGBMC. • Student Special Lecture: Students have the opportunity to invite an outstanding scientist to give the Student Special Lecture. • Award: Each year, a student is awarded a travel fellowship to attend a scientific meeting. • International meetings: Scientific meetings are organized on a regular basis and provide our younger researchers with the opportunity to meet outstanding speakers from different fields. • Guides: Three guides have been developed to facilitate a newcomers’ life: a welcome guide, living in Strasbourg, information guide about the University and the Graduate School and fellowships. • International Cine-Club: Launched in December 2007, the Cine-Club invites staff to discover and learn from cultures that are represented at IGBMC. Movies are selected and introduced by a student/postdoc from abroad that is willing to share some cultural aspects about his or her country. • French and English lunches: Once a month, the SPB arranges a lunch to allow participants to practice their language skills: speaking either French with native French speakers or English with English natives. • Social events: The SPB often organizes parties and sport events to bring together students and post-docs to strengthen bonds and to promote interactions in a different setting. • Lunches with prominent scientists: Students have the opportunity to lunch with the Special Seminar speakers in an informal setting. • Internal seminars and retreats: Internal seminars give students and post-docs the opportunity to present all of their work to IGBMC. Retreats organized by the scientific Programme offer a more informal setting to present findings and to discuss and share knowledge with more senior scientists. • Seminars: We organize career development seminars for our community. • Retreats: In June 2010, the SPB organized the first retreat for students and post-docs in a relaxed setting in the Vosges Mountains.< The SPB organizes the following activities: • Poster sessions: On a monthly basis, students and post-docs present their research projects. Staff from core facilities and platforms are also invited to present their activities. • Scientific workshops: Three workshops are organized every year with the assistance of platforms and researchers of the Institute. Topics includes: Microarray Analysis; Bioinformatics in the post-genomics area; 3D Structure Visualization and Functional Genomics. Two of these workshops have been part of the training within the IGBMC International PhD Programme since 2009. The International PhD Programme Programme Coordination: Dr. Astrid Lunkes PhD Programme Committee: Dr. Elisabeth Georges-Labouesse, Dr. Sophie Jarriault, Dr. Hélène Puccio, Dr. Evi Soutoglou, Dr. Laszlo Tora, Dr. Bernardo Reina-San-Martin, Dr. Marat Yusupov Student affairs: Dr. Katia Befort The PhD Programme Administrator: Laetitia Gonzalez Students & Post-doc Board Coordination & communication: Irène Yujnovsky Workshop: Fabrice Klein Alumni & guides: Pierre Antony, Johann Boehm Information package: Angelita Simonetti, Thomas Hussenet Web site: Florent Colin, Pierre-Eric Lutz Poster sessions: Thomas Daniel, Jérôme Mastio, Adrien Rousseau Events: Sara Milosevic, Elisabeth Daguenet, Martin Moune-Dimala 5 Unravelling the mystery of cell differentiation "Understanding how cells differentiate and how they organize in a 3-dimensional space should enable us to find tools to regenerate human tissues, or to reduce the risk of cancer". M. Labouesse The Development and Stem Cells Programme features 12 teams using the fruit fly, zebrafish, worm, chicken and the mouse as models of interest. The unifying theme of the Programme is cell plasticity, either at the nuclear level as it pertains to the mechanisms that control stem cell maintenance and cell fate reprogramming, or at the cellular level, with regard to the mechanisms that control cell shape changes during morphogenesis. While we all focus on fundamental issues, most of us develop models of human diseases such as, cardiovascular diseases, retinal degeneration, scoliosis, Crohn’s disease, diabetes, infertility and cancer, among others. Stem cells and reprogramming Currently two- thirds of the teams aim to define the genetic and epigenetic Programmes Development & Stem cells Mouse and chicken •Pancreas development and diabetes •Integrin signaling in organ morphogenesis •Retinoic acid signaling and neuronal cell fate •Role of retinoic acid signaling in germ cell specification • Progenitor germ cells and iPS •Somitogenesis and PSM extension •Epigenetic marks in maintenance of blastomere identity Zebrafish •Somitogenesis •Blood flow and cardiovascular development •Cilia in development Fruit fly •Role of miRNAs in neural differentiation •Collective cell migration •Choice between glial and neuronal differentiation Worm •Cell fate reprogramming •Impact of cell division on cellular potential •Secretion and lumen formation •Forces in embryonic morphogenesis Physics and biology •In vitro modeling of cell shape change •Cytokinesis in fission yeast that characterize the toti/pluripotence of stem cells. Some specific questions are “How do early mouse blastomeres remain totipotent?”, “how are pancreas endocrine cells made different from each other?” “How does a pluripotent progenitor restrict its potential to generate only glial or neuronal progenitors?” “How do germ cells differentiate into male and female gametes?” “How do muscle 13 Groups precursors arise from the 23 Staff scientists presomitic mesoderm?”, “how does a cell reprogram 31 Postdoctoral fellows its identity?” The different 23 PhD students labs approach these ques tions through a combination 33 Engineers/Technicians of genetic and cell biology 1 Assistant analyses in mouse, chicken, zebrafish, fly, worm, as well as high throughput proteomic and sequencing methods. Morphogenesis Other teams aim to understand how cells communicate during late stages of embryogenesis when organs achieve their final functional shape. Imaging and cell biology are central to the approaches developed and more recently, the input of physics has become key to these developments. Some specific questions being asked are “How do cells collectively migrate to their final positions?” “How do cells interact with the extracellular matrix to define organ shape?” “How does a cell change its shape?” and “What is the impact of forces on organ formation?” n 7 highlights • Central Nervous System Patterning: We revealed that retinoic acid acts antagonistically to FGFs, but is required to potentiate Sonic hedgehog signaling, during elongation of embryonic trunk structures and patterning of the neural tube. shaping the future Roles of retinoic acid in mouse development and physiology Further work will aim at elucidating common molecular mechanisms operating during neurogenesis within the developing telencephalon and the sensory neurons. This work will also contribute to an understanding of molecular pathways under retinoic acid control that are involved in neural progenitor renewal, fate and differentiation. The pathophysiological consequences of manipulating retinoic acid production or RAR/RXR signaling in specific cell populations will also be investigated. < ●Sensory Organ Development: Several molecular and cellular alterations resulting from invalidated retinoic acid signaling within the embryonic retina, the inner ear, and the olfactory placode were characterized in our laboratory. 1 probing complexity Development of the vertebrate nervous system results from an interplay between all major embryonic signaling pathways and requires coordination of many embryonic events including axial patterning, lateral inhibition, progenitor fate determination, axonal guidance, and ectomesenchymal transformation giving rise to neural crest. Sensory systems are formed by direct out-growth and evagination of brain neuroepithelium in the case of the retina, and the inductive processes leading to the formation of ectodermal placodes generating the lens, the embryonic inner ear (otocyst), and the prospective olfactory epithelium. Our team is interested in deciphering some of the highly pleiotropic functions of the vitamin A derivative retinoic acid, and its nuclear receptors (RARs/RXRs), in these processes. We are using the mouse and chicken systems to invalidate specific actors of this signaling pathway using targeted gene inactivation, as well as pharmacological and siRNA approaches. This work permits us to show that the fine tuning of the activity of two classes of enzymes catalyzing the synthesis (retinaldehyde dehydrogenases), or the catabolism (CYP26 cytochromes), of retinoic acid in specific cell populations, is critical for the development of the central nervous and sensory systems. < Pascal Dollé Studying developmental genes: When «collinearity» leads to complexity ●Brain Development and Physiology: We unravelled specific roles of retinoid signaling in higher brain functions including motor control, cognitive functions and emotional processing. We found that RARbeta signaling controls the development of a subpopulation of dopaminoceptive neurons in the striatum involved in motor control, and that RXRgamma inactivation results in depressive behavior due to impaired dopamine D2 receptor function in the nucleus accumbens. Krzyzosiak A, Szyszka-Niagolov M, Wietrzych M, Gobaille S, Muramatsu S, Krezel W. Retinoid X Receptor Gamma Control of Motivated Behaviours involves Dopaminergic Signalling in Mice. Neuron. 66:908-920. (2010). ●Mouse Models: Novel conditional alleles were generated to study the function of the main retinol dehydrogenase acting during development, and to assess the roles of retinoids produced by specific neuronal populations and by the meninges. n 1- Analysis of neuronal progenitor subpopulations in the developing cerebral cortex. 2- Distribution of stem cells (green) among differentiating heart myocardial cells (red). Lin SC, Dollé P, Ryckebüsch L, Noseda M, Zaffran S, Schneider MD, Niederreither K. Endogenous retinoic acid regulates cardiac progenitor differentiation. Proc Natl Acad Sci U S A. 107(20):9234-9. (2010). Ribes V, Le Roux I, Rhinn M, Schuhbaur B, Dollé P. Early mouse caudal development relies on crosstalk between retinoic acid, Shh and Fgf signalling pathways. Development. 136(4):665-76. (2009). Niederreither K, Dollé P. Retinoic acid in development: towards an integrated view. Nat Rev Genet. 9(7):541-53. (2008). Pascal Dollé received his medical degree in 1992 at Louis Pasteur University in Strasbourg, France. Following a research internship on developmental biology and embryology with Denis Duboule and Pierre Chambon, he obtained a PhD in1992 on the developmental expression features of murine Hox genes and was one of the pioneers of the study of knock-out mice in this gene family. In parallel, Pascal worked on the characterization of retinoic acid receptors function during development. In 1992, he joined Pierre Chambon’s team and characterized several functions of RALDH2, a key enzyme involved in retinoic acid synthesis during embryogenesis. In 2001, he created his own team whose current focus is on the development of the nervous system and sensory organs. n 2 9 highlights • We have found that α6β4 integrin in the hemidesmosome is essential for epidermal/dermal attachment. This integrin and laminin ligands are implicated in genetic forms of epidermolysis bullosa in human patients. • By studying double-mutant mice for integrins to overcome functional redundancy, we have been able to reveal novel roles for this family in tissue morphogenesis processes such as formation of the apical ectodermal ridge in the limb and neural tube closure. 1 probing complexity Tissue morphogenesis and function involve changes in cell shape, cell adhesion and migration. These events which implicate cell-cell or cell-extracellular matrix (ECM) interactions are tightly regulated. We are interested by cell-ECM interactions mediated by the integrin family of adhesion receptors. By binding specific ECM ligands through their extracellular domains and recruiting signaling molecules and cytoskeletal elements in the cytoplasm, integrins serve as sensors of the milieu for the cells and participate in the building of the appropriate cellular responses. Perturbations of integrins/ECM lead to severe pathological conditions (developmental defects, congenital muscular dystrophies, skin blistering and cancer). Our goal is to understand the cellular and molecular mechanisms of integrin activity in vivo and address questions such as tissue and stage requirements of integrins, ligand specificity in vivo, signaling pathways, relevance to human pathologies. To this end, we have mostly used mouse genetics.< shaping the future Integrin function and signaling in tissue morphogenesis, integrity and homeostasis Currently we concentrate our efforts on the role of integrins in the epithelial tissues of skin and intestine. Of interest is the fact that in adults these two tissues also undergo tissue renewal and are submitted to mechanical forces. Both epithelia possess hemidesmosomes. These anchoring junctions at the basal pole of cells are molecular complexes which play a crucial role in the control of epithelial cell shape and polarity. In particular, they ensure the link with the basement membrane outside the cell and the keratin cytoskeletal network inside the cells. These junctions are also important in tissue integrity during stress. Using mouse genetics and cellular approaches, we hope to gain insight about the signaling pathways that are implicated in the formation and function of these junctions.< Elisabeth Georges-Labouesse Cellular and development biology in vivo • Our team was the first to elucidate the role of an integrin in brain and retinal layer organization. • We have found that the Nck interacting kinase (NIK) is a molecular partner for integrin β1. Our recent studies suggest that it is the α5β1 integrin heterodimer, a fibronectin receptor, which interacts with NIK, notably during neuronal migration. n Marchetti G, Escuin S, van der Flier A, De Arcangelis A, Hynes RO, Georges-Labouesse E. Integrin alpha5beta1 is necessary for regulation of radial migration of cortical neurons during mouse brain development Eur J Neurosci. 31:399-409. (2010). Wu C, Ivars F, Anderson P, Hallmann R, Vestweber D, Nilsson P, Robenek H, Tryggvason K, Song J, Korpos E, Loser K, Beissert S, Georges-Labouesse E, Sorokin LM. Endothelial basement membrane laminin alpha5 selectively inhibits T lymphocyte extravasation into the brain. Nat Med. 15(5):519-27. (2009). 1- Localisation of integrin α6 at the basal pole of the basal keratinocytes and hair follicle in skin. A mouse skin section was stained with an antibody against α6 integrin (green). DAPI staining (blue) labels nuclei. 2- Plectin (red) and actin cytoskeleton (green) of a migrating Caco2 cell. Rodius S, Indra G, Thibault C, Pfister V, Georges-Labouesse E. Loss of alpha6 integrins in keratinocytes leads to an increase in TGFbeta and AP1 signaling and in expression of differentiation genes. J Cell Physiol 212(2):439-49. (2007). Haubst N, Georges-Labouesse E, De Arcangelis A, Mayer U, Gotz M. Basement membrane attachment is dispensable for radial glial cell fate and for proliferation, but affects positioning of neuronal subtypes. Development 133(16):3245-54. (2006). After earning a master’s degree in Virology, Elisabeth Georges-Labouesse obtained her PhD at the Pasteur Institute UPMC (Paris) on cutaneous papillomavirus and analyzed the viral Programme, which depended on epidermal cell differentiation steps. She became a CNRS research associate in 1985 and was appointed a post-doctoral position in 1989 at MIT, Cambridge, US with Richard Hynes. While conducting her research at MIT, she focused on the role of extracellular matrix proteins in tissue architecture by using mutant mice. Her findings showed that fibronectin was involved in somite organization. In 1996 she discovered the essential role of a6 integrin in hemidesmosomes for dermal-epidermal attachment. In 2010 Elisabeth’s work continues to focus on the role of integrins in development and the comprehension of the integrin signaling pathway in vivo, using in-utero electroporation, while equally studying the mechanisms of epithelium organization and maintenance of their integrity in skin and intestine. n 2 11 highlights • Cell differentiation and plasticity: We identified the glial lineages and the glial determinant, Glide/Gcm transcription factor, necessary to induce gliogenesis and sufficient to reprogram cells towards the glial fate. We determined Gcm molecular cascade and provided evidence for evolutionary conservation. 1 probing complexity A major challenge in developmental biology is to understand how cell diversity is generated and how cells interact to build the sophisticated metazoan architecture. In humans, defects in such events lead to severe pathologies, from mental retardation to cancer. Evolutionary conservation, sophisticated genetics and simple organization make the fly an ideal tool to study these events in vivo and in vitro, at cellular resolution. The nervous system constitutes one of the most complex tissues, made of neurons and glia of different types. These cells arise from multipotent precursors or stem cells. The analysis of the signals controlling stem cell differentiation and reprogramming has recently drawn much attention, due to potential medical applications. Our goal is to study the molecular and the epigenetic events controlling cell differentiation and reprogramming. During development, many cells move as cohorts to attain their final destination and shape both tissues and organs. Distinct homeostatic cell interactions control directionality, coordination and integrity of the migratory unit. This dynamic event can hardly be tackled in vivo in large animals. We have developed a simple model to analyze the cellular and molecular features of this collective behavior.< shaping the future Cellular and molecular mechanisms of nervous system differentiation The integration of complex molecular cascades and cell interactions secures the differentiation of the diverse cell types and tissues that compose a multicellular organism. Modern biology tackles the molecular, epigenetic and cellular mechanisms underlying these events with unprecedented resolution in space and time. We are convinced that understanding how cells enter a specific differentiative pathway and attain their appropriate position requires a multidisciplinary approach that straddles genetics, molecular and cellular biology, imaging and informatics. Our in vivo and in vitro studies will also address the conservation of these pathways throughout evolution. < • Collective cell migration: We identified a glial migratory chain in the fly nervous system and specific cell interactions required for collective migration by genetic, ablation and time-lapse approaches. Glia interacts with the neuronal substrate to gain directionality. Also, glial cells at the chain tip (pioneers) explore the environment and trigger the movement of following glia. In addition, homotypic interactions within the migratory unit control the extent of migration and chain integrity. Angela Giangrande Drosophila’s brain • An animal model for human pathologies: We characterized a molecular pathway linked to the Fragile X Mental Retardation Protein, the mutation of which induces most frequent cognitive defects in humans. We also created a transgenic model for a severe and frequent form of human retinal degeneration (Retinitis Pigmentosa). n Su-chun Ho M, Chen H, Chen M, Jacques C, Giangrande A, and Chien C-T. Gcm protein degradation mediated by F-box E3 ligases suppresses proliferation of glial progenitors. PNAS. 106:6778-83. (2009). 1- Pupal eye triple labeled with glia (red), photoreceptor (green) and axonal (blue) markers. 2- Neural and glial labeling on pupal wing. Aigouy B, Lepelletier L, and Giangrande A. Chain migration requires pioneer cells. J Neurosci. 28:11635-41. (2008). Soustelle L, Trousse F, Jacques C, Ceron J, Cochard P, Soula C, and Giangrande A. Neurogenic role of Gcm transcription factors is conserved in chicken spinal cord. Development. 134:625-634. (2007). De Iaco R, Soustelle L, Kammerer M, Sorrentino S, Jacques C, Giangrande A. Huckebein mediated autoregulation of Glide/ Gcm triggers glia specification. EMBO J. 25:244-254. (2006). 2 After studying thalassemia at Bari University in Italy, Angela Giangrande joined G. Richards’ team in 1984 and obtained her PhD in 1988 on transcription regulation in Drosophila. During her postdoc research with J. Palka at the University of Washington (Seattle) she began a long fascination with the development of the Drosophila nervous system where she discovered the origin of adult peripheral glial cells. Since 1992, she has led a team on nervous system differentiation and has studied molecular and cellular associated mechanisms. She uses the Drosophila as a model to study conserved mechanisms and molecules. On glial differentiation, her work led to the identification and characterization of the glial determinant Glide/Gcm (1996). She now focuses on the mechanisms controlling cell plasticity and collective cell migration. Her research on human disease modeling led to the discovery of the link between the Fragile X Mental Retardation protein and actin remodeling (2003). A transgenic model of retinal degeneration (2005), has also been developed. Angela coordinated the imaging facility until 2009. n 13 highlights 1 probing complexity Future strategies for cell replacement therapies and regenerative medicine strongly depend on our knowledge of the detailed mechanisms that control the differentiation of multipotent stem cells into highly specialized cells. Along these lines of research, our goal is to understand how pancreatic and intestinal endocrine cells are generated from endodermal stem/progenitor cells during embryogenesis towards adult life. Pancreatic endocrine cells are clustered in islets within the exocrine tissue and include insulinsecreting beta cells that control glucose homeostasis and become destroyed in Type-1 diabetes. Intestinal endocrine cells, also called enteroendocrine cells, are closely related endocrine cells, found within the intestinal epithelium, which secrete a variety of hormones promoting pancreatic insulin secretion to regulate various aspects of digestion. Particularly, we focus on the role of transcription factors and signals in the control of cell fate choices and the acquisition of the generic and specific properties of islet and enteroendocrine cell during organogenesis. We hope that our studies will contribute to the development of a cell based therapy in diabetes, as well as to understand the mechanisms underlying the pathophysiology of islet and enteroendocrine hormone failure in the human.< • Cell fate choice: We made the seminal discovery that the transcription factor Neurogenin3 (Ngn3) is the master gene controlling endocrine cell fate choices both in the embryonic pancreas and intestine. Mice without Ngn3 lack islet and enteroendocrine cells are diabetic and die postnatally. • Differentiation Programme: We determined the transcriptome of islet progenitors leading to the identification of Insm1 and Rfx6 transcription factors as two downstream effectors of Ngn3 endocrinogenic function. We reported for the first time that Rfx6 is essential for proper islet cell development. • Beta cell neogenesis: We contributed to the demonstration that new beta cells can arise, de novo, from progenitor cells in the injured adult mouse pancreas. Mellitzer, G., Beucher, A., Lobstein, V., Michel, P., Robine S., Kedinger, M., and Gradwohl, G. Loss of enteroendocrine cells in mice alters lipid absorption and glucose homeostasis, and impairs postnatal survival. Journal of Clinical Investigation. 120:1708-21. (2010). shaping the future Control of endocrine cell differentiation in the pancreas and intestine An important bottleneck in the generation of beta cells from human ES cells is the inability to obtain functional, glucose responsive, insulin-secreting beta cells in vitro. For this reason, our current studies emphasize unappreciated signaling pathways and mechanisms of islet progenitor cell maturation. We also generated mouse models mimicking human beta cell deficiency to determine whether mouse and human beta cells differentiate according to similar genetic pathways and to explore mechanisms of beta cell regeneration in the adult pancreas. In the intestine we further investigate the role of enteroendocrine cells/hormones in the maintenance of the homeostasis of the intestinal epithelium and in the control of nutrient absorption and energy metabolism.< Gérard Gradwohl Mechanisms of stem/progenitor cells differentiation • Signals: We demonstrated that retinoic acid signaling is required for the normal development of the dorsal pancreas. • Enteroendocrine cells: We generated the first mouse model with a specific ablation of enteroendocrine cells and discovered that enteroendocrine cells/hormones are essential for life and control intestinal crypt cell homeostasis as well as lipid absorption. n Soyer, J., Flasse, L., Raffelsberger, W., Beucher, A., Orvain, C., Peers, B., Ravassard, P., Vermot, J., Voz, M. L., Mellitzer, G. and Gradwohl, G. Rfx6 is an Ngn3-dependent winged helix transcription factor required for pancreatic islet cell development. Development 137:203-212. (2010). 1- Enteroendocrine progenitor cells (Ngn3-eYFP in yellow) in intestinal crypts of adult mouse small intestine. 2- Insulin-producing beta cells (red) in the adult mouse pancreas express Rfx6 transcription factor (green nuclei). Xu,X., D’Hoker,J., Stange,G., Bonne,S., De Leu,N., Xiao,X., Van de,C.M., Mellitzer,G., Ling,Z., Pipeleers,D., Bouwens,L., Scharfmann,R., Gradwohl,G., and Heimberg,H. Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas. Cell. 132:197-207. (2008). Martin,M., Gallego-Llamas,J., Ribes,V., Kedinger,M., Niederreither,K., Chambon,P., Dolle,P., and Gradwohl,G. Dorsal pancreas agenesis in retinoic acid-deficient Raldh2 mutant mice. Dev. Biol. 284:399-411. (2005). As a PhD student at the IGBMC (Strasbourg), working on DNA repair, Gérard Gradwohl showed that the Zinc-finger domains of Poly (ADP-ribose) polymerase detect DNA strand breaks (1990). He then became interested in development biology and relocated as postdoc to the Mount Sinai Hospital (Toronto, Canada) where he studied, in 1992, TIE2, a tyrosine kinase receptor that controls angiogenesis in the mouse embryo. Gérard then joined the team of Francois Guillemot at the IGBMC on the role of bHLH transcription factors in neurogenesis. In 2000, he showed that Neurogenin3, one of the factors studied, was also key for pancreatic beta cell differentiation, a discovery which promoted the creation of his Inserm team at Hautepierre Hospital (Strasbourg). His group then determined that neurogenin3 also controls endocrine fate decision in intestinal stem cells. In 2008, he joined the IGBMC to develop his research on the regulation of endocrine cell differentiation in the pancreas and intestine. n 2 15 highlights 1 probing complexity Our focus is to understand how neurons are specified within uniform fields of cells. We use the power of Drosophila genetics to address this question. The regulation of the two key proneural genes Achaete and Scute provide the identity and the position of most of the sensory organs within the peripheral nervous system. Achaete and Scute encode bHLH transcription factors, which, together with the Zn-finger transcription factor GFI/Senseless, confer the potential to develop a neural fate to cells. The general architecture of the peripheral nervous system, also called prepattern, is controlled by specific transcription factors. These bind to enhancers common to both Achaete and Scute which in turn promote proneural expression in small fields of cells, the proneural clusters, at reproducible antero-posterior and dorso-lateral coordinates. The spacing between neuron precursors is achieved by a mechanism known as lateral inhibition, which involves the Notch receptor and its ligand Delta. We recently showed that several decisions mediated by Notch and Delta are fine-tuned and controlled by a common set of microRNAs. Collectively, these mechanisms define the positions and identities of each sensory organ; similar mechanisms are conserved in human haematopoiesis and might be at play in the etiology of T-ALL leukemia. < • Our genetic screens led us to characterize several zinc finger-containing transcription factors (the GATA factor Pannier, the FOG homologue Ushaped, the LIM proteins Islet and dLMO) that together define the prepattern for the median thorax. • We have highlighted the importance of the Ldb homologue Chip in stabilizing enhancerpromoter communications during Achaete/Scute transcription. Moreover, we showed that dLMO acts as an activator, while Islet and Ushaped behave as negative regulators, of the Pannier/Chipdependent Achaete/Scute expression. Both Ushaped and Pannier recruit the dCtBP protein and its partner HDAC1 in a repressive mechanism involving alteration of local chromatin structure, while Islet repression seems to mediate competition for Chip. Heitzler P. Biodiversity and non canonical Notch signaling. In «Notch signaling». (R. Kopan, Ed),Elsevier Inc., San Diego. Current Topics in Developmental Biology. 92:427-451. (2010). shaping the future Genetic and molecular analysis of early neurogenesis in Drosophila melanogaster The regulation of Achaete/Scute by Islet seems to depend on the specific enhancer context. In order to clarify how enhancer position and context can lead to different outcomes, we have developed sensors in Achaete 5’UTR that allow us to measure in vivo variations in enhancer-promoter co-operations. We have also identified a novel enhancer, Scutellar, that appears to mediate such variation in Achaetae/Scute expression Currently, we are exploring three mir-9 and three mir-9* products as regulators of a clock-pulsed feedback loop of Notch. With the use of new tools, we are performing a unique genetic analysis to reveal the individual contribution and expression patterns for each mir hairpin. Similar mechanisms may be involved in acute leukemia and several non-Hodgkins lymphomas in human. We will perform microarray assays to check whether specific miRNA may influence early LMO2- or Notch-dependent steps of T-cell differentiation in mouse.< Pascal Heitzler Formal genetics as a vocation • The main dLMO isoform, named dLMO-PA, controls both neurogenesis and wing development. dLMO is homologous to the leukaemogenic factor LMO2 that is found in similar transcription complexes in human. We found that an ancient family of miRNAs (mir-9) regulates dLMO abundance, and in turn controls neurogenesis in Drosophila. Thus, as for LMO2, dLMO stoichiometry is critical in vivo. n Biryukova I, Asmar J, Abdesselem H & Heitzler P. Drosophila mir-9a regulates wing development via fine-tuning of LIM only factor, dLMO. Dev Biol. 327:487-496. (2009). Biryukova I & Heitzler P. Drosophila C-terminal binding protein, dCtBP is required for sensory organ development and sharpens a proneural transcriptional activity of GATA factor, Pnr. Dev Biol. 323:64-75. (2008). 1- MicroRNA expression in imaginal disc. 2- MicroRNA expression in brain. Pascal Heitzler developed very early on an interest for genetics. His encounter with Pat Simpson in 1985 confirmed his vocation and he joined her team for his PhD on the analysis of developmental genetics on sensory organ patterns Drosophila (1993). Together they were the first to reveal the role of Notch/Delta as receptor/ligand during lateral inhibition. In 1994, he obtained a post-doc position at the IGBMC and revealed a feedback loop mechanism in Notch-Delta signaling. He then discovered a genetic network, including GATA, FOG, LIM-HD, Ldb, LMO, SWI/SNF transcription factors which are involved in sensory bristle prepatterning. In 2001, he became a team leader and described the role of Chip in proneural enhancerpromoter communications, as well as a new non-canonical Notch pathway. His recent projects focus on miRNAs that control all the described processes. n Asmar J, Biryukova I & Heitzler P. Drosophila dLMOPA isoform acts as an early activator of achaete/ scute proneural expression. Dev Biol. 316:487-497. (2008). Biryukova I & Heitzler P. The Drosophila LIM-Homeodomain Islet protein antagonizes proneural cell specification in the peripheral nervous system. Dev Biol. 288:559-570. (2005). 2 17 highlights 1 probing complexity How differentiated cells can change their identity is a fascinating question in biology and has implications for the development of regenerative medicine strategies. Numerous examples of cellular plasticity in physiological, experimental and pathological settings exist, but how a differentiated cell can change its identity remains unknown. Our research tackles this important question by employing a powerful and innovative model, the nematode C. elegans. We study in vivo cell reprogramming events at the single cell level. Specifically, we focus on a differentiated rectal cell (named Y) that during normal development changes its identity into a motoneuron (named PDA). We notably investigate the cellular and molecular events underlying Y-to -PDA cellular reprogramming. Our system also allows us to identify the molecular players and to dissect the cellular requirements during cell type conversion in vivo. Comparing different cell plasticity events, we are assessing what key aspects have been conserved in cell reprogramming. Our integrated approach will contribute to unraveling the key mechanisms that allow a differentiated cell to become plastic and change its identity. This knowledge has significant therapeutic implications, as it will further our understanding as to how certain cancers arise, and will improve our ability to reprogram cells for regenerative medicine purposes. < • Cellular potential: We have found that a pulse of Notch signal is necessary for the Y cell to acquire the competence to switch identity. • Direct cell type conversion occurs in vivo through distinct intermediate cellular steps, not via a mixed cellular identity. Remarkably, Y-toPDA conversion, which occurs in the absence of cell division, involves as a first step the complete erasure of the initial identity. Importantly, this dedifferentiated state has limited cellular potential. This suggests that unlike reprogramming induced in vitro, natural in vivo reprogramming is much more constrained. • Technical development: We have developed Deep Mapping, an innovative method using next generation sequencing, which allows mutation identification without any prior SNP Zuryn S., Le Gras S., Jamet K. and Jarriault S. Deep Mapping: A novel mutagen-based method for direct mapping and mutation identification using deep sequencing, in Press Genetics. (2010). shaping the future In vivo analysis of cellular plasticity in C. elegans The aim of our work is to systematically address the cellular and molecular mechanisms that allow a differentiated cell to be reprogrammed and change identity. Such knowledge has profound implications for the elaboration of therapeutic strategies, both in cancerology and in cellular therapy. Many technical obstacles remain in complex organisms to address these questions in vivo, in particular because it is virtually impossible to track cells as they get reprogrammed in mammals. Our novel experimental model, combined with integrated molecular and cellular in vivo approaches allows us to bypass these limitations and bring new insights into cellular plasticity. We will focus on the mechanisms that make a specific cell, rather than its neighbors, competent to change identity and the cellular mechanisms that are used to allow its conversion. The comparison of the mechanisms used in different direct reprogramming events will be key for our understanding of cellular plasticity in vivo. knowledge. Deep Mapping is a powerful, yet a conceptually simple strategy that is easy to apply to different model organisms. • Impact of cell division on the cellular potential: Comparison of reprogramming events in C. elegans suggests that different cellular strategies are used to erase the initial identity: retraction from the tissue, which resembles an EMT, or asymmetric cell division. n Jarriault S. LIN-12/Notch signaling: Induction, lateral specification and interaction with the EGF/Ras pathway. Handbook of Cell Signaling 3rd Edition (Eds. R.A. Bradshaw and E.A. Dennis). Elsevier Academic Press. :1891-1895. (2009). 1- Visualization of the adherens junctions that shape the posterior of the worm, including the rectal area. 2- Electron microscopy picture of the rectal area of a young C. elegans larva, showing Y cell epithelial cell. Ventral is to the bottom. Jarriault S.*, Schwab Y. & Greenwald I. A C. elegans model for epithelial-neuronal transdifferentiation. PNAS, 105(10) : 3790-5 *, corresponding author. 5/3/08 « Must Read », « Top 10 Developmental Biology Papers » of Faculty 1000. (2008). Jarriault S. and Greenwald I. Functional interchangeability of ADM-4/Tace and SUP-17/Kuzbanian in C. elegans. Developmental Biology. 287(1):1-10 (2005). < Sophie Jarriault Cellular plasticity and reprogramming in C. elegans Sophie Jarriault obtained her PhD in Alain Israël’s laboratory at the Pasteur Institute (Paris) where she worked on the transduction of the Notch signal in mammals. In 1998, her research led to an original model for signal transduction involving cleavage of the Notch1 transmembrane receptor and release of the intracellular part. She did her postdoc research at Columbia University (New York), where she worked with Iva Greenwald on vulva organogenesis in C. elegans. She notably, involved the COREST corepressor and the LSD1 demethylase in downstream modulation of Notch-mediated target genes expression. In 2006, she was awarded the Young Investigator Starting Grant (ATIP-CNRS) as well as other grants from the ARC, FRM and AFM to start her own team at IGBMC. Sophie chose to focus her research on cellular plasticity. At Present, her team works on the mechanisms of cellular reprogramming in C. elegans, and has established the worm as a powerful animal model to study such processes in a physiological context. n 2 19 highlights • Cell polarity: We previously analysed genes that maintain epithelial polarity and adherens junction integrity. • Forces in morphogenesis: We identified several pathways that control myosin II activity and found that myosin II is mainly active in a subset of cells. Modelling in collaboration with physicists has confirmed our genetic data. 1 probing complexity Our team’s research is based on the interface of cell and developmental biology, focusing on epithelial tissues, which form the architecture of most of our organs. These highly polarized cells that have specific junctional complexes, play an essential role during embryonic morphogenesis, as well as later on in physiology. The goal of our research is to explain how embryos form and grow to generate organs with different properties and shapes. We are dissecting the processes that create a lumen, secrete specific proteins to distinct surfaces, or bring cell shape changes. Many organs include several cell types of epithelial and non-epithelial origins; an important focus of our studies is to understand how these cells interact with each other to achieve higher order tissue morphogenesis. In parallel, we intend to use the knowledge gained from studies in embryos to reveal the causes of pathologies involving structures, such as the hemidesmosome, that are present in C. elegans and mammals. We are mainly using the model C. elegans, combining molecular genetics and modern imaging methods to capitalize on the speed of development and simplicity of this system. < shaping the future Forces and signals in tissue morphogenesis Today, in connection with the analysis of cell shape changes and secretion, my lab is focusing on the input of physical forces in morphogenesis. My view is that physics is at the crossroads of cell and developmental biology, and will provide major input to interpret key biological processes. It offers very important perspectives for the decade to come. More generally, we are reaching a point where quantitative analysis and modelling is becoming essential in biology. To accomplish this, in the coming years, we will progress along two axes: ● Analyze the role of forces in epithelial morphogenesis, including their input on trafficking ● Study how osmotic pressure affects membrane tension and lumen shape.< Michel Labouesse • Hemidesmosomes (HDs): We have contributed to define the nature of HD-like junctions between the epidermis and the ECM and revealed how the spectraplakin VAB-10 maintains epidermal integrity against mechanical stress during morphogenesis. More recently, we could identify proteins that promote HD biogenesis and found that HDs provide a platform for mechanotransduction during morphogenesis. Through epithelial cell differentiation • Trafficking: We characterized the first transmembrane complex acting in apical trafficking. By doing so, we showed that apical secretion of some protein cargos involves the release of exosomes from multivesicular bodies, which is mediated by the V0 sector of the V-ATPase. Zahreddine H*, Zhang H*, Diogon M, Nagamatsu Y, Labouesse M. CRT-1/calreticulin and the E3-ligase EEL-1/ HUWE1 control hemidesmosome maturation in C. elegans development. Curr. Biol. 20:322-327. (* co-first) (2010). • New technique: We designed a protocol to easily bridge time-lapse imaging with electron microscopy (CLEM). n Gally C, Wissler F, Zahreddine H, Quintin S, Landmann F, Labouesse M. Myosin II regulation during C. elegans embryonic elongation: LET-502/ROCK, MRCK-1 and PAK1, three kinases with different roles. Development. 136:3109-19. (Issue cover). (2009). 1- Closing the gap between fast fluorescence videomicroscopy and high resolution electron microscopy through Correlative Light and Electron Microscopy. 2- A 4-colour view of the main C. elegans hemidesmosome components and underlying muscles. Ciarletta, P., Ben Amar, M., and Labouesse, M. Continuum model of epithelial morphogenesis during C. elegans embryonic elongation. Philos Transact A Math Phys Eng Sci. 367:3379-400. (2009). Liégeois, S., Benedetto, A,, Garnier, J.M., Schwab, Y. and Labouesse, M. The V0 ATPase mediates apical secretion of Hedgehog-related proteins through exosomes. J. Cell Biol. 173:949-961. (rated Must Read by F1000). (2006). Having an engineering degree in maths and physics from Ecole Polytechnique, Michel Labouesse began a career in biology and obtained his PhD in genetics in Piotr Slonimski’s laboratory (Gif-sur-Yvette, 1983). His work dealt with the control of mitochondrial gene expression by nuclear genes. Inspired by Sydney Brenner’s work on the nematode C. elegans, he decided to switch to this model in 1989 during his postdoc at MIT. Supervised by H. Robert Horvitz, he was initially interested in understanding how two cells become different during asymmetric cell division. The Zn-finger transcription factor LIN-26 that characterized turned out to be essential for the differentiation of the entire skin. Michel began his research at IGBMC in 1992 and went on to work on epithelial morphogenesis. Today, he leads the Development and Stem Cells Programme. n 2 21 highlights 1 probing complexity Retinoic acid (RA), the biologically active metabolite of vitamin A, exerts a wide range of cellular effects by regulating proliferation, differentiation and apoptosis. At the cellular level, the action of RA is controlled through the modulated expression of cellular enzymes producing or degrading RA, retinaldehyde dehydrogenases (RALDH) or cytochrome P450 hydroxylases (CYP26), respectively. Then, acting in an hormone-like manner, RA binds to nuclear receptors (RAR) that function as transcriptional regulators through controlling, usually in the form of heterodimers with rexinoid receptors (RXR), the expression of RA target genes. Among the various tissues whose physiology depends upon vitamin A action, the seminiferous epithelium of the testis represents the most remarkable paradigm to investigate the pleiotropic effects of RA in vivo, as it integrates the problematic of stem cell renewal, cell proliferation, switching from mitotic to meiotic cell division, programmed cell death and paracrine signaling. Using a combination of innovative genetic, pharmacological and molecular approaches in the mouse, we are studying the cellular and molecular mechanisms that underlie the capabilities of RA to promote spermatogonia differentiation and beyond the differentiation of normal stem cells in vivo.< • Two distinct, sequential, sources of RA involved in spermatogonia differentiation. RA synthesized by somatic Sertoli cells is indispensable only during the prepubertal wave of spermatogenesis. The subsequent differentiation of spermatogonia requires RA synthesized by their progenies, the meiotic spermatocytes, a mechanism which might be instrumental to the perpetuation of the spermatogenetic cycle. • A catabolic barrier protects spermatogonia from untimely differentiation. The peritubular myoid cells that surround the seminiferous epithelium isolate spermatogonia from extratubular sources of RA through synthesizing the RA-degrading enzymes, CYP26. • A model of non-canonical mechanism of action for RAR. As the pathological phenotypes resul- Mascrez B, Ghyselinck NB, Chambon P, Mark M. A transcriptionally silent RXRalpha supports early embryonic morphogenesis and heart development. Proc. Natl. Acad. Sci. USA. 106:4272-4277. (2009). shaping the future Retinoic acid signalling pathways driving stem spermatogonia ontogenesis and differentiation The seminiferous epithelium comprises somatic, supporting, Sertoli cells that contribute to the niche environment directing the fate of spermatogonia. Outside their niche, spermatogonia can become multipotent and therefore represent possible tools for cellular therapy. We propose to identify the genetic networks and to characterize the molecular mechanisms through which RA, notably in Sertoli cells, controls spermatogonia proliferation and differentiation. Our preliminary data indicate that expression of the few genes known to be involved in stem spermatogonia homeostasis is not altered upon impairment of RA function. Thus, RA acts on yet uncovered genetic cascades, the understanding of which will open new perspectives on stem cells with promising therapeutic potential.< Norbert Ghyselinck At the face of Molecular Genetics and Developmental Biology ting from the ablation of RAR and of RXR in Sertoli cells are really different, the canonic RAR:RXR heterodimers do not operate in these cells. This probably also applies to spermatogonia. • A novel switch which controls the choice between mitosis and meiosis. STRA8 protein is involved in the process that leads to stable commitment of differentiated spermatogonia to the meiotic cell cycle. n Mark M, Jacobs H, Oulad-Abdelghani M, Dennefeld C, Féret B, Vernet N, Codreanu CA, Chambon P, Ghyselinck NB. STRA8-deficient spermatocytes initiate, but fail to complete, meiosis and undergo premature chromosome condensation. J. Cell Sci. 121:3233-3242. (2008). 1- Immunostaining of spread nuclei to visualize chromosomes during meiosis 2- Histological section showing pathological accumulation of lipids in the seminiferous epithelium Vernet N, Dennefeld C, Guillou F, Chambon P, Ghyselinck NB, Mark M. Prepubertal testis development relies on retinoic acid but not rexinoid receptors in Sertoli cells. EMBO J. 25:5816-5825. (2006). Mark M, Ghyselinck NB, Chambon P. Function of retinoid nuclear receptors: lessons from genetic and pharmacological dissections of the retinoic acid signaling pathway during mouse embryogenesis. Annu. Rev. Pharmacol. Toxicol. 46:451-480. (2006). Manuel Mark Mark Manuel obtained an MD degree from Strasbourg University and defended his PhD in 1988 on extracellular matrix functions during embryonic development. After joining Pierre Chambon’s team in 1992 he became involved in the phenotypic analysis of Hox gene mutant mice. In 1993 he initiated collaboration with Norbert Ghyselinck who was also a member of Chambon’s team. Norbert Ghyselinck received his PhD in 1991 from Clermont-Ferrand University. His research focused on the regulation of gene expression by nuclear receptors. In 2004 he was appointed a Research Director of the CNRS. Together, Mark and Ghyselinck formed a team at IGBMC to decipher retinoic acid signaling pathways during differentiation of male stem cells. In 2006, they brought to light non-canonic mechanisms of actions for retinoic acid receptors in vivo. n 2 23 highlights 1 probing complexity We are interested in the mechanisms controlling the formation of the body of vertebrates during embryogenesis. In particular, we have been focusing on the process of axis elongation and on segmentation whereby a periodic series of anatomical structures such as vertebrae are formed during organogenesis. Our work relies on developmental biology studies in chicken, mouse and zebrafish embryos combining genetic approaches with genomic strategies such as transcriptomics or high throughput sequencing and bioinformatics as well as sophisticated in vivo imaging. Our goal is to understand the basic principles underlying these morphogenetic processes. We are actively collaborating with physicists and mathematicians to attempt to model these complex molecular and cellular processes in the embryo. Equally we are interested in the clinical relevance of these findings and are exploring the molecular basis of spine patterning defects such as scoliosis in humans. Lastly, we are moving into the field of regenerative medicine, translating our understanding of differentiation of the muscle and vertebral lineages into in vitro strategies to differentiate mouse and human embryonic or reprogrammed stem cells for cellular therapies of degenerative diseases such as Duchenne muscular dystrophy. < • Segmentation clock : We have identified a molecular oscillator associated with the process of rhythmic production of vertebral precursors in the embryo. The pulsatile signal generated by the oscillator travels progressively along the embryo thus generating the spatial periodicity of the future vertebrae. This led us to propose a comprehensive paradigm accounting for vertebrate segmentation. • Axis elongation : We have shown the role of Hox genes in the collinear control of mesodermal cell ingression and proposed a mechanism akin to diffusion involving a gradient of random cell motility to explain the striking terminal elongation process resulting in body formation in the embryo. • Bilateral symmetry : We have demonstrated that a novel pathway downstream of retinoic acid is Bénazéraf, B., Francois, P., Baker, R.E., Denans, N., Little, C.D., & Pourquié, O., A random cell motility gradient downstream of FGF controls elongation of an amniote embryo Nature. in press (2010). shaping the future Development of muscle and vertebrae My lab is now trying to understand the mechanisms controlling axis formation and patterning at the genomic level. We have set out to identify the gene regulatory networks involved in these processes focusing on the molecular basis of the oscillator controlling segment production and on the control of differentiation of the muscle and vertebral lineages. Our work now relies more and more on quantitative approaches and modeling at the interface with physics. In parallel, we are developing in vitro systems of these processes using embryonic or reprogrammed stem cells, with the goal of using these systems to shape strategies for therapeutical approaches of diseases of the musculoskeletal axis.< Olivier Pourquié All about embryo patterning required to buffer the desynchronizing effect of the leftright machinery which controls the asymmetrical development of organs such as heart or liver. This mechanism maintains the bilateral symmetry of cervical somites. • Segment number : We have identified a mechanism involved in the control of vertebral number and have demonstrated that in snakes, acceleration of the segmentation clock accounts for the increase in vertebral count. n Vilhais-Neto, G.C., Maruhashi, M., Smith, K.T., Vasseur-Cognet, M., Peterson, A.S., Workman, J.L., & Pourquie, O., Rere controls retinoic acid signalling and somite bilateral symmetry. Nature. 463(7283):953-957 (2010). 1- Somite organization highlighting the actin-rich apical side of epithelial cells (green), nucleus (blue), basal side (red). 2- Segmental gene expression in embryo. HN-Cadherin (green), actin (red), nucleus (blue). Dorsal view. Gomez C, Ozbudak EM, Wunderlich J, Baumann D, Lewis J & Pourquié O. (2008). Control of Segment number in vertebrate embryos. Nature. 454(7202):335-9. (2008). Dequeant, M.L., Glynn, E., Gaudenz, K., Wahl, M., Chen, J., Mushegian, A., & Pourquie, O., A complex oscillating network of signaling genes underlies the mouse segmentation clock. Science. 314(5805):1595-1598 (2006). Olivier Pourquié graduated as an engineer from the National Institute of Agronomy and completed his PhD and postdoc work in developmental biology with Nicole le Douarin at the College de France. In 1996, he became an independent group leader at the Institut de Biologie du Développement de Marseille (IDBM). There, he published the first evidence of a molecular oscillator which controls segmentation of the vertebral precursors, the segmentation clock. This discovery was acknowledged as one of the 24 milestones in developmental biology over the past 100 years by the magazine Nature. In 2002, he joined the Stowers Institute for Medical Research (US) and became a Howard Hughes Medical Institute Investigator in 2005. In 2009, he returned to France and became the director of the IGBMC. His research is focused on the development of muscle and vertebrae, using in vivo strategies such as mouse genetics and microsurgery or in vitro approaches based on embryonic stem cells. n 2 25 highlights • Collective effects in vitro: We showed an instability of molecular motors by developing a new acto-myosin motility assay. The filaments go spontaneously back and forth, which suggests an oscillatory mechanism at play in cells. • Mechanosensing in cell adhesion: We demonstrated that focal contacts and cell-cell contacts are undergoing elongation upon force application. This reinforcement mechanism explains key aspects of cell motility and tissue organisation. 1 probing complexity Cells are traditionally viewed as a soup of chemical reactions. However cell shape is also the result of self-assembly phenomena; the physical laws of matter provide the appropriate framework for understanding shapes of cells and tissues. Within the cells, the cytoskeleton dynamics and its associated Rho pathways give the proper unit for understanding cell motility, cell division and cell shapes within tissues. We use interdisciplinary approaches to address these phenomena in cell culture. The physical framework is inspired from soft matter physics. The biological side is adapted from cell biology for changing specifically cell shapes with cytoskeleton drugs, genetic modifications, and mechanical constraints. The chemical part consists in designing new agents for altering cell motility and cell division. In the long term, our research may provide new ideas for understanding cancer. Being interdisciplinary in nature, our group is located both at The Science and Supramolecular Engineering Institute in Strasbourg for the physical/chemical themes as well as at the IGBMC for those of cell biology. We use classical cell lines for fibroblasts and epithelium, along with fission yeast as a model system for cytokinesis. Microfabrication plays a key role for our measurements of forces, as well as for ordering cells in our experiments.< • Soft Matter Physics for Cells: We developed quantitative models for explaining the shapes of adhesive contacts and the shapes of yeast cells. The geometrical relations are derived from physical laws demonstrated in soft matter systems. shaping the future Laboratory of Cell Physics Our research is articulated around three axes : ● Rectification of individual cells’ motion : By using surface patterning, we are seeking to show that the directed motion of cells can be a physical mechanism with no chemical gradients. Some synthetic molecules targeted against the actin cytoskeleton are also tested in collaboration with the laboratory of J.-M. Lehn at ISIS. ● Dynamics of monolayer : We are studying the evolution of cells monolayer under the microscope and in silico; we translate images of contacts into local constraints in order to explain and predict the changes in local and global shapes of cells monolayers. Extensions for this project address the distributions of constraints within developing embryos with several groups at IGBMC, in particular the teams of O. Pourquié and M. Labouesse. ● Cell division : We are characterizing the closure of the cytokinetic ring in a chemical physics way by using a new method of observation, combining microfabrication, genetics, and optical observations. A technology transfer based on our new method is being developed for a new high-throughput screening device for anti-cancer drugs.< • The cytokinetic ring closure: By developing a new approach, we outlined a mechanism for explaining the closure of the cytokinetic ring in fission yeast. The action of molecular motors promotes the ring closure. n Riveline D., P. Nurse, Injecting yeast, Nature Methods. 6:513. (2009). Riveline D. Explaining lengths and shapes of yeast by scaling arguments. PLoS One. 4(7):e6205. (2009). Brevier J, Montero D, Svitkina T, Riveline D. The asymmetric self-assembly mechanism of adherens junctions: a cellular push-pull unit. Phys Biol. 5(1):016005. (2008). Brevier J., M. Vallade, D. Riveline, Force-extension relationship of cell-cell contacts, Phys. Rev. Lett. 98:268101. (2007) 2 1- Reinforcement in cells. Cells probe their environment by pulling on adhesion areas. 2- Microfabrication and Biology. By imposing the location of individual yeast cells, we can study self-assembly mechanisms and develop new methods such as the first yeast injection shown here. Daniel Riveline Physics to explain roles of molecular motors in biology Daniel Riveline graduated with a master’s degree in physics and then received a PhD at the Curie Institute (Paris). He focused on actin-myosin molecular motors with J. Prost and A. Ott and in 1997 he revealed the oscillatory nature of these motors. Next he obtained a post-doc position at the Weizmann Institute (Israel) with A. Bershadsky where he was trained in cellular biology and studied cell adherence to the extracellular matrix. In 1999, he created his own group in cellular adhesion and the structure of chromosomes. Demonstrating cell mechanosensory mechanisms for focal contacts and intercellular contacts. A sabbatical in P. Nurse’s team at the Rockefeller University followed where he focused on cytokinetic ring closure and developed a new method to observe this process. In 2009, he created the Laboratory of Cell Physics, a joint laboratory between the Institut de science d’ingénierie supramoléculaires (ISIS) and the IGBMC. n 27 highlights • Reprogramming: In an effort to delve into the mechanisms governing epigenetic reprogramming, we have identified new histone modifications and uncovered putative roles for histone variants in this process. shaping the future Epigenetics and cell fate in early mammalian development The proteins that regulate chromatin structure in the early embryo, or the way in which the chromatin is remodeled throughout preimplantation development are largely unknown. Our efforts during the coming years will be devoted to understand : ● how the structure of the chromatin is established at the beginning of embryonic development and how it supports totipotency. ● which players are involved in chromatin remodeling during early mammalian development. ● the role of these molecules in reprogramming and in formation of the pluripotent compartment of the inner cell mass.< • Cell fate: We have determined that by manipulating the epigenetic information of a cell we can regulate its fate. We also revealed that histone modifications, in particular arginine methylation, can regulate pluripotency in the early mouse embryo. 1 probing complexity Research in our group focuses on understanding how early mouse development is regulated by chromatin-mediated changes in gene regulation, that is, by epigenetic information. In particular, we are interested in understanding how the transitions in cell potency and cell fate are regulated by chromatin-mediated processes. We use the mouse embryo as a model because this is one of the few systems where it is possible to explore the foundations of totipotency and differentiation. Indeed, the zygote, which is the product of fertilization of an oocyte by sperm, has an inherent capacity to form all cell types in an organism. Chromatinmediated changes in gene regulation have to ensure the plasticity required for undertaking such an essential task during development. Our projects will help us to understand how chromatin structure is progressively modified to restrict cell fate determination with the consequent loss of totipotency. Our work will also allow new insights in understanding the biology of the pluripotent stem cells, in particular on their origin and development. From a broader perspective, deciphering the basic mechanisms underlying the earliest steps of mammalian development is essential to understand early aspects of embryonic development, human reproduction and stem cell biology. < Maria Elena Torres-Padilla Epigenetics in cellular differentiation • Fertility: By using genetic approaches, we have shown that a protein related to the TATA-binding protein, TBP2, is essential for female fertility and oocyte growth. TBP2 plays its part by regulating the transcriptional Programme of the oocyte as well as chromatin condensation. n Gazdag E1. Santenard A1. Ziegler-Birling C. Altobelli G. Poch O. Tora L* Torres-Padilla ME* TBP2 is essential for germ cell development by regulating transcription and chromatin condensation in the oocyte. Genes and Development. 23(18):2210-23. (2009). 1Equal contribution. *Corresponding authors. Daujat S, Weiss T, Mohn F, Lange U. C, Ziegler-Birling C, Zeissler U, Lappe M, Schübeler D, Torres-Padilla ME, Schneider R. H3K64 trimethylation marks heterochromatin and is dynamically remodeled during developmental reprogramming. Nature Structural Molecular Biology; 16(7):777-81. (2009). Santenard A. Torres-Padilla ME. Epigenetic reprogramming in mammalian reproduction: contribution from histone variants. Epigenetics. 4(2):80-84. (2009). 1- Mouse blastocyst a day before implantation. Blue DNA, yellow cell-cell boundary. 2- Male and female pronuclei undergoing the first mitosis. In 2002, Maria Elena Torres-Padilla obtained her PhD at the Pasteur Institute (Paris) on the role of the nuclear receptor HNF4 in hepatic differentiation. Notably, she demonstrated the role of chromatin factors on the specific action of splicing variants of HNF4. An interest in chromatin led Marie-Elena to become focused on its study in early development transitions. She did post-doctorate research at the Gurdon Institute (Cambridge University, UK) with Magdalena Zernicka-Goetz in close collaboration with Tony Kouzarides. They were able to show that cell fate and pluripotency are controlled by epigenetic information in the early embryo. At the end of 2006, Maria Elena Torres-Padilla joined Laszlo Tora’s team, noted for its biochemical approach on the analysis of transcrition mechanisms, to expand her biochemical skills. At the end of 2008, she created her own team at the IGBMC on epigenetic mechanisms in early mouse development. n 2 Torres-Padilla ME. Parfitt D.E. Kouzarides T. and Zernicka-Goetz M. Histone arginine methylation regulates pluripotency in the early mouse embryo. Nature. 445(7124):214-218 (2007). 29 highlights 1 probing complexity During embryonic development, cells act as force sensors and can actively trigger physical changes in order to maintain the harmonious body-plan patterning and growth. Defects in these processes can cause catastrophic developmental abnormalities, in particular in the cardiovascular system where blood flow is generating shear forces essential for cardiogenesis. We use live imaging techniques, cell biology and genetic analysis to study the dynamics and the roles of biological flow during the development of the zebrafish and the Danio rerio. Our goals are to address the physical stimuli and the molecular/genetic mechanisms that specify cell responses to flow forces during embryogenesis. In particular, we are interested in understanding the relationship between physical forces and tissue organization during endothelium maturation. Also, we investigate how cells alter and maintain their physical environment to control morphogenesis. To address this question, we study the activity of beating cilia during embryogenesis using novel imaging techniques to visualize and analyze cilia mediated flow. < • Addressing the dynamics of development: the cardiovascular system is under the influence of complex stimuli generated by flowing blood. We identified the physical stimuli involved in controlling valvulogenesis during heart development and found a flow responsive gene linking blood flow and valve morphogenesis • Mixing classical approaches in genetics with light imaging to address cilia activity during development: We discovered that manipulating cilia dynamics using mRNA knock down affects inner ear formation. The use of fast imaging uncovered the mechanism of action of cilia during this process • Developing approaches to probe flow in vivo: We used techniques to measure flow and explored cilia mediated flow in the embryo. In particular, we used fast imaging combined with optical shaping the future Mecano-genetic interplays and embryonic morphogenesis We aim at investigating the relationship between the viscous forces generated by blood flow and endothelial tissue organization. Endothelial cells express different genes in response to the flow patterns they experience. Our working hypothesis is that blood flow influences cardiovascular development by acting on cell identity and cell motility. We generated a flow responsive reporter in order to explore endothelial cell responses to shear forces during embryonic blood vessel development. This line will allow us to address precisely the flow-tissue interaction during vascular development and to address the endothelial cell behavior in response to flow using live imaging. We use molecular biology and genetic approaches to address the gene networks activated by blood flow. < Fluid dynamics applied to development approaches to analyze cilia mediated flow in the left-right organizer and in the developing inner ear • Using multidisciplinary approaches to address embryogenesis: In collaboration with physicists, engineers and mathematicians, we proposed new models, approaches and hypothesis to solve problems related to flow. n Vermot J, Forouhar AS, Liebling M, Wu D, Plummer D, Gharib M, Fraser SE. Reversing blood flows act through klf2a to ensure normal valvulogenesis in the developing heart. PLoS Biol. e1000246. 7(11) (2009). 1- Intersegmental vessels and their flow dynamics revealed by a transgenic line labeling red blood cell (red) and endothelial cell response (white). 2- Head vascular network in the Zebrafish embryo labeled using the fli:gfp line. Colantonio JR*, Vermot J*, Wu D, Langenbacher AD, Fraser S, Chen JN, Hill KL. The dynein regulatory complex is required for ciliary motility and otolith biogenesis in the inner ear. (*equal authors). Nature. 457:205-9. (2009). Supatto W, E. Fraser S, and Vermot J. An all-optical method for probing microscopic flows in living tissues. Biophysical J. 95(4):L29-31. (2008). Vermot J, Fraser SE, Liebling M. Fast fluorescence microscopy for imaging embryonic development. HFSP journal. 2:143-155. (2008). Julien Vermot In 2003, Julien Vermot completed his PhD with P. Dollé at the IGBMC in development biology and genetics. In 2004, he became a member of O. Pourquié’s team at The Stowers Institute for Medical Research in Kansas City, USA to supplement his PhD work. Next, he joined the group of S. Fraser at the Beckman Imaging Center of the California Institute of Technology (Pasadena, USA) as a long term HFSP fellow in 2005. This experience allowed him to become familiar with a broad range of live imaging techniques that he would employ to address the role of cilia driven flow and blood flows during embryogenesis. Julian notably demonstrated the role of biological flow produced by cilia in inner ear development, as well as the importance of reversing flow during heart valve formation (2009). The accumulation of is research experience offered new prospects and he returned to France in 2009 to create his own team in the IGBMC, where he focuses on the role of fluid forces on embryogenesis. n 2 31 highlights 1 probing complexity Primordial germ cells (PGC) represent a unique class of cells. They are the embryonic precursors of the gametes, ensuring propagation of the gene pool to the next generation and the continuation and evolution of the species. Our main focus is to decipher the molecular mechanisms that account for maintenance of their pluripotency. Indeed, PGC’s can be considered as totipotent cells since following fertilization they can generate all cell types and yield a fully functional organism. Accordingly, most if not all, pluripotent genes are expressed by PGCs. Our goal is to identify actors involved in the process of the PGC’s ontogeny, to study their physiological functions and to challenge them as pluripotency reprogramming factors. We also study the genetics of infertility in humans. Specifically, we aim to identify genes affecting human gametogenesis and therefore potentially being involved on the PGC’s ontogeny. Based on our expertise in mouse and human embryonic stem (ES) cells, we are setting up a new platform dedicated to the production of pluripotent stem cells, such as ES or induced pluripotent stem (iPS) cells. This is done in parallel with the creation of a Biobank of human somatic cells that serve as pathological models. < • PGC’s ontogeny: we have identified a new gene Tex19.1 specifically expressed by PGC and pluripotent stem cells, that is essential for the formation of male gametes. The protein presents two well conserved domains of unknown function. • Pluripotency: Starting from the human ES cell model, we have recently cloned a gene coding for a specific pluripotent stem cell protease, which is also expressed by germ cells. • Genetics of infertility: Studying two families, we have cloned two genes involved in the formation of the acrosome. We are now analyzing a third family with brothers suffering from azoospermia. • Pathological models: We have generated the second largest (worldwide), collection of human shaping the future Primordial germ cells’ (PGC) ontogeny Being responsible for the in vitro fertilization ward of the hospital, we believe that it is of fundamental importance to conduct research as a continuum from basic to clinical and from clinical to basic. In this context, we hope that our work will highlight some of the fundamental mechanisms in the PGC’s ontogeny, contribute to the understanding of pluripotency and will have a clinical outcome. Indeed, the study of genes involved in such processes will notably clarify the biology of PGC and pluripotency but should allow significant advances in the production of pluripotent stem cells that can be used either as cellular models to study the physiopathology of diseases or as a tool for cell therapeutic, or for pharmaceutical screening. We also aim to produce cellular models for the research community by building a biobank of iPS cells. < Stéphane Viville Primordial germ cells, first step in reproductive biology embryonic stem cells carrying genetic disease genes. In addition, we have implemented the reprogramming technology and produced dozens of iPS cells from normal or pathological tissues. n P Tropel, J Tournois, J Côme, C Varela, C Moutou, P Fragner, M Cailleret, Y Laâbi, M Peschanski, S Viville High efficiency derivation of human embryonic stem cell lines following pre-implantation genetic diagnosis. In Vitro Cellular & Developmental Biology– Animal 2010 46(3-4):376-85. Epub Mar 9. (2010). Madanl B, Madanl V, Weber O, Tropel P, Bluml C, Kieffer E, Viville S, Fehling HJ. The strictly pluripotencyassociated gene Dppa4 is dispensable for embryonic stem cell identity and germ cell development, but essential for embryogenesis. Mol. Cell. Biol. 29(11):3186-203. (2009). 1- Immunofluorescence detection of Oct4 (red) or Tex19.1 (green) and merge image. Top at 8 cells stage, bottom at blastocyst stage. 2- In situ hybridization detecting Tex19.1 mRNA on male (top) or female embryonic gonads (bottom). Kuntz S, Kieffer E, Bianchetti L, Lamoureux N, Fuhrmann G, Viville S. Tex19, a mammalian specific protein, with a restricted expression in pluripotent stem cells and germ line. Stem Cells 26: 734-744. (2008). Dam AHDM, Koscinski I, Kremer JAM, Moutou C, Jaeger A-S, Oudakker AR, Tournaye H, Charlet N, Lagier-Tourenne C, van Bokhoven H, Viville S. Homozygous mutation in SPATA16 is associated with male infertility in human globozoospermia. Am. J. Hum. Genet. 81:813-820. (2007). After Stéphane Viville received a degree in Pharmacy, he completed his PhD in immune tolerence with Diane Mathis and Christophe Benoist’s team in Strasbourg. In 1993, he demonstrated the fundamental role of the invariant chain. Stéphane undertook his post-doc research with Azim Surani’s team in Cambridge, (UK) and working on genomic imprinting, proved the role of PEG1 in maternal behavior. In 1995, he created the Preimplantation Genetic Diagnosis Center at the Strasbourg Hospital and also taught classes at the Faculty of Medicine while working on retinoic acid with Manuel Mark’s group. In 2003, he formed his own team at the IGBMC, focusing on primordial germ cell ontogeny. In 2008, his team identified a new gene involved in pluripotency (Tex19) and a human mutation that influences acrosome development (SPATA16). Since 2004, he has led the Reproductive Biology Ward at the Strasbourg Hospital. n 2 33 Regulation of gene expression, signalling and cancer " The idea is to study in the widest sense the transcription control, in normal, physiological but also pathological states ". I. Davidson The fundamental mechanisms regulating gene expression have been one of the major research areas of the IGBMC, and its forerunner the LGME, for more than 30 years. Since the initial purification and characterization of RNA polymerase II by Pierre Chambon and colleagues, a consistent theme has been the identification of the machinery required for basal and regulated transcription. The isolation of two major transcription factors TFIIH and TFIID proved a major advance in understanding the close link between transcription, DNA repair and human disease and highlighted the importance of the preinitiation complex in developmental and cell-specific gene regulation. Currently, the Programme has incorporated the study of epigenetics and chromatin dynamics, RNA metabolism as well as high throughput genomic and proteomics technologies as it pursues its quest to understand the regulation of gene expression. The study of nuclear receptors, transcription factors whose activity is regulated by a diverse set of hor- Functional Genomics & Cancer • Fundamental mechanisms of transcription, RNA metabolism and DNA repair : identification and characterisation of complexes involved in transcription, RNA degradation and DNA repair, role of nuclear and cellular organisation in the control of these processes. • Epigenetic modifications, histone variants and chromatin dynamics during differentation : purification and characterization of complexes involved in the enzymatic modification of histones and deposition of histone variants in chromatin, role of these factors in control of gene expression in differentiation and development. • Physiopathology and molecular biology of nuclear receptors : development of mouse models for study of nuclear receptors and their cofactors in metabolism and disease, identification and characterisation of nuclear receptor cofactor complexes, high throughput genomic approaches to identify target genes and pathways. • Signalling pathways, gene expression and cancer : integrative approaches to link basal factors, DNA repair, nuclear receptors and other transcription regulatory factors and signalling pathways to deregulated gene expression involved in malignant transformation, development of novel paradigms for cancer therapy through targeting epigenetic enzymes, understanding the basis of tumor-cell selective apoptosis. Human cancers : molecular characterisation of factors overexpressed in breast and other human cancers, discovery, and testing of novel prognostic markers and therapeutics. mones and metabolites and of their cofactors is the second major theme of the Programme. The function of several receptors, principally estrogen, glucocorticoid, vitamin D3 and retinoic acid receptors in development, physiology and disease are studied by germ line and spatio-temporal somatic mutation in an extensive set of recombinant mouse models. In recent years, this approach has revealed the importance of retinoic acid signaling in the development of hepatocellular carcinoma, and vitamin 13 Groups D signaling in atopic 32 Staff Scientists dermatitis and asthma. Complementary to the 42 Postdoctoral fellows in vivo studies are the 53 PhD students biochemical charac terization of receptor 34 Engineers/Technicians cofactors and the mole1 Assistant cular basis of cross-talk with other signalling pathways. A recurrent and transversal theme throughout the Programme is how the normal gene regulatory mechanisms are deregulated during malignant transformation. The Programme has underlined the contribution of basal factors, transactivators, epigenetics and nuclear receptors in the etiology and treatment of various cancers. Bringing together the teams working on functional genomics and on cancer in one Programme aims to strengthen the interaction around this common theme. The recruitment of new groups in epigenetics, nuclear organization and DNA repair underscores the commitment of the Programme to pursue the study of the links between fundamental nuclear processes and cancer. n 35 highlights • Cancer: Using genetic approaches, we have developed a mutant mouse line (IkL/L) in which Ikaros expression is knocked down to 10% of normal levels. We have shown that Ikaros exerts its tumor suppressor activity by repressing the transcription of target genes associated with the Notch pathway in developing T cells. Notch activation is therefore an early and critical event during T-cell leukemogenesis in IkL/L mice. Moreover, we have provided evidence that the Ikaros gene is mutated in human T-ALLs. 1 probing complexity Hematopoiesis is characterized by the development of hematopoietic stem cells into pluripotent progenitor cells and finally into mature blood cells. This orderly and continuous process involves self-renewal, cell fate choice, differentiation and proliferation. Transcription factors are responsible for coordinating these processes but their expression must be tightly regulated to prevent hematological malignancies. In this context, we are interested in the following questions. How do transcription factors control hematopoiesis ? How do alterations in their function contribute to leukemogenesis ? In the past, we have shown that retinoic acid receptor alpha and PU.1 play important roles in granulocyte and erythroid differentiation, respectively. Our current studies focus on the Ikaros family of transcription factors, which act mainly as transcriptional repressors through association with chromatin remodeling complexes. Ikaros (Ikzf1) is a zinc finger protein expressed by all hematopoietic cells. Ikaros is crucial for lymphocyte differentiation and homeostasis and also functions as a tumor suppressor in T and B cells. Furthermore, loss of Ikaros has been detected in human T- and B-cell acute lymphoblastic leukemias (ALLs). The molecular mechanisms responsible for these activities remain unclear.< shaping the future Hematopoiesis and Leukemogenesis in the Mouse Today we are dissecting the molecular pathways regulated by the Ikaros family. We will apply this knowledge and our tools to understand how Ikaros family members control hematopoietic cell differentiation, stem cell function and chromatin remodeling. We are using the Ikaros-deficient T-ALL model to address fundamental issues regarding T-cell leukemogenesis, such as the existence and identity of leukemia initiating cells and the role of the Notch receptors. < Philippe Kastner Susan Chan Two converging careers • Hematopoiesis: In the B-cell lineage, we have shown that Ikaros blocks the development of immature pro/preB cells in the bone marrow and controls isotype selection during class switch recombination in mature splenic B cells. In the dendritic cell (DC) lineage, we have shown that Ikaros is specifically required for the differentia- Susan Chan obtained her Bachelor’s degree at the University of California, Berkeley and her PhD in immunology at the University of Pennsylvania in the U.S. In 1991, she moved to Strasbourg to studyT-cell differentiation as tion of a subset of DCs called plasmacytoid dendritic cells. A Marçais, R Jeannet, L Hernandez, J Soulier, F Sigaux, S Chan and P Kastner. Genetic inactivation of Ikaros is a rare event in human T-ALL. Leukemia Res. 4:426-429. (2010). • Ikaros family members: More recently, we have generated the first knock-out mouse line for Helios (Ikzf2), another member of the Ikaros family. We are currently investigating how Helios plays a role in hematopoiesis. n a post-doctoral fellow with Diane Mathis and Christophe Benoist. There, she met Philippe Kastner, a scientist in Pierre Chambon’s lab. A graduate of Ecole Polytechnique, he was working at the time on retinoic acid receptors and developing mutant mouse models to study their function. In 1997, they started a new team together at the IGBMC. Q Cai, A Dierich, M Oulad-Abdelghani, S Chan and P Kastner. Helios deficiency has minimal impact on T cell development and function. J Immunol. 183:2303-2311. (2009). Now they study how transcription factors M Sellars, B Reina-San-Martin, P Kastner and S Chan. Ikaros controls isotype selection during class switch recombination. J Exp Med. 206:1073-1087. (2009). development. n E Kleinmann, AS Geimer Le Lay, M Sellars, P Kastner and S Chan. Ikaros represses the transcriptional response to Notch signaling in T-cell development. Mol Cell Biol. 28:7465-7475. (2008). regulate hematopoiesis and how mutations in 2 these proteins promote leukemia 1- Specific gene expression signatures define various subtypes of murine T-cell leukemias. 2- Activation of Notch1 transcription from an intragenic promoter in Ikaros-deficient T-cell leukemias: visualization by ChIP-seq of histone H3 acetylation over the Notch1 gene in IkL/L tumors with an intact Notch1 gene (black) or with a deletion of the Notch1 promoter (red). 37 highlights • The TAF4 subunit of transcription factor TFIID plays an important role in diverse signaling pathways as an essential co-activator for CREB and the RARs. Genetic study of TAF4null MEFs and its inactivation in the mouse epidermis revealed novel functions in regulating the TGF-beta and EGF signaling pathways and highlighted its role as a cell autonomous and cell non-autonomous tumor suppressor. 1 probing complexity The main focus of our work over the last several years has been to understand the role of TFIID subunits such as TAF4 or paralogues such as TAF4b, TRF2 or TAF7L in gene expression. The study of TAF4 function in cells derived from genetically modified mice in vitro and tissue-specific knockouts in vivo revealed novel facets of its function as a regulator of multiple signaling pathways and cell proliferation, as well as a tumor suppressor. The study of TAF4 as a co-activator for the retinoic acid receptors (RARs), allowed us to decipher the molecular mechanism of cross-talk between the RA and TGF-beta signaling pathways that promotes cell proliferation. We completed this cycle of experiments by using ChIP-chip to identify RAR occupied loci and to target genes revealing the importance of cell-specific binding site occupancy in determining the biological response to RA. More recently, we were responsible for setting up high throughput sequencing techniques at the IGBMC. The implementation of these technologies has profoundly altered our experimental Programme over the last few years allowing us to ask different sets of questions. We have begun to use these techniques to understand the role of various transcription factors in developmental and pathological processes. < shaping the future Structure and function of the general transcription factor TFIID The development of high-throughput genomic technologies (ChIP-seq, RNA-seq) facilitates the characterization of complex gene regulatory networks. We are using these approaches to address the functions of transcription factors such as MITF, its co-factors and target gene products in the proliferation of malignant melanoma, the TEAD family in muscle differentiation and regeneration and the RARs and RAR-regulated transcription factors in neuronal differentiation of ES cells and tumor suppression. We are also pursuing our characterization of TAF4 through the generation of novel tissue-specific knockout mice. We have identified a novel TAF4-containing TFIID sub-complex that is abundant in several cell types and are investigating its potential function by comparing the genomic occupancy of TAF4 and TBP in these cells. < Irwin Davidson Regulation of transcription in differentiation and cancer • We characterized the molecular basis of the cross-talk between the RA and TGF-beta signaling pathways in MEFs. RA induces TGF-beta dependent autocrine growth by directly activating the expression of the TGF-beta3 ligand and the secreted mitogen connective tissue growth factor (CTGF). Transient RA treatment is sufficient to set up a positive feedback loop between these two mitogens. Delacroix L, Moutier E, Altobelli G, Legras S, Poch O, Choukrallah M-A, Bertin I, Jost B, and Davidson I. Cell-specific interaction of retinoic acid receptors with target genes in mouse embryonic fibroblasts and embryonic stem cells. Mol. Cell Biol. 30: 231-244. (2010). After obtaining his PhD at the University of Glasgow on the herpes virus (1985), Irwin Davidson became a post-doc fellow with Pierre Chambon’s group in Strasbourg, working on the identification of transcription factors interacting with the SV40 enhancer. • We used ChIP-chip and transcriptomics/RNA-seq to identify RAR binding sites and target genes in MEFs and ES cells. More than 750 RAR-occupied promoters were identified, in which the vast majority were cell specifically occupied. Our results reveal that differences in the chromatin landscape regulate accessibility of RARs to their target loci modulating the repertoire of target genes that can be regulated and the biological effects of RA. n In 1990, he became interested in the mechanisms responsible for transcription activation and in 1993 he participated in the identification of the TFIID factor and its role as a co-activator. Irwin then focused on cloning genes and encoding different TFIID subunits through biochemical, structural and genetic approaches. He also discovered the important role of the histone fold motif in TFIID organization. At the beginning of 2000, he began to study the role of Fadloun, A., Kobi, D., Delacroix, L., Dembélé, D., Michel, I., Lardenois, A., Tisserand, J., Losson, R., Mengus, G., and Davidson, I. Retinoic acid induces TGFb-dependent autocrine fibroblast growth. Oncogene. 27(4):477-89. (2008). TBP and its paralogueTRF2 in the mouse, where he 1- Mislocalisation of H1T2 in TRF2-null mouse haploid spermatids. 2- Senescent human 501 melanoma cells. Fadloun, A., Kobi, D., Pointud, J-C., Kumar, I., Teletin, M., Bole-Feysot, C., Testoni, B., Mantovani, R., Metzger, D., Mengus, G., and Davidson, I. The TFIID subunit TAF4 regulates keratinocyte proliferation and has cell-autonomous and non-cell-autonomous tumour suppressor activity in mouse epidermis. Development. 134(16):2947-58. (2007). demonstrated the specific role of TRF2, in transcription regulation in male germ cells. This work also led to the discovery of novel histone variants and chromatin-associated proteins that specify nuclear domains within the developing spermatozoa. Irwin’s group characterized the structure and functions of theTFIID subunitTAF4 and revealed its role in various signaling pathways. n 2 39 highlights 1 probing complexity Transcription, one of the key steps of gene expression in response to different stimuli of the organism like stress or hormones, requires a combination of factors. The deleterious action of physical or chemical agents that create lesions in the DNA disrupts the expression of genes. If these lesions are not removed by efficient repair systems they will be at the origin of mutations that can lead to cancer and aging. TFIIH, a multi-subunit complex that we are studying plays a pivotal role in both the transcription of genes and their repair. Mutations in some of the subunits of TFIIH are responsible for three genetic diseases (xeroderma pigmentosum (XP), trichotyodystrophy (TTD) and Cockayne syndrome (CS)), the phenotypes of which result from defects in both DNA repair and gene expression. For instance, TTD patients present brittle hairs and nails, that cannot account from a defect in DNA repair and that can be recapitulated in mouse TTD models. With the help of biochemical, genetic and cellular biology we study these diseases and consequences of deregulation of gene expression and repair in various cellular systems and animal models.< • Over the last five years we have dissected the role of each of the TFIIH subunits and explained the molecular defects in transcription and repair responsible for the phenotypes of XP and TTD patients mutated in TFIIH subunits. We have recently identified the tenth subunit of TFIIH and demonstrated its specific involvement in DNA repair; mutation in this subunit results in TTD group A disorder. We showed that the CAK module was not part of TFIIH when removing DNA lesions but was specifically devoted to the transcription function of TFIIH. • In transcription, we showed that CAK was not only involved in the phosphorylation of the RNA polymerase II but also in the phosphorylation of several nuclear receptors e.g. retinoic acid, peroxisome proliferator-activated and thyroid hormone receptors. The phosphorylation Ueda, T., Compe, E., Catez, P., Kraemer, K.H., and Egly, J.M. Both XPD alleles contribute to the phenotype of compound heterozygote xeroderma pigmentosum patients. J Exp Med. 206:3031-3046. (2009). shaping the future Genome expression and repair We aim to: ● better understand the mechanisms of gene expression and DNA repair and to elucidate the dynamic interactions between these two pathways. ● explain the biochemical phenotypes of transcription/ repair syndromes, focusing on the hormonal dys-regulation that leads to these various phenotypes. In addition, it will allow us to gain extensive insights into the physiology of nuclear hormones. ● employ sophisticated technologies to obtain mechanistic details on DNA repair. These include : - Screening of genomic siRNA libraries to identify new DNA repair factors - Follow up TTD/XPD diseases in vivo in animal models using a robotic biopsy collection and Computed Tomography (CT) coupled with 3-D tissue reconstruction in collaboration with IRCAD (Strasbourg). ● further our understanding of molecular effects induced by anti-cancer agents, in collaboration with a pharmaceutical company.< of these nuclear receptors by TFIIH is required for their recruitment and stabilization to their DNA cognate sequence, leading to the ligand-dependent transactivation of their responsive genes. Mutation in XPD subunit of TFIIH (as found in XP-D patients) destabilize the interaction between the CAK complex and the core TFIIH, leading to nuclear receptor phosphorylation defect and impaired transactivation of specific genes. n Coin, F., Oksenych, V., Mocquet, V., Groh, S., Blattner, C., and Egly, J.M. Nucleotide excision repair driven by the dissociation of CAK from TFIIH. Mol Cell. 31:9-20. (2008). 1- Coomassie blue staining of the highly purified nucleotide excision repair (NER) factors required to remove DNA damages. 2- Transactivation mechanism mediated by the phosphorylation of nuclear receptors by TFIIH. Compe, E., Malerba, M., Soler, L., Marescaux, J., Borrelli, E., and Egly, J.M. Neurological defects in trichothiodystrophy reveal a coactivator function of TFIIH. Nat Neurosci. 10:1414-1422. (2007). Coin, F., De Santis, L.P., Nardo, T., Zlobinskaya, O., Stefanini, M., and Egly, J.M. p8/TTD-A as a RepairSpecific TFIIH Subunit. Mol Cell. 21:215-226. (2006) Frédéric Coin Jean-Marc Egly Transcribing and repairing DNA. Educated in biochemistry, Jean-Marc Egly became interested in two main cellular mechanisms, DNA transcription and repair. Besides their fundamental character, his works contributed to many breakthroughs for the comprehension of cancerogenesis and rare genetic disorder mechanisms. In 1985, he started his team in Strasbourg. In 2006, he was appointed president of the ARC scientific council and, in 2007, special advisor for the general director of Inserm. Frédéric Coin was a PhD student in JeanMarc Egly’s team and revealed in 1998 that mutations on helicases XPD and XPB observed in Xeroderma pigmentosum affected the transcriptional function of TFIIH. In 2004 Frédéric isolated the 10th subunit of TFIIH involved in trichothiodystrophy (TTD). Their work then continued on the link between genome expression and repair. The team also showed TFIIH factor implication in Rift valley viral infection in collaboration with the Pasteur Institute. n 2 41 highlights • Nuclear receptor ligand action: Identification of the structural and mechanistic basis of ligand action, including agonists, inverse agonists, and neutral, partial and full antagonists • Description of the mechanistic basis and cancer therapeutic potential of two apoptogenic rexinoid-based therapeutic paradigms. This work allows for the preparation of clinical studies. 1 probing complexity Our interest is to understand the mechanisms of signal transduction and epigenetic (de)regulation in normal cells and cancer, which are relevant for the development of novel therapeutic paradigms. Based on our expertise on nuclear receptors we first develop global systems biology approaches to deconvolve receptor signaling complexity and aberrant (epigenetic), signaling upon tumorigenic transformation and study the mechanisms and therapeutic potential of cancer-selective apoptosis pathways induced by various signaling drugs and epigenetic modulators. Our studies involve a plethora of genome-wide technologies and the corresponding development of bioinformatics tools. One emphasis of our work is to decipher the mechanisms underlying the cancer-selectivity of apoptogenic TRAIL signaling. We use the obtained knowledge to devise novel therapeutic tools and study the contribution of this pathway to the action of (epi-) drugs. Studies on the role of the senescence-mediated secretome on TRAIL action and the epigenetic alterations during cellular senescence address the link between cancer and aging. Our work benefits from the integration and coordination of several national and European consortia, which provide the necessary multidisciplinary input and a close link to the local hospital (HUS). < shaping the future From nuclear receptor action to novel paradigms for cancer therapy action We will expand our bioinformatics competence to facilitate large-scale data analyses from genome-wide studies, which is an inevitable component of future biomedical research. By such global analyses we will identify sub-routines and key networks in a given programme, such as estrogens or retinoic acid signaling, integrating the (epigenetic) action of, and crosstalk between co-regulators. We will decrypt the molecular and structural features underlying the tumor-selectivity of the TRAIL pathway. There is no doubt that an understanding of this evolutionarily developed tumor defence system will lead to an entirely novel class of cancer therapeutics, devoid of the (geno) toxicity of current chemo/radiotherapy. We will continue using our expertise to generate novel (epi-) drugs for improved cancer therapies.< Towards novel anticancer therapies • Identification of the convergence of several divergent cancer therapeutics, including retinoids and HDAC inhibitors, on the TRAIL pathway. • Discovery of the first triple-action epi-drug (patented) for cancer therapy and demonstration of strong anticancer activity and drug action in tumor cells of animal models treated in vivo. Pavet, V., Beyrath, J., Pardin, C., Morizot, A., Lechner, M.C., Briand, J.P., Wendland, M, Maison. W., Fournel, S., Micheau, O., Guichard, G. and Gronemeyer, H. (2010). Multivalent DR5-selective peptides activate the TRAIL death pathway and exert tumoricidal activity in vivo. Cancer Res. 70:1101-10. (2010). Hinrich Gronemeyer • Discovery of the crosstalk between histone acetyltransferases and methyltransferase CARM1, which specifies estrogen sub-programmes and is altered during breast cancer progression. n Germain, P., Gaudon, C., Pogenberg, V., Sanglier, S., Van Dorsselaer, A., Royer, C.A., Lazar, MA., Bourguet, W. & Gronemeyer, H. (2009). Differential action on coregulator interaction defines inverse retinoid agonists and neutral antagonists. Chemistry & Biology. 16:479-89. (2009). 1-MS275 induces apoptosis in the blasts of individuals with acute myeloid leukemia. 2- Epigenetic information provided by chromatin modifications and non-coding RNAs contributes to transcriptional regulation and genomic architecture. Shankaranarayanan, P., Rossin, A., Khanwalkar, H., Jacobson, A., de Lera, A. R., Altucci, L., and Gronemeyer, H. (2009). Growth factor-antagonized rexinoid apoptosis involves permissive PPAR /RXR heterodimers to activate the intrinsic death pathway by NO. Cancer Cell. 16:220-31 (2009). Chemist by training, Hinrich Gronemeyer developed a strong interest in nuclear receptor (NR) signaling during his PhD studies on the chromatin-regulatory functions of the steroid hormone ecdysone, using a photochemical approach. In 1980, he joined Pierre Chambon’s team in Strasbourg as a post-doc fellow, where he purified and contributed to cloning of the progesterone and glucocorticoid receptors. After two years at the Ludwig Institute for Cancer Research, Hinrich returned to Strasbourg to start his own group. In 1989 he predicted the existence of NR co-regulators and in 1996 he cloned TIF2 and established the first 3D structures of a ligand-binding domain with Dino Moras. Thereafter, Hinrich’s work has focused increasingly on therapeutic applications, discovering in 2001 the TRAIL connection. In collaboration with chemists and the pharmaceutical industry, his team developed several novel therapeutic paradigms. Since 2005, Hinrich is the coordinator of the Epigenetic Treatment of Neoplastic Disease (EPITRON) consortium. n 2 43 highlights • Chromatin remodeling: We discovered the molecular mechanism of chromatin remodeling by drosophila NURF. NURF remodels chromatin by sliding nucleosomes along DNA. • Gene regulation: We have shown that the histone variant macroH2A directly binds PARP-1 and inactivates transcription by down-regulating PARP-1 enzymatic activity. 1 probing complexity The main research activity of our group is to study the role of histone variants and their deposition machineries in the epigenetic control of human genome activity at the genomewide level. Among the various epigenetic memory mechanisms, the local replacement of canonical histones within the nucleosome by variant histones has the potential to affect considerably the activity of the corresponding genomic regions. Indeed, nucleosomes bearing histone variants have distinct structures and functional activities in vitro and some histone variants are incorporated at specific genomic locations. Our laboratory is focusing on the role of histone variants in gene regulation and genome integrity. We have recently implicated macroH2A in PARP-1 enzymatic activity and transcription regulation (Ouararhni et al., 2006), and discovered a new link between histone variants, transcription factors and noncoding RNA (unpublished results). This association with transcription factors and non-coding RNA is novel and will certainly help us to understand how chromatin domains are established and how epigenetic information is stored and transmitted to daughter cells. Alteration of these epigenetic marks is associated with developmental disorders and cancer. shaping the future Chromatin and epigenetic regulation Our major goal is to study the epigenetic mechanisms underlying gene regulation using mammalian cells as a model system. More specifically, we are interested in addressing how histone variants are deposited at specific chromatin loci and how this impacts chromatin structure and gene regulation. We are also interested in the role of non-coding RNA in histone deposition. The involvement of non-coding RNAs in targeting molecules adds another level of complexity to chromatin regulation. Interestingly, histone variants deposition complexes contain several non-coding RNAs. However, the mechanism by which these RNAs function is unknown. One of our future aims will be to elucidate how these interactions influence histone deposition and gene regulation.< Ali Hamiche A chromatin biochemist • Histone deposition: We have identified and characterized the first vertebrate histone chaperone, involved in the deposition of CenpA at centromeres. We have investigated in detail the molecular mechanism of the histone variant H3.3 deposition and found that the death-associated pro- Drané P, Ouararhni K, Depaux A, Shuaib M and Hamiche A. The death-associated protein DAXX is a novel histone chaperone involved in the replicationindependent deposition of H3.3. Genes Dev. 24(12):1253-65. (2010). tein DAXX and the chromatin-remodeling factor ATRX are crucial components of the H3.3 deposition machinery. Our data argues that DAXX functions as a histone chaperone, involved in the replication-independent deposition of H3.3, thus linking apoptosis to gene regulation. Our findings also provide a clue as to how mutations in the ATRX gene lead to the human genetic diseases of a-thalassemia and X-linked mental retardation. n n Shuaib M, Ouararhni K, Dimitrov S and Hamiche A, (2010). HJURP Binds CENP-A via a highly conserved N-terminal domain and mediates its deposition at centromeres. Proc. Natl. Acad. Sci. USA. 107(4):1349-54. (2010). 1- Specific marking of chromatin by histone variants. 2- Schematic representation of the assembly of the core histones into the nucleosome. Ouararhni K, Hadj-Slimane R, Ait-Si-Ali S, Robin P, Mietton F, Harel-Bellan A, Dimitrov S, Hamiche A. The histone variant mH2A1.1 interferes with transcription by down-regulating PARP-1 enzymatic activity. Genes Dev. 20(23):3324-36. (2006). Ali Hamiche graduated from the Pierre and Marie Curie University (Paris VI), where he also obtained a PhD on chromatin structure and function. Recruited in 1996 as a Research Scientist with Hélène RichardFoy’s team (LBME, Toulouse), he worked on conformational transitions of the nucleosome during transcription. In 1998 he received a Human Frontier postdoctoral fellowship and joined Carl Wu’s laboratory at the National Cancer Institute, (NIH) in Bethesda, MD, US. In 1999 he discovered the mechanism of chromatin remodeling by the Drosophila NURF complex . In 2003, he joined the André Lwoff Institute (Villejuif) as a young group leader to work on histone variants, and two years later he joined the IGBMC to create a new team that currently focuses on the same topic. n < 2 45 highlights 1 probing complexity The last decade has seen an enormous rise in the interest for nuclear receptors (NRs), because of their central role in the coordination of development and homeostasis, through their ability to transduce hormonal signals into modulation of gene activity. Our main goal is to study, under physiological and pathophysiological conditions in the whole organism, the function and interdependence of signaling pathways that are regulated by various nuclear receptors [e.g. the retinoid X receptors (RXRs), the retinoic acid receptors (RARs), the peroxisome proliferator-activated receptors (PPARs), the vitamin D3 receptor (VDR), the oestrogen receptors (ERs), the androgen receptor (AR) and the glucocorticoid receptor (GR)] in different organs (e.g. skin, lung, intestine, skeletal muscle and reproductive organs). In that respect, we have used our site-directed cell-specific temporally-controlled somatic mutagenesis method, which allows the generation of somatic mutations in the mouse of any given gene, at any chosen time and in any specific cell type. Interestingly, as this genetic tool is also of great value to generate mouse models of human cancers, the knowledge gained from their analyses should contribute to the development of new cancer therapies. < • Through phenotypic analyses of various mouse mutants, we have revealed that RXR/RAR and RXR/VDR heterodimers, as well as GR, control the expression of the cytokine “thymic stromal lymphopoietin (TSLP)” in epidermal keratinocytes, and demonstrated that this cytokine plays a key role in initiating the development of a skin and systemic disease mimicking human atopic dermatitis. • Our studies also revealed that enhanced TSLP expression in mouse epidermal keratinocytes aggravates experimental allergic asthma, indicating that keratinocyte-produced TSLP may represent an important factor in the “atopic march” that links atopic dermatitis and asthma. • By analyzing mutant mice in which PPARβ is selectively ablated in skeletal muscles, we have demonstrated that this nuclear receptor is instru- Z. Zhang, P. Hener, N. Frossard, S. Kato, D. Metzger, M. Li and P. Chambon Thymic stromal lymphopoietin overproduced by keratinocytes in mouse skin aggravates experimental asthma. Proc. Natl. Acad. Sci. U.S.A. 106:1536-1541. (2009). shaping the future Genetic dissection of nuclear receptor signaling in the mouse Our projects are aimed at further characterizing the physiological role of nuclear receptors, the mechanism of their action at the molecular, cellular and organismal levels, and their implication in the pathogenesis of human diseases. We will analyze in skeletal muscles the functional role of AR, GR and PPARs, and of some of their co-regulators (SRC-1, TIF2 and SRC-3; NCoR and SMRT), in order to characterize the signaling pathways controlled by these factors and to identify new drug targets to prevent or treat metabolic and muscle diseases. The role played by TSLP and nuclear receptors in the pathogenesis of atopic dermatitis and asthma will be further investigated, as well as their function in intestinal epithelial cells, where they could be involved in the control of the immune response to commensal bacteria. This latter study and related studies should reveal some of the molecular mechanisms through which the dialog between the microbiota and intestinal epithelial cells contributes to homeostasis and disease of the intestine. < Pierre Chambon Daniel Metzger All about nuclear receptors mental in myocytes to the maintenance of oxidative fibers, and have shown that fiber type switching is likely to be the cause, and not the consequence of metabolic diseases such as obesity and diabetes. • By ablating the tumor suppressor gene PTEN selectively in epithelial cells of the mouse prostate, we have generated a model mimicking human invasive prostatic adenocarcinoma. n C. K. Ratnacaram, M. Télétin, M. Jiang, X. Meng, P. Chambon and D. Metzger. Temporally-controlled ablation of PTEN in adult mouse prostate epithelium generates a model of invasive prostatic adenocarcinoma. Proc. Natl. Acad. Sci. U. S. A. 105:2521-2526. (2008). 1- Immunohistochemical detection of RyR1 (Green) and DHPRβ (red) in mouse tibialis muscle. Nuclei are stained with DAPI (blue). 2- Immunohistochemical detection of TSLP in mouse epidermal keratinocytes. M. Schuler, F. Ali, C. Chambon, D. Duteil, J-M. Bornert, A. Tardivel, B. Desvergne, W. Wahli, P. Chambon and D. Metzger PGC1α expression is controlled in skeletal muscles by PPARβ, whose ablation results in fiber type switching, obesity and type 2 diabetes. Cell Metabolism 4:407-414. (2006). Pierre Chambon was ranked fourth among most prominent life scientists for the 19832002 period. Honorary Professor at the Collège de France and Emeritus Professor at the Faculté de Médecine, he was the founder and first Director of the IGBMC and the Institut Clinique de la Souris (ICS). During his PhD training performed under Pierre Chambon’s supervision, Daniel Metzger revealed a striking conservation of the molecular mechanisms underlying transcriptional activation across eukaryotes. He was recruited in 1995 at the CNRS, and established CreERT2, an efficient system to perform spatio-temporally controlled targeted somatic mutagenesis in the mouse, which is now used worldwide. Daniel then applied this mouse mutagenesis system to investigate the role of nuclear receptors in vivo. These studies brought major insight on the functions of nuclear receptors, notably in intermediary metabolism and skeletal muscle. He is presently co-leading with Pierre Chambon a research group on functions of nuclear receptors in homeostasis and diseases. n 2 47 highlights • The DNA damage sensors Parp1 and Parp2 contribute to the resolution of AID‐induced DNA breaks during CSR by facilitating DNA repair by the alternative‐NHEJ pathway and by suppressing oncogenic translocations. • The transcription factor Ikaros plays a key role in establishing a transcriptional competition between switch regions in individual cells that is critical for isotype specification during CSR. 1 probing complexity The functional properties of antibodies are shaped during immune responses through somatic hypermutation (SHM) and class switch recombination (CSR). SHM modifies antibody affinity, while CSR modulates antibody effector functions by replacing the antibody isotype expressed. These reactions are unusual among somatic cells in that they are initiated by DNA damage and both have been implicated in the development of cancer. They are triggered by Activation Induced Cytidine Deaminase (AID), an enzyme that deaminates cytidine residues in DNA. B lymphocytes are particularly prone to malignant transformation and it has been proposed that DNA damage incurred during CSR or SHM may account for this cancer susceptibility. Such damage might be due to bystander gene targeting by AID or alternatively to aberrant DNA repair. AID has the potential to inflict significant collateral DNA damage. Nevertheless, most B cells expressing AID do not suffer widespread mutation or chromosome instability and only a few non‐immunoglobulin genes are known to be mutated. Therefore, it appears that specific regulatory mechanisms restrict this potential to the appropriate cell type and loci. Despite great progress, how specific AID targeting is achieved in vivo and how AID‐induced DNA damage is accurately repaired is not known.< shaping the future Molecular biology of B cells SHM and CSR are initiated by programmed DNA damage induced by AID. This type of damage is normally restricted to antibody genes. However, aberrant targeting of AID to nonimmunoglobulin genes and/or abnormal repair of AID induced lesions may account for the increased propensity for malignant transformation in B cells. Our projects will provide insight into the molecular mechanisms that control AID activity and that enforce the accurate repair of AID‐induced DNA damage. Furthermore they will allow us to further understand the complex problem of double stranded DNA break detection, signaling and repair. The conclusions drawn from these projects will not only foster our future studies on B cell repertoire diversification, but will also have important implications for the fields of DNA repair and cancer. < Bernardo Reina B cell diversification and humoral immunity • AID associates with KAP1 and HP1 in a complex that is tethered in vivo to transcribed switch regions bearing the H3K9me3 mark. These results provide a novel and original AID targeting mechanism that accounts for the epigenetic modifications induced at the immunoglobulin heavy chain locus during CSR. • We have developed an innovative tool that allows us to homogeneously induce double stranded DNA breaks at a single genomic locus of known sequence, and for the first time, to isolate and identify novel factors recruited into DNA repair foci. n Robert, I., Dantzer, F. and Reina‐San‐Martin, B. Parp1 facilitates alternative NHEJ, whereas Parp2 suppresses IgH/c‐ myc translocations during immunoglobulin class switch recombination. J Exp Med. 206:1047‐56. (2009). Sellars, M., Reina‐San‐Martin, B., Kastner, P. and Chan, S. Ikaros controls isotype selection during immunoglobulin class switch recombination. J Exp Med. 206:1073‐87. (2009). 1- Histological section of a lymph node showing germinal centers, the sites of B cell receptor diversification by SHM and CSR. 2- Collateral damage to the genome triggered by AID expression. Robbiani, D.F., Bothmer, A., Callen, E., Reina‐San‐Martin, B., Dorsed, Y., Difilippantonio, S., Bolland, D.J., Chen, H.T., Corcoran, A.E., Nussenzweig, A. and Nussenzweig, M.C. AID is required for the chromosomal breaks in c‐myc that lead to c‐myc/IgH translocations. Cell. 135:1028‐38. (2008). Reina‐San‐Martin, B., Chen, J., Nussenzweig, A. and Nussenzweig, M.C. Enhanced intraswitch region recombination during immunoglobulin class switch recombination in 53BP1‐/‐ B cells. Eur J Immunol. 37:235‐9. (2007). Bernardo Reina obtained a PhD in Immunology at the Pasteur Institute (Paris). There he revealed that Trypanosoma cruzi (etiological agent of Chagas’ Disease) produces a B cell mitogen that enhances the susceptibility of the host and proposed a novel and alternative vaccination strategy that can be generalized to other pathogens of medical importance (2000). Bernardo then relocated to New York, USA for a postdoctoral stay with Michel Nussenzweig at the Rockfeller University and the Howard Hughes Medical Institute (HHMI) and began working on somatic hypermutation (SHM) and class switch recombination (CSR), two mechanisms initiated by Activation Induced cytidine Deaminase (AID), which are essential for establishing highly specific and adapted humoral responses. He implicated the DNA damage response pathway in the mechanism of CSR (2001) and proposed that its role is to induce chromatin conformational changes that facilitate long-range recombination (2003-2005). In 2006, he created his own group at the IGBMC. n 2 49 highlights • Cancer and metabolism: We have identified MMP11 as an unusual matrix metalloproteinase that favors cancer cell invasion through the reprogramming of cancer-associated adipocytes (CAAs) towards fibroblast-like cells. 1 probing complexity The natural history of breast cancer involves progression through clinical stages starting with abnormal epithelial proliferation, progressing into in situ and invasive carcinomas, concluding in metastatic disease. The progression from in situ to invasive carcinoma is a critical step in terms of prognosis and relies on cancer and normal cells present in the tumor. In this context, MMP11 identification as a secreted mesenchymal factor associated with invasive breast cancer was a pioneer finding in the field of invasion. Using a variety of cellular and mouse models of tumorigenesis, MMP11 expression by stromal cells was shown to be crucial in the invasive process. Moreover, prognostic studies indicated its association with a poor patient outcome. Indeed, the Oncotype DX (Genomic Health, Redwood City, CA) that predict breast cancer relapse includes MMP11 dosage. Breast cancer is a heterogeneous disease that is currently classified into 3 main classes: luminal (estrogen receptor (ER) and progesterone receptor (PR) positive), HER2 (a member of the epidermal growth factor (EGF) receptor family) positive and basal-like, which is primarily PR, ER and HER2 negative (ie, triple negative). By comparing benign and malignant breast biopsies, we have identified several genes including pS2/ TFF1 expressed in luminal tumors and TRAF4, MLN51 and STARD3 which are specifically expressed in HER2 tumors. All of these genes are expressed by cancer cells and our research is focused on the function of the respective proteins. < • Cancer and mRNA metabolism: We have characterized the RNA binding protein MLN51 as a member of the exon junction complex (EJC) that controls gene expression at the mRNA level, and showed that MLN51 is essential for the survival of cells under stress. shaping the future Molecular and cellular biology of breast cancer We study breast cancer via the functional characterisation of selected genes and pathways. Obesity is a poor prognosis factor. We have shown a connection between MMP11, cancer progression and adipose tissue. We are investigating the molecular mechanisms behind MMP11 action on cancer-associatedadipocytes (CAAs). TFF1, alias pS2, is a secreted factor. Despite being a classic ER-induced gene, its role in breast cancers remains unclear. We are exploring the molecular basis of TFF1 function. HER2-positive breast cancer, usually classified as high risk, is hardly a homogeneous disease. Indeed, some patients do not experience relapse, and HER2-targeted therapies show variable response. TRAF4, MLN51 and STARD3 are overexpressed in HER2-positive tumors. Their molecular functions in normal and malignant conditions are under examination. They may help to define HER2-positive tumor subsets. Altogether, our studies will reveal targets and molecular pathways that will help to understand the natural history of breast cancer and will be translated into the clinic. < Marie-Christine Rio Development & progression of breast cancer • Cell polarity: We have demonstrated that the TRAF4 adaptor protein is essential during development and acts in epithelial cells as a tight junctionassociated dynamic signaling component. • Translational research: Recently, in collaboration with the Breast Clinical Center in Strasbourg, we showed that hormone-dependency is underestimated in pregnancy-associated breast cancers and should be considered for patient management. n Tumor necrosis factor receptor-associated factor 4 is a dynamic tight junction-related shuttle protein involved in epithelium homeostasis. Kédinger V, Alpy F, Baguet A, Polette M, Stoll I, Chenard MP, Tomasetto C, Rio MC. PLoS One. 3(10):e3518; (2008). 1- Transverse section of a human mammary gland normal lobule showing honeycomb tight junction structure with TRAF4 (green) co-localizing with occludin (red) .2- X-ray computed tomography (microCT) of metastasis development (green) in the lungs of live mouse. Matrix metalloproteinase-11/stromelysin-3 exhibits collagenolytic function against collagen VI under normal and malignant conditions. Motrescu ER, Blaise S, Etique N, Messaddeq N, Chenard MP, Stoll I, Tomasetto C, Rio MC. Oncogene. 27(49):6347-55. (2008). The exon-junction-complex-component metastatic lymph node 51 functions in stress-granule assembly. Baguet A, Degot S, Cougot N, Bertrand E, Chenard MP, Wendling C, Kessler P, Le Hir H, Rio MC, Tomasetto C. J Cell Sci. 120(Pt 16):2774-84. (2007). Catherine Tomasetto Catherine Tomasetto met Marie-Christine Rio in 1989 while she was a PhD student in Pierre Chambon’s laboratory. Marie-Christine had conducted clinical research for developing tests for hormonal receptor dosage in prostate cancer at the Strasbourg Hospital. She had worked since 1982, on pS2/TFF1, a marker for breast cancer hormonodependence. In 1990, the team demonstrated the role of Matrix Metalloproteinase 11 (MMP11), a paracrin factor synthesized by normal stromal cells into the tumor. The same year, Catherine went to the Dana-Farber Cancer Institute in Boston, US for a post-doc position. She focused on a tumor suppressor for breast cancer and used subtractive hybridization screening. In 1993, she returned to Marie-Christine’s team and applied this technology to patient tumor biopsies. Currently, they lead a team together at the IGBMC and are interested in tumor development and progression. n 2 51 highlights • Retinoic acid induces the rapid activation of the p38MAPK/MSK1 pathway through non genomic effects. Subsequently, RARs become phosphorylated at two residues through a coordinated cascade starting with the phosphorylation by MSK1 of a serine located in the ligandbinding domain. This phosphorylation increases the ability of RARs to interact with the cdk7/cyclinH subcomplex of TFIIH with a downstream consequence on the phosphorylation of a serine located in an N-terminal proline-rich motif. 1 probing complexity We work at the interface of cellular and molecular biology focusing on retinoic acid, the active metabolite of vitamin A, which controls cell proliferation and differentiation through nuclear receptors. These receptors, RARs, work as ligand-dependent transcriptional regulators but complexity came with the finding that RARs are targets for post-translational modifications such as phosphorylations, which turned out to be crucial for their transcriptional activity. Our goal is to dissect the mechanisms and consequences of RARs phosphorylation. We analyze how phosphorylation controls the dynamics of RAR-target genes’ transcription. We also focus on how phosphorylation interferes with ubiquitination for RARs transcriptional activity and degradation. Given that many tumoral processes are characterized by aberrant kinase signaling pathways, we are engaged in a large analysis of RARs phosphorylation in cancer cell lines and tumor samples in correlation with their sensitivity to retinoic acid. The final important focus of our studies is to define how phosphorylation controls the activity of RARs during cell differentiation and embryonic development.< shaping the future Nuclear retinoic acid receptors phosphorylation and cross-talk with signaling pathways The importance of RARs phosphorylation stands in contrast to the difficulty of detecting this modification and of analyzing its biological significance. The challenge in the next years to come is to combine cellular, molecular, biophysical and computer modeling approaches to investigate : ● the phosphorylation state of RARs in different cell types. The purpose is to correlate aberrant RAR phosphorylation states with cancer ● the conformational changes induced by phosphorylation in order to predict changes in the interaction of RARs with protein partners or DNA ● the role of RARs phosphorylation during development by using induced pluripotent stem cells obtained from mouse embryonic fibroblasts and reexpressing RARs mutated at the phosphorylation sites in a RAR null background. < Cécile Rochette-Egly From cellular to molecular biology • Phosphorylation regulates the transcription of RAR-target genes via the control of RARs recruitment to gene promoters and via RARs ubiquitination and degradation by the proteasome. • New coregulators with SH3 domains and interacting with the N-terminal proline-rich motif have been cloned and shown to interact with RARs in a phosphorylationdependent manner. Bruck N, Vitoux D, Ferry C, Duong V, Bauer A, de Thé H, Rochette-Egly C. A coordinated phosphorylation cascade initiated by p38MAPK/MSK1 directs RARalpha to target promoters. EMBO J. 28(1):34-47. (2009). • RARs phosphorylation is altered in several breast cancer cells that are resistant to the antiproliferative effect of retinoic acid. n Bour G, Lalevée S, Rochette-Egly C. Protein kinases and the proteasome join in the combinatorial control of transcription by nuclear retinoic acid receptors. Trends Cell Biol. 17(6):302-9. (2007). 1- Nuclear localization of RARalpha (green) in breast cancer cells. 2- Aberrant cytosolic localization of RARalpha (green) in stromal fibroblasts from invasive breast carcinoma. Gaillard E, Bruck N, Brelivet Y, Bour G, Lalevée S, Bauer A, Poch O, Moras D, Rochette-Egly C. Phosphorylation by PKA potentiates retinoic acid receptor alpha activity by means of increasing interaction with and phosphorylation by cyclin H/cdk7. Proc Natl Acad Sci U S A.103(25):9548-53. (2006). After earning a diploma in Pharmacy and completing her residency in Paris, Cécile Rochette-Egly obtained a PhD in Cell Biology in Villejuif in 1978. At that time her work dealt with cell signaling and cyclic nucleotide second messengers. In 1979 she joined Inserm as a Research Associate and in 1980 she obtained a Honorary Doctorate of Science on the role of cyclic nucleotide in biological functions. In 1988, she joined Pierre Chambon’s team in Strasbourg to work on nuclear retinoic acid receptors (RARs). Since 1992 she is an INSERM Research Director and in 1994 she moved to the IGBMC and became the leader of her own team on RARs phosphorylation. She showed that retinoic acid receptors are targets for phosphorylation cascades which turned out to be crucial for their activity. n 2 53 highlights • A new cellular compartment: We have identified and characterized new cytoplasmic structures involved in mRNA decay that contain, among others, decapping enzymes. • Hidden transcripts: We have identified a new poly(A) polymerase and have shown that it is involved in a RNA quality control pathway that targets a new type of transcript: Cryptic Unstable Transcripts (CUTs). 1 probing complexity The control of gene expression allows cells and organisms to adapt to changing conditions and to drive internal Programmes such as cell cycle or development. Gene expression is an intricate multistep pathway in eukaryotes, allowing for multiple layers of regulations, but this process is also error prone. Thus, potentially deleterious aberrant transcripts need to be rapidly eliminated. Our goal is to understand how the RNA decay process contributes both to regulated gene expression and to the elimination of defective transcripts. Several pathways implicated in the degradation of eukaryotic RNAs have been identified. If several or most of the enzymes involved in this process are now known, the mechanisms activating specific enzymes in defined conditions remain to be elucidated. For example, we would like to decipher the mechanisms allowing cells to discriminate bona fide mRNA from defective transcripts or how the stability of specific RNA is regulated. This requires the characterization of the various protein complexes implicated in RNA decay and of their dynamic interactions. Additional complexity arises from the connection of mRNA decay to other cellular processes. Integrated approaches are thus necessary to decipher globally the contribution of mRNA decay to gene expression control.< shaping the future Protein networks and complexes regulating eukaryotic mRNA decay RNA decay remains a complex and poorly understood process involved both in the constant renewal of mRNA used for protein synthesis and in the elimination of defective transcripts. Enzymes involved in this process act in general turnover and RNA quality control pathways. We will now concentrate on the mechanism leading to activation of these factors. Using mechanistic, cellular and integrated approaches, several lines of research will be pursued using both yeast and mammalian cells as model systems: (i) How do cells distinguish aberrant from functional RNAs? (ii) How is mRNA decay regulated? (iii) Can we decipher the dynamic protein interaction networks underlying RNA decay? (iv) How is RNA decay integrated at the cellular level (e.g., with transcription and translation)? < Bertrand Seraphin • Protein complexes mediating RNA decay: Using a combination of approaches including innovative protein purification and mass spectrometry strategies, we have contributed to the characterization of the organization, structure, function and activities of various protein complexes involved in eukaryotic mRNA decay such as the And RNA disappears exosome and the Exon Junction Complex (EJC) core. Lebreton A, Tomecki R, Dziembowski A, Séraphin B. Endonucleolytic RNA cleavage by a eukaryotic exosome. Nature. 456:993-996. (2008). • Regulating gene expression at a post-transcriptional level: We have characterized yeast and human deadenylases and some of their regulators to understand how this process is integrated in the cell and contributes to the control of protein production. n Mauxion F, Faux C, Séraphin B. The BTG2 protein is a general activator of mRNA deadenylation. EMBO J. 27:1039-1048. (2008). 1- mRNA decay bodies (Dcp bodies or P bodies) in HEK293 cells: red immunodetection of hDcp1a; blue: DNA/nuclei. 2- Monitoring the effect of mRNA decay factors on translation termination: different yeast mutants harboring a reporter for stop codon readthrough turn from white to red (right), or grow better (left), on appropriate media. Dziembowski A, Lorentzen E, Conti E, Séraphin B. A single subunit, Dis3, is essentially responsible for yeast exosome core activity. Nat Struct Mol Biol. 14:15-22. (2007). Wyers F, Rougemaille M, Badis G, Rousselle JC, Dufour ME, Boulay J, Régnault B, Devaux F, Namane A, Séraphin B, Libri D, Jacquier A. Cryptic pol II transcripts are degraded by a nuclear quality control pathway involving a new poly(A) polymerase. Cell. 121:725-737. (2005). After studying at the ENS, Bertrand Seraphin obtained his PhD in 1988 at Curie Institute in Orsay on mitochondrial intron splicing, particularly on the role of a protein from a new family, the DEAD Boxes. From 1987 to 1989, while he was a post-doctorate at Brandeis University in Boston, USA with Professor Rosbash, he studied yeast nuclear splicing and brought to light new splicing complexes (commitment complexes). In 1991, he joined the EMBL, with Iain Mattaj and then became a team leader. His work on characterization of splicing complexes (Sm proteins, snRNP U1 et U6, etc.) brought him to develop a protein complexes purification strategy, the “Tandem Affinity Purification” (TAP method). In 2000, he integrated the CGM (Gif-sur-Yvette) where he studied RNA decay, discovering new enzymes, a new cellular compartment and new nonannotated transcripts: CUTs (Cryptic Unstable Transcripts). His research has been continuing at the IGBMC since 2009. n 2 55 highlights • 3D tracking of the motion of DSBs in living cells has shown that free chromosome ends have limited motion and they are unable to roam the cell nucleus. • The repair protein Ku80 is involved in keeping the free chromosome ends aligned. • Analysis of translocation partners of unrepaired chromosome breaks has shown that they preferentially undergo translocations with neighboring chromosomes. 1 probing complexity Each cell in the human body receives thousands of DNA lesions per day. DNA lesions can interfere with genome replication and transcription, and if they are not repaired or are repaired incorrectly, they lead to mutations that may threaten cell viability. The most deleterious DNA breaks are the Double Strand Breaks (DSBs) because unfaithful repair can lead to the formation of cancerous chromosomal translocations. It is poorly understood why translocations between chromosomes recur at specific break points in the genome and even less is known about how ends from different DSBs meet in the cell nucleus. It was recently shown that broken chromosome ends are positionally stable and unable to roam the cell nucleus and that unrepaired DSBs preferentially undergo translocations with neighboring chromosomes. In our group we are using a unique cell system to induce DSB at a specific chromosomal location and to follow the fate of damaged DNA in living cells in real time. Our goal is to investigate the dynamics of DSBs in relation to the surrounding chromatin structure and nuclear architecture and to test how this is related to their repair and their involvement in the formation of chromosomal translocations.< shaping the future Cell Biology of genome integrity We are interested in the role of chromatin structure and nuclear architecture in the repair of Double Strand Breaks and the formation of chromosomal translocations. Our current objectives are: ● to visualize the dynamics of DNA damage response and the formation of chromosomal translocations in living cells. ● to study the role of chromatin structure and nuclear architecture in DNA damage response and formation of chromosomal translocations. ● to identify novel chromatin related proteins that are involved in DNA repair. The insights gained from these in vivo studies will have a significant impact on our understanding of how the organization of repair in the context of a highly compacted chromatin and compartmentalized nucleus contributes to prevention of genomic instability. < Evi Soutoglou Nuclear architecture and chromatin structure in DNA repair • DNA repair factors are spreading around the DSBs and they form microscopically visible structures known as repair foci. Mimicking of repair foci by immobilization of a single repair factor to chromatin has shed light into their critical role in the activation and the amplification of the DNA damage response. • siRNA screen in mammalian cells revealed several chromatin related proteins that are involved in DNA damage response and DNA repair (unpublished). n DNA repair: easy to visualize, difficult to elucidate. Nagy Z, Soutoglou E. Trends Cell Biol. 19(11):617-29. (2009). Evi Soutoglou obtained her PhD at the Institute of Molecular Biology and Biotechnology (IMBB) in Crete, (Greece) on transcription regulation in 2002. She then spent a short post-doc with Laszlo Tora’s group at the IGBMC. In 2003, she joined the National Institute of Health (NIH) and worked with Tom Misteli on nuclear architecture and chromatin dynamics in DNA repair. Evi created a system to visualize DNA double strand breaks in living cells. In 2009, she returned to the IGBMC to set up her own team. n The emerging role of nuclear architecture in DNA repair and genome maintenance. Misteli T, Soutoglou E. Nat Rev Mol Cell Biol. 10(4):243-54. (2009). 1- Chromatin decondensation induced by tethering of the repair factor MDC1 shown in 3D. 2- Dual color FISH with chromosome 3 (green) and chromosome 19 (red) paint probes in NIH 3T3 cells. Activation of the cellular DNA damage response in the absence of DNA lesions. Soutoglou E, Misteli T. Science. 320(5882):1507-10. (2008). Positional stability of single double-strand breaks in mammalian cells. Soutoglou E, Dorn JF, Sengupta K, Jasin M, Nussenzweig A, Ried T, Danuser G, Misteli T. Nat Cell Biol. 9(6):675-82. (2007). 2 57 highlights • We identified and characterized several novel subunits of the human and Drosophila general transcription factor, TFIID. We described that different TFIID complexes exist in the cells and that these complexes regulate transcription differentially. • We discovered and characterized novel TATA binding protein (TBP)-like factors, such as TLF (also called TRF2) and TBP2 (also called TRF3) and our work suggested that that these factors play a role as cell-type specific initiation factors. shaping the future Chromatin modifications and regulation of gene expression during differentiation We characterize the activities of several mammalian transcription factor complexes to get more insights in their function during cell growth and differentiation. The combination of different multidisciplinary approaches (i.e. ChIP-seq, proteomic, cell biology, imaging, and bioinformatics) will lead to the identification of genes and pathways regulated by these complexes and their function in mammalian cell differentiation and development. Our studies will also help to find ways to interfere with the action of these transcription factors, when needed in pathological situations. We anticipate that the results of our research will have a major impact on the field and will potentially lead to a paradigm for contemporary metazoan transcription regulation and multiprotein complex research.< LÁszlÓ Tora Studying transcription factors and coactivators • We demonstrated a tight coupling between initiation of transcription and the 3’ maturation of premRNAs. 1 probing complexity Our main research interest is to study how specific protein coding genes are turned on and off in the nucleus of a given cell, during growth, differentiation and development. We use biochemical, genetic, cell biology, imaging and structural approaches to study the problem of control of gene expression in different metazoan organisms. The lab’s research is broadly centred on three different axes, which all aim to better understand RNA polymerase II transcription: (i) the role of different proteins belonging in the TATA box binding protein (TBPs) protein family, (ii) the role of TBP-associated factor (TAF)-containing complexes (i.e. TFIID and SAGA); and (iii) the role of histone acetyl transferase (HAT)-containing chromatin remodelling coactivator complexes (i.e. SAGA and ATAC) in gene regulation. Our current and planned activities aim to better understand gene regulatory processes in different cellular systems and also during the development of intact vertebrate organisms. Our work on the different transcription factors also contributes to a better understanding of how a polyglutamine expansion in a SAGA subunit causes a neurodegenerative disorder (i.e. Spinocerebellar ataxia type 7) or how the deregulation of the deubiquitination function of human SAGA causes metastatic cancers.< • We discovered and characterized a new human histone acetyl transferase-containing coactivator Zhao Y., Lang G., Ito S., Bonnet J., Metzger E., Sawatsubashi S., Suzuki E., Le Guezennec X., Stunnenberg H.G., Aleksey Krasnov, Sofia G. Georgieva, Schüle R., Takeyama K.-I., Kato S, Tora L.* and Devys D.* A TFTC/ STAGA module mediates histone H2B deubiquitination, nuclear receptor activation and counteracts heterochromatin silencing. Molecular Cell. 29:92-101. (2008). complex (called TFTC or SAGA) playing a role in chromatin remodelling. Motivated to explore the importance of this factor in diseases, our findings have helped to uncover the mechanisms by which a polyglutamine expansion in a subunit of SAGA leads to photoreceptor dysfunction in a mouse model of SCA7. n Kurshakova M.M., Krasnov A.N., Kopytova D., Shidlovskii Y.V., Nikolenko J.V., Nabirochkina E.N., Spehner D., Schultz P., Tora L.* and Georgieva S.G.* SAGA and a novel Drosophila export complex anchor efficient transcription an mRNA export to NPC. EMBO J. 26:4956-4965. (2007). Helmlinger D., Hardy S., Abou-Sleymane G., Eberlin A., Bowman A., Gansmüller A., Picaud S., Zoghbi H.Y., Trottier Y., Tora L.* and Devys D.* GlutamineExpanded Ataxin-7 Alters TFTC/STAGA Recruitment and Chromatin Structure Leading to Photoreceptor Dysfunction. Plos Biology. 4(3):0432-0445. (2006). 2 1- Detection of specific subunits of a histone acetyltransferase complex (ATAC) by immuno fluorescence in mouse cells by using antibodies against Ada2a (red) and Ada3 (green). Merged images together with DNA (blue) are shown in the left panels. 2- Heat map representing density matrix after k-means clustering of genome wide transcription factor binding sites. Binding densities of four histone acetyl transferases (PCAF, CBP, MOF and TIP60) were clustered according to p300 binding loci. After graduating from the Eötvös Lorànd University in Budapest with a PhD in Biochemistry and Molecular Biology in 1985, Laszlo Tora did his post-doc in Strasbourg to study nuclear receptors, first with M. Bellard and then with Pierre Chambon. Between 1988 and 1989, he participated in the discovery that estrogen and progesterone nuclear receptors have two distinct activation domains. Since 1993, he has led his own team at the IGBMC and has worked on transcription regulation mechanisms. In 1994, his team identified and characterized three subunits of the TFIID transcription factor and showed that different TFIID complexes have a different regulation activity. In 1997, Laszlo demonstrated the coupling between transcription initiation and polyadenylation. He then found TFTC and proved its role in chromatin remodeling in 1998. In 2000 and 2004, he discovered and characterized new TBP-like factors. n 59 highlights • From functional characterization, therapeutic target validation and high throughput screening, we have identified a compound, XRP44X, which inhibits the Ras oncogene – Net transcription factor signalling pathway (Wasylyk et al., 2008) and metastasis in pre-clinical cancer models (patent application). 1 probing complexity Our mission is to create knowledge that could be beneficial for the treatment of cancer in the near future. We are interested in studying the cancer-related functions of novel genes that we have identified, as well as known oncogenes and tumour suppressors. This involves studying their mechanisms of action, validating them as targets for high throughput screens (HTS), and establishing their usefulness as markers for diagnosis and prognosis. We are studying several types of human cancer, including head and neck squamous cell carcinoma (HNSCC) and prostate cancer (CaP). HNSCC is particularly frequent in France; it accounts for 12% of deaths by cancer in males, and 5-year survival is about 30%. Prostate cancer is now the most frequent cancer in men (350,000, 20% in Europe). Treatment of primary tumours leads to initial remission that later progress to invasive growth and metastasis. The reasons for relapse are not known and are the topic of many ongoing research projects. There is an urgent need to understand the biological mechanisms of cancer progression, in order to develop new therapies and markers that could be used to successfully treat cancer patients.< • We have characterized a novel gene, TTLL12, whose expression increases with prostate cancer progression (Wasylyk et al., 2010), validated it as a target for HTS (patent application), developed an assay, and identified hits in a screen of a worldwide diversity set of compounds. shaping the future Molecular and cellular biology of cancer We aim to: ● complete the characterization of XRP44X in pre-clinical cancer models of metastasis and characterize the novel signaling pathway that is affected. ● use the hits from the screen of the world diversity set of compounds to develop compounds that will be useful to explore TTLL12 functions and establish whether they are effective for tumour therapy. ● study the functions of the genes in the gene-expression signature of future metastasis and use them to develop new treatments (Ano1). ● validate our model for intimate crosstalk between HIF1alpha and Net, and use this knowledge to develop treatments for hypoxia related diseases.< Biology applied to cancer • Gene expression signatures have been identified that distinguish subgroups of HNSCC associated with poor and good prognosis (Rickman et al., 2008 and Jung et al., 2010, respectively). • We have postulated a model for intimate crosstalk between signaling pathways involving the master regulator of the hypoxic response, HIF1alpha, and the transcription factor Net (Gross et al., 2008; Serchov et al., 2010). n n Wasylyk C, Zambrano A, Zhao C, Brants J, Abecassis J, Schalken JA, Rogatsch H, Schaefer G, Pycha A, Klocker H, Wasylyk B. Tubulin tyrosine ligase like 12, link to prostate cancer through tubulin post-translational modification and chromosome ploidy. Int J Cancer. (2010). 1 + 2 - Colocalisation of TTLL12 with filaments. TTLL12 (red, 1 & 2) colocalises with vimentin (green, 1) and microtubules (not shown), which may to related to its effects on tubulin modification, mitotic duration, chromosome number instability and cancer. Jung A, Briolat J, Millon R, de Reyniès A, Rickman D, Thomas E, Abecassis J, Clavel C and Wasylyk B. Biological and clinical relevance of transcriptionnally active human papillomavirus (HPV) infection in oropharynx squamous cell carcinoma. Int J Cancer. 126(8):1882-94. (2010). Wasylyk C, Zheng H, Multon MC, Debussche L and Wasylyk B. Inhibition of the Ras-Net (Elk-3) pathway and tumour growth in vivo by a novel pyrazole. Cancer Res. 68(5):1275-83. (2008). Bohdan Wasylyk After obtaining a PhD in Biochemistry at Glasgow University on DNA structure and DNA interactions with RNA polymerase during transcription, Bohdan Wasylyk was awarded an EMBO Fellowship to join Pierre Chambon’s laboratory in Strasbourg in 1975. He worked on eukaryotic RNA polymerases and regulation of transcription by nucleosomes and promoter elements (TATA sequences and enhancers). Bohdan was appointed CNRS Research Associate and focused his work on the regulation of gene expression by oncogenes (Ras, MDM2, etc.) and tumour suppressors (p53). He has focused on particular cancers, including head and neck squamous cell carcinoma and prostate cancer. Bohdan participated in HTS’s for small molecule therapeutics that target p53, Ras and TTLL12. This has resulted notably in the identification of XRP44X. n 2 61 How the architecture of biological systems sheds light on their function " We aim at determining biomolecular structures at the atomic level to position these molecules in functional complexes and to analyze or predict their motion. " P. Schultz Our Programme aims at understanding the mechanisms of cellular processes by correlating the structural and the functional properties of their macromolecular components. The regulation of gene expression in a broad sense, ranging from transcription regulation, chromatin structure, DNA topology to messenger RNA translation, forms the core scientific activity of our community. These fundamental biological processes are of major importance for public health and the research conducted. These impact on our understanding of human diseases, drug target identification and drug optimization, for example, the studies on nuclear hormone receptors and their co-regulators have strong implications for cancer, osteoporosis, obesity and type II diabetes. INTEGRATED STRUCTURAL BIOLOGY The research teams are highly interconnected since multiple experimental approaches have to be combined to address challenging projects. Highly specialized structure determination methods such as X-ray and neutron • The structures of eukaryotic transcription factors and co activators illuminate the mechanisms of transcriptional regulation and explain the action of hormones. • The ribosome machinery, determined in different functional states, explains the translation regulation pathways and the inhibition strategies of antibiotics. diffraction and diffusion, NMR, cryo electron microscopy and more recently, imaging are associated with multi parametric data analysis and modeling approaches developed in the bio-computing teams. Recombinant and endogenous biological sample production, purification and biophysical characterization are key players in the structure determination pipeline supported and de8 Groups veloped in our Programme. 29 Staff scientists 24 Postdoctoral fellows The vision of structural biology for the next years will 21 PhD students pave the way towards inte25 Engineers/Technicians grated cellular structural biology with the aim of addres1 Assistant sing multifactorial effects in the personalized context of a unique human being. Our contribution to this evolution translates as the multi-resolution analysis of large complexes from their atomic structure to their in-cell distribution and interactome, in the investigation of the dynamics of biological processes at all levels of complexity and in hybrid data integration for knowledge production. In line with this commitment, the future Center of Integrated Biology will host one of the two French nodes of INSTRUCT, the European infrastructure for integrative structural biology. n • Chromatin and DNA modifying enzymes are drug targets for human diseases. Our studies contribute to the better understanding of drug action and guide rational drug design. • Integrative bioinformatics and genomics combine evolutionary, ontologic and omics data to extract knowledge. • We are committed to the development of new conceptual methodologies and novel modes of instrumentation to lead us towards tomorrow’s discoveries. 63 highlights • PRMTs: Co-activator-associated arginine methyltransferase 1 (CARM1 also known as PRMT4) methylates a large variety of proteins which are vital to gene expression. Our high resolution structures of several isolated modules of CARM1 have highlighted molecular switches that expand our understanding of how CARM1 regulates its biological activities. Furthermore, complementary functional studies have provided clues on the mode of binding of arginine-containing substrate or product peptides to CARM1. 1 probing complexity Chromatin structure is a major barrier to all nuclear processes and its modulation is essential for correct cell growth. There is now clear evidence that defects in chromatin modulation are responsible for a wide range of diseases, including cancer. Epigenetic modifications have a large impact on chromatin structure, acting in synergy with ATP-dependent remodelers, histone variants and histone chaperones to regulate nuclear mechanisms. By combining state-of-the-art biochemical and crystallographic techniques, we aim to understand chromatin remodeling at the molecular level. Three main targets are the current focus of our research: protein arginine methyltransferases (PRMTs), histone chaperones and histone deacetylases (HDACs). Furthermore, we are also developing structurebased rational approaches to characterize small molecule modulators of the biological activities of our targets since they are frequently involved in human diseases. Our team also participates in collaborative research on several related projects which require our specific expertise. Present collaborations concern the structural studies of complexes of cancer-related targets such as the Translationally Controlled Tumor Protein (TCTP) and the mRNA of human histone H4.< shaping the future Structural biology of epigenetic targets Most epigenetic players involved in the regulation of gene expression are multi-protein complexes. We are addressing the challenge of characterizing in molecular terms the biologically relevant complexes (that contain our targets) by developing a structural biology-based approach, integrating identification, production, purification, biophysical characterization and structure determination. Combining multi-resolution structural analysis with biological-functional studies will allow us to gain a better understanding on how these complexes function in a biological context. A structure-based approach will also be used to discover, generate and validate novel epigenetic regulators, since the availability of such compounds is at the heart of new therapies against the expanding number of diseases shown to depend on epigenetic mechanisms. < Jean Cavarelli Understanding biology at the atomic scale • Histone chaperones: The structure of the C-terminal part of Spt6 revealed a non-canonical tandem SH2 domain essential for Spt6 interaction with the hyperphosphorylated RNA Polymerase II CTD. Furthermore, structural characterization of the interaction between Spt6 and Iws1 leads to specific mutants affecting yeast viability and we are deciphering the role of each partner in vivo. Bieniossek, C., Nie, Y., Frey, D., Olieric, N., Schaffitzel, C., Collinson, I., Romier, C., Berger, P., Richmond, T.J., Steinmetz, M.O., Berger I. Automated unrestricted multigene recombineering for multiprotein complex production. Nat. Methods. 6:447-450. (2009). • Technological developments: Besides our scientific commitments, our development of multi-expression tools and our structural and crystallographic expertise is essential to tackle our internal and collaborative projects. n Troffer-Charlier N., Cura V., Hassenboehler P. , Moras D. and Cavarelli, J., Functional insights from structures of coactivator-associated arginine methyltransferase 1 domains. EMBO J. 26(20):4391-4401. (2007). 1- Structure of the PRMT catalytic domain of CARM1with bound SAH at 2.2 Å. Two monomers are shown here (residues 140-480 of mouse CARM1) building an active dimer. 2- Model of the Spt6/Iws1 complex. Romier, C., James, N., Birck, C., Cavarelli, J., Vivarès, C., Collart, M.A., Moras, D. Crystal structure, biochemical and genetic characterization of yeast and E. cuniculi TAFII5 N-terminal domain: implications for TFIID assembly. J. Mol. Biol. 368:1292-1306. (2007). Klein, F.A.C., Atkinson, R.A., Potier, N., Moras, D. and Cavarelli, J. Biochemical and NMR mapping of the interface between CREB-binding protein and ligand binding domains of nuclear receptors: beyond the LXXLL motif, J. Biol. Chem. 280:5682-5692. (2005). Educated in chemistry, Jean Cavarelli changed his focus to biology and obtained a PhD in Structural Biophysics at the University of Strasbourg with Dino Moras’s team, focusing on crystallographic studies of proteinnucleic acid complexes (1987). He spent two years as a post-doctoral fellow at Purdue University in West-Lafayette, Indiana, USA where he solved the structure of an insect virus in 1989. Beginning as Associate Professor and then Full Professor in 1996 at the University of Strasbourg, he worked for a long time on aminoacyl-tRNA synthetasecomplexes before focusing on epigenetics targets, notably PRMTs. He has participated in several collaborative projects on proteins related to human diseases, nuclear receptor coactivators and transcription factors. The research of his team is currently focused on epigenetic targets. He is also strongly involved in teaching, leading several Programmes in structural biology and structural bioinformatics at the University. n 2 3 65 highlights • Identification of dynamically coupled amino acids in the I-domain of the integrin LFA-1 uncovers an allosteric network that participates in the complex signal transduction mechanism of integrins. The calculations show that active compounds can interfere with this dynamically coupled network and result in the perturbation of signal transmission. 1 probing complexity Molecular interactions that occur between biological molecules and their ligands are underlying the vast array of processes that take place in a living organism are. We are trying to understand and characterize the molecular recognition processes using computational approaches that permit the characterization of the molecule-level dynamics and aim to understand the thermodynamics of molecular recognition processes and to further comprehend the structural and dynamic consequences of such interactions. For example, we are trying to elucidate how molecular signals are transmitted in biomolecular systems. Recent applications focus on the nuclear receptor superfamily of ligand-activated transcription factors which play a primordial role in complex processes such as cell differentiation, development, and homeostasis. We study the interplay between ligand binding, structural changes and allostery in these receptors. Other proteins under study include the integrin cell adhesion proteins. These projects are developed in collaboration with experimental groups at the IGBMC and elsewhere. These studies also provide important information that can be exploited in rational drug design projects that are developed at the interface of molecular biology, biophysics and organic synthesis groups.< shaping the future Biocomputing Our goal will be to use computational tools to study structure-function relationships of macromolecular complexes implicated in transcription regulation, in collaboration with other IGBMC teams involved in structural biology, experimental biophysics and bioinformatics. We will use atomic-level simulation studies of nuclear receptors to further our understanding of the molecular mechanisms of ligand recognition and allosteric communication in these proteins. Specifically, we shall study the role of phosphorylation-mediated structural and dynamical changes, the associated molecular mechanisms and implications for allosteric communication. We will also work towards the modeling of large macromolecular assemblies using hybrid computational and experimental methods. < Annick Dejaegere Computation for biology • A dynamically coupled amino acid network is identified in the ligand-binding domain of human peroxisome proliferator-activated receptor-g nuclear receptor protein. This network was shown to be influenced by the presence of agonist molecules. • Free energy decomposition of protein-protein and protein-ligand complexes provides insight into the molecular mechanisms of supramolecular assembly and pinpoints highlights interactions that can be used as starting points for drug design. n G. Moroy, A. Dejaegere, R.H. Stote, “Molecular basis for BH3 domain recognition in the BCL-2 protein family: Identification of conserved hot-spot interactions”. J. Biol. Chem. 284:17499-17511. (2009). C. Browning, E. Martin, C. Loch, JM Wurtz, D. Moras, R.H. Stote, A.P. Dejaegere, I.M.L. Billas, “Critical Role of Desolvation in the binding of 20-Hydroxyecdysone to the ecdysone receptor”. J. Biol. Chem. 282:32924-32934. (2007). 1- Representation of functional motions in the I-domain of the integrin LFA-1. 2- Representation of the dimer of the ligand binding domains of the nuclear receptors RAR and RXR. Analysis of the stabilizing amino acids at the dimer interface shows a conserved motif (highlighted in surface representation). T. Gaillard, E. Martin, E. San Sebastian, F. P. Cossio, X. Lopez, A. Dejaegere, R. H. Stote, “Comparative Normal Mode Analysis of LFA-1 Integrin I-domains” J. Mol. Biol. 374:231-249. (2007). V. Lafont, M. Schaefer, R. H. Stote, D. Altschuh, A. Dejaegere « Protein-protein recognition and interaction hot spots in an antigen-antibody complex: free energy decomposition identifies “efficient amino acids”. » Proteins. 67:418-434. (2007). Annick Dejaegere obtained a PhD in Physical Chemistry at the Free University of Brussels in Belgium. In 1989, she studied interactions in aromatic molecules with Jacques Reisse. She joined Martin Karplus’ team for her post-doc at Harvard University, USA. She worked on the reactivity of phosphates and proposed an original interpretation on the role of solvation in the reactivity of these molecules. In 1993, she joined the NMR group of JeanFrançois Lefèvre at the (Louis-Pasteur) University of Strasbourg, and developed predictive models of chemical shift for NMR structure determination. In 1995, her work highlighted original links between DNA backbone structure and chemical shift. In 2002, she joined IGBMC where she leads a team dedicated to the computational study of protein complexes. n 2 3 67 highlights • We have elucidated the solution structure of several domains of transcription factor TFIIH and have studied their interaction properties. In particular, we have shown that the self-association properties of p8, are closely linked to trichothiodystrophy. shaping the future Biomolecular Nuclear Magnetic Resonance The availability of a new 700 MHz spectrometer equipped with a high sensitivity probe opens new perspectives for the group. Efforts to describe disordered and functionally important regions of proteins will be continued. Particular emphasis will be put on the study of N-terminal regions of several nuclear receptors, aiming at understanding the rules that govern their evolution. Efforts will also be devoted to study molecular interactions within large complexes such as nucleosomes or within cellular environments.< • The solution structure of the C-terminal zinc finger domain of the oncoprotein E6 was elucidated. The structure displays a novel fold and suggests a model of interaction with PDZ domains. 1 probing complexity NMR spectroscopy provides invaluable insights into the structural and dynamical features of biomolecular systems. We are taking advantage of NMR, combined with other structural biology methods, to investigate molecular properties that are important to achieve a given biological function. Recent topics which have been addressed by the group include the study of several structural domains of transcription factors such as TFIIH and SAGA. The group is also involved in the study of the Human Papilloma Virus E6 protein and its interaction with host PDZ domains. Often, the molecular plasticity of proteins is crucial for their function, as illustrated by the self-association properties of p8, the smallest subunit of TFIIH or by the role of flanking regions of PDZ domains in protein-peptide interactions. Recently, novel experimental and theoretical tools have been developed to study the disordered states of proteins that are involved in signaling mechanisms. These tools are currently used to study AB domains of nuclear receptors such as RARg and to describe their modification upon phosphorylation.< Bruno Kieffer Understanding the intimate nature of proteins • Two homologous zinc finger domains of the Deubiquitination complex of SAGA were studied. Despite a common zinc binding motif, the two domains exhibited distinct secondary structures and functional properties. In particular, it was found that the SCA7 zinc finger of ATXN7 protein was able to bind nucleosomes with high affinity. • The concept of fractal dimension has been suggested as a functionally relevant property to describe disordered segments of proteins. n Augé S, Schmit PO, Crutchfield CA, Islam MT, Harris DJ, Durand E, Clemancey M, Quoineaud AA, Lancelin JM, Prigent Y, Taulelle F, Delsuc MA. NMR measure of translational diffusion and fractal dimension. Application to molecular mass measurement. J Phys Chem B. 113:1914-1918. (2009). Vitorino M., Coin F., Zlobinskaya O., Atkinson A.R., Moras D., Egly J.-M., Poterszman A., Kieffer B. Solution structure and self-association properties of the p8 TFIIH subunit responsible for trichothiodystrophy. J Mol Biol. 368(2):473-480. (2007). 1- Structure of the TFIIH MAT1 RING domain illustrating the dialog between structure and sequence 2- Structure of the TFIIH p8 subunit and the corresponding 1H-15N HSQC spectrum Nominé Y., Masson M., Charbonnier S., Zanier K., Ristriani T., Deryckère F., Weiss E., Orfanoudakis G., Kieffer B., Travé G. Structural and functional analysis of papillomavirus E6 oncoprotein: New insights in the molecular pathways of HPV-mediated pathogenesis. Molecular Cell. 21(5):665-678. (2006). Bruno Kieffer obtained his PhD on the NMR of proteins with Jean-François Lefèvre’s team at Strasbourg University in 1992 and relocated to Oxford University for postdoctoral research where he resolved the first structure of the surface protein CD59 whilst in the group of Iain Campbell. On his return Bruno was appointed Associate Professor at the ESBS in Strasbourg where besides teaching bioinformatics and biophysics, his research continued employing NMR to study bio-molecular behavior and function. In collaboration with the other teams of the Institute and the IGBMC, Bruno worked on transcription factors such as TFIIH and SAGA and successfully determined the solution structure of the PH domain of the TFIIH p62 sub-unit, and three years later, the 10th subunit, called p8. Continuing his close professional relationship with the ESBS, Bruno elucidated the structure of the human papillomavirus E6 oncoprotein. This research led to a registered drug patent for medical protein control. n 2 3 69 highlights In the past few years we have made major progress in the study of bacterial translation initiation complexes and human nuclear receptor complexes: • We have trapped a 70S ribosome initiation complex with initiation factor IF2 and revealed the conformational changes of IF2 and of the ribosome occurring upon GTP-hydrolysis. 1 probing complexity Transcription and translation are fundamental molecular mechanisms of gene activity regulation with profound implications for human health. The ligand-dependent transcriptional regulation by nuclear receptors bound to DNA response elements involves the transient assembly of large co-regulator complexes. These trigger chromatin remodeling and facilitate the assembly of the general transcription machinery on the promoter of the target gene. Gene expression is also regulated at the level of protein synthesis, for example, by protein factors that bind to the ribosome during the translation initiation, elongation and termination phase. The initiation phase is strongly regulated by factors and also by the mRNA itself and well-characterized reaction intermediates of the initiating ribosomal nano-machinery are potential targets for antibiotics. Both transcription and translation complexes represent large, transient macromolecular assemblies that we investigate by using an integrative structural biology approach with crystallography and cryo-electron microscopy forming the core.< shaping the future Large complexes involved in gene expression In the coming years we will focus on understanding the function of large macromolecular complexes involved in transcription and translation. Our particular aim is to determine the functional and structural determinants of ligand-dependent transcription activation of nuclear receptor and co-regulators and to address the mechanism of action of chromatin associated complexes. We will also explore the specific differences between the pro- and eukaryotic protein synthesis machineries. For this, we will pursue an integrative structural biology approach, combining, biochemistry, bio-physics and bio-informatics, with software developments for three-dimensional reconstruction and multi-state image processing as well as an enhanced integration of multi-scale tools. < Bruno Klaholz • We have also visualized for the first time the 30S initiation complex in the presence of the initiator tRNA and initiation factors IF1 and IF2, revealing the cooperative stabilization of the tRNA in this first event of gene decoding during protein synthesis. Integrative biology to understand the mechanisms of gene expression • We have also achieved major advances in the study of the architecture of a full nuclear receptor complex, while previously structural studies have been limited to the individual ligand-binding and DNA-binding domains. A. G. Myasnikov, A. Simonetti, S. Marzi, B. P. Klaholz. Structure-function insights into prokaryotic and eukaryotic translation initiation. Curr. Op. Struct. Biol. 9:300-309. (2009). • Furthermore, we have shown the important role of mRNA structure as illustrated by the transient ribosome-entrapment by a folded mRNA. Notably, sequence and structure analysis suggests the existence of a conserved site on the ribosome for binding regulatory mRNAs. n A. Simonetti, S. Marzi, A. G. Myasnikov, A. Fabbretti, G. Yusupova, M. Yusupov, C. O. Gualerzi, B. P. Klaholz. Structure of the 30S translation initiation complex. Nature. 455:416-420. (2008). 1- Structure of the 30S translation initiation complex. 2- The conserved site on the ribosome for binding regulatory mRNAs during translation preinitiation S. Marzi, A. G. Myasnikov, A. Serganov, C. Ehresmann, P. Romby, M. Yusupov & B. P. Klaholz. Structured mRNAs regulate translation initiation by binding to the platform of the ribosome. Cell. 130:1019–1031. (2007). A. G. Myasnikov, S. Marzi, A. Simonetti, A. M. Giuliodori, C. O. Gualerzi, G. Yusupova, M. Yusupov, B. P. Klaholz. Conformational transition of initiation factor 2 from the GTP- to GDP-bound state visualized on the ribosome. Nat. Struct. Mol. Biol. 12:1145-1149. (2005). 2 Early on, Bruno Klaholz became interested in molecular recognition. During his PhD he worked with Dino Moras’ team at the IGBMC and became involved in crystallography to determine the three-dimensional structure of nuclear receptors and to analyze their specific interactions with pharmaceutical ligands. In 2001, he began his post-doc at Imperial College, London and extended his knowledge to single particle cryo-electron microscopy, allowing him to visualize the 3D structure of molecules in their native state. At that time he investigated the molecular function of ribosomes during termination of protein synthesis. In 2003, he returned to the IGBMC and subsequently created his own research team with a focus on the molecular mechanisms that regulate gene expression. For the study of these large complexes he favors an integrated approach using electron microscopy, crystallography, biochemistry and bioinformatics. Through advanced image processing and statistical analysis he also developed approaches to separate different structural states in a single sample. n 3 71 highlights • The functional relevance of two mutually exclusive signature motifs of the ligand binding domain that partition the nuclear receptor superfamily into two classes with specific molecular pathways is illustrated by the allosteric control of retinoic acid receptor (RAR) activity by phosphorylation. shaping the future Expression of genetic information Three aspects of transcription regulation by nuclear hormone receptors will be investigated: • The role of DNA in the correct positioning of the receptors and the architecture of their complexes with cofactors. • The molecular mechanisms that control the binding of cofactor proteins. • The structural aspects of NHR action at the chromatin level. Integrative structural biology tools will be used and developed to characterize structural and functional aspects both in vitro and in the cell. For the vitamin D and retinoids receptors, the investigation will be extended to the tissue and animal model (mouse) levels. < • Role of the ligand and drug design: i) the work on the insect ortholog USP of RXR provides novel insights into receptor-ligand binding evolution and dynamics; ii) a structural water channel has been characterized and used to design a more potent superagonist for the Vitamin D Receptor. 1 probing complexity Our goal is to understand the molecular mechanisms that control gene expression in human cells and animal models. We are focusing on the study of transcriptional regulation by nuclear hormone receptors (NHR), using integrative structural biology approaches to decipher the structural basis of the communication between nuclear receptors, DNA and components of the basal transcription machinery. NHR-mediated transcription is regulated by ligands and post-translational modifications. Understanding the role of these factors in molecular control mechanisms (ie. allosteric regulation of the activity), has important implications for pharmacological applications (drug design). Today’s integrative structural biology demands multiscale structural data and their translation into functional knowledge. We use X-ray crystallography, EM and NMR techniques to determine structure and dynamics at different levels of detail, ranging from atomic resolution of individual proteins or small complexes to tomographic analysis of large particles. Macromolecular complexes can be relatively stable (e.g. ribosomes) or transient. For the latter, the temporal component is crucial and reconstitution of the proper sequence of events in the cell using light microscopy tools is an essential part of our studies.< Dino Moras Molecular mechanisms of gene expression • The structural basis for understanding the role of DNA in the spatial organization of NHR heterodimers in functional transcription complexes with cofactors has been investigated using a combination of solution techniques. Several structures have been determined. • The first structure of HIV integrase in complex with DNA and the cellular cofactor LEDGF/p75 was determined by EM. Michel F, Crucifix C, Granger F, Eiler S, Mouscadet JF, Korolev S, Agapkina J, Ziganshin R, Gottikh M, Nazabal A, Emiliani S, Benarous R, Moras D, Schultz P, Ruff M. Structural basis for HIV-1 DNA integration in the human genome, role of the LEDGF/P75 cofactor. EMBO J. 28:980-91. (2009). • The X-ray structure of p8/Tfb5 in complex with another TFIIH subunit was elucidated, providing a molecular basis for trichothiodystrophy, a rare disease. n Hourai S., Rodrigues L.C., Antony P., Reina-SanMartin B., Ciesielski F., Magnier B.C., Schoonjans K., Mourino A., Rochel, N., Moras D. Structure-based design of a superagonist ligand for the vitamin D nuclear receptor. Chem. Biol. 15:383-392. (2008). 1- RXR/RAR on the RARβ2 promotor within a nucleosome.. 2- The HIV-1 integrase in complex with a cellular co-factor (LEDGF), viral and cellular DNA. Iwema T., Billas, I.M.L., Beck Y., Bonneton F., Nierengarten H., Chaumot A., Richards G., Laudet V., Moras D. Structural and functional characterization of a novel type of ligand-independent RXR-USP receptor. EMBO J. 26:3770-3782. (2007) Yusupova G., Jenner L., Rees B., Moras D., Yusupov M. Structural Basis for messenger RNA movement on the ribosome. Nature. 444:391-394. (2006) DIno Moras graduated with a degree in Chemistry, then went on to study structural biology as a post-doctorate fellow with Michael Rossmann at Purdue University, West Lafayette, Indiana, USA. Notably, he participated to the discovery of the Rossmann Fold in 1974. Dino returned to France in 1975 and created a team at the Institute of Chemistry in Strasbourg. In 1980, he founded the Crystallography and Structural Biology Department at the IBMC. In 1990, he discovered the partition of amino-acyl-tRNA synthetases in two classes and determined the first threedimensional structure of a class II tRNAsynthetase. Dino then worked on nuclear receptors in collaboration with Pierre Chambon and elucidated the first crystal structure of the retinoic acid receptor. From 2007 to 2009, he was Director of the IGBMC. n 2 3 73 highlights • AlExSys (Alignment Expert System) drives optimal strategies for high quality multiple alignment and analysis of complete sequences by dissecting the relationships between Programme strengths/ weaknesses and the information content of extensive set of sequences. 1 probing complexity Our research is focused on the management of the ubiquitous «data overload» from today’s high-throughput technologies. We use complementary bottom-up and topdown approaches to study the behaviour and evolution of biological systems, such as “hyperstructures” (macromolecular complexes, organelles, viruses…) or biological networks (metabolic, transcriptional, interaction as well as developmental or disease-related networks…). In the bottom-up approach, we study basic components and integrate the data to detect relevant patterns (e.g., proteins and their interactions within a complex). In the top-down approach, we establish our knowledge of the system and attempt to disassemble it, e.g. to study normal and abnormal (disease) processes. This involves the development of: - Original algorithms and software based on an evolutionary approach to analyse hierarchical systems in the light of their conservation and distribution in eukarya, - New data system architectures suitable for computer and data grids to allow rapid retrieval, organization and exploration of raw data and information and to extract hidden knowledge, - Bioinformatics pipelines for quality control, integration, analysis and real time maintenance of interconnected genomics data with the goal of understanding disease origins and identifying and developing new therapeutic targets.< • SM2PH-db involves automatically launched and updated cascade of data processing and analyses embedded in the BIRD system and Decrypthon data and computer grid to allow the study and interpretation of the molecular consequences of mutations in the context of all known human monogenetic diseases. shaping the future Evolutionary systems biology Addressing the complexity of biological systems will involve automatic value-added data processing and analysis, requiring extensive computational power as well as rapid access to dispersed but synchronized data resources. In this context, our research will focus on the development of original bioinformatics solutions deployed on advanced computer and data technologies to ensure automated, updated and sustained analyses. Our approach of comparison and modeling of biological systems will take advantage of the postulate “what is important is generally conserved”: i.e., billions of years of evolution and selection constitute a discriminating filter and a unique source of information for interpreting what the new data actually mean and how they can be used to address questions that need to be answered. < Toward integrative bioinformatics • The ortho-proteogenomics approach combines original sequence analysis tools with proteomics approaches highlighting the extensive sequence error rate in well studied proteomes and allowing the creation of Programme cascades for automatic prediction of sequence-error and proteome-level curation. Bard N, Bolze R, Caron E, Desprez F, Heymann M, Friedrich A, Moulinierr L, Nguyen NH, Poch O Toursel T (2010) Decrypthon Grid – Grid resources dedicated to neuromuscular disorders in “Studies in Health Technology and Informatics», IOS Press, in press. (2010). Olivier Poch • Following our study of the Bardet-Biedl Syndrome and the characterization of the BBS10 and BBS12 genes, extensive analysis and genetic diagnosis of hundreds of families allowed the identification of 28 novel mutations. This study highlights the burden of private mutations in an extensively heterogeneous disease and confirms that BBS1, BBS10 and BBS12 are the most frequently mutated genes. n Aniba MR, Poch O, Marchler-Bauer A, Thompson JD, (2010) AlexSys: a knowledge-based expert system for multiple sequence alignment construction and analysis Nucleic Acids Res. in press. (2010). 1- Derivation of the mean square error in the Multidimensional fitting algorithm 2- Pathways of PtdIns5P synthesis and transformation : a)Schematic representation of phosphoinositides, fatty acids are represented only for PtdIns with co-evolving complexes highlighted. b) Phylogenetic distribution of proteins implicated in the PtdIns5P metabolism in 39 eukaryotic organisms c) Inferred PtdIns5P metabolism in different organisms. Friedrich A, Garnier N, Gagnière N, Nguyen H, Albou LP, Biancalana V, Bettler E, Deléage G, Lecompte O, Muller J, Moras D, Mandel JL, Toursel T, Moulinier L, Poch O. SM2PH-db: an interactive system for the integrated analysis of phenotypic consequences of missense mutations in proteins involved in human genetic diseases. Hum Mutat. 31(2):127-35. (2010). Muller J, Stoetzel C, Vincent MC, Leitch CC, Etal...Poch O, Mandel JL, Dollfus H. Identification of 28 novel mutations in the Bardet-Biedl syndrome genes: the burden of private mutations in an extensively heterogeneous disease. Hum Genet. 127(5):583-93. (2010). Having been simultaneously at the Pasteur Institute and the IGBMC, Olivier Poch obtained a PhD from Strasbourg University on the determination of rabies virus genes sequence in 1987. He then focused on the analysis of the viral protein sequences and predicted that all monomeric polymerases from all living organisms may share a common unifying fold. In 1992, he became interested in the experimental study of the yeast cell cycle while he still worked on bioinformatics analysis, notably on tRNA synthetases where he contributed to the discovery of 2 classes in this ancestral protein family. In 1997, he joined the IGBMC to create a Bioinformatics laboratory and develop original approaches for multiple sequence alignment and comparative genomics programmes. He then focused on more integrative approaches working on functional genomics data, ontology development, clustering algorithms and recently, turned toward data mining and knowledge extraction approaches. n 2 3 75 highlights • TFIID flexibility: We have characterized major conformational changes of the transcriptional coactivator TFIID and found that the presence of the Taf2 subunit stabilizes an active conformation. • Activation of transcription: The cryoEM structure of a complex containing TFIID, TFIIA and the Rap1 activator assembled on a ribosomal gene promoter revealed the role of TFIIA in the activation process. TFIIA is part of a molecular switch that changes conformation upon interaction with the activator and triggers initiation complex assembly. 1 probing complexity Our team deciphers the three-dimensional organization of macromolecular complexes by using high resolution cryoElectron Microscopy (EM) to visualize single molecules in their native state and image analysis to extract signal from noise. We study how basal transcription factors interact with the promoter of protein-encoding genes and analyze the transmission of activation signals mediated by cis-acting transcription factors towards the basal transcription machinery in order to trigger increased transcription. The study of different intermediate states reveals snapshots of dynamic processes that highlight the conformational changes of these nanomachines during transcription initiation. These approaches were applied to several molecular systems including the SAGA and TFIID coactivators, but also to other systems such as TFIIH, TFIIE, RNA Polymerase I, DNA Topoisomerase II or HIV integrase, in collaboration mainly with IGBMC team leaders. As part of an integrated vision of cellular structural biology we analyze the organization of transcription within the nuclei of rod photoreceptors. The biological system used is a mouse model of a human disease: the type 7 Spinocerebellar Ataxia. Chromatin organization, histone modification and transcriptional activity were correlated in this context. < shaping the future Architecture of nucleoprotein systems by 3-D Electron Microscopy Future developments in the image analysis of large datasets of eukaryotic transcription complexes will reveal finer morphological details as well as a larger number of discrete conformational states. This wealth of information will allow us to fit more accurately atomic structure into our cryo EM maps and to reveal the dynamic processes underling transcription initiation. The analysis of frozen hydrated cell sections aims at positioning the transcription process in its cellular context and to visualize the molecular complexes in their functional chromatin environment. More complex in vitro systems will be established and studied to determine the structures of integrated biological architectures taking into account transcription factors in their chromatin context. < Patrick Schultz Visualizing molecules to understand their function • In vivo organization of transcription: The correlation of histone modifications, chromatin compaction and active RNA polymerase positions places the transcriptional scene within the cell nucleus. We found that transcription occurs in decondensed chromatin at a fixed distance from the heterochromatin interface. Papai G, Tripathi MK, Ruhlmann C, Layer JH, Weil PA and Schultz P. TFIIA and the transactivator Rap1 cooperate to commit TFIID for transcription initiation, Nature. 465(7300):956-60. (2010). • Mechanism of viral DNA integration: In collaboration with Marc Ruff we determined a 3-D model of the HIV-1 Integrase in the presence of DNA molecules. The cryoEM structures reveal the detailed molecular architecture of the integration complex. n Papai G, Tripathi MK, Ruhlmann C, Werten S, Crucifix C, Weil PA, Schultz P. Mapping the initiation binding Taf2 subunit in the structure of hydrated yeast TFIID. Structure. 17(3):363-373. (2009). 1- Three-dimensional model of the HIV integrase (blue) complexed to the cellular co-factor LEDGF (grey) in interaction with the viral DNA (modeled in two distinct conformations in gold) and the cellular DNA (red). 2- Extended chromatin fiber (red) visualized in an electron tomogram of a mouse rod photoreceptor nuclei. Dense heterochromatin is depicted in green whereas the euchromatin domain appears in blue. Michel F, Crucifix C, Granger F, Mouscadet JF, Korolev S, Gottik M, Nazabal A, Emiliani S, Benarous R, Moras D, Schultz P and Ruff M. Mechanism for viral DNA integration in the human genome by the HIV-1 integrase and role of human LEDGF. EMBO J. 28(7):980-991. (2009). Thauvin C, Rickling S, Schultz P, Célia H, Meunier S, Mioskowski C (2008) Carbon nanotubes as templates for polymerized lipid assemblies. Nat Nanotechnol. 3(12):743-8. (2008). Patrick Schultz graduated with a degree in Molecular Biology and Genetics from Strasbourg University and obtained his PhD in 1987 on the organization of chromatin working in Pierre Oudet’s team. During his postdoc at EMBL (Heidelberg), he had the opportunity to use cryo Electron Microscopy which opened a new prospect for visualizing molecules in their native, hydrated, environment. When he returned to Strasbourg he focused on the structural studies of multiprotein complexes involved in transcription and in its regulation. In 1991 he published the first 3-D model of a eukaryotic RNA polymerase. In 1994, the newly created IGBMC brought him closer to X-ray crystallography and NMR and since then he has tried to integrate these complementary structural approaches. Following Pierre Chambon’s pioneering work on transcription factors, he identified a structural model for TFIIH in 2000 and worked on the comprehension of the structure and the function of the TFIID complex. More recently, his research has turned towards understanding chromatin structure and the transcription units organization within the cellular environment. n 2 3 77 highlights • We have determined the first crystal structure of eukaryotic 80S ribosome from yeast. We have determined structures of several bacterial 70S ribosome complexes containing tRNAs and different mRNAs. We have proposed a mechanism of mRNA movement on the ribosome during translation. shaping the future Ribosomes X-ray crystallography of the yeast ribosomes will allow us to investigate the mechanism of regulation of translation in eukaryotes. This study will create a basis for the beginning of x-ray study of translation system in human cells. We will continue the investigation of the mechanism of translocation by x-ray structure determination of different ribosome functional complexes from bacteria and yeast. New antibiotics developed for inhibition of the ribosome activity in bacteria and fungi will be studied by x-ray crystallography. < • Our study of codon-anticodon interactions of tRNAPhe containing hypermodified nucleoside in the anticodon loop at position 37 (ms2i6A37) have shown how natural modification is involved in preventing frame-shifting and stabilizing mRNA-tRNA interactions. 1 probing complexity The ribosome is a large ribo-nucleoprotein complex with total molecular mass of 2 500 000 Dalton in bacteria and 3 300 000 Dalton in yeast. Several components contribute to the greater weight and complexity of eukaryotic ribosomes: RNA expansion segments that are inserted into the evolutionary conserved rRNA core and additional 25 proteins. The ribosome performs protein synthesis using a messenger RNA template. It mediates the interactions between mRNAs and tRNAs and it catalyses the peptide bond formation. Our group has developed methods for x-ray study of bacterial 70S ribosome from the extreme thermophilic microorganism Thermus thermophilus and recently for the 80S ribosome from the yeast Saccharomyces cerevisiae. The goal of our group is to study structure and mechanism of protein biosynthesis in bacteria, yeast and humans. < Marat Yusupov All about the ribosome • High resolution structures show that tRNA in the A and P sites communicate through a protein rich environment suggesting a mechanism for the control of these tRNAs position/function by the ribosome. • We have identified the network of ribosomal elements involved in proofreading of tRNA by comparing three crystal structures of the 70S ribosome with an empty A site or the A site occupied by cognate or near-cognate tRNA. n Jenner L, Demeshkina N, Yusupova G and Yusupov M (2010) Structural rearrangements of the ribosome at the tRNA proofreading step. Nat Struct Mol Biol. in press. Jenner L, Demeshkina N, Yusupova G, Yusupov M. Structural aspects of messenger RNA maintenance by the ribosome. Nat Struct Mol Biol. 17:555-560. (2010). 1- Crystals of the ribosome from yeast Saccharomyces cerevisiae diffracting to 4.2A resolution. 2- 80S ribosome from yeast Saccharomyces cerevisiae. 40S subunit in blue, 60S subunit in yellow-green. RNA expansion segments in red. Yusupova G, Jenner L, Rees B, Moras D, Yusupov M Structural basis for messenger RNA movement on the ribosome. Nature. 444:391-394. (2006). In 1978, Marat Yusupov began his research at the Institute for Protein Research in Pushchino, Russia. In 1986, he obtained his PhD on the study of ribosome structure. He then supervised a research team at the Institute for Protein Research and collaborated with Dino Moras for ribosome x-ray study. In 1996, he joined the University of California in Santa Cruz, USA and continued his work on the ribosome. In 2001, he made a major discovery with Harry Noller, Jamie H. D Cate and Gulnara Yusupova and determined the complete structure of the ribosome. This work largely contributed to the Nobel Prize Award in 2009 to Ada E. Yonath,Thomas A. Steitz and Venkatraman Ramakrishnan. Since then, he dedicates himself to the study of ribosome functioning and translation mechanisms. He became the leader of the Ribosome Team at the IGBMC in 2001. n 2 3 79 From the disease to the gene and vice-versa, pathomechanims and therapeutic strategies " We explore the genetic bases of physiological and pathological processes, and use genetic mouse models to understand human disorders." B. Kieffer Translational medicine & neurogenetics This Programme brings together nine research groups. Most of them address neural dysfunctions and all of the teams share a common interest in integrative mouse biology. Six independent teams constitute the human genetics component founded and animated by JeanLouis Mandel (Professor of Human Genetics at the Collège de France). This component of the Programme is focused on deciphering the mechanisms underlying several monogenic diseases affecting the nervous system or skeletal muscle. Projects include the identification of genes involved and of their mutations, as well as analysis of the normal function of cognate proteins and pathomechanisms responsible for the clinical phenotype, as a prerequisite for the development of novel therapeutic strategies. Among the transversal goals shared by several teams are the development of high-throughput sequencing approaches for the efficient identification of mutated genes and molecular epidemiology studies in heterogeneous genetic diseases. Strong links have been established with clinicians (patient recruitment, genotypephenotype studies) and with patients associations. Many teams are involved in teaching (Medical and Life Sciences faculties, College de France) and bringing science closer to the lay public. Annual meetings on important topics in Human Genetics Mouse models of human diseases : X-linked mental retardation, Coffin-Lowry Syndrome and mental retardation, Bardet-Biedl syndrome, Myotonic dystrophies, Friedreich and other recessive ataxias, Myotubularin-associated neuromuscular diseases, Polyglutamine and Huntington disease, Aneuploidies and Down syndrome, Chronic pain, Drug abuse, Depression, Atherosclerosis and thrombosis, Metabolic syndrome. Cellular processes involved in pathogenicity : Cytoskeletal proteins and membrane trafficking, intracellular cell signaling and kinases, Fe-S cluster biogenesis and cell metabolism, protein/RNA aggregation and cellular toxicity, mRNA transport, DNA repair, neurotransmission and membrane receptors, prostaglandin and leukotriene signaling are organized at the College de France. A novel team directed by Yann Hérault has joined recently and addresses the genetic bases of human chromosome 21 deficiencies with a main focus on the Down Syndrome. The research addresses phenotype-genotype relationships and gene dosage effects. Experimental strategies involve chromosomal engineering of the mouse genome and behavioral analysis of genetic mouse models. This team works in close collaboration with human geneticists. The team headed by Brigitte 9 Groups Kieffer explores the function of peptidic neuromodulatory 24 Staff scientists systems in complex behaviors. 17 Postdoctoral fellows The principle approaches include gene targeting in mice, 30 PhD students pharmacology and behavior, 21 Engineers/Technicians fluorescence imaging and electrophysiology. Focus is on the 1 Assistant opioid system and associated signaling pathways with a major goal being to decipher the molecular bases of pain control, hedonic homeostasis and emotional behaviors. This research impacts in areas of pathological pain, drug abuse, stress and mood disorders. This team has strong links with both molecular biologists and behavioral psychologists, and has recently established an international associated laboratory with the Scripps Research Institute (La Jolla, USA). Finally, a cardiovascular component is developing. A team headed by Jean-Etienne Fabre investigates signaling mechanisms leading to atherothrombosis and has developed unique mouse models to study the disease in vivo. A new team led by Romeo Ricci has been recruited recently. This team studies signal transduction mechanisms underlying the metabolic syndrome. n 81 highlights • CUG and CCUG expanded repeats sequester the MBNL1 splicing factor. We found that MBNL1 sequestration in DM patients leads to alternative splicing of the BIN1 pre-mRNA. In collaboration with Jocelyn Laporte’s team, we found that BIN1 splicing mis-regulation results in T-Tubules and muscle weakness in DM patients, cell and mouse models. 1 probing complexity The RNA gain-of-function diseases are a novel class of human genetic disorders in which expansion of repeated nucleotides are toxic at the RNA level. These diseases include Congenital and Adult type 1 and 2 Myotonic Dystrophies (DM), Fragile X-Associated Tremor/Ataxia Syndrome (FXTAS), Spinocerebellar Ataxia 10 (SCA10) and possibly SCA12 and Huntington’s disease-like type 2. These autosomal dominant genetic diseases are caused by expanded tri-, tetra- or penta-nucleotide repeats that are transcribed but are not exported, and accumulate in pathogenic nuclear RNA aggregates that sequester specific RNA-binding proteins, leading to molecular changes ultimately resulting in the pathological symptoms. While the general paradigm of these diseases is now established, very little is known on the detailed molecular mechanisms involved in these disorders. Our goal is to elucidate the molecular causes of CGG, CUG, CCUG and AUUCU RNA toxicity in FXTAS, DM1, DM2 and SCA10 patients, respectively. We are particularly focusing on the RNA binding proteins sequestered by these repeats and the cellular consequences of such sequestration.< shaping the future Physiopathology of the RNA gain-offunction diseases We will pursue the identification of the RNA binding proteins which are sequestered within the RNA aggregates in RNA gain-of-function patients. Notably, we just found that the FOX1 and FOX2 splicing factors co-localize with CCUG aggregates in DM2 patients. Furthermore, we discovered that the expression of specific mi-RNAs is altered in skeletal muscle and heart of Myotonic Dystrophic patients. We will study the molecular mechanism and the consequences of these alterations. We are currently developing in vitro and in cellulo mediumand high-throughput screens to identify pharmacological compounds able to reverse the toxic effects of expanded CUG, CCUG or CGG repeats. If successful, this work would constitute a first step toward identifying compounds able to reverse the pathogenesis in RNA gain-of-function diseases. < • We identified a novel function of MBNL1 as a regulator of micro RNA processing. Consequently, the processing of the micro RNA miR1 is altered in DM patients, resulting in cardiomyocyte dysfunctions. specific alternative splicing events regulated by SAM68 are altered in brain samples of FXTAS patients. • We identified that expanded CGG repeats sequester the DROSHA/ DGCR8 complex. DROSHA is the enzyme responsible for pre-micro-RNA processing. Consequently in DROSHA sequestration, we observed a global alteration of the micro RNA processing resulting in neuronal cell death in FXTAS patients. n Dhaenens CM, Schraen-Maschke S, Tran H, Vingtdeux V, Ghanem D, Leroy O, Delplanque J, Vanbrussel E, Delacourte A, Vermersch P, Maurage CA, Gruffat H, Sergeant A, Mahadevan M, Ishiura S, Buee L, Cooper TA, Caillet-Boudin ML, Charlet-B N, Sablonniere B, Sergeant N. Overexpression of MBNL1 fetal isoforms and modified splicing of Tau in the DM1 brain: two individual consequences of CUG trinucleotide repeats. Exp Neurol. 210(2):467-78. (2008). Charlet-B. N., Feuerhahn S., Kong S., Ziserman H., Conaway J., Conaway R., Egly J.M. RNA polymerase II bypass of oxidative DNA damage is regulated by transcription elongation factors. EMBO. J. 25(23):5481-91. (2006). Nicolas Charlet-Berguerand Fight against RNA aggregates • We found that expanded CGG repeats sequester the SAM68 splicing factor in FXTAS patients, cell and mouse models. Consequently, Sellier C., Rau F., Liu Y., Tassone F., Hukema RK., Gattoni R., Schneider A., Richard S., Willemsen R., Elliott DJ., Hagerman PJ., Charlet-B. N. Sam68 sequestration and partial loss of function are associated with splicing alterations in FXTAS patients. EMBO. J. January. In Press. (2010). Human Genetics 1- Co-localization of DHPR (green) and BIN1 (red) in mouse isolated muscle fiber. 2- Co-localization of BIN1 (green) and RYR1 (red) in human myotube. DNA is labeled in blue (DAPI) After having spent three years in Paris at The Centre de Génétique Moléculaire (CGM) with J. Marie’s team, followed by three more years in T. A. Cooper’s laboratory, Nicolas Charlet-Berguerand obtained his PhD in Paris in 2003 on splicing, applied to human diseases and in particular to myotonic dystrophies. In 2003, he began his post-graduate research on DNA repair in Jean-Marc Egly’s laboratory at the IGBMC. In 2006, he returned to the study of myotonic dystrophies and created a team dedicated to this topic at the IGBMC. Today, his research focuses also on Fragile X Tremor Ataxia Syndrome (FXTAS) a genetic disorder that also reveals, as in myotonic dystrophies, toxic RNA aggregates. His team studies these diseases and searches their associated proteins. They also test chemical compounds able to suppress the toxic effects and restore normal functions in patient cells. n 2 83 highlights • We have developed a new model for studying in vivo atherothrombosis and have developed new methods for investigating vasoconstriction in mice, and for quantifying ruptures of atherosclerotic plaques. 1 probing complexity Atherothrombosis, the occurrence of thrombosis on atherosclerotic plaques, is the main cause of myocardial infarction and stroke, the leading causes of death in developed societies. Atherosclerotic plaques are complex lesions of the vascular wall, provoked by oxidized lipid deposition which initiates and maintains local inflammation. Inflammation is itself a complex process that involves numerous lipid mediators, and its role in atherothrombosis is poorly delineated. To probe this complex picture, we focus our studies on the involvement of the arachidonic pathway in atherothrombosis. These molecules have the potential to interfere with the plaque vulnerabilization, with the vessel reactivity and with the blood platelet behaviour. We examine in mice how each prostaglandin and leukotriene produced by the plaque contributes either to the plaque rupture or to local thrombosis through a direct action on platelets. In the end, we hope to get a whole picture of the effects of these inflammatory mediators on atherothrombosis. Understanding the inflammation-thrombosis relationship will improve the way atherothrombosis is prevented and also might have important implications in other diseases where inflammation is complicated by thrombosis, such as cancer and auto-immune diseases. < • We have revealed in our laboratory that murine and human plaques produce significant amounts of PGE2 and we identified the PGE2/ EP3 pathway as aggravating atherothrombosis. As a consequence, we demonstrated that blocking EP3 allows inhibition of atherothrombosis without altering haemostasis, i.e. without increasing the risk of bleeding. shaping the future Atherosclerosis and thrombosis We need to keep developing our in vivo tools in order to improve atherothrombosis detection and quantification and are currently developing a method using MRI in collaboration with a German team for both experimental and clinical purposes. Our ultimate goal is to be able to describe by which pathways the whole inflammatory machinery impacts atherothrombosis. When we have delineated the roles of the main prostaglandins and leukotrienes, we will follow three directions: • keep investigating the arachidonic pathway through less known molecules (lipoxins, epoxy-derived compounds) • investigating the role of the sphingosin/ceramide axis • examining the involvment of cytokines. These studies should allow us to draw a global picture of the inflammation-thrombosis link. < Jean-Etienne Fabre Understanding cardiovascular diseases • Two classical inflammatory vasoconstrictors, LTC4 and TXA2 were used to demonstrate that vasoconstriction can break atherosclerotic plaques, however only when they are vulnerable. We showed that only TXA2 is produced by atherosclerotic plaques. Magnetic resonance molecular imaging of thrombosis in an arachidonic acid mouse model using an activated platelet targeted probe. Klink A, Lancelot E, Ballet S, Vucic E, Fabre JE, Gonzalez W, Medina C, Corot C, Mulder WJ, Mallat Z, Fayad ZA. Arterioscler Thromb Vasc Biol. 30(3):40310. (2010). • In plaques, we detected fair amounts of leukotriene B4, a powerful chemoattractor for neutrophils. We currently test the hypothesis that neutrophils degrade the fibrous cap by releasing their proteases, linking infectious diseases to the plaque rupture. We demonstrated that neutrophils can enter the plaques and release their enzymatic content. n Antagonists of the EP(3) Receptor for Prostaglandin E(2) Are Novel Antiplatelet Agents That Do Not Prolong Bleeding. Singh J, Zeller W, Zhou N, Hategen G, Mishra R, Polozov A, Yu P, Onua E, Zhang J, Zembower D, Kiselyov A, Ramírez JL, Sigthorsson G, Bjornsson JM, Thorsteinsdottir M, Andrésson T, Bjarnadottir M, Magnusson O, Fabre JE, Stefansson K, Gurney ME. ACS Chem Biol. 4(2):115-126. (2009). Vascular wall-produced prostaglandin E2 exacerbates arterial thrombosis and atherothrombosis through platelet EP3 receptors. Gross S, Tilly P, Hentsch D, Vonesch JL, Fabre JE. J Exp Med. 204(2):311-20 (2007). 1- SEM picture showing the rupture of an aortic murine atherosclerotic plaque. Inside the fracture, activated platelets are mounting the thrombotic response. After working for ten years as a Cardiologist in an intensive care unit, Jean-Etienne Fabre worked for the pharmaceutical industry for three years at LAFON Laboratories. In 1995, he joined J.M. Isner’s laboratory at Tuft University in Boston, (USA) to work on angiogenesis. There he studied the effects of conversion enzyme inhibitors on angiogenesis. He then joined B.H. Koller’s team at the University of North Carolina, Chapel Hill USA. There he showed that P2Y1 was an ATP receptor in blood platelets and discovered the existence of LTB4 transcellular metabolism. Jean-Etienne also proved that PGE2 promoted thrombosis in vivo. He obtained his PhD in 2000 from Strasbourg University. In 2001, he was contacted by Pierre Chambon and joined the IGBMC to focus on the link between explosive thrombosis and inflammation of atherosclerotic plaques. n 2 85 highlights 1 probing complexity Mental retardation (MR), defined as major deficits in cognitive functions manifesting early in life, originates from highly heterogeneous causes, that prominently include single gene mutations. We study several of these genes trying to decipher their function at the molecular, cellular and central nervous system level, as well as the consequence of their deficit in neurons and on cognitive functions in the mouse. We study the FMR1 gene mutated in the Fragile X MR syndrome, the most common cause of inherited MR, as well as two protein-kinase encoding genes, RSK2 and PAK3, responsible for X linked forms of syndromic (Coffin-Lowry) or non-syndromic MR. We are also involved in genetic studies of the Bardet-Biedl syndrome, a pleiotropic ciliopathy, and in genetic diagnosis applications related to MR. Our FMR1 studies focus on the processes involved in mRNA transport and local translation in dendrites that appear important for synaptic plasticity. Another focus is the role of signal transduction pathways that include RSK kinases and PAK kinases and their role in regulation of neurotransmission and in synaptic plasticity. Knowledge of the precise functions of these proteins will contribute to the better understanding of molecular pathomechanisms leading to MR and may open paths towards better treatments.< • Protein networks: With B. Bardoni, we identified several protein interactors of FMRP, notably the conserved CYFIP proteins, that interact with Rac1 GTPase and the WAVE complex, key regulators of cytoskeletal actin remodeling. • Metabolism of neuronal mRNAs (Dr. Hervé Moine): We showed that FMRP binds specifically mRNAs containing G(uanine)-quartet motif and also a triple stem-loop motif in SOD1 mRNA. We found that FMRP regulates its own mRNA splicing and the formation of mRNA stress granules. We recently showed that the G-quartet motifs present in the 3’ untranslated region of key neuronal mRNAs act as «zipcode» for their localization to dendrites (unpublished). • Signal transduction: Found increased level of cortical dopamine in our Rsk2 deficient mice that is caused by increased tyrosine hydroxylase (TH) activity. We provided evidence that loss of neurons due Bechara EG, Didiot MC, Melko M, Davidovic L, Bensaid M, Martin P, Castets M, Pognonec P, Khandjian EW, Moine H, Bardoni B. A novel function for fragile X mental retardation protein in translational activation. PLoS Biol. 7(1):e16. (2009). shaping the future Mechanisms of monogenic forms of mental retardation Gaining a thorough insight into the mechanisms underlying MR will contribute to better understand the processes of normal cognition, notably the molecular events and signaling pathways implicated in activity-dependent local changes that take place in neuronal processes. Our major goals are to : ● define and understand the molecular signals that are involved in controlling mRNA transport and local protein synthesis in neurons, and how they impact fragile X pathomechanisms. ● understand the mechanisms leading to increased apoptosis and to AMPA receptor dysfunction in Rsk2-KO mice and define the contribution of each of these deregulations to the cognitive dysfunction. ● define the functional interactions between PAK3, Rsk2 and FMRP. In collaboration with the teams of M. Koenig and J. Laporte, and with the DNA diagnostic lab (Strasbourg Hospital), we will develop high-throughput sequencing strategies for gene identification and molecular diagnosis in MR. < to impaired apoptosis and a defect in AMPA neurotransmission and plasticity might contribute to cognitive dysfunction in Rsk2 KO mice. • Gene discovery: Contributed to the identification of two major genes mutated in Bardet-Biedl syndrome that defined a novel chaperonin family (collaboration with the team of Pr. H. Dollfus), and to the definition of a new microdeletion syndrome (del16p11.2) associated to developmental delay and obesity (with the DNA diagnostic lab at Strasbourg Hospital, and a large international collaboration). n Didiot MC, Subramanian M, Flatter E, Mandel JL, Moine H. Cells lacking the fragile X mental retardation protein (FMRP) have normal RISC activity but exhibit altered stress granule assembly. Mol Biol Cell. 20:428-37. (2009). 1- The G(uanine) quartet structure is a binding site for the Fragile X Mental Retardation Protein on neuronal mRNAs (left and middle). Presence of a G-quartet-FMRP binding site in the 3’-untranslated region of a mRNA enables its dendritic localization in cortical neurons (green signal on right panel). 2- Dendritic spines of hippocampal neurons revealed by actin (red) and Neurabin2 (green) staining. DAPI staining of the nuclei (blue). Didiot MC, Tian Z, Schaeffer C, Subramanian M, Mandel JL, Moine H. The G-quartet containing FMRP binding site in FMR1 mRNA is a potent exonic splicing enhancer. Nucleic Acids Res. 36:4902-12. (2008) Marques Pereira P, Gruss M, Braun K, Foos N, Pannetier S, Hanauer A. Dopaminergic system dysregulation in the mrsk2 KO mouse, an animal model of the Coffin-Lowry syndrome. J Neurochem. 107:1325-34. (2008). Human Genetics Jean-Louis Mandel André Hanauer Understanding the X-linked mental retardations Jean-Louis Mandel earned an MD and MSc and taught genetics at the Strasbourg University before being nominated Professor at the Collège de France in 2003. His research in human genetics has brought major contributions, notably to our understanding of neuromuscular genetic diseases. In 1991, his team notably identified unstable mutations involving nucleotide triplet repeat expansions responsible for the Fragile X syndrome. From 2002 to 2009, he directed the IGBMC jointly with Dino Moras. Today, he leads the Genetic Diagnosis Laboratory of the Strasbourg Hospital and is in charge of a team with André Hanauer on X-linked mental retardation. André Hanauer obtained his PhD in 1989 and is Associate Professor at the University of Strasbourg. In 1996, he identified the gene responsible for Coffin-Lowry’s syndrome. The team he conducts jointly with Jean-Louis Mandel has made major contributions to the understanding of functional and genetic aspects of the Rsk2 gene that is responsible for Coffin-Lowry syndrome. n 2 87 highlights • The Aneuploid zoo: a comprehensive series of mouse model for Aneuploidies linked to human chromosome 21: Homologous and syntenic regions of the Human chromosome 21 (Hsa21) are found on murine chromosomes 16, 17 and 10. We used chromosomal engineering, a strategy we pioneered a few years ago, in order to develop a comprehensive series of new mouse models carrying segmental aneuploidies for all the regions that are homologous to Hsa21 in the mouse. 1 probing complexity Our goal is to identify dosage sensitive genes and we focus our interests on a few aneuploid syndromes: the Down syndrome (DS), a frequent aneuploidy affecting 1 newborn out of 700, identified as the consequence of Trisomy 21 and the rarer Monosomy 21 (M21), caused by the loss of one copy of chromosome 21 (Hsa21). Despite prenatal diagnosis, the incidence of DS is still high partly due to the increase in the maternal age and to the social disparity in the prenatal follow-up. DS is associated with mental retardation, heart defect and a large panel of dysmorphologies affecting the face and the skeleton Changes in metabolism and increased frequencies of some pathologies such as leukemia, autism, epilepsy and early onset Alzheimer disease are also observed. A few individuals with DS alterations carry a segmental duplication instead of a complete trisomy. The fifty cases described in the literature are very informative but still not enough to understand further the phenotype-genotype relationship. Thus we decided to use the mouse as a model system in order to identify dosage sensitive genes (or other genetic units) that could have a direct or indirect effect on the DS phenotype. < shaping the future Physiopathology of aneuploidy, gene dosage effect and Down syndrome We plan to focus on three objectives: (1) To address the phenotype–genotype relationship integrating human data; (2) To identify candidate genes and pathways involving dosage sensitive genes responsible for DS and M21 syndromes and (3) To validate new therapeutic approaches in preclinical models with the aim of facilitating the life of people with DS. Furthermore, this study will challenge the balance hypothesis underlying dosage sensitivity suggesting that the stoichiometry of the subunits participating in multiprotein complexes have to be maintained to perform their biological function. In addition the contribution of the noncoding sequences and of the chromosomal organization of the aneuploid phenotypes will be carefully evaluated. Besides this main project, we are working with human geneticists to study new aneuploid syndromes in the mouse to develop new strategies to evaluate the contribution of copy number variation to human disease and physiology. < • Tackling the genotype-phenotype relationship in DS models: With this aneuploid zoo for Hsa21 we are performing a comprehensive phenotypic analysis for behavior, cognition, morphology and physiology in parallel with gene expression profiling. This strategy led us to identify the consequence of Monosomy 21 on the inflammatory and lung functions and the contribution of some telomeric part of Hsa21 to the DS behavioral impairments. Lopes Pereira, P., Magnol, L., Sahún Abizanda, I., Brault, V., Duchon, A., Prandini, P., Gruart, A.,, Bizot, J-C, Chadefaux-Vekemans, B., Deutsch, S., Trovero, F., María Delgado-García, J., Antonarakis, S.E., Dierssen, M. and Herault Y. A new mouse model for the trisomy of the Abcg1-U2af1 region revals the complexity of the combinatorial genetic coded of Down syndrome Hum. Mol. Genet. 18:4756-4769. (2009). Yann Hérault Mice models to study gene dosage effects • Contribution of regions and candidate genes to aneuploid disorders: These studies identified several genetic loci contributing to DS phenotypes, acting either positively or negatively on specific functions. This hypothesis allows us to explain part of the phenotypic variability observed in DS patients. Our current analysis of the contribution of additional regions will help to understand further the physiopathology of the DS. n 1- Pyramidal neurons from the hippocampus. 2- Tackling the hippocampal changes in DS models involved in learning and memory. Dierssen, M., Herault, Y., Estivill, X. Aneuploidy: from a Physiological Mechanism of Variance to Down Syndrome. Physiol. Rev. 89:887-920. (2009). Duchon, A., Besson, V., Lopes Pereira, P., Magnol, L., and Hérault, Y. Inducing segmental aneuploid mosaicism in the mouse through Targeted Asymmetric Sister Chromatid Event of Recombination (TASCER). Genetics. 180:51-59. (2008). Yann Hérault graduated from the Ecole Normale Supérieure and obtained his PhD on the transcription regulation of clusterin at the ENS in Lyon in 1993. He then became interested in developmental biology and joined Duboule’s laboratory at the University of Geneva in order to study Hox genes regulation. In 1998, he created the TAMERE method, an in vivo chromosome engineering tool. While working on functional redundancy, he became interested in Down’s syndrome, a disease also due to gene dose effects. He created a resource of trisomic mice models for almost the entire Down’s syndrome homolog genes in mouse. From 2000 to 2009, he participated and led the Transgenesis and Archiving of Animal Models unit in Orléans. He joined the IGBMC in 2010 as the director of the Mouse Clinic Institute and leads a team aiming at mapping genotypes / phenotype relations in Down’s syndrome. n 2 89 highlights 1 probing complexity Opiates have been used for thousands of years for their pain-relieving and rewarding properties. Opiates produce their potent effects by activating opioid receptors in the brain, thereby highjacking a complex neuromodulatory system. Opioid receptors (mu, delta, kappa and nociceptine/orphanineFQ) are normally stimulated by endogenous peptides to control pain and stress responses, as well as emotional and addictive behaviors. Our goal is to tackle the role of opioid and related neuropeptide systems in brain physiology, as well as neurological and psychiatric disorders. We address molecular mechanisms underlying the development of chronic pain and associated affective disorders and investigate molecular adaptations that develop under repeated exposure to drugs of abuse, and contribute to craving and relapse. We explore potential roles of these receptors in other areas of psychiatric disorders, and identify neurons where the receptors operate to control behavioral responses. Finally, we use genome-wide approaches to discover novel molecular targets for drug abuse research. Our laboratory uses gene targeting approaches in mice (knockout, conditional knockout and knock-in), behavioral analysis, pharmacology, as well as receptor and cellular imaging to achieve these goals. < • We have demonstrated that a single receptor protein, the mu opioid receptor, mediates both therapeutic and adverse effects of clinically useful opiates. This receptor also mediates rewarding properties of other drugs of abuse, and natural rewards. • We have discovered an unexpected role of delta opioid receptors in anxiety and depression, which has important clinical implications. We have uncovered unanticipated and opposing contributions of mu and delta receptors in selfcontrol, opening novel avenues towards understanding behavioral inhibition. • We have identified novel genes, whose role in addictive behaviors is yet unknown. • We have created knock-in mice where delta opioid receptors are directly visible in vivo. This Olmstead C., Ouaggazzal A-M. and Kieffer B. L. Mu and delta opioid receptors oppositely regulate motor impulsivity in the signaled nose poke task Cited in the Editor’s choice section of Science PLoS ONE 4(2):984-985. (2009). shaping the future Opioid systems and brain function Our goals are to : ● futher develop site-specific gene knock-out in targeted neurons to identify receptor populations governing addictive behaviors and pain relief. ● continue to study delta and nociceptin orphanin peptide receptors (NOP) as potential therapeutic targets in inflammatory pain, mood disorders, and cognitive deficits. ● explore the role of novel genes in cognitive, emotional and addictive behaviors. ● continue to investigate the abstinent brain at behavioral, genetic and imaging levels. ● develop novel mutant mice with fluorescent receptors together with dynamic neural imaging approaches, to elucidate molecular bases of receptor adaptations and receptor-receptor interactions in vivo. < Brigitte Kieffer Specialist in opioid receptors unique tool has allowed us to describe receptor anatomy throughout the nervous system at cellular and subcellular levels, and to investigate real-time receptor trafficking in live neurons and to understand behavioral desensitization mechanisms. This is a ground-breaking approach in the field of G protein-coupled receptor biology and drug discovery. n Scherrer G, Imamachi N, Cao YQ, Contet C, Mennicken F, O’Donnell D, Kieffer BL and Basbaum AI. Dissociation of the opioid receptor mechanisms that control mechanical and thermal pain. Cell 137:1148-59. (2009). 1- Fluorescently-labelled opioid receptors visible in the brain hippocampus of genetically engineered mice. 2- Direct opioid receptor visualization in live neurons Pradhan A., Becker J., Scherrer G., Tryoen-Toth P., Filliol D., Matifas A., Massotte D., Gavériaux-Ruff, C and Kieffer B. L. In vivo delta opioid receptor internalization controls behavioral responses to agonists. PLoS ONE 4(5):e5425. (2009) Scherrer G., Tryoen-Tóth P., Filliol D., Matifas A., Laustriat D., Cao Y. Q., Basbaum A. I., Dierich D., Vonesh J.-L., GavériauxRuff C. and Kieffer B. L. Knock-in mice expressing fluorescent delta opioid receptors uncover G protein coupled receptor dynamics in vivo Proc. Natl. Acad. Sci. USA. 103:9691-6. (2006) As Assistant Professor at the University of Strasbourg, Brigitte Kieffer isolated, in 1992, the first gene encoding an opioid receptor paving the way towards understanding genetic bases of opioid-controlled behaviors. Using gene targeting in mice she demonstrated that a single receptor protein, the mu opioid receptor, mediates both potent analgesic and addictive effects of morphine. She also discovered anxiolytic and antidepressant activities of the delta opioid receptor. Her work has important implications in basic neurosciences and the biomedical community. From 1994 to 2006 she was a Professor at the Faculty of Pharmacy in Strasbourg and is currently a Research Director at Inserm. She has also been a visiting Professor at University of California, Los Angeles since 1998. She joined the IGBMC with her team in 2001. In 2004, she received the Loundsberry Award from joined French and US Academies of Science and became a EMBO Member in 2009. n 2 91 highlights • Identification of the genes defective in Friedreich ataxia and ataxia due to Coenzyme Q10 deficiency, both coding for novel mitochondrial proteins (frataxin and ADCK3) supporting assembly of components of the respiratory chain. Construction and characterization of the first murine and cellular models of Friedreich ataxia for preclinical drug screenings. 1 probing complexity Recessive progressive ataxias represent a heterogeneous group of neurological disorders characterized by the degeneration of the cerebellum and/or the spinocerebellar and posterior columns of the spinal cord. Albeit ataxia is the prominent symptom, recessive progressive ataxias often combine additional features such as peripheral neuropathy, pyramidal and/or extrapyramidal signs, nystagmus, oculomotor apraxia, ophthalmoplegia, optic atrophy, deafness, mental retardation, and epilepsy, as well as non-neurological features. Due to these associated signs and symptoms, differential diagnosis is often difficult with related disorders such as spastic paraplegias with cerebellar involvement, sensory ataxic neuropathies, congenital ataxias, metabolic ataxias, periodic ataxias, cerebellar hypoplasias, and the CACH and Joubert syndromes. We have opted to tackle the very high clinical and genetic heterogeneity of the recessive ataxias by linkage studies of large consanguineous families referred to us by collaborating neurologists worldwide. Today, at least 15 genes of recessive progressive ataxias have been identified, of which seven were identified by or in collaboration with our group. Mutations of these genes account for about 50 to 60% of the cases, indicating that many rare forms of recessive ataxias remain to be identified. < shaping the future Recessive ataxias The publication of the first and subsequent drafts of the human genome sequence has allowed the identification of an increasing number of genes and mutations causing mendelian disorders. It has also revealed part of the incredible complexity of these disorders, due to underestimated genetic and phenotypic heterogeneity. A future impact of the human genome project will be the availability of personalized human genome sequencing (2nd and 3rd generation high-throughput sequencing methods). In addition to raising great concerns about ethical issues, this possibility paves the way to molecular diagnosis for clinical practice of (almost) any mendelian disorder. Our aim is to develop applications of the next generation highthroughput sequencing for diagnostic purposes and further unravel the complexity of the disorders for which DNA samples of patients are referred to us.< • Identification of novel forms of inherited ataxias that associate cerebellar degeneration, sensorimotor peripheral neuropathy and oculomotor apraxia. Identification of the corresponding genes, coding for novel nuclear proteins, aprataxin and senataxin, involved in DNA repair and RNA maturation. Michel Koenig A crossed career between medicine and genetics for rare diseases diagnosis • Identification of the genes defective in Giant Axonal Neuropathy, Ataxia with isolated Vitamin E Deficiency, Marinesco-Sjögren Syndrome and Salih Ataxia, coding for cytosolic proteins involved in various functions of membranes and cytoskeleton. M. Assoum, M.A. Salih, N. Drouot, D. H’Mida-Ben Brahim, C. Lagier-Tourenne, A. AlDrees, S.A. Elmalik, T.S. Ahmed, M.Z. Seidahmed, M.M. Kabiraj, M. Koenig Rundataxin, a novel protein with RUN and diacylglycerol binding domains, is mutant in a new recessive ataxia. Brain. (In press) (2010). • We identified all disease genes by the pioneering use of homozygosity mapping and high density genetic marker analysis. Their identification had immediate and important applications for diagnosis of recessive ataxias in clinical practice. n M. Anheim, B. Monga, M. Fleury, P. Charles, C. Barbot, M. Salih, J.-P. Delaunoy, M. Fritsch, L. Arning, M. Synofzik, L. Schöls, J. Sequeiros, C. Goizet, C. Marelli, I. Le Ber, J. Koht, J. Gazulla, J. De Bleecker, M. Mukhtar, N. Drouot, L. Ali-Pacha, T. Benhassine, M. Chbicheb, A. M’Zahem, A. Hamri, B. Chabrol, J. Pouget, R. Murphy, M. Watanabe, P. Coutinho, M. Tazir, A. Durr, A. Brice, C. Tranchant and M. Koenig. Ataxia with oculomotor apraxia type 2: clinical, biological and genotype/phenotype correlation study of a cohort of 90 patients. Brain. 132: 2688-2698. (2009). C Lagier-Tourenne, M Tazir, LC López, CM Quinzii, M Assoum, N Drouot, C Busso, S Makri, L Ali-Pacha, T Benhassine, M Anheim, D Lynch, C Thibault, F Plewniak, L Bianchetti, C Tranchant, O Poch, S DiMauro, JL Mandel, MH Barros, M Hirano, M Koenig. ADCK3, an ancestral kinase, is mutated in a form of recessive ataxia associated with Coenzyme Q10 deficiency. Am J Hum Genet. 82:661-672. (2008). Human Genetics 1- Expression of gene identified in Cos cells (electronic microscopy) 2- Gene Identification, linkage, sequencing & light Scanner Michel Koenig trained in medicine and biology. In 1986, he obtained a PhD in genetics on the human X and Y chromosomes cartography. He went on to pursue his post-doc with Louis Kunkel’s team at the Harvard Medical School, USA until 1989. At that time, this team had just discovered the Duchenne muscular dystrophy gene and Michel Koenig contributed to its characterization. In 1990 he obtained his MD and integrated Jean-Louis Mandel’s team to work on recessive ataxias. The gene responsible for Friedreich’s ataxia was identified in 1996 and enabled the development of mouse models. As a result, many other recessive ataxia genes were identified. Since 1996, he works with the Strasbourg Hospital and his discoveries are now used by neurologists for rare genetic disorder diagnoses. Michel has led his own team at the IGBMC on recessive ataxias since 2002. n 2 93 highlights • We identified and characterized the myotubularins family, phosphoinositides phosphatases mutated in several neuromuscular diseases and implicated in endocytosis and autophagy. • We identified the first mutations in amphiphysin 2 (BIN1), previously described as a tumor suppressor, in autosomal centronuclear myopathy. Mutations either impact on its membrane remodelling property or disrupt its binding with dynamin 2, another protein mutated in such diseases. 1 probing complexity We study rare and severe neuromuscular disorders caused by mutations in proteins affecting organelles and membrane trafficking. Our principle focus is on three families of proteins that regulate or are regulated by membrane lipids: the phosphoinositide phosphatase myotubularins, the membrane remodelling amphiphysins and the membrane fissioning GTPase dynamins. Both myotubularins and dynamin 2 are mutated in centronuclear myopathies, severe congenital myopathies characterized by mislocalisation of nuclei and in Charcot-Marie-Tooth peripheral neuropathies, suggesting a common mechanism. While focusing on these genetic diseases, our approaches are multidisciplinary and encompass the identification of the implicated genes by high-throughput sequencing, the study of the molecular and cellular functions of these proteins in cells and in C. elegans, the validation of mammalian disease models, and the use of viral vectors (AAVs) for pathophysiology studies and preclinical therapeutic trials. In parallel, we study the function of these proteins in skeletal muscle under normal and pathological conditions through the development of novel imaging methods (correlative microscopy and in vivo imaging) in close contact with the IGBMC platforms. < shaping the future Mechanisms of neuromuscular diseases In order to better understand the molecular basis of severe neuromuscular diseases and to propose possible therapeutic targets and approaches, we aim at: ● identifying additional genes implicated in neuromuscular diseases through the development of high-throughput sequencing strategies. Such approaches are at the heart of the future personal medicine based on the knowledge of our own genome. ● characterizing the link between membrane remodeling and organelle positioning and the implication of the cytoskeleton. This work should point to novel pathways implicated in the late maturation and structural maintenance of skeletal muscle fibers. ● using viral transfer in mouse to generate and rescue muscle weakness. ● validating confocal macroscopy for non-invasive monitoring of muscle in mammals. < • Together with the IGBMC imaging platform, we validated integrated methods for high pressure freezing and correlative light-electron microscopy, linking dynamic live cell imaging to 3D ultrastructure. • We developed and transferred most of the existing molecular diagnostic strategies for the different forms of centronuclear myopathies to the Genetic Diagnosis lab at Strasbourg Hospital. n Al-Qusairi L, Weiss N, Toussaint A, Berbey C, Messaddeq N, Kretz C, Sanoudou D, Beggs AH, Allard B, Mandel JL, Laporte J, Jacquemond V, Buj-Bello A. Defective excitation-contraction coupling in muscle fibres lacking myotubularin phosphoinositide phosphatase. Proc Natl Acad Sci U S A. 106 (44):18763-18768. (2009). Nicot AS, Toussaint A, Tosch V, Kretz C, Wallgren-Pettersson C, Iwarsson I, Kingston H, Garnier JM, Biancalana V, Oldfors A, Mandel JL, Laporte J. Mutations in amphiphysin 2 (BIN1) disrupt interaction with dynamin 2 and cause autosomal recessive centronuclear myopathy. Nature Genetics. 39:1134-1139. (2007). Jocelyn Laporte Interdisciplinarity for the study on neuromuscular diseases • Using a mouse model for X-linked centronuclear myopathy, our group showed that the muscle weakness is linked to T-tubules and calcium homeostasis defects. Spiegelhalter C, Tosch V, Hentsch D, Koch M, Kessler M, Schwab Y, Laporte J. From dynamic live cell imaging to 3D ultrastructure: integrated methods for high pressure freezing and correlative light-electron microscopy. PLoS ONE 5(2):e94. (2010). Human Genetics 2 1- Co-localisation between the early endosome antigen 1 protein (green) and the Phosphoinositide 3-monophosphate detected with a biosensor (red). DNA in blue. 2- Homozygous mutation of the BIN1 gene in a consanguineous family. Above : chromatopherogram, below : pedigree of the affected family. After completing studies in technical formation at the Strasbourg School of Biotechnology (ESBS), Jocelyn Laporte did his PhD with Jean-louis Mandel and identified in 1996 the myotubularin gene (MTM1) involved in centronuclear myopathies. He became a team leader at the IGBMC in 2007. The same year, his team brought to light the involvement of amphiphysin 2 in myopathies. His team’s research is based on a interdisciplinary approach from human genetics to therapeutic approaches through protein functional analysis and animal models development. Currently his team participates in the validation of new sequencing and imaging technologies. The team’s research is both fundamental and applied and they work closely with the laboratory for genetic disease diagnosis at the Strasbourg Hospital. n 95 highlights • Through conditional knockout approaches, we have developed the first mouse models for FRDA, which reproduce important progressive features of the human disease. With these models, we have shown that the primary event in the disease is the deficiency in mitochondrial Fe-S proteins followed by a secondary mitochondrial iron accumulation. More recently, we have provided the first direct evidence that frataxin is necessary for the proper assembly of extra-mitochondrial Fe-S proteins in mammalian tissues, both cytosolic and nuclear. 1 probing complexity The laboratory is dedicated to unraveling the causes and mechanisms of progressive recessive ataxias, which are neurodegenerative disorders that affect the cerebellum and/or the spinal cord. Recessive ataxias represent a heterogeneous set of severely disabling neurological disorders estimated to affect 1/20,000 individuals in Europe. Friedreich ataxia (FRDA), the most common recessive ataxia, is characterized by progressive gait and limb ataxia associated with hypertrophic cardiomyopathy and an increased incidence in diabetes. The disease is due to severely reduced levels of frataxin, a highly conserved mitochondrial protein thought to be involved in multiple iron-dependent mitochondrial pathways. In particular, frataxin is proposed to be involved in iron-sulfur (Fe-S) cluster biosynthesis. Fe-S clusters are critical redox active prosthetic groups that are present in proteins involved in numerous essential cellular processes ranging from nuclear genome stability, protein translation to mitochondrial metabolism. Mitochondria contain a complex system for assembly of Fe/S metal centers and their insertion into proteins. In addition to unravelling the pathophysiology of the disease, we aim to understand the molecular mechanism of mammalian Fe-S cluster biogenesis, with a particular interest in the role of frataxin. Our strategy combines human genetics, biochemistry, cell biology and animal models. < shaping the future Biology and physiology of recessive ataxia The objectives of the laboratory are to further elucidate the molecular mechanism underlying neuronal dysfunction in FRDA and XLSA/A, two recessive ataxia linked to Fe-S cluster deficit and ARCA2, a newly discovered recessive ataxia linked to Coenzyme Q10 deficit. We will develop animal models for ARCA2 and XLSA/A, as well as neuronal cellular models for the three diseases. In particular, the recent technical advances in the generation of induced pluripotent stem cells (iPS) from somatic cells provide a powerful tool to create disease-specific cellular models. Furthermore, to uncover common pathways among recessive ataxia leading to neuronal dysfunction, comparative transcriptomic and proteomic analysis will be performed. In parallel, we will continue functional studies to further unravel the molecular steps of Fe-S biogenesis and coenzyme Q10 biogenesis, two fundamental pathways in the mitochondria. < • We have developed different cell models based either on antisense strategy reproducing the quantitative defect found in patients or on expression of frataxin carrying disease-causing mutation. These models exhibit proliferation defects associated with specific biochemical features of the disease and are therefore good models for drug screening. • We have shown that idebenone, a CoQ10 analog in clinical use, has a significant effect on the cardiac function and the life span of the murine cardiac model. n Calmels N., Schmucker S., Wattenhofer-Donzé M., Martelli A., Vaucamps N., Reutenauer L., Messaddeq M., Bouton C., Koenig M., Puccio H. The first cellular models based on frataxin missense mutations that reproduce spontaneously the defects associated with Friedreich ataxia. PlosOne. 4(7):e6379. (2009) 1- Mitochondrial dysregulation and iron accumulation in cardiac tissue from mouse model of Friedreich ataxia. 2- Human neurons (red: bIII-tubulin) and astrocytes (green: GFAP) differentiated from induced pluripotent stem cells (iPS) derived from adult fibroblasts. Schmucker S, Argentini M., Carelle-Calmels N., Martelli A, Puccio H. The in vivo mitochondrial two-step maturation of human frataxin. Human Molecular Genetics. 17(22):3521-31. (2008). Martelli A., Wattenhofer-Donzé M., Schmucker S., Bouvet S., Reutenauer L., and Puccio H. Frataxin is essential for extramitochondrial Fe-S cluster proteins in mammalian tissues. Human Molecular Genetics. 16(22):2651-8. (2007). Hélène Puccio Understanding the molecular mechanisms of recessive ataxia • By combining in vitro and in vivo approaches, we have recently demonstrated that frataxin interacts with the preassembled core complex involved in the early steps of de novo Fe-S biogenesis, providing a new understanding of the sequential steps of Fe-S biosynthesis in vivo. Schmucker S and Puccio H. Understanding the molecular mechanisms of Friedreich’s ataxia to develop therapeutic approaches. Human Molecular Genetics. 19(R1):R103-10. (2010). Human Genetics After obtaining her PhD in Genetics from Harvard University, USA in 1998, Hélène Puccio joined the group of Michel Koenig at the IGBMC to work on the molecular pathogenesis of Friedreich ataxia (FRDA). In 2001, she was appointed Inserm research associate and has since been the principal investigator on the FRDA project. Her research focuses on understanding the physiopathology of FRDA through the development of animal and cellular models and on the development of therapeutic approaches. She was awarded the Pediatric Pathology Prize in 2005 and also received the label "Equipe FRM". In 2007, she obtained the prestigious Young Investigator ERC award, and in 2008 she received the Dr. Jean Toy Prize from the Academy of Sciences and was promoted Inserm Research Director. n 2 97 highlights • Polyglutamine expansion and short polyglutamine tract have similar structural properties in vitro. This contradicts the hypothesis that polyglutamine can adopt a toxic structure only beyond a specific length and rather suggests that toxicity increases with polyglutamine length and manifests when cellular mechanisms against toxicity are overwhelmed. 1 probing complexity Polyglutamine diseases are ten adult-onset genetic disorders that result in the degeneration of selected brain areas. They are caused by an expansion of unstable CAG trinucleotide repeats coding for toxic polyglutamine (polyQ) expansions in ubiquitously expressed disease proteins. Understanding the molecular and cellular mechanisms underlying polyQ toxicity and neurodegeneration is essential to develop effective treatments against these incurable diseases. To this end, our strategy combines biochemistry, biophysics, cell biology and analysis of mouse models of two polyQ disorders: Huntington’s disease (HD) and Spinocerebellar ataxia type 7 (SCA7). Our plans are : 1) to study the interaction and aggregation properties of proteins harboring polyQ expansion to understand why they become neurotoxic ; 2) to study signaling pathways and gene expression to understand how affected neurons respond to toxic stress and how they degenerate; 3) to investigate the mechanism of instability of CAG repeats in HD, which generates larger expansions associated with increased protein toxicity and is particularly important in the striatum, the primary target in HD. Our ultimate goal is to identify effective treatments for polyQ disorders. < • Our SCA7 transgenic mouse recapitulates the retinal degeneration affecting the patients. Expression profile analysis of SCA7 mouse retina reveals that transcriptional alterations compromise the photoreceptor differentiation status and function. shaping the future Pathogenic mechanisms of polyglutamine expansion diseases We aim to understand the relationship between the polyQ structure, aggregration and toxicity and are using aggregation modulators as tools to delineate the aggregation mechanisms and combine theses with biological, structural and computational approaches. In the SCA7 mouse PolyQ toxicity induces not only photoreceptor death involving apoptotic and non-apoptotic mechanisms but also proliferation, which may partially compensate for photoreceptor loss. We are studying the molecular mechanisms underlying these exceptional degenerative and potentially regenerative processes in this model. To further understand the mechanisms underlying CAG instability in HD, we combine in vitro and in vivo approaches and investigate the role of Parp1, which coordinates BER (Base Excision Repair) by facilitating the remodeling of chromatin and the recruitement of BER factors to DNA lesions. < Yvon Trottier Understanding the Huntington disease • Polyglutamine aggregation triggers a cellular stress response involving the activation of AP1 transcriptional pathway in the SCA7 mouse retina. AP1 activation correlates with repression of genes involved in photoreceptor function. Inhibiting AP1 in the SCA7 mouse delays the retinal phenotype. • Stoichiometry of the DNA repair proteins DNA Polymerase ß and Flap endonuclease 1, which are involved in Base Excision Repair (BER), contributes to the tissue selectivity of CAG instability in HD mice. n Yefimova MG, Messaddeq N, Karam A, Jacquard C, Weber C, Jonet L, Wolfrum U, Jeanny JC, Trottier Y. Polyglutamine toxicity induces rod photoreceptor division, morphological transformation and death in Spinocerebellar ataxia 7 mouse retina. Neurobiol Dis. (in press) (2010). 1- SCA7 retinal section showing aggregates (green) of polyglutamineexpanded ataxin-7 in photoreceptor nuclei (red, left). 2- Electron micrograph showing structured amyloid-like fibers formed by polyglutamines in vitro. Goula AV, Berquist BR, Wilson DM 3rd, Wheeler VC, Trottier Y, Merienne K. Stoichiometry of base excision repair proteins correlates with increased somatic CAG instability in striatum over cerebellum In Huntington’s disease transgenic mice. PLoS Genet. 5(12):e1000749. (2009). Klein F, Pastore A, Masino L, Zeder-Lutz G, Nierengarten H, Oulad-Abdelghani M, Altschuh D, Mandel JL, Trottier Y. Pathogenic and non-pathogenic polyglutamine tracts have similar structural properties: towards a length dependent toxicity gradient. J. Mol. Biol. 371:235-244. (2007). Human Genetics Originally from Canada, Yvon Trottier obtained his PhD in 1992 at Laval University in Québec. In 1992 he joined the laboratory of Jean-Louis Mandel, who had just uncovered the disease-causing triplet repeat expansions, to work as a postdoc on Huntington’s Disease. In 1995, he characterized an antibody able to detect polyglutamine expansions and used it as a tool to identify two more polyglutamine disorders. In 2002, he studied the proteolysis mechanism of the Huntington’s disease protein. His recent work on Spinocerebellar ataxia type 7 enabled him to show that in response to polyglutamine toxicity, neurons undergo different cell fates including dedifferentiation, proliferation and death. Since 2007, he has led his own team at IGBMC on pathogenic mechanisms of polyglutamine expansion diseases. n 2 99 MOUSE CLINICAL INSTITUTE ¨ The mouse model has become the premier model to better understand the fundamental mechanisms responsible for complex biological phenomena.¨ Y. Hérault Institut CLINIQUE DE LA SOURIS The Institut Clinique de la Souris (ICS) is a research infrastructure of excellence for translational research and functional genomics. Founded in 2002 by Pierre Chambon and operated by Inserm, CNRS, and the Univeristy of Strasbourg, it provides a comprehensive set of specialized services to academic and industrial users and is a major player in the European post-genomic era programs. The close interaction with the IGBMC strongly contributes to the development and design of new techniques and functional assays. The ICS combines the capacity of generating mutant mice on a large scale with a high-throughput and comprehensive phenotypic analysis of the animals. The Institute’s phenotyping platforms are adapted for the study of genetically engineered mouse models and genetic reference populations but can also be used for preclinical studies including the validation of therapeutic targets as well as pharmacological and toxicological studies in the mouse. The three highly interactive departments Genetic Engineering and Model Validation, Phenotyping and Mouse Facility generate approximately 200 genetically modified mice per year. Phenotypic analysis cover hematology, immunology, inflammation, blood chemistry, metabolism, cardiovascular, respiratory, sensory organs, cognition, learning, beha- vior, reproduction, morphology etc.. The ICS plays a central role in large-scale infrastructures, both on a national level with CELPHEDIA and on a European level through the European Infrastructure Programme INFRAFRONTIER. Likewise, the ICS is a major player in various European funded programs oriented towards mouse genetics such as EUComm, EUmodic, EMMA. In joining the InternaIBiSA tional Mouse Phenotyping ConsorISO 9001/2008 in progress tium, the ICS contributes to de6 years of partnership with cipher mammalian gene functions pharmaceutical companies in order to better understand huMore than 250 mutagenesis man physiology and disease. or phenotyping projects The services of the ICS will ultiper year mately help the scientific commuPartner in 14 EU funded nity to use the mouse to develop a projects since 2002 complete functional annotation of 110 Staff members the human genome and to employ this to better understand human diseases and their underlying physiological and pathological basis. < The ICS is a national infrastructure closely linked to the IGBMC. Its mission is to : ● provide a service platform combining a large scale capacity for generating mouse mutants followed by subsequent high-throughput and comprehensive phenotypic analysis of the animals ● facilitate access to engineering, analysis and distribution of mouse models for the scientific community ● initiate and advance relevant in-house research and development programs to support the activities of ICS in the domains of mutagenesis, transgenesis, phenotyping, bioinformatics, and data analysis. ● promote training both internally and for users in order to guarantee the most efficient procedure in compliance with ethics and animal welfare. ● be a reference centre for mouse functional genomics and preclinical research 101 bioinformatiCS ¨ Previously, we worked mainly on genome sequences, however today the field of investigation has widened. There is not only one, but many Bioinformatics.¨ F. Plewniak platforms The IGBMC Bioinformatics technical platform platform provides bioinformatics resources including up-to-date databases, bioinformatics software, efficient computer systems, large storage capacity and expertise capable of addressing biological problems from basic sequence analysis to integrative and high-throughput computational biology. In sympathy with the IGBMC’s research interests, we are mainly involved in comparative genomic and bioinformatic studies of transcription related to research on human pathologies, such as cancer or genetic diseases. This includes promoter analysis, for example, phylogenetic foot-printing, in relation to transcriptomics data analysis, as well as studies of evolutionary events or mutation effects in human and animal model organisms. The platform is located within the IGBMC premises in close proximity to the biologists. The platform benefits from a particularly favo- rable environment to develop its expertise both in biology and computer science. The dual competence of its staff members permits the offering of a high standard of expertise in bioanalysis, as well as database or software development services. For all projects we guarantee that the underlying ISO 9001 scientific problem is properly understood and addressed using IBiSA appropriate solutions. 35 projects/year The platform is a member of the Réseau National des plate-formes 5 Staff members Bio-Informatiques (ReNaBi) and was recognized as an operational platform by IBiSA (Infrastructures Biologie Santé et Agronomie) in 2009. The Bioinformatics platform Quality Management System according to ISO 9001 norm was certified in June 2007. < Access to bioinformatics resources The bioinformatics resources provided by the platform include: • Over 30 public databases (Genbank, Uniprot, Protein Data Bank,Unigene,...) • All prokaryotic complete genomes available from the NCBI • 41 selected eukaryotic complete genomes • Locally developed and third party software Expertise in bioinformatics and bio-analysis This activity includes both short-term consultancy projects and the participation of the platform as a partner to longterm funded projects. Research and development Apart from the above services, an important mission of the platform is to develop innovative solutions for bioinformatics and bio-analysis questions. 103 HIGH-THROUGHPUT cell-based SCREENING "High throughput cell-based screening technologies accelerate the phase of identification of genes responsible for a particular phenotype or response, by delivering an immediate functional characterization" L. Brino The IGBMC High-Throughput Cell-based Screening platform, created in 2006, is one of the major high-throughput RNAi screening facilities in France. We participate in the global project realization beginning with: cellular process optimization, assay miniaturization, image analysis and development even before the screening campaign is started. Primary and secondary phenotype screening and validations are performed by the facility. We are equipped with a BSL2+ biologicalcontainment facility for mammalian cell culture, high-throughput cell transfection and post-processing cell manipulation. Currently, assay read-outs are performed either on the fully automated GE Life Sciences INCELL1000 analyzer or the Berthold High throughput MITHRAS LB940. Together with the IGBMC Bioinformatics platform, we have developed a robust biostatistical tool (RReportGenerator software) for the analysis of large cellular multiparameter datasets as well as downstream data enrichment solutions for comprehensive phe- notype analyses. So far, we have performed more than ten RNAi-based screening projects and we are currently investigating the chromatin function in DNA damage repair processes. Our siRNA libraries target diverse human and mouse gene subsets such as the kinome/phosphatome or the druggable genome IBiSA in process and we have recently acquired ISO 9001 a Qiagen human genome-wide in process siRNA library. The platform is 65,000 analyses developing a Quality Managein 2009 ment System (ISO9001 norm) and has been identified by IBiSA 5 Staff members for the coming round of qualification and support. The platform is largely open to French and foreign academic laboratories. < • Expertise in mammalian cell transfection and RNAi screening design. • Participates in the preparation of applications for research projects that aim to conduct phenotypic screening. • Develops innovative high-throughput cell-based applications. • Participates in the development of high-content analysis solutions in biostatistics and bioinformatics. • Strengthens and accelerates the approach “From Genes to Drugs”. imaging and microscopy ¨ Over 20 years, the imaging service has acquired a unique expertise. Inventive and creative, it develops prototypes that can be industrialized. ¨ J-L Vonesch The Imaging Center was created in 1992 to deal with the increasing demands and the complexity of biological imaging at the IGBMC. Recognized as an IBiSA platform, this facility is not only devoted to the IGBMC but reaches out to both the local and French scientific community. The Imaging staff, organized as two groups for light and electron microscopy, has complementary expertise and experience that ranges from biology and physics to computing. This experience and expertise allows the platform to offer a wide variety of equipments and approaches both in terms of service, teaching and development. The light microscopy group (7 engineers) offers an open access to a range of imaging systems, such as epifluorescent and confocal microscopes and macroscopes. Image analysis stations are also offered to the users of the platform. The specialists of the group assist the researchers in more complex experiments where specific expertise is needed, such as Ftechniques (FRAP, FRET/FLIM). More than 80% of the experiments in the facility are performed on living samples with specific setups tailored for functional imaging, time lapse microscopy, and fast multiphoton and confocal imaging. The electron microscopy group (4 engineers and a technician) has extensive experience in studying the ultrastructure of various cell types and tissues in normal and pathological conditions. With an expertise in sample preparation ranging from conventional techniques to the use of cryomethods, such as high pressure IBiSA freezing and freeze-substitution, 100 projects per year the facility can process almost any 40 imaging systems type of biological sample, from • 10 confocal cellular extracts to tissue biopsies. microscopes Besides the ultrastructural analy• 2 TEM, • 1 SEM sis, the group also performs protein localization at high resolution 25-30 publications by immuno EM labeling. per year The two groups also offer com13 Staff members bined and integrated approaches such as correlated light and electron microscopy.< The Imaging Center is an open facility that offers integrative imaging approaches to trained and untrained users: • Development of new tools and protocols in collaboration with research teams, to address challenging biological questions, such as micromanipulation and live imaging, photo-ablation, correlative light and electron microscopy. • Further development of the ultra-wide-field photonic imaging and confocal macroscope designed at the Center, to visualize and quantify biological processes at the centimeter scale. • Industrial partnership (Leica Microsystems, COMAT). • Continuum of imaging techniques between light and electron microscopy (confocal microscopy and macroscopy, infra and far-red two photon microscopy, fast confocal microscopy, ultrastructural analysis). • Image processing and analysis • Teaching programs and training workshops at the University with national and international organizations. 105 MICROARRAYs & DEEP SEQUENCING " Our techniques allow the study of genetic and molecular differences between the normal and pathological states." C.Thibault The IGBMC Microarray and Deep Sequencing platform, created in 2000, is one of the major microarray facilities in France. We are equipped with Affymetrix and Agilent microarray technologies and we run 1,500 to 2,000 arrays annually for a wide variety of transcriptomic and genomic applications. In October 2008, the platform invested in high-throughput sequencing technology, and implemented an Illumina GAII sequencing system. Since its installation, we have completed more than 50 projects and generated over 3 terabases of sequences. Importantly, our platform also provides support to researchers for data analysis. Indeed, we have the expertise (statistic and bioinformatic) and the infrastructure (server and dedicated software) to conduct microarray and sequencing data analysis. Our platform is open to French and foreign academic laboratories, and data obtained have contributed to numerous publications, many of them in the best international journals (82 publications as of February 2010). The platform has been involved in several large national programs, such as the Tumor Identity Card (CIT) program from the Ligue Nationale Contre le Cancer, and the RESOGEN program from the French Center for Genomic ISO 9001 Research. It has also been quaIBiSA lified as ¨ National platform ¨ by the ¨ Réseau Inter-Organism 150 Projects ¨ (RIO) and also by the IBISA per year since 2003. Lastly, because the 11 Staff members platform has heavily invested in quality, it became one of the first French technology platforms to obtain the ISO9001 certificate in 2007, which was renewed in 2010. < Our mission is to provide researchers with state of the art high throughput technologies for analysing gene expression and regulation. We provide array based applications such as Affymetrix microarrrays for gene expression profiling, miRNA expression, SNP genotyping, and CNV analysis. We also provide Illumina high throughput sequencing applications such as ChIP-seq, RNA-seq and whole genome or targeted re-sequencing. Full services involve all steps from quality checks of starting material, labelling and hybridisations for array applications, library preparations and sequencing, and data analysis. Support for in-depth bioinformatics and statistical data analysis for array and sequencing approaches is also available. STRUCTURAL BIOLOGY & GENOMICS ¨ This platform is the technological backbone for the production and characterization of functional complexes and will be an essential player in the future challenges in structural cell biology.¨ D. Busso The focus of the Structural Biology and Genomics platform (SBGP) is to provide technological resources to investigate complex biological systems, that range from single proteins to assemblies of proteins and nucleoprotein complexes. Ultimately this will contribute to an integrated view of cellulars systems. The objective of the platform is to accelerate the pace of discoveries by providing integrated high-throughput approaches to investigate eukaryotic macromolecular complex systems related to human health by using a continuum of state-of-the-art technologies. These technologies range from gene cloning to structure determination and ultimately to cellular resolution. The SBGP was registered by the RIO coordination in 2003 and was confirmed in 2006 and 2008 by IBiSA. This national label implies that the platform is open to the scientific community and is implicated in the important mission of training both scientists and technical staff within collaborative projects. Moreover, the tight synergies between the SBGP and the IGBMC facilities (Bioinformatics, Mass Spectroscopy, Cell Imaging) create an attractive scientific center for academic and industrial researchers. Thus, providing integrated structural and functional studies on systems that are important to IBiSA the understanding of biological pathways and human diseases. HTP procedures A major commitment of the Quality statements SBGP is to implement cutting edge technologies and to deveEuropean networks lop methodologies for protein 8 Staff members production and characterization that contribute to the technological backbone of the Strasbourg node in the European Integrative Structural Biology Infrastructure Program (INSTRUCT). < The SBGP offers high-level support to its users, ranging from gene cloning to structure determination. It notably makes use of the following methods and cutting edge technologies: • Automated cloning (Gateway, SLIC, restriction, …) and mini-expression screening protocols using Tecan liquid handling systems. • Protein production in various expression systems (bacteria, insect cells, mammalian cells). • Parallel purification of proteins, nucleic acids and their complexes using Akta Xpress and Akta Purifier FPLC chromatographic systems. • Macromolecule characterization using different biophysical methodologies (DLS, AUC, ITC). • Automated crystallization of macromolecules using a Cartesian nano-volume liquid handling robot. • Structure elucidation on crystals (X-rays with Micromax rotating anode from Rigaku) or in solution (EM, SAXS; NMR). So far, the platforms contributions have been published in ten articles. 107 Core facilities Core facilities • Mass Spectrometry Facility • Core service for Flow Cytometry & Fluorescence-Activated Cell Sorting Claudine Ebel • Animal house Head Animal House: Elisabeth Metzger Responsible Sanitary: Armelle Van Es The Animal House, which includes the ICS, serves around 25 research groups at the IGBMC and accommodates approximately 105,000 mice. Other than its animal maintenance roles and the technical assistance granted to researchers, the animal house has specific activities such as the importation and exportation of animals, the rederivation and the cryopreservation of mouse lines. • Cell Culture Facility Betty Heller The facility maintains more than 2,000 cell lines which include normal and tumoral cell lines from several origins. It produces 5,000 T-75 cm2 cell flasks, freezes 6,000 cryotubes, and prepares 5,000L of media per year. The facility also manages a L3 laboratory for virus experiments. This service allows investigators to perform multi-parameter flow cytometry analysis and sorting of cells and particles in liquid suspension based on size, granulosity and color of fluorescence. Two state-of-the-art analyzers (FACS, Calibur for 4 color analysis, LSR II for 15-color analysis) and two high-speed sorters (FACSVantage SE option DIVA with 8 color capacity, FACSAria II with 15 color capacity) are available to IGBMC scientists and the Strasbourg scientific community. The core service also provides protocols and procedures for numerous applications. • Histopathology & Embryology Service Stéphanie Muller Mouse models are readily accessible to post-mortem analyses at any time during the course of a disease, including the stages preceding its clinical onset. Thus, histological analysis of mutant and/or treated mice provide a powerful means to draw on pathophysiological scenarios. •Transgenic Facility Adeline Page Andrée Dierich The mass spectrometry facility provides the IGBMC community with standard and dedicated proteomic analyses for the characterization of proteins, including recent technological developments. Among others, this includes protein identification, protein modification detection and measurement of the molecular masses of intact protein and non-covalent complexes. The service is equipped with two ionic trap mass spectrometers (LTQ XL and LTQ Velos), an ESI-TOF mass spectrometer for analysis of molecular masses (MicrOTOF Focus) and a MALDI-TOF (Reflex IV) apparatus. The Transgenic Facility generates mouse models for in vivo gene function analysis by: • Classical transgenesis (micro-injection of transgenes) • Homologous recombination in mouse embryonic stem cells. • Tetraploid aggregation chimeras • Production of monoclonal & polyclonal antibodies Mustapha Oulad-Abdelghani (monoclonal) Gilles Duval (polyclonal) Our service produces up to 40 monoclonal and 200 polyclonal antibodies per year for in-house and external teams. The service can use recombinant proteins or polypeptide coupled to ovalbumine for immunization of the mouse, rabbit or rat. It also provides advice to design recombinant or synthetic antigens suitable to specific application. • The Baculovirus Service Isabelle Kolb-Cheynel and Nathalie Troffer-Charlier The Baculovirus Expression Facility provides investigators with equipment, expertise and custom services for protein production in insect cells. Services include: generation, amplification and maintenance of high-titer baculovirus stocks, analytical scale productions for protein-protein interaction studies, optimization of protein expression and preparative scale productions.< • Peptide synthesis Pascal Eberling Peptide synthesis is performed on a 433A peptide synthesizer (ABI), using Fmoc chemistry. Crude peptides are purified by inverse phase HPLC. End products are checked and characterized by HPLC and mass spectrometry. 109 Facts & figures Facts & figures Statistics until June 30th A major part of IGBMC staff (62%) is located within research teams: these are statutary scientists, post-docs, PhD students and technical staff. Engineers and technicians with high levels of expertise are employed on our platforms and our core facilities al- A major part of IGBMC’s funding (87%) comes through research grants. Our researchers apply to charities (AFM, FRM...), national public funding agencies (ANR, INCA...) and/or European or International Programmes (FP7, NIH, HFSP...). The other 13% comes from institutional funding (CNRS-Inserm-Université de Strasbourg). Research funding 12% 3% 5% Staff allocations (except ICS) 13% 15% Institutional funding 3% 12% Public funding 18% 46% Charities Research teams European Union funding IGBMC common International support Platforms Industrial contracts & licences Core Facilities Investments & services ICS 5% 6% Nationalities of staff scientists Distribution of statutary and non-statutary staff Distribution of staff scientists 7% 32% 17% 33% 67% lowing the pooling of resources: they represent 26% of our staff and work for both internal and external research teams and/or industrial laboratories. The remaining 12% cover administrative (Purchasing, Human Resources, Finance...) , IT Service and Building Maintenance. 13% 12% 33% 55% 62% The IGBMC is an international center with 48 different nationalities represented among its personnel. All activities in the Institute are carried out in English. 768 employees (including ICS) 43 groups 4 research programmes 5 platforms + ICS 31% Non-Statutary staff Group leaders PhD students Statutary staff Staff scientists Master students France More than 200 publications/year EU (except France) 57 million Euros budget Others 14,000 m² of laboratories Post-docs 111 staff scientists Development & Stem Cells Coordinator : LABOUESSE Michel Assistant: BRONNER Sylvianne DOLLE Pascal Role of retinoic acid in mouse development Scientists BLOCH-ZUPAN Agnès DOLLE Pascal KREZEL Wojciech RHINN Muriel Postdoctoral fellows PASCHAKI Marie PhD Students ETTER Guillaume KRZYZOSIAK Agnieszka LAUGEL Virginie RATAJ Monika Engineers/Technicians FRAULOB Valérie SCHUHBAUR Brigitte Master Student SCHUH Mélanie HEITZLER Pascal Genetic and molecular analysis of early neurogenesis in Drosophila melanogaster Scientists HEITZLER Pascal Postdoctoral fellows BIRYUKOVA Inna Engineers/Technicians ACKERMANN Claudine Master Students CHOONG Kee-Fong SALLOUM Anastasia JARRIAULT Sophie In vivo analysis of cellular plasticity in C. elegans Scientists JARRIAULT Sophie Postdoctoral fellows AHIER Arnaud HAJDUSKOVA Martina ZURYN Steven PhD Students DANIELE Thomas Engineers/Technicians GEORGES-LABOUESSE Elisabeth FISCHER Nadine Integrin function and signaling in Master Students tissue morphogenesis, integrity Marie-Charlotte Morin and homeostasis Scientists LABOUESSE Michel DE ARCANGELIS Adèle Forces and signals in tissue GEORGES-LABOUESSE Elisabeth morphogenesis PhD Students Scientists TOSSE Lindzy GALLY Christelle Engineers/Technicians LABOUESSE Michel PFISTER Véronique QUINTIN Sophie SIEBERT Stéphanie Postdoctoral fellows KOLOTUEVA Irina GIANGRANDE Angela OSMANI Naël Cellular and molecular ZHANG Huimin mechanisms of nervous system PhD Students differentiation APAYDIN Ahmet Scientists Engineers/Technicians GIANGRANDE Angela RODRIGUEZ David Postdoctoral fellows Master Student KOMONYI Orban PASTI Gabriella LANEVE Pietro POPKOVA Anna MARK Manuel/GHYSELINCK PhD Students Norbert BERZSENYI Sara Retinoic acid signalling ERKOSAR Berra pathways driving stem sperFLICI Hakima matogonia ontogenesis and KARATAS Omer Faruk differentiation KUMAR Arun Scientists Engineers/Technicians GHYSELINCK Norbert ARBOGAST Nadine MARK Manuel DIEBOLD Céline Postdoctoral fellows MURA Carole GELY PERNOT Aurore PAPALINI Giulia JACOBS Hugues PhD Students GRADWOHL Gérard RAVERDEAU Mathilde Control of endocrine cells Engineers/Technicians differentiation in the pancreas DENNEFELD Christine and intestine FERET Betty Scientists KLOPFENSTEIN Muriel GRADWOHL Gérard Master Student MELLITZER Georg BOUROUINA Imane Rym Postdoctoral fellows BEUCHER Anthony MERLE Carole PhD Students NIVLET Laure PICCAND Julie Engineers/Technicians MEUNIER Aline POULET Martine Master Student STRASSER Perinne POURQUIE Olivier Development of muscle and vertebrae Scientists KNOCKAERT Marie POURQUIE Olivier REBAGLIATI Michael Postdoctoral fellows BENAZERAF Bertrand CADETE VILHAIS NETO Gonçalo CHAL Jérôme MARUHASHI Mitsuji OGINUMA Masayuki SU Cheng Wen WAHL Matthias PhD Students ANTONI Bernadette BERA Agata DE MOT Laurane DENANS Nicolas KROL Aurélie Engineers/Technicians DALI Soraya GARNIER Jean-Marie KENNEDY Leif MONCUQUET Philippe PACE Jennifer PLASSAT Jean-Luc TASSY Olivier Master Student GUENNOUN Rym RIVELINE Daniel Laboratory of Cell Physics Scientists RIVELINE Daniel Engineers/Technicians HOEL Antonin TORRES-PADILLA Maria Elena Epigenetics and cell fate in early mammalian development Scientists TORRES PADILLA Maria Elena Postdoctoral fellows BURTON Jonathan Adam FADLOUN Anas MIYANARI Yusuke PhD Students SANTENARD Angèle Engineers/Technicians ZIEGLER-BIRLING Céline Master Student BENDER Ambre VERMOT Julien Mecano-genetic interplays and embryonic morphogenesis Scientists VERMOT Julien Postdoctoral fellows GOETZ Jacky MOJZISOVA Halina PhD Students HECKEL Emilie Engineers/Technicians GESCHIER Sandrine ROTH Stéphane VIVILLE Stéphane Primordial germ cells’ ontogeny Scientists KOSCINSKI Isabelle TELETIN Marius VIVILLE Stéphane Postdoctoral fellows CELEBI Catherine EL RAMY GHOSN Rosy PhD Students ELINATI Elias JUNG Laura Engineers/Technicians ANDRE Cécile SKORY Valérie TROPEL Philippe Master Students FOSSARD Camille TARABAY Yara staff scientists Functional Genomics & Cancer Coordinator: DAVIDSON Irwin Assistant: GASSERT Valerie CHAN Susan/ KASTNER Philippe Hematopoiesis and Leukemogenesis in the mouse Scientists CHAN Susan KASTNER Philippe KIRSTETTER Peggy Postdoctoral fellows CAI Qi ORAVECZ Attila PhD Students APOSTOLOV Apostol KAVERI Deepika LE LAY Anne-Solen MACIAS Beatriz Alejandra MASTIO Jérôme Engineers/Technicians MARCHAL Patricia Master Student VESIN Rose-Marie DAVIDSON Irwin Structure and function of the general transcription factor TFIID Scientists DAVIDSON Irwin MARTIANOV Igor MENGUS Gabrielle Postdoctoral fellows ALPERN Daniil KOBI Dominique LANGER Diana PhD Students BENHADDOU Ataaillah CHOUKRALLAH Mohamed-Amin MOUTIER Emmanuel STRUB Thomas Engineers/Technicians MICHEL Isabelle YE Tao Master Student URBAN Sylvia EGLY Jean-Marc/COIN Frédéric Genome expression and repair Scientists COIN Frédéric COMPE Emmanuel EGLY Jean-Marc Postdoctoral fellows EBEROVA Jitka HASHIMOTO Satoru LE MAY Nicolas VELEZ-CRUZ Renier VITRENKO Yakov PhD Students GIRAUDON Christophe ILTIS Izarn KORETS Roman KRISTENSEN Hans-Ulrik SINGH Amita TRABOULSI Hussein ZADORIN Anton ZHOVMER Alexander Engineers/Technicians BRAUN Cathy CATEZ Philippe LARNICOL Annabel MARTEL Fernand PREVE Brigitte Master Student ZIANI Salim GRONEMEYER Hinrich From nuclear receptor action to novel paradigms for cancer therapy action Scientists GRONEMEYER Hinrich Postdoctoral fellows CESCHIN Danilo KEDINGER Valérie LUND Per MENDOZA PARRA Marco Antonio PATTABHIRAMAN Shankara-Narayanan PAVET-PORTAL Valeria PORTAL Maximiliano PhD Students KHANWALKAR Harshal MUELLER Cathrin VJETROVIC Jelena WALIA Mannu Kamalraj Engineers/Technicians ERB Cathie LIEB Michèle PAULI Cecilia VALLET Judith VAN GOOL Wouter YUJNOVSKY Irene Master Students BORIES Pierre WEISS Mélanie HAMICHE A . Chromatin and epigenetic regulation Scientists HAMICHE Ali RAMAIN Catherine RAMAIN Philippe Postdoctoral fellows DEPAUX Arnaud OUARARHNI Khalid PAPIN Christophe PhD Students OBRI Arnaud OURY Julien SHUAIB Muhammad TRIPATHI Vivek YETTOU Guillaume Engineers/Technicians DANIEL Dorothée LOSSON R. Scientists CAMMAS Florence Postdoctoral fellows RICLET Raphaël PhD Students GRABER Céline HERQUEL Benjamin Engineers/Technicians CERVINO Margarita LEROUGE Thierry METZGER/CHAMBON Genetic dissection of nuclear receptor signaling in the mouse Scientists CHAMBON Pierre KRUST Andrée LI Mei METZGER Daniel Postdoctoral fellows HUA Guoqiang JIANG Hua MUKHERJI Atish SURJIT Milan PhD Students DUTEIL Delphine GALI RAMAMOORTHY Thanuja GANTI Krishna Priya LEYVA CASTILLO Juan Manuel Engineers/Technicians BORNERT Jean-Marc FRIEDMANN Laetitia GARGOWITSCH Laëtitia HENER Pierre HUC Magali MEYER Tania Master Students BAGCI Hakan TALEB Fatima REINA SAN MARTIN Bernardo Molecular biology of B cells Scientists REINA SAN MARTIN Bernardo Postdoctoral fellows ROBERT Isabelle ROTTNER Mathilde PhD Students JEEVAN RAJ Beena Patricia MILOSEVIC Sara SCHIAVO Ebe Engineers/Technicians HEYER Vincent Master Student OUBRAHAM Lila RIO Marie-Christine / TOMASETTO Catherine Molecular and cellular biology of breast cancer Scientists ALPY Fabien DALI YOUCEF Nassim RIO Marie-Christine TOMASETTO Catherine Postdoctoral fellows BUACHE Emilie PhD Students DAGUENET Elisabeth LEGUEUX François ROUSSEAU Adrien SIMOES David TAN Jinxiang Engineers/Technicians STOLL Isabelle, technicienne WENDLING Corinne, ingénieur ROCHETTE-EGLY Cécile Nuclear retinoic acid receptors phosphorylation and Crosstalk with signaling pathways Scientists EGLY Cécile Postdoctoral fellows ALTANOUZY Ziad DUONG Vanessa PANKOTAI-BODO Gabriella PhD Students FERRY Christine PISKUNOV Aleksandr SAMARUT Eric Engineers/Technicians GAOUAR Samia LUTZING Régis SERAPHIN Bertrand Protein networks and complexes regulating eukaryotic mRNA decay Scientists MAUXION Fabienne SERAPHIN Bertrand Postdoctoral fellows DREUMONT Natacha GAS LOPEZ Maria KOLESNIKOVA Olga PhD Students RISPAL Delphine VAN DEN ELZEN Antonia Engineers/Technicians FAUX Céline GAUDON-PLESSE Claudine MULLER Benjamin SOUTOGLOU Evi Cell Biology of genome integrity Scientists SOUTOGLOU Evi Postdoctoral fellows NAGY Zita PANKOTAI Tibor PhD Students HOFFBECK Anne-Sophie LEMAITRE Charlène Engineers/Technicians BONHOMME Céline FURST Audrey TORA Laszlo Chromatin modifications and regulation of gene expression during differentiation Scientists DEVYS Didier TORA Laszlo Postdoctoral fellows BALLARINO Monica FOURNIER Marjorie HELMRICH Anne KARMODIYA Krishanpal UMLAUF David PhD Students BONNET Jacques GYENIS Akos KREBS Arnaud LANG Guillaume RISS Anne Engineers/Technicians SCHEER Elisabeth STIERLE Matthieu WASYLYK B. Molecular and cellular biology of cancer Scientists WASYLYK Bohdan PhD Students LI Yadong SEMENCHENKO Kostyantyn Engineers/Technicians WASYLYK Christine Attached researcher: DU MANOIR Stanislas Integrated Structural Biology Coordinator : SCHULTZ Patrick Assistant: NEY Anne CAVARELLI Jean Structural biology of epigenetic targets Scientists CAVARELLI Jean CURA Vincent ROMIER Christophe WURTZ Jean-Marie Postdoctoral fellows HASSENBOEHLER Pierre MAREK Martin PhD Students DIEBOLD Marie Laure LEPROULT Emeline LIENHART Yann MAILLIOT Justine Engineers/Technicians TROFFER-CHARLIER Nathalie DEJAEGERE-STOTE Annick Biocomputing Scientists DEJAEGERE-STOTE Annick STOTE Roland Postdoctoral fellows BROCHET Xavier SCHWARZ Benjamin PhD Students AMAL Ismail FERRARIO Maria Giovanna KIEFFER Bruno Biomolecular Nuclear Magnetic Resonance Scientists DELSUC Marc-André KIEFFER Bruno LEBARS Isabelle Postdoctoral fellows RAMIREZ RAMOS Juan Ramon QUINTERNET Marc PhD Students TANTY Matthieu Engineers/Technicians LING Claude KLAHOLZ Bruno Large complexes involved in gene expression Scientists KLAHOLZ Bruno OURJOUMTSEV Alexandre Postdoctoral fellows KRISHNAGIRI VENKATASUBRAMANIAN Srividhya MALETTA Massimiliano MANICKA Sankar Narayanan MYASNIKOV Alexander ORLOV Igor SIMONETTI Angelita PhD Students TORCHY Morgan Engineers/Technicians HAZEMANN Isabelle MENETRET Jean-François 113 staff scientists & PLATFORMS MORAS Dino Expression of genetic information Scientists BILLAS-MASSOBRIO Isabelle DOCK-BREGEON Anne-Catherine MORAS Dino POTERSZMAN Arnaud ROCHEL-GUIBERTEAU Natacha RUFF Marc Postdoctoral fellows ABDULRAHMAN Wassim ANTONY Pierre BRELIVET Yann LEVY Nicolas OSZ-PAPAI Judit RADU Laura TAKACS Maria PhD Students ALBOU Laurent Philippe MAILLOT Benoît MARTINEZ ZAPIEN Denise SCHAETZEL Aurélie UCHIKAWA Emiko Engineers/Technicians EILER Sylvia PELUSO-ILTIS Carole POCH Olivier Evolutionary systems biology Scientists POCH Olivier LECOMPTE Odile MULLER Jean THOMPSON Julie WICKER Nicolas Postdoctoral fellows NGUYEN Ngoc Hoan PhD Students ANNO Yannick-Noël ANIBA Mohamed Radhouane LINARD Benjamin LUU Tien Dao Engineers/Technicians MOULINIER Luc POIDEVIN Laëtitia RIPP Raymond SCHULTZ Patrick Architecture of Nucleoprotein Systems by 3-D Electron Microscopy Scientists DRILLIEN Robert LAMOUR Valérie SCHULTZ Patrick Postdoctoral fellows PAPAI Gabor PRADEAU AUBRETON Karine PhD Students KIZILYAPRAK Caroline PAPILLON Julie Engineers/Technicians CRUCIFIX Corinne DURAND Alexandre RUHLMANN Christine SIMONI Albin SPEHNER Danièle YUSUPOV Marat Ribosomes Scientists JENNER Lasse Bohl YUSUPOV Marat YUSUPOVA Gulnara Postdoctoral fellows BEN SHEM Adam DEMESHKINA Natalia MELNIKOV Sergey PhD Students GARREAU DE LOUBRESSE Nicolas Engineers/Technicians DUCLAUD Sylvie Attached Scientists: PODJARNY Alberto COUSIDO-SIAH Alexandra MITSCHLER André Translational medicine & Neurogenetics Coordinator : KIEFFER Brigitte Assistant: BRONNER Sylvianne CHARLET-BERGUERAND Nicolas Physiopathology of the RNA gain-of-function diseases Scientists CHARLET BERGUERAND Nicolas Postdoctoral fellows SELLIER Chantal PhD Students FREYERMUTH Fernande RAU Frédérique Engineers/Technicians FISCHER-HUMMEL Marie-Christine FUGIER Charlotte HICKEL Pierre FABRE Jean-Etienne Atherosclerosis and thrombosis Scientists FABRE Jean-Etienne Postdoctoral fellows BOUCHAREB-DAHOU Rihab SLIMANI Farid Engineers/Technicians TILLY Peggy HANAUER André/ MANDEL Jean-Louis Mechanisms of monogenic forms of mental retardation Scientists HANAUER André MANDEL Jean-Louis MOINE Hervé Postdoctoral fellows BOHM Johann PhD Students MEHMOOD Tahir SCHNEIDER Anne TABET Ricardos VASLI Nasim Engineers/Technicians PANNETIER Solange FLATTER Eric HAUMESSER Nicolas HERAULT Yann Physiopathology of aneuploidy, gene dosage effect and Down syndrome Scientists BRAULT Véronique HERAULT Yann Postdoctoral fellows MARECHAL Damien RAVEAU Matthieu SALEH Abdelsalam Engineers/Technicians CHEVALIER Claire DUCHON Arnaud NALESSO Valérie KIEFFER Brigitte The opioid system and brain function Scientists BECKER Jérôme BEFORT Katia GAVERIAUX-RUFF Claire KIEFFER Brigitte MASSOTTE Dominique OUAGAZZAL Abdel-Mouttalib ROUX Michel STEPHAN Aline Postdoctoral fellows DARCQ Emmanuel LE MERRER Julie NOZAKI TAKAHASHI Chihiro PETERSCHMITT Yvan SEBAI Sarra PhD Students CHU SIN CHUNG Paul DEL BOCA Carolina DENIZ Sercan FAGET Lauren GARDON Olivier LUTZ Pierre-Eric REZAI Xavier WEIBEL Raphaël Engineers/Technicians ERBS Eric FILLIOL Dominique KOEBEL Pascale MATIFAS Audrey REISS David ROBE Anne KOENIG Michel Recessive ataxias Scientists KOENIG Michel Postdoctoral fellows ASSOUM Mirna Engineers/Technicians DROUOT Nathalie LAPORTE Jocelyn Mechanisms of neuromuscular disease Scientists LAPORTE Jocelyn Postdoctoral fellows COWLING Belinda D’ALESSANDRO Manuela HNIA Karim KOUTSOPOULOS Olga ROYER ZEMMOUR Barbara PhD Students AMOASII Leonela TOSCH Valérie ZIVKOVIC Ivana Engineers/Technicians KOCH Catherine KRETZ Christine PUCCIO Hélène Biology and Physiology of recessive ataxia PUCCIO Hélène Postdoctoral fellows BOYER Frédéric MARTELLI Alain TESCHNER Julia WATTENHOFER-DONZE Marie PhD Students COLIN Florent LICITRA Floriana SCHMUCKER Stéphane Engineers/Technicians REUTENAUER Laurence VAUCAMPS Nadège Master HICK Aurore TROTTIER Yvon Pathogenic mechanisms of polyglutamine expansion diseases Scientists MERIENNE Karine TROTTIER Yvon Postdoctoral fellows KLEIN Fabrice PhD Students DAVRANCHE Aurélien GOULA Agathi-Vasiliki KARAM Alice Engineers/Technicians WEBER Chantal Master EL KHOURY Rita Technical support & administration PLATFORMS Bioinformatics PLEWNIAK Frédéric BIANCHETTI Laurent GEOFFROY Véronique RAFFELSBERGER Wolfgang SIGUENZA Sophie OULAD-ABDELGHANI Mustapha, Antibodies Monoclonal ANDRES Valérie JUNG Nicole Microarrays and Deep Sequencing THIBAULT-CARPENTIER Christelle ALUNNI Violaine BAMBA Géraldine COLAS Ingrid DEMBELE Doulaye HANAUER Antoine HEROUARD-MOLINA Cathy JOST Bernard KEIME Céline LE GRAS Stéphanie VICAIRE Serge TROFFER-CHARLIER Nathalie, Baculovirus KOLB-CHEYNEL Isabelle VIALLE Chantal Structural Biology and Genomics Platform BUSSO Didier BIRCK Catherine GRANGER Florence LITT Alain MCEWEN Alastair POUSSIN-COURMONTAGNE Pierre SALIM Loubna TROESCH Edouard Imaging Center VONESCH Jean-Luc HENTSCH Didier SCHWAB Yannick MESSADDEQ Nadia BOEGLIN Marcel DEISS Alexandre FAUNY Jean-Daniel HERGUEUX Josiane KESSLER Pascal KOCH Marc LUTZ Yves SPIEGELHALTER Coralie WEICKERT Jean-Luc High Throughput Cell-based Screening Platform BRINO Laurent FISCHER Benoit FROIDEVAUX Laure MOUNE-DIMALA Martin WEISS Amélie Cores Facilities VAN ES Armelle, Veterinary METZGER Elisabeth, Head of Animal House BLONDELLE Asmae CHRIST Lydia DELAPORTE Claude DIETRICH Holly DUFOUR Stephane EISENMANN Aurélie FALCONE Sylvie GENDRON Michaël HACHEM Rabiaa HIRLIMANN Anne-Lise JAWAD Agouna KHADRAOUI Hafid KORCHI Hafid KORCHI Nordine MAGNANT William MEMEDOV Fatima POIROT Martine RICHERT Mourad SIEGEL Sabine VINCENT Alexandre ZINK Nicolas DUVAL Gilles, Antibodies Polyclonal MEMEDOV Djemalj HELLER Betty, Cell culture & Cell Bank BOSSENMEYER Patricia CHRISTOFFEL Ghislaine FELS Karine FERANDEL Noëlle MASTIO Leslie RIESTERER Hélène SIGNOUR Aline PAGE Adeline, Head of Mass spectrometry CHAVANT Virginie RUFFENACH Frank EBEL Claudine, Cell Sorting EBERLING Pascal, Peptide synthesis ICS HERAULT Yann, ICS Director SORG-GUSS Tania, Technical Director ICS Management Support BLONDELLE Eric CHEBBOUB Djaouida FRICKER Bastien NAGRE Isabelle PENSAVALLE Joëlle RIO-ZENNER Fanny SELLOUM Mohammed ICS Bioinformatics VASSEUR Laurent, Head of Bioinformatics DEBOUZY Guillaume LEBLANC Sophie MORO Anne-Isabelle ICS Genetic Engineering and Model Validation PAVLOVIC Guillaume, Head of Mutagenesis, Transgenesis & Molecular Phenotyping Department AUGE Fabrice BIRLING Marie-Christine CARADEC Claudia CAYROU Pauline CHARTOIRE Nathalie DIERICH Andrée DREYER Dominique EGLINGER Yolande ERBS Valérie ESSABRI Karim HELMSTETTER-VERDOT Cindy JACQUOT Sylvie KLEISS Charlotte LINDNER Loïc LORENTZ Romain LUPPI Laurence MERTZ Annelyse QUEUCHE Danielle ROTH Christelle ROUSSEAU Valérie SCHWOERER Marie-Jeanne SEITZ Thierry VENTEO Lydie WALLERICH Sandrine ICS Phenotyping SORG-GUSS Tania, Head of Phenotyping AMANN Grégory AUBURTIN Aurélie AUVRAY Sandy BECHAKRA Malik BECKER Julien BEDU Elodie BUNZ Isabel CES Aurélia CHAMPY Marie-France COMBE Roy DING Thomas EL FERTAK Lahcen FOUGEROLLE Jean-Victor GOETZ-REINER Patrice GUIMOND Alain KOUTSEFF Alexis LALANNE Valérie LE MARCHAND Elise LECOCQ Muriel LUX Aline MEZIANE Hamid MITTELHAEUSER Christophe MONTIAL Marina MOULAERT David MULLER Stéphanie PETER Emilie PETIT-DEMOULIERE Benoît PHAM THI BICH Hanh POUILLY Laurent RIET Fabrice ROUSSEAU Stéphane SUTTER-WOLTER Anne TILLY Isabelle WAGNER Christel WEBER Bruno WENDLING Olivia ICS Mouse Facility GONCALVES DA CRUZ Isabelle, Veterinary AYADI Abdelkader, Head of Animal Facility Department ALI-HADJI Dalila ANDRE Philippe BAM’HAMED Chaouki BANQUART-OTT Nadine BOUR Raphaël BRIGNON Sophie CHARLES Philippe DELANGLE Benoît EL FERTAK Leila ENNAH Hamid FISCHER Fabienne FISCHER Natacha GRUBER Frédéric HEMMERLE Mathieu KUJATH Christelle KURTZ Caroline LANTZ-CHAMPY Charlotte LAEUFER Laurent LEGEAY Sandrine MELLUL Peggy MOKNI Mourad OHLMANN Thierry SCHMITT Raphaël SCHOEDEL Christophe SEILER Stéphane TOUBARI Chadia UZUN Ibrahim VINCENT Cindy WALCH Laëtitia WETZSTEIN Eric WIECROCK Cyrille WIECROCK Ludovic ZANINELLO Fabienne ZANINELLO Nathalie Management POURQUIE Olivier, Director of IGBMC BENSEL Nicolas, Director of Logistic & Scientific Operations LUNKES Astrid, Director of Scientific Affairs SCHAEFFER Katell, Director of Finance and Administration Management Assistance BENARROCH Laurence GONZALEZ Laetitia KAUFFMANN Dominique MEYER Estelle SCHUBEL Françoise THORNTON Laura Reception ACKER Evelyne MENNA Martine Human Ressources Department PASQUIER Lydia, Head of Human Ressources ACKER Dominique FISCHER Joëlle LENTZ Brigitte LESECQ Florence METAIS Bénédicte PETILLON Armelle ROTH Catherine TOUSSAINT Elisabeth Communication ESTEBAN-POURQUIE Silvia, Head of Communication BOUR Sandra LEGRAND Elodie Accounting/Controlling RAMOND Marie-Jeanne, Head of Finance PINCHON Frédérique, Head of Controlling HOFFER Stéphane Head of Accounting CARBONNIER Laëtitia GAUPP Cindy GONTHIER Patricia GRUFFAZ Agnès HECKER Annick MUNIER Brigitte RACK Nicole REGNERY Véronique UFFLER Sylvie Purchasing / Stock MATHON Peggy CHARLES Christelle FASSEL Serge GERBER Philippe KIENZLER Elise MULLER Catherine REINE Valérie REPIS Christian SIGWALT Jessica TAMAYO Sandrine WETZSTEIN Eric Technical Department KOLB Claude, Head of Technical Department UNTEREINER Christophe, Deputy/Technical Department BEAUJEAN Frédéric KUNTZ Pascal LANG Doris POIMBOEUF-MAHIEU Marc SCHERMULY Jean-Claude TANTON Patrick TAUBERT Serge VATTE Christophe VONSCHEIDT Régis WEIL Luc House Maintenance BINTEIN Cathy BORSCHNECK Denise CRABEY Karine EL YAMANI Hassan GREDLER Sylvie HAOUALA Ilias HEITZ Nadine LAACHARI Mohammed PERAL Louisette PLANCHAT Catherine SCHMITT Sylvie STIEFFEL Emma Valorisation/Grant Management BROOKS Steve DAUZET Frédéric Systems Architect/Administrator MOUTAUX Tony OFFNER Michel IT Service FRITZ Rémy, Head of IT Service MALLOUH Véronique CERDAN Stéphane RONGVAUX Youry SEITH Guillaume TOUSSAINT Jean-Luc UGE Serge VERLEY Philippe ZOUIOUIECH Agathe Hygiene Safety BIELLMANN Dominique Library MARTINA Hélène PENEY Marie-Thérèse 115 prizes & distinctions 2010, Pediatric Pathology Research Senior Prize 2009, Jocelyn LAPORTE 2008, CNRS Bronze Medal, Bruno KLAHOLZ 2010, Fondation Gairdner Prize (Canada), Pierre CHAMBON 2008 Nominated Inventor of the Year by the European Patent Office, Bertrand SÉRAPHIN 2010, Eurosystem Young Investigator Award, Sophie JARRIAULT 2007, Madeleine Lecoq Prize, Académie des Sciences, Julie THOMPSON-MAALOUM 2009, Ligue Contre le Cancer René et Andrée Duquesne Prize, Jean-Marc EGLY 2007, Jules Martin Prize, Académie des Sciences, Daniel METZGER 2009, INSERM Innovation Prize, Jean-Marie GARNIER 2009, Fondation Schlumberger pour la Recherche et l’Enseignement Prize, Sophie JARRIAULT 2009, Fondation pour la Recherche Médicale Marguerite Delahautemaison Prize, Cécile ROCHETTE-EGLY 2009, Fondation pour la Recherche Médicale Comité Alsace Scientific Prize, Maria-Elena TORRES-PADILLA 2009, Fondation pour la Recherche Médicale Comité Alsace Scientific Prize, Bernardo REINA SAN MARTIN 2007, Prix de la Recherche de la Fondation pour la Recherche Medicale Comité Alsace, Catherine TOMASETTO 2006, Lacassagne Prize of Collège de France, Filippo RIJLI 2006, EMBO Young Investigator, Bruno KLAHOLZ 2006, Grand Prix de la Fondation pour la Recherche Médicale, Jean-Louis MANDEL 2005, Pediatric Pathology Research Senior Prize, Hélène PUCCIO 2005, Jean Valade Award, Johan AUWERX 2005 Cristal CNRS, Julie THOMPSON-MAALOUM 2009, Académie Nationale de Médecine Prize, Jean-Louis MANDEL 2005, Helmholtz Humboldt Research Prize, Laszlo TORA 2009, Académie Nationale de Médecine Henry et Mary-Jane Mitjaville Prize, Jean-Marc EGLY 2005, Grand Prix Victor Noury, Académies des Sciences , Olivier POURQUIÉ 2009, Académie des Sciences Fondation scientifique franco-taïwanaise prize, Angela GIANGRANDE 2005, Edouard Van Beneden de l’Académie Royale de Belgique, Olivier POURQUIÉ 2009, Gutenberg Prize, Bruno KLAHOLZ 2005, Grand Prix Ruban Rose de la Recherche, Marie-Christine RIO 2008, Fondation pour la Recherche Médicale Comité Alsace Scientific Prize, Sophie JARRIAULT 2004, Richard Lounsbery, Académie des Sciences, France et USA Brigitte KIEFFER 2008, Prix Jean et Madeleine Schaeverbeke de la Fondation de France, Adèle DE ARCANGELIS 2004, Prix Harland Winfield Mossman en Biologie du Développement, American Association of Anatomists, Olivier POURQUIÉ 2008, Dr. Jean Toy Prize, Académie des Sciences, Hélène PUCCIO 2008, Charles-Louis de Saulses de Freycinet Prize, Académie des Sciences, Frédéric COIN 2004, Prix d’Honneur INSERM, Pierre CHAMBON 2004, Paul Basset Award, Catherine TOMASETTO 2004, IPSEN Neuronal Plasticity, Jean-Louis MANDEL 2004, Ipsen Endocrine communication and regulation, Pierre CHAMBON 2004, Grand Prix INSERM 2004, Jean-Marc EGLY 2004, ESHG Young Scientist Award, Aurora PUJOL publications Until June 2010 Amsen E, Alfred J, Pourquie O (2010) The Node: a place to discuss, debate and deliberate developmental biology. Development 137: 2251 Anheim M, Fleury M, Monga B, Laugel V, Chaigne D, Rodier G, Ginglinger E, Boulay C, Courtois S, Drouot N, Fritsch M, Delaunoy JP, Stoppa-Lyonnet D, Tranchant C, Koenig M (2010) Epidemiological, clinical, paraclinical and molecular study of a cohort of 102 patients affected with autosomal recessive progressive cerebellar ataxia from Alsace, Eastern France: implications for clinical management. Neurogenetics 11: 1-12 2004, Albert Lasker Award for Basic Medical Research, Pierre CHAMBON Antony P, Sigueiro R, Huet T, Sato Y, Ramalanjaona N, Rodrigues LC, Mourino A, Moras D, Rochel N (2010) StructureFunction Relationships and Crystal Structures of the Vitamin D Receptor Bound 2alpha-Methyl-(20S,23S)- and 2alpha-Methyl-(20S,23R)-epoxymethano1alpha,25-dihydroxyvitamin D(3). J Med Chem 53: 1159-71 2003, INSERM Innovation prize «Methodology», Jean-Luc VONESCH Arai Y, Gradwohl G, Kameda Y (2010) Expression of neuropeptide Y and agoutirelated peptide in the hypothalamic arcuate nucleus of newborn neurogenin3 null mutant mice. Cell Tissue Res 340: 137-45 2004, Edwin B. Astwood Award, Paolo SASSONE-CORSI Audo I, Manes G, Mohand-Said S, Friedrich A, Lancelot ME, Antonio A, Moskova-Doumanova V, Poch O, Zanlonghi X, Hamel CP, Sahel JA, Bhattacharya SS, Zeitz C (2010) Spectrum of rhodopsin mutations in French autosomal dominant rod-cone dystrophy patients. Invest Ophthalmol Vis Sci 51: 3687-700 Ayoub, C, Wasylyk, C, Li Y, Thomas E, Marisa L, Robé A, Roux M, Abecassis J, de Reyniès A and Wasylyk B. 2010 ANO1 amplification and expression in HNSCC with a high propensity for future distant metastasis and its functions in HNSCC cell lines. BJC, In press Baret JC, Beck Y, Billas-Massobrio I, Moras D, Griffiths AD (2010) Quantitative cell-based reporter gene assays using droplet-based microfluidics. Chem Biol 17: 528-36 Ben Khedher S, Zouari N, Messaddeq N, Schultz P, Jaoua S. Overproduction of Delta-Endotoxins by Sporeless Bacillus thuringiensis Mutants Obtained by Nitrous Acid Mutagenesis. Curr Microbiol. 2010 May 20. [Epub ahead of print] PubMed PMID: 20490495. Berge C, Froloff N, Kalathur RK, Maumy M, Poch O, Raffelsberger W, Wicker N (2010) Multidimensional fitting for multivariate data analysis. J Comput Biol 17: 723-32 Bich C, Bovet C, Rochel N, Peluso-Iltis C, Panagiotidis A, Nazabal A, Moras D, Zenobi R (2010) Detection of nucleic acid-nuclear hormone receptor complexes with mass spectrometry. J Am Soc Mass Spectrom 21: 635-45 Bieniossek C, Nie Y, Frey D, Olieric N, Schaffitzel C, Collinson I, Romier C, Berger P, Richmond TJ, Steinmetz MO, Berger I. Automated unrestricted multigenerecombineering for multiprotein complex production. Nat Methods. 2009 Jun;6(6):447-50. Bonnet J., Wang Y-H., Spedale G., Atkinson R.A., Romier C., Hamiche A., Pijnappel W.W.M. P., Timmers H.Th. M., Tora L., Devys D. and Kieffer B. (2010) Structural plasticity of SCA7 domains defines their differential nucleosome binding properties. EMBO Reports, In press. Börjesson A, Lagerquist M, Liu C, Shao R, Windahl S, Karlsson C, Sjögren K, Movérare-Skrtic S, Antal M, Krust A, Mohan S, Chambon P, Sävendahl L, Ohlsson C. The role of estrogen receptor-alpha in growth plate cartilage for longitudinal bone growth. J Bone Miner Res. 2010 Jun 18. Costello P, Nicolas R, Willoughby J, Wasylyk B, Nordheim A, Treisman R. J Immunol. 2010 Jun 16. [Epub ahead of print]PMID: 20554967 [PubMed - as supplied by Publisher] Ternary Complex Factors SAP-1 and Elk-1, but Not Net, Are Functionally Equivalent in Thymocyte Development. Bossenmeyer-Pourie, C., S. Blaise, G. Pourie, C.Tomasetto, S. Audonnet, S. Ortiou, V. Koziel, M. C. Rio, J. L. Daval, J. L. Gueant, and B. Beck, 2010, Methyl donor deficiency affects fetal programming of gastric ghrelin cell organization and function in the rat: Am J Pathol, v. 176, p. 270-7. Cronin T, Raffelsberger W, Lee-Rivera I, Jaillard C, Niepon ML, Kinzel B, Clerin E, Petrosian A, Picaud S, Poch O, Sahel JA, Leveillard T (2010) The disruption of the rod-derived cone viability gene leads to photoreceptor dysfunction and susceptibility to oxidative stress. Cell Death Differ 17: 1199-210 Boumlic A, Nomine Y, Charbonnier S, Dalagiorgou G, Vassilaki N, Kieffer B, Trave G, Mavromara P, Orfanoudakis G (2010) Prevalence of intrinsic disorder in the hepatitis C virus ARFP/Core+1/S protein. Febs J 277: 774-89 Brasse D, Mathelin C, Leroux K, Chenard MP, Blaise S, Stoll I, Tomasetto C, Rio MC. Matrix metalloproteinase 11/stromelysin-3 exerts both activator and repressor functions during the hematogenous metastatic process in mice. Int J Cancer. 2010, In press (+ cover) Budin, G., M. M. Dimala, V. Lamour, P. Oudet, C. Mioskowski, S. Meunier, L. Brino, and A. Wagner, 2010, A chemical labeling strategy for proteomics under nondenaturing conditions: Chembiochem, v. 11, p. 79-82. Cammas L, Trensz F, Jellali A, Ghyselinck NB, Roux MJ, Dolle P (2010) Retinoic acid receptor (RAR)-alpha is not critically required for mediating retinoic acid effects in the developing mouse retina. Invest Ophthalmol Vis Sci 51: 3281-90 Calippe B, Douin-Echinard V, Delpy L, Laffargue M, Lélu K, Krust A, Pipy B, Bayard F, Arnal JF, Guéry JC, Gourdy P. 17{beta}-Estradiol Promotes TLR4-Triggered Proinflammatory Mediator Production through Direct Estrogen Receptor {alpha} Signaling in Macrophages In Vivo. J Immunol. 2010 Jun 16. Canto C, Jiang LQ, Deshmukh AS, Mataki C, Coste A, Lagouge M, Zierath JR, Auwerx J (2010) Interdependence of AMPK and SIRT1 for metabolic adaptation to fasting and exercise in skeletal muscle. Cell Metab 11: 213-9 Charlot C, Dubois-Pot-Schneider H, Serchov T, Tourrette, Y, Wasylyk B. Posttranslational modifications and sub-cellular localization of Ets transcription factors: possiblr connections with cancer and involvement in the hypoxic response. In Transcription Factors Methods and Protocols Series: Methods in Molecular Biology , Vol. 647 Higgins, Paul J. (Ed.) 2010, Approx. 375 p. 93 illus., 7 in color., Hardcover ISBN: 978-1-60761-737-2 Cheng L, Guo J, Sun L, Fu J, Barnes PF, Metzger D, Chambon P, Oshima RG, Amagai T, Su DM (2010) Postnatal Tissuespecific Disruption of Transcription Factor FoxN1 Triggers Acute Thymic Atrophy. J Biol Chem 285: 5836-47 Claeys KG, Maisonobe T, Bohm J, Laporte J, Hezode M, Romero NB, Brochier G, Bitoun M, Carlier RY, Stojkovic T (2010) Phenotype of a patient with recessive centronuclear myopathy and a novel BIN1 mutation. Neurology 74: 519-21 Cobucci-Ponzano B, Guzzini L, Benelli D, Londei P, Perrodou E, Lecompte O, Tran D, Sun J, Wei J, Mathur EJ, Rossi M, Moracci M (2010) Functional characterization and high-throughput proteomic analysis of interrupted genes in the archaeon Sulfolobus solfataricus. J Proteome Res 9: 2496-507 Degletagne C, Keime C, Rey B, de Dinechin M, Forcheron F, Chuchana P, Jouventin P, Gautier C, Duchamp C. Transcriptome analysis in non-model species: a new method for the analysis of heterologous hybridization on microarrays. BMC Genomics. 2010 May 31;11(1):344. Delacroix, L., E. Moutier, G. Altobelli, S. Legras, O. Poch, M. A. Choukrallah, I. Bertin, B. Jost, and I. Davidson, 2010, Cell-specific interaction of retinoic acid receptors with target genes in mouse embryonic fibroblasts and embryonic stem cells: Mol Cell Biol, v. 30, p. 231-44. De Graaf P., Mousson F., Geverts B., Scheer E., Tora L., Houtsmuller A.B. and Timmers H. Th. M. (2010) Chromatin interaction of TATA-binding protein (TBP) is dynamically regulated in human cells in human cells. Journal of Cell Science, In press. Drane P, Ouararhni K, Depaux A, Shuaib M, Hamiche A (2010) The death-associated protein DAXX is a novel histone chaperone involved in the replication-independent deposition of H3.3. Genes Dev 24: 1253-65 Dreumont, N., C. F. Bourgeois, F. Lejeune, Y. Liu, I. E. Ehrmann, D. J. Elliott, and J. Stevenin, 2010, Human RBMY regulates germline-specific splicing events by modulating the function of the serine/arginine-rich proteins 9G8 and Tra2-{beta}: J Cell Sci, v. 123, p. 40-50. Dreumont N, Hardy S, Behm-Ansmant I, Kister L, Branlant C, Stevenin J, Bourgeois CF (2010) Antagonistic factors control the unproductive splicing of SC35 terminal intron. Nucleic Acids Res 38: 1353-66 Eduardo-Canosa S, Fraga R, Sigüeiro R, Marco M, Rochel N, Moras D, Mouriño A. Design and synthesis of active vitamin D analogs. J Steroid Biochem Mol Biol. 2010 Mar 24 Edwards MM, Mammadova-Bach E, Alpy F, Klein A, Hicks WL, Roux M, Simon-Assmann P, Smith RS, Orend G, Wu J, Peachey NS, Naggert JK, Lefebvre O, Nishina PM. Mutations in Lama1 disrupt retinal vascular development and inner limiting membrane formation. J Biol Chem. 2010 Mar 5;285(10):7697-711. Fabre, J. E., and M. E. Gurney, 2010, Limitations of current therapies to prevent thrombosis: a need for novel strategies: Mol Biosyst, v. 6, p. 305-15. Facca S, Cortez C, Mendoza-Palomares C, Messadeq N, Dierich A, Johnston AP, Mainard D, Voegel JC, Caruso F, Benkirane-Jessel N (2010) Active multilayered capsules for in vivo bone formation. Proc Natl Acad Sci U S A 107: 3406-11 Fidelak J, Ferrer S, Oberlin M, Moras D, Dejaegere A, Stote RH. Dynamic correlation networks in human peroxisome proliferator-activated receptor-gamma nuclear receptor protein. Eur Biophys J. 2010 May 23. Fourcade S, Ruiz M, Guilera C, Hahnen E, Brichta L, Naudi A, Portero-Otin M, Dacremont G, Cartier N, Wanders R, Kemp S, Mandel JL, Wirth B, Pamplona R, Aubourg P, Pujol A (2010) Valproic acid induces antioxidant effects in X-linked adrenoleukodystrophy. Hum Mol Genet 19: 2005-14 Friedrich A, Garnier N, Gagniere N, Nguyen H, Albou LP, Biancalana V, Bettler E, Deleage G, Lecompte O, Muller J, Moras D, Mandel JL, Toursel T, Moulinier L, Poch O (2010) SM2PH-db: an interactive system for the integrated analysis of phenotypic consequences of missense mutations in proteins involved in human genetic diseases. Hum Mutat 31: 127-35 Gales A, Conduche A, Bernad J, Lefevre L, Olagnier D, Beraud M, Martin-Blondel G, Linas MD, Auwerx J, Coste A, Pipy B (2010) PPARgamma controls dectin-1 expression required for host antifungal defense against Candida albicans. PLoS Pathog 6: e1000714 Gallagher S.K., P. Witkovsky, M.J. Roux, M.J. Low, V. Otero-Corchon, S.T. Hentges, J. Vigh. b-endorphin in the mouse retina. J Comp Neurol 2010 Aug 518(15):3130-48. Genet, C., A. Strehle, C. Schmidt, G. Boudjelal, A. Lobstein, K. Schoonjans, M. Souchet, J. Auwerx, R. Saladin, and A. Wagner, Structure-activity relationship study of betulinic acid, a novel and selective TGR5 agonist, and its synthetic derivatives: potential impact in diabetes: J Med Chem, v. 53, p. 178-90. Germain M, De Arcangelis A, Robinson SD, Baker M, Tavora B, D’Amico G, Silva R, Kostourou V, Reynolds LE, Watson A, Jones JL, Georges-Labouesse E, Hodivala-Dilke K (2010) Genetic ablation of the alpha 6-integrin subunit in Tie1Cre mice enhances tumour angiogenesis. J Pathol 220: 370-81 Gilliver SC, Emmerson E, Campbell L, Chambon P, Hardman MJ, Ashcroft GS (2010) 17Beta-estradiol inhibits wound healing in male mice via estrogen receptoralpha. Am J Pathol 176: 2707-21 Guilini C, Urayama K, Turkeri G, Dedeoglu DB, Kurose H, Messaddeq N, Nebigil CG (2010) Divergent roles of prokineticin receptors in the endothelial cells: angiogenesis and fenestration. Am J Physiol Heart Circ Physiol 298: H844-52 Hacke K, Rincon-Orozco B, Buchwalter G, Siehler SY, Wasylyk B, Wiesmuller L, Rosl F (2010) Regulation of MCP-1 chemokine transcription by p53. Mol Cancer 9: 82 Halter B, Gonzalez de Aguilar JL, Rene F, Petri S, Fricker B, Echaniz-Laguna A, Dupuis L, Larmet Y, Loeffler JP. Oxidative stress in skeletal muscle stimulates early expression of Rad in a mouse model of amyotrophic lateral sclerosis. Free Radic Biol Med. 2010 Apr 1;48(7):915-23. Epub 2010 Jan 14.PMID: 20079427 [PubMed in process]Related citations Heitzler P. (2010) Biodiversity and noncanonical Notch signaling. In «Notch signaling». CTDB 92, 427-451 (R. Kopan, Ed). Elsevier Inc., San Diego. Hikake T, Hayashi S, Chambon P, Watanabe H, Iguchi T, Sato T. Differential involvement of insulin-like growth factor-I and estrogen on prolactin cells in the mouse anterior pituitary. Exp Biol Med (Maywood). 2010 Jun 24. Host L, Anglard P, Romieu P, Thibault C, Dembele D, Aunis D, Zwiller J (2010) Inhibition of histone deacetylases in rats self-administering cocaine regulates lissencephaly gene-1 and reelin gene expression, as revealed by microarray technique. J Neurochem 113: 236-47 117 publications Until June 2010 Hussenet T, Dali S, Exinger J, Monga B, Jost B, Dembelé D, Martinet N, Thibault C, Huelsken J, Brambilla E, du Manoir S. SOX2 is an oncogene activated by recurrent 3q26.3 amplifications in human lung squamous cell carcinomas. PLoS One. 2010 Jan 29;5(1):e8960. Hussenet,T., D. Dembele, N. Martinet, J. M. Vignaud, and S. du Manoir, 2010, An adult tissue-specific stem cell molecular phenotype is activated in epithelial cancer stem cells and correlated to patient outcome: Cell Cycle, v. 9, p. 321-7. Jenner LB, Demeshkina N, Yusupova G, Yusupov M (2010) Structural aspects of messenger RNA reading frame maintenance by the ribosome. Nat Struct Mol Biol 17: 555-60 Jimenez-Lara, A. M., A. Aranda, and H. Gronemeyer, 2010, Retinoic acid protects human breast cancer cells against etoposide-induced apoptosis by NFkappaB-dependent but cIAP2-independent mechanisms: Mol Cancer, v. 9, p. 15. Joseph A, Hess RA, Schaeffer DJ, Ko C, Hudgin-Spivey S, Chambon P, Shur BD (2010) Absence of estrogen receptor alpha leads to physiological alterations in the mouse epididymis and consequent defects in sperm function. Biol Reprod 82: 948-57 Joseph A, Shur BD, Ko C, Chambon P, Hess RA (2010) Epididymal hypo-osmolality induces abnormal sperm morphology and function in the estrogen receptor alpha knockout mouse. Biol Reprod 82: 958-67 Jung AC, Briolat J, Millon R, de Reynies A, Rickman D, Thomas E, Abecassis J, Clavel C, Wasylyk B (2010) Biological and clinical relevance of transcriptionally active human papillomavirus (HPV) infection in oropharynx squamous cell carcinoma. Int J Cancer 126: 1882-94 D. E. Kainov, V. Cura, M. Vitorino, H. Nierengarten, P. Poussin, B. Kieffer, J. Cavarelli and A. Poterszman, Structure determination of the minimal complex between Tfb5 and Tfb2, two subunits of the yeast transcription/DNA-repair factor TFIIH: a retrospective study, Acta Cryst. (2010) D66, 745-755. Kainov DE, Selth LA, Svejstrup JQ, Egly JM, Poterzsman A (2010) Interacting partners of the Tfb2 subunit from yeast TFIIH. DNA Repair (Amst) 9: 33-39 Klink A, Lancelot E, Ballet S, Vucic E, Fabre JE, Gonzalez W, Medina C, Corot C, Mulder WJ, Mallat Z, Fayad ZA (2010) Magnetic resonance molecular imaging of thrombosis in an arachidonic Acid mouse model using an activated platelet targeted probe. Arterioscler Thromb Vasc Biol 30: 403-10 Kobi D, Steunou AL, Dembele D, Legras S, Larue L, Nieto L, Davidson I (2010) Genome-wide analysis of POU3F2/BRN2 promoter occupancy in human melanoma cells reveals Kitl as a novel regulated target gene. Pigment Cell Melanoma Res 23: 404-18 Koch M, Diebold ML, Cavarelli J, Romier C (2010) Crystallization and preliminary crystallographic analysis of eukaryotic transcription and mRNA export factor Iws1 from Encephalitozoon cuniculi. Acta Crystallogr Sect F Struct Biol Cryst Commun 66: 207-10 Kolotuev, I., Y. Schwab, and M. Labouesse, 2010, A precise and rapid mapping protocol for correlative light and electron microscopy of small invertebrate organisms: Biol Cell, v. 102, p. 121-32. Krebs A. R., Demmers J., Karmodiya K., Chang N.C., Chang A.C. and Tora L. (2010) ATAC and Mediator coactivators form a stable complex and regulate a set of non-coding RNA genes. EMBO Reports, Jul;11(7):541-7. A. Krzyzosiak, M. Szyszka-Niagolov, M. Wietrzych, S. Gobaille, S. Muramatsu, W. Krezel. Retinoid X Receptor Gamma Control of Affective Behaviors Involves Dopanimergic Signaling in Mice. Neuron 66, 908-920, 24 june 2010 Lahmar I, Guinard M, Sauer A, Marcellin L, Abdelrahman T, Roux M, Mousli M, Moussa A, Babba H, Pfaff AW, Candolfi E (2010) Murine neonatal infection provides an efficient model for congenital ocular toxoplasmosis. Exp Parasitol 124: 190-6 Landles C, Sathasivam K, Weiss A, Woodman B, Moffitt H, Finkbeiner S, Sun B, Gafni J, Ellerby LM, Trottier Y, Richards WG, Osmand A, Paganetti P, Bates GP (2010) Proteolysis of mutant huntingtin produces an exon 1 fragment that accumulates as an aggregated protein in neuronal nuclei in huntington disease. J Biol Chem 285: 8808-23 Kalogeropoulou M, Voulgari A, Kostourou V, Sandaltzopoulos R, Dikstein R, Davidson I, Tora L, Pintzas A (2010) TAF4b and Jun/ activating protein-1 collaborate to regulate the expression of integrin alpha6 and cancer cell migration properties. Mol Cancer Res 8: 554-68 Lapillonne H, Kobari L, Mazurier C, Tropel P, Giarratana MC, Zanella-Cleon I, Kiger L, Wattenhofer-Donzé M, Puccio H, Hebert N, Francina A, Andreu G, Viville S and Douay L (2010) Red blood cells generation from human induced pluripotent stem cells: perspectives for transfusion medicine. Haematologica, In press Kastner P, Chan S, Vogel WK, Zhang LJ, Topark-Ngarm A, Golonzhka O, Jost B, Le Gras S, Gross MK, Leid M. Bcl11b represses a mature T-cell gene expression program in immature CD4(+)CD8(+) thymocytes. Eur J Immunol. 2010 Jun 11. Le May N, Mota-Fernandes D, VelezCruz R, Iltis I, Biard D, Egly JM (2010) NER factors are recruited to active promoters and facilitate chromatin modification for transcription in the absence of exogenous genotoxic attack. Mol Cell 38: 54-66 Kemmel V, Klein C, Dembele D, Jost B, Taleb O, Aunis D, Mensah-Nyagan AG, Maitre M. A single acute pharmacological dose of gamma-hydroxybutyrate (GHB) modifies multiple gene expression patterns in rat hippocampus and frontal cortex. Physiol Genomics. 2010 Jan 26. Li G, Margueron R, Ku M, Chambon P, Bernstein BE, Reinberg D (2010) Jarid2 and PRC2, partners in regulating gene expression. Genes Dev 24: 368-80 Kerr N, Pintzas A, Holmes F, Hobson SA, Pope R, Wallace M, Wasylyk C, Wasylyk B, Wynick D (2010) The expression of ELK transcription factors in adult DRG: Novel isoforms, antisense transcripts and upregulation by nerve damage. Mol Cell Neurosci 44: 165-77 Kizilyaprak C, Spehner D, Devys D, Schultz P (2010) In vivo chromatin organization of mouse rod photoreceptors correlates with histone modifications. PLoS One 5: e11039 Marcais A, Jeannet R, Hernandez L, Soulier J, Sigaux F, Chan S, Kastner P (2010) Genetic inactivation of Ikaros is a rare event in human T-ALL. Leuk Res 34: 426-429 Marchetti G, Escuin S, van der Flier A, De Arcangelis A, Hynes RO, GeorgesLabouesse E (2010) Integrin alpha5beta1 is necessary for regulation of radial migration of cortical neurons during mouse brain development. Eur J Neurosci 31: 399-409 Marsaud V, Tchakarska G, Andrieux G, Liu JM, Dembele D, Jost B, Wdzieczak-Bakala J, Renoir JM, Sola B (2010) Cyclin K and cyclin D1b are oncogenic in myeloma cells. Mol Cancer 9: 103 Marty C, Lacout C, Martin A, Hasan S, Jacquot S, Birling MC, Vainchenker W, Villeval. Myeloproliferative neoplasm induced by constitutive expression of JAK2V617F in knock-in mice. Mavrakis M, Pourquie O, Lecuit T (2010) Lighting up developmental mechanisms: how fluorescence imaging heralded a new era. Development 137: 373-87 McClelland V, Cullup T, Bodi I, Ruddy D, BujBello A, Biancalana V, Boehm J, Bitoun M, Miller O, Jan W, Menson E, Amaya L, Trounce J, Laporte J, Mohammed S, Sewry C, Raiman J, Jungbluth H (2010) Vici syndrome associated with sensorineural hearing loss and evidence of neuromuscular involvement on muscle biopsy. Am J Med Genet A 152A: 741-7 Melberg A, Kretz C, Kalimo H, WallgrenPettersson C, Toussaint A, Bohm J, Stalberg E, Laporte J (2010) Adult course in dynamin 2 dominant centronuclear myopathy with neonatal onset. Neuromuscul Disord 20: 53-56 Mellitzer G, Beucher A, Lobstein V, Michel P, Robine S, Kedinger M, Gradwohl G (2010) Loss of enteroendocrine cells in mice alters lipid absorption and glucose homeostasis and impairs postnatal survival. J Clin Invest 120: 1708-21 Meng X, Leyva ML, Jenny M, Gross I, Benosman S, Fricker B, Harlepp S, Hébraud P, Boos A, Wlosik P, Bischoff P, Sirlin C, Pfeffer M, Loeffler JP, Gaiddon C. A ruthenium-containing organometallic compound reduces tumor growth through induction of the endoplasmic reticulum stress gene CHOP. Cancer Res. 2009 Jul 1;69(13):5458-66. Mikesch JH, Gronemeyer H, So CW (2010) Discovery of Novel Transcriptional and Epigenetic Targets in APL by Global ChIP Analyses: Emerging Opportunity and Challenge. Cancer Cell 17: 112-114 Morgan H, Beck T, Blake A, Gates H, Adams N, Debouzy G, Leblanc S, Lengger C, Maier H, Melvin D, Meziane H, Richardson D, Wells S, White J, Wood J; EUMODIC Consortium, de Angelis MH, Brown SD, Hancock JM, Mallon AM & collaborators. EuroPhenome: a repository for high-throughput mouse phenotyping data. Nucleic Acids Res, 2010, 38 (Database issue): D577-85 Lin SC, Dolle P, Ryckebusch L, Noseda M, Zaffran S, Schneider MD, Niederreither K (2010) Endogenous retinoic acid regulates cardiac progenitor differentiation. Proc Natl Acad Sci U S A 107: 9234-9 Livolsi A, Niederhoffer N, Dali-Youcef N, Rambaud C, Olexa C, Mokni W, Gies JP, Bousquet P (2010) Cardiac muscarinic receptor overexpression in sudden infant death syndrome. PLoS One 5: e9464 Morsut L, Yan KP, Enzo E, Aragona M, Soligo SM, Wendling O, Mark M, Khetchoumian K, Bressan G, Chambon P, Dupont S, Losson R, Piccolo S. Development. 2010 Jun 23. Negative control of Smad activity by ectodermin/ Tif1{gamma} patterns the mammalian embryo. Development. 2010 Jun 23. Müller F., Zaucker A. and Tora L. (2010) Developmental regulation of transcription initiation: more than just changing the actors. Current Opinion in Genetics & Development, 20; In press. Losson R, Nielsen AL (2010) The NIZP1 KRAB and C2HR domains cross-talk for transcriptional regulation. Biochim Biophys Acta 1799: 463-468 Muller J, Creevey CJ, Thompson JD, Arendt D, Bork P (2010) AQUA: automated quality improvement for multiple sequence alignments. Bioinformatics 26: 263-5 Muller J, Stoetzel C, Vincent MC, Leitch CC, Laurier V, Danse JM, Helle S, Marion V, Bennouna-Greene V, Vicaire S, Megarbane A, Kaplan J, Drouin-Garraud V, Hamdani M, Sigaudy S, Francannet C, Roume J, Bitoun P, Goldenberg A, Philip N, Odent S, Green J, Cossee M, Davis EE, Katsanis N, Bonneau D, Verloes A, Poch O, Mandel JL, Dollfus H (2010) Identification of 28 novel mutations in the Bardet-Biedl syndrome genes: the burden of private mutations in an extensively heterogeneous disease. Hum Genet 127: 583-93 Muller-Steffner H, Kuhn I, Argentini M, Schuber F (2010) Identification of the Nglycosylation sites on recombinant bovine CD38 expressed in Pichia pastoris: their impact on enzyme stability and catalytic activity. Protein Expr Purif 70: 151-7 Nagy, Z., A. Riss, S. Fujiyama, A. Krebs, M. Orpinell, P. Jansen, A. Cohen, H. G. Stunnenberg, S. Kato, and L.Tora, 2010, The metazoan ATAC and SAGA coactivator HAT complexes regulate different sets of inducible target genes: Cell Mol Life Sci, v. 67, p. 611-28. Nakade K., Wasylyk B., Yokoyama K.K. (2010) Epigenetic regulation of p16Ink4a and Arf by JDP2 in cellular senescence Niculescu C, Ganduli-Indra G, Pfister V, Dupé V, Messadeq N, De arcangelis A and Georges-Labouesse E (2010) Conditional ablation of intergrin alpha-6 in mouse epidermis leads to sjin fragility and inflammation. European J. Cell Biology Oksenych, V., and F. Coin, 2010, The long unwinding road: XPB and XPD helicases in damaged DNA opening: Cell Cycle, v. 9, p. 90-6. Orpinell M., Fournier M., Riss A., Nagy Z., Krebs A.R. and Tora L. (2010) The ATAC acetyl transferase complex controls mitotic progression by targeting non-histone substrates. EMBO Journal, 2010 Jun 18. Ozbudak EM, Tassy O, Pourquie O. (2010) Spatiotemporal compartmentalization of key physiological processes during muscle precursor differentiation. Proc Natl Acad Sci U S A 107: 4224-9 Pankotai T., Popescu C., Martin D, Grau B., Zsindely N., Bodai L., Tora L., Ferrús A. and Boros I. (2010) Genes of the ecdysone biosynthesis pathway are regulated by the ATAC histone acetyl transferase complex in Drosophila. Mol. Cell Biol, 2010 Jun 28. Papai G, Tripathi MK, Ruhlmann C, Layer JH, Weil PA, Schultz P (2010) TFIIA and the transactivator Rap1 cooperate to commit TFIID for transcription initiation. Nature 465: 956-60 Pavet V, Beyrath J, Pardin C, Morizot A, Lechner MC, Briand JP, Wendland M, Maison W, Fournel S, Micheau O, Guichard G, Gronemeyer H (2010) Multivalent DR5 Peptides Activate the TRAIL Death Pathway and Exert Tumoricidal Activity. Cancer Res 70: 1101-10 Pavlowsky A, Gianfelice A, Pallotto M, Zanchi A, Vara H, Khelfaoui M, Valnegri P, Rezai X, Bassani S, Brambilla D, Kumpost J, Blahos J, Roux MJ, Humeau Y, Chelly J, Passafaro M, Giustetto M, Billuart P, Sala C (2010) A postsynaptic signaling pathway that may account for the cognitive defect due to IL1RAPL1 mutation. Curr Biol 20: 103-15 Pelaez IM, Kalogeropoulou M, Ferraro A, Voulgari A, Pankotai T, Boros I, Pintzas A (2010) Oncogenic RAS alters the global and gene-specific histone modification pattern during epithelial-mesenchymal transition in colorectal carcinoma cells. Int J Biochem Cell Biol 42: 911-20 publications Until June 2010 Petit-Haertlein I, Blakeley MP, Howard E, Hazemann I, Mitschler A, Podjarny A, Haertlein M (2010) Incorporation of methyl-protonated valine and leucine residues into deuterated ocean pout type III antifreeze protein: expression, crystallization and preliminary neutron diffraction studies. Acta Crystallogr Sect F Struct Biol Cryst Commun 66: 665-9 Sharif T, Auger C, Alhosin M, Ebel C, Achour M, Etienne-Selloum N, Fuhrmann G, Bronner C, Schini-Kerth VB (2010) Red wine polyphenols cause growth inhibition and apoptosis in acute lymphoblastic leukaemia cells by inducing a redox-sensitive up-regulation of p73 and down-regulation of UHRF1. Eur J Cancer 46: 983-94 Pradeau-Aubreton K, Ruff M, Garnier JM, Schultz P, Drillien R. (2010) Vectors for recombinational cloning and gene expression in mammalian cells using modified vaccinia virus Ankara. Anal Biochem. 404, 103-105 Shimura E, Hozumi N, Kanagawa O, Metzger D, Chambon P, Radtke F, Hirose S, Nakano N (2010) Epidermal gammadelta T cells sense precancerous cellular dysregulation and initiate immune responses. Int Immunol 22: 329-40 Procter JB, Thompson J, Letunic I, Creevey C, Jossinet F, Barton GJ (2010) Visualization of multiple alignments, phylogenies and gene family evolution. Nat Methods 7: S16-25 Qiu, J. J., X. Lu, B. B. Zeisig, Z. Ma, X. Cai, S. Chen, H. Gronemeyer, D. J. Tweardy, C. W. So, and S. Dong, 2010, Leukemic transformation by the APL fusion protein PRKAR1ARAR{alpha} critically depends on recruitment of RXR{alpha}: Blood, v. 115, p. 643-52. Reichman, S., R. K. Kalathur, S. Lambard, N. Ait-Ali, Y. Yang, A. Lardenois, R. Ripp, O. Poch, D. J. Zack, J. A. Sahel, and T. Leveillard, 2010, The homeobox gene CHX10/VSX2 regulates RdCVF promoter activity in the inner retina: Hum Mol Genet, v. 19, p. 250-61. Rochel N, Hourai S, Moras D. Crystal structure of hereditary vitamin D-resistant rickets-Associated mutant H305Q of vitamin D nuclear receptor bound to its natural ligand. J Steroid Biochem Mol Biol. 2010 Apr 18 Rocher-Ros V, Marco S, Mao JH, Gines S, Metzger D, Chambon P, Balmain A, Saura CA (2010) Presenilin modulates EGFR signaling and cell transformation by regulating the ubiquitin ligase Fbw7. Oncogene 29: 2950-61 Rosselot, C., L. Spraggon, I. Chia, E. Batourina, P. Riccio, B. Lu, K. Niederreither, P. Dolle, G. Duester, P. Chambon, F. Costantini, T. Gilbert, A. Molotkov, and C. Mendelsohn, 2010, Non-cell-autonomous retinoid signaling is crucial for renal development: Development, v. 137, p. 283-92. Saadaoui I, Al-Thani R, al-Saadi F, Belguith Ben hassan N, Abdelkefi-Mesrati L, Schultz P, Rouis S and Jaoua S (2010) Characterization of Tunisian Bacillus thuringiensis strains with abundance of kurstaki subspecies harbouring insecticidal activities against the lepidopteran inscet Ephestia kuehniella. Current Microbiology Schmucker S, Puccio H (2010) Understanding the molecular mechanisms of Friedreich’s ataxia to develop therapeutic approaches. Hum Mol Genet 19: R103-10 Sellier C, Rau F, Liu Y, Tassone F, Hukema RK, Gattoni R, Schneider A, Richard S, Willemsen R, Elliott DJ, Hagerman PJ, Charlet-Berguerand N (2010) Sam68 sequestration and partial loss of function are associated with splicing alterations in FXTAS patients. EMBO J 29: 1248-61 Sene A, R Tadayoni, T Pannicke, A Wurm, B El Mathari, R Benard, MJ Roux, D Yaffe, D Mornet, A Reichenbach, JA Sahel, A Rendon. Functional implication of Dp71 in osmoregulation and vascular permeability of the retina (2009). PLoS One Oct 7;4(10):e7329 Shuaib M, Ouararhni K, Dimitrov S, Hamiche A (2010) HJURP binds CENP-A via a highly conserved N-terminal domain and mediates its deposition at centromeres. Proc Natl Acad Sci U S A 107: 1349-54 Serchov T, Dubois-Pot-Schneider H, Charlot C, Roesl F, Wasylyk B. J Biol Chem. 2010 Apr 28. [Epub ahead of print]PMID: 20427288 [PubMed - as supplied by publisher] Involvement of Net and HIF1alpha in distinct yet intricately linked hypoxia induced signalling pathways. Soyer, J., L. Flasse, W. Raffelsberger, A. Beucher, C. Orvain, B. Peers, P. Ravassard, J. Vermot, M. L. Voz, G. Mellitzer, and G. Gradwohl, 2010, Rfx6 is an Ngn3-dependent winged helix transcription factor required for pancreatic islet cell development: Development, v. 137, p. 203-12. Spiegelhalter C, Tosch V, Hentsch D, Koch M, Kessler P, Schwab Y, Laporte J (2010) From Dynamic Live Cell Imaging to 3D Ultrastructure: Novel Integrated Methods for High Pressure Freezing and Correlative LightElectron Microscopy. PLoS One 5: e9014 Tora L, Timmers HT (2010) The TATA box regulates TATA-binding protein (TBP) dynamics in vivo. Trends Biochem Sci 35: 309-314 Timmers HT, Tora L (2010) The spectacular landscape of chromatin and ncRNAs under the Tico sunlight. EMBO Rep 11: 147-9 Tora L. and Timmers H. Th. M. (2010) The TATA-box regulates TBP dynamics in vivo. TIBS. 35, 309-314. Tosch V, Vasli N, Kretz C, Nicot AS, Gasnier C, Dondaine N, Oriot D, Barth M, Puissant H, Romero NB, Bönnemann CG, Heller B, Duval G, Biancalana V, Laporte J. Novel molecular diagnostic approaches for X-linked centronuclear (myotubular) myopathy reveal intronic mutations. Neuromuscul Disord. 2010 Jun;20(6):375-81. Trivella A, Gaillard T, Stote RH, Hellwig P (2010) Far infrared spectra of solid state aliphatic amino acids in different protonation states. J Chem Phys 132: 115105 Tropel P, Tournois J, Come J, Varela C, Moutou C, Fragner P, Cailleret M, Laabi Y, Peschanski M, Viville S (2010) High-efficiency derivation of human embryonic stem cell lines following pre-implantation genetic diagnosis. In Vitro Cell Dev Biol Anim 46: 376-385 Tosco A, Monti MC, Fontanella B, Montefusco S, D’Andrea L, Ziaco B, Baldantoni D, Rio MC, Marzullo L (2010) Copper binds the carboxy-terminus of trefoil protein 1 (TFF1), favoring its homodimerization and motogenic activity. Cell Mol Life Sci 67: 1943-55 Vegeto, E., S. Cuzzocrea, C. Crisafulli, E. Mazzon, A. Sala, A. Krust, and A. Maggi, 2010, Estrogen receptor-alpha as a drug target candidate for preventing lung inflammation: Endocrinology, v. 151, p. 174-84. Vilhais-Neto GC, Maruhashi M, Smith KT, Vasseur-Cognet M, Peterson AS, Workman JL, Pourquie O (2010) Rere controls retinoic acid signalling and somite bilateral symmetry. Nature 463: 953-7 Virely C, Moulin S, Cobaleda C, Lasgi C, Alberdi A, Soulier J, Sigaux F, Chan S, Kastner P, Ghysdael J (2010) Haploinsufficiency of the IKZF1 (IKAROS) tumor suppressor gene cooperates with BCR-ABL in a transgenic model of acute lymphoblastic leukemia. Leukemia 24: 1200 Walter A, Etienne-Selloum N, Brasse D, Khallouf H, Bronner C, Rio MC, Beretz A, Schini-Kerth VB. Intake of grape-derived polyphenols reduces C26 tumor growth by inhibiting angiogenesis and inducing apoptosis. FASEB J. 2010, In press Warther D, Bolze F, Leonard J, Gug S, Specht A, Puliti D, Sun XH, Kessler P, Lutz Y, Vonesch JL, Winsor B, Nicoud JF, Goeldner M (2010) Live-Cell One- and Two-Photon Uncaging of a Far-Red Emitting Acridinone Fluorophore. J Am Chem Soc 132: 2585-2590 Wasylyk C, Zambrano A, Zhao C, Brants J, Abecassis J, Schalken JA, Rogatsch H, Schaefer G, Pycha A, Klocker H, Wasylyk B. Int J Cancer. 2010 Feb 16. [Epub ahead of print]PMID: 20162578 [PubMed - as supplied by publisher] Tubulin tyrosine ligase like 12, link to prostate cancer through tubulin post-translational modification and chromosome ploidy. Wicker N (2010) Perfect sampling algorithm for small mxn contingency tables. STATISTICS AND COMPUTING 20: 57-61 Wilhelm E, Kornete M, Targat B, VigneaultEdwards J, Frontini M, Tora L, Benecke A, Bell B (2010) TAF6delta orchestrates an apoptotic transcriptome profile and interacts functionally with p53. BMC Mol Biol 11: 10 Yefimova MG, Messaddeq N, Karam A, Jacquard C, Weber C, Jonet L, Wolfrum U, Jeanny JC, Trottier Y. Polyglutamine toxicity induces rod photoreceptor division, morphological transformation and death in Spinocerebellar ataxia 7 mouse retina. Neurobiology of Disease (2010), In press Yoo JH, Bailey A, Ansonoff M, Pintar JE, Matifas A, Kieffer BL, Kitchen I (2010) Lack of genotype effect on D1, D2 receptors and dopamine transporter binding in triple MOP-, DOP-, and KOP-opioid receptor knockout mice of three different genetic backgrounds. Synapse 64: 520-527 Zahreddine H, Zhang H, Diogon M, Nagamatsu Y, Labouesse M (2010) CRT-1/ Calreticulin and the E3 Ligase EEL-1/HUWE1 Control Hemidesmosome Maturation in C. elegans Development. Curr Biol 20: 322-327 Zanier K, Ruhlmann C, Melin F, Masson M, Ould M’hamed Ould Sidi A, Bernard X, Fischer B, Brino L, Ristriani T, Rybin V, Baltzinger M, Vande Pol S, Hellwig P, Schultz P, Trave G (2010) E6 proteins from diverse papillomaviruses self-associate both in vitro and in vivo. J Mol Biol 396: 90-104 Zhang H, Labouesse M (2010) The making of hemidesmosome structures in vivo. Dev Dyn 239: 1465-76 Zhang, S.O., Mathur, S., Hattem, G., Tassy O., Pourquie, O., Sex-dimorphic gene expression and ineffective dosage compensation of Z-linked genes in gastrulating chicken embryos. BMC Genomics 11, 13 (2010). Zhang X, Oulad-Abdelghani M, Zelkin AN, Wang Y, Haikel Y, Mainard D, Voegel JC, Caruso F, Benkirane-Jessel N (2010) Poly(L-lysine) nanostructured particles for gene delivery and hormone stimulation. Biomaterials 31: 1699-706 Zhovmer A, Oksenych V, Coin F (2010) Two sides of the same coin: TFIIH complexes in transcription and DNA repair. ScientificWorldJournal 10: 633-43 Zullo A, Iaconis D, Barra A, Cantone A, Messaddeq N, Capasso G, Dollé P, Igarashi P, Franco B. Kidney-specific inactivation of Ofd1 leads to renal cystic disease associated with upregulation of the mTOR pathway. Hum Mol Genet. 2010 May 17. Pereira PM, Schneider A, Pannetier S, Heron D, Hanauer A (2010) Coffin-Lowry syndrome. Eur J Hum Genet 18: 627-33 119 publications 2009 Abdulrahman, W., M. Uhring, I. KolbCheynel, J. M. Garnier, D. Moras, N. Rochel, D. Busso, and A. Poterszman, 2009, A set of baculovirus transfer vectors for screening of affinity tags and parallel expression strategies: Anal Biochem, v. 385, p. 383-5. Afonine, P. V., R. W. Grosse-Kunstleve, A. Urzhumtsev, and P. D. Adams, 2009, Automatic multiple-zone rigid-body refinement with a large convergence radius: J Appl Crystallogr, v. 42, p. 607-615. Albou, L. P., B. Schwarz, O. Poch, J. M. Wurtz, and D. Moras, 2009, Defining and characterizing protein surface using alpha shapes: Proteins, v. 76, p. 1-12. Alpy, F., F. Legueux, L. Bianchetti, and C. Tomasetto, 2009, [START domain-containing proteins: a review of their role in lipid transport and exchange.]: Med Sci (Paris), v. 25, p. 181-191. Al-Qusairi, L., N. Weiss, A.Toussaint, C. Berbey, N. Messaddeq, C. Kretz, D. Sanoudou, A. H. Beggs, B. Allard, J. L. Mandel, J. Laporte, V. Jacquemond, and A. Buj-Bello, 2009, T-tubule disorganization and defective excitation-contraction coupling in muscle fibers lacking myotubularin lipid phosphatase: Proc Natl Acad Sci U S A, v. 106, p. 18763-8. Alvarez, S., H. Khanwalkar, R. Alvarez, C. Erb, C. Martinez, F. Rodriguez-Barrios, P. Germain, H. Gronemeyer, and A. R. de Lera, 2009, C3 halogen and c8’’ substituents on stilbene arotinoids modulate retinoic Acid receptor subtype function: ChemMedChem, v. 4, p. 1630-40. Alvarez, S., R. Alvarez, H. Khanwalkar, P. Germain, G. Lemaire, F. Rodriguez-Barrios, H. Gronemeyer, and A. R. de Lera, 2009, Retinoid receptor subtype-selective modulators through synthetic modifications of RARgamma agonists: Bioorg Med Chem, v. 17, p. 4345-59. Anheim, M., B. Monga, M. Fleury, P. Charles, C. Barbot, M. Salih, J. P. Delaunoy, M. Fritsch, L. Arning, M. Synofzik, L. Schols, J. Sequeiros, C. Goizet, C. Marelli, I. Le Ber, J. Koht, J. Gazulla, J. De Bleecker, M. Mukhtar, N. Drouot, L. Ali-Pacha, T. Benhassine, M. Chbicheb, A. M’Zahem, A. Hamri, B. Chabrol, J. Pouget, R. Murphy, M. Watanabe, P. Coutinho, M. Tazir, A. Durr, A. Brice, C.Tranchant, and M. Koenig, 2009, Ataxia with oculomotor apraxia type 2: clinical, biological and genotype/phenotype correlation study of a cohort of 90 patients: Brain, v. 132, p. 2688-98. Anheim, M., C. Lagier-Tourenne, G. Stevanin, M. Fleury, A. Durr, I. J. Namer, P. Denora, A. Brice, J. L. Mandel, M. Koenig, and C. Tranchant, 2009, SPG11 spastic paraplegia : A new cause of juvenile parkinsonism: J Neurol, v. 256, p. 104-8. Aniba, M. R., S. Siguenza, A. Friedrich, F. Plewniak, O. Poch, A. Marchler-Bauer, and J. D.Thompson, 2009, Knowledge-based expert systems and a proof-of-concept case study for multiple sequence alignment construction and analysis: Brief Bioinform, v. 10, p. 11-23. Argmann, C., R. Dobrin, S. Heikkinen, A. Auburtin, L. Pouilly, T. A. Cock, H. Koutnikova, J. Zhu, E. E. Schadt, and J. Auwerx, 2009, Ppargamma2 is a key driver of longevity in the mouse: PLoS Genet, v. 5, p. e1000752. Atanassov, B. S., Y. A. Evrard, A. S. Multani, Z. Zhang, L.Tora, D. Devys, S. Chang, and S. Y. Dent, 2009, Gcn5 and SAGA regulate shelterin protein turnover and telomere maintenance: Mol Cell, v. 35, p. 352-64. Aulehla, A., and O. Pourquie, 2009, More than patterning--Hox genes and the control of posterior axial elongation: Dev Cell, v. 17, p. 439-40. Balamotis, M. A., M. A. Pennella, J. L. Stevens, B. Wasylyk, A. S. Belmont, and A. J. Berk, 2009, Complexity in transcription control at the activation domain-mediator interface: Sci Signal, v. 2, p. ra20. Balayssac, S., S. Trefi, V. Gilard, M. MaletMartino, R. Martino, and M. A. Delsuc, 2009, 2D and 3D DOSY 1H NMR, a useful tool for analysis of complex mixtures: application to herbal drugs or dietary supplements for erectile dysfunction: J Pharm Biomed Anal, v. 50, p. 602-12. Beau-Faller, M., M. Legrain, A. C. Voegeli, E. Guerin, T. Lavaux, A. M. Ruppert, A. Neuville, G. Massard, J. M. Wihlm, E. Quoix, P. Oudet, and M. P. Gaub, 2009, Detection of K-Ras mutations in tumour samples of patients with non-small cell lung cancer using PNA-mediated PCR clamping: Br J Cancer, v. 100, p. 985-92. Bechara, E. G., M. C. Didiot, M. Melko, L. Davidovic, M. Bensaid, P. Martin, M. Castets, P. Pognonec, E. W. Khandjian, H. Moine, and B. Bardoni, 2009, A Novel Function for Fragile X Mental Retardation Protein in Translational Activation: PLoS Biol, v. 7, p. e16. Beck,Y., C. Delaporte, D. Moras, G. Richards, and I. M. Billas, 2009, The ligand-binding domains of the three RXR-USP nuclear receptor types support distinct tissue and ligand specific hormonal responses in transgenic Drosophila: Dev Biol, v. 330, p. 1-11. Berdasco, M., S. Ropero, F. Setien, M. F. Fraga, P. Lapunzina, R. Losson, M. Alaminos, N. K. Cheung, N. Rahman, and M. Esteller, 2009, Epigenetic inactivation of the Sotos overgrowth syndrome gene histone methyltransferase NSD1 in human neuroblastoma and glioma: Proc Natl Acad Sci U S A, v. 106, p. 21830-5. Bieniossek, C., Y. Nie, D. Frey, N. Olieric, C. Schaffitzel, I. Collinson, C. Romier, P. Berger, T. J. Richmond, M. O. Steinmetz, and I. Berger, 2009, Automated unrestricted multigene recombineering for multiprotein complex production: Nat Methods, v. 6, p. 447-450. Bigliardi, P. L., D. J. Tobin, C. GaveriauxRuff, and M. Bigliardi-Qi, 2009, Opioids and the skin--where do we stand?: Exp Dermatol, v. 18, p. 424-30. Billon-Gales, A., C. Fontaine, C. Filipe, V. Douin-Echinard, M. J. Fouque, G. Flouriot, P. Gourdy, F. Lenfant, H. Laurell, A. Krust, P. Chambon, and J. F. Arnal, 2009, The transactivating function 1 of estrogen receptor {alpha} is dispensable for the vasculoprotective actions of 17{beta}-estradiol: Proc Natl Acad Sci U S A, v. 106, p. 2053-2058. Birling, M. C., F. Gofflot, and X. Warot, 2009, Site-specific recombinases for manipulation of the mouse genome: Methods Mol Biol, v. 561, p. 245-63. Biryukova, I., J. Asmar, H. Abdesselem, and P. Heitzler, 2009, Drosophila mir-9a regulates wing development via fine-tuning expression of the LIM only factor, dLMO: Dev Biol. Bonuccelli, G., M. C. Casimiro, F. Sotgia, C. Wang, M. Liu, S. Katiyar, J. Zhou, E. Dew, F. Capozza, K. M. Daumer, C. Minetti, J. N. Milliman, F. Alpy, M. C. Rio, C.Tomasetto, I. Mercier, N. Flomenberg, P. G. Frank, R. G. Pestell, and M. P. Lisanti, 2009, Caveolin-1 (P132L), a Common Breast Cancer Mutation, Confers Mammary Cell Invasiveness and Defines a Novel Stem Cell/MetastasisAssociated Gene Signature: Am J Pathol, v. 174, p. 1650-62. Bouffard, C., S. Viville, and B. M. Knoppers, 2009, Genetic diagnosis of embryos: clear explanation, not rhetoric, is needed: Cmaj, v. 181, p. 387-91. publications 2009 Bourgo, R. J., H. Siddiqui, S. Fox, D. Solomon, C. G. Sansam, M. Yaniv, C. Muchardt, D. Metzger, P. Chambon, C. W. Roberts, and E. S. Knudsen, 2009, SWI/SNF deficiency results in aberrant chromatin organization, mitotic failure, and diminished proliferative capacity: Mol Biol Cell, v. 20, p. 3192-9. Chorro, L., A. Sarde, M. Li, K. J. Woollard, P. Chambon, B. Malissen, A. Kissenpfennig, J. B. Barbaroux, R. Groves, and F. Geissmann, 2009, Langerhans cell (LC) proliferation mediates neonatal development, homeostasis, and inflammation-associated expansion of the epidermal LC network: J Exp Med, v. 206, p. 3089-100. Doisne, J. M., L. Bartholin, K. P. Yan, C. N. Garcia, N. Duarte, J. B. Le Luduec, D. Vincent, F. Cyprian, B. Horvat, S. Martel, R. Rimokh, R. Losson, K. Benlagha, and J. C. Marie, 2009, iNKT cell development is orchestrated by different branches of TGF-beta signaling: J Exp Med, v. 206, p. 1365-78. Bovet, C., B. Plet, M. Ruff, S. Eiler, F. Granger, A. Panagiotidis, R. Wenzel, A. Nazabal, D. Moras, and R. Zenobi, 2009, Towards high-throughput identification of endocrine disrupting compounds with mass spectrometry: Toxicol In Vitro, v. 23, p. 704-9. Ciarletta, P., M. Ben Amar, and M. Labouesse, 2009, Continuum model of epithelial morphogenesis during Caenorhabditis elegans embryonic elongation: Philos Transact A Math Phys Eng Sci, v. 367, p. 3379-400. Dolle, P., 2009, Developmental expression of retinoic acid receptors (RARs): Nucl Recept Signal, v. 7, p. e006. Bruck, N., D. Vitoux, C. Ferry, V. Duong, A. Bauer, H. de The, and C. Rochette-Egly, 2009, A coordinated phosphorylation cascade initiated by p38MAPK/MSK1 directs RARalpha to target promoters: Embo J, v. 28, p. 34-47. Buehler, A., L. Urzhumtseva, V. Y. Lunin, and A. Urzhumtsev, 2009, Cluster analysis for phasing with molecular replacement: a feasibility study: Acta Crystallogr D Biol Crystallogr, v. 65, p. 644-50. Cai, Q., A. Dierich, M. Oulad-Abdelghani, S. Chan, and P. Kastner, 2009, Helios deficiency has minimal impact on T cell development and function: J Immunol, v. 183, p. 2303-11. Calmels, N., H. Seznec, P. Villa, L. Reutenauer, M. Hibert, J. Haiech, P. Rustin, M. Koenig, and H. Puccio, 2009, Limitations in a frataxin knockdown cell model for Friedreich ataxia in a high-throughput drug screen: BMC Neurol, v. 9, p. 46. Calmels, N., S. Schmucker, M. Wattenhofer-Donze, A. Martelli, N. Vaucamps, L. Reutenauer, N. Messaddeq, C. Bouton, M. Koenig, and H. Puccio, 2009, The first cellular models based on frataxin missense mutations that reproduce spontaneously the defects associated with Friedreich ataxia: PLoS One, v. 4, p. e6379. Canto, C., Z. Gerhart-Hines, J. N. Feige, M. Lagouge, L. Noriega, J. C. Milne, P. J. Elliott, P. Puigserver, and J. Auwerx, 2009, AMPK regulates energy expenditure by modulating NAD(+) metabolism and SIRT1 activity: Nature, v. 458, p. 1056-1060. Catena, R., E. Escoffier, C. Caron, S. Khochbin, I. Martianov, and I. Davidson, 2009, HMGB4, a Novel Member of the HMGB Family, Is Preferentially Expressed in the Mouse Testis and Localizes to the Basal Pole of Elongating Spermatids: Biol Reprod, v. 80, p. 358-66. J. Cavarelli. Détermination des structures 3D des macromolécules biologiques par diffraction des rayons X, I, Techniques de l’Ingénieur, Traité Analyse chimique et Caractérisation, (2009), P1 1110, 1-11. J. Cavarelli. Détermination des structures 3D des macromolécules biologiques par diffraction des rayons X, II, Techniques de l’Ingénieur, Traité Analyse chimique et Caractérisation, (2009), P1 1111, 1-19. Chang, L. Y., A. M. Mir, C.Thisse, Y. Guerardel, P. Delannoy, B.Thisse, and A. HarduinLepers, 2009, Molecular cloning and characterization of the expression pattern of the zebrafish alpha2, 8-sialyltransferases (ST8Sia) in the developing nervous system: Glycoconj J, v. 26, p. 263-75. Chavey, C., G. Lazennec, S. Lagarrigue, C. Clape, I. Iankova, J. Teyssier, J. S. Annicotte, J. Schmidt, C. Mataki, H. Yamamoto, R. Sanches, A. Guma, V. Stich, M. Vitkova, B. Jardin-Watelet, E. Renard, R. Strieter, A. Tuthill, G. S. Hotamisligil, A. Vidal-Puig, A. Zorzano, D. Langin, and L. Fajas, 2009, CXC ligand 5 is an adipose-tissue derived factor that links obesity to insulin resistance: Cell Metab, v. 9, p. 339-49. Ciaudo, C., N. Servant, V. Cognat, A. Sarazin, E. Kieffer, S. Viville, V. Colot, E. Barillot, E. Heard, and O. Voinnet, 2009, Highly dynamic and sex-specific expression of microRNAs during early ES cell differentiation: PLoS Genet, v. 5, p. e1000620. Cignarella, A., C. Bolego, V. Pelosi, C. Meda, A. Krust, C. Pinna, R. M. Gaion, E. Vegeto, and A. Maggi, 2009, Distinct roles of estrogen receptor-alpha and beta in the modulation of vascular inducible nitric-oxide synthase in diabetes: J Pharmacol Exp Ther, v. 328, p. 174-82. Cler, E., G. Papai, P. Schultz, and I. Davidson, 2009, Recent advances in understanding the structure and function of general transcription factor TFIID: Cell Mol Life Sci, v. 66, p. 2123-34. Cossee, M., C. Lagier-Tourenne, C. Seguela, M. Mohr, F. Leturcq, H. Gundesli, J. Chelly, C. Tranchant, M. Koenig, and J. L. Mandel, 2009, Use of SNP array analysis to identify a novel TRIM32 mutation in limb-girdle muscular dystrophy type 2H: Neuromuscul Disord, v. 19, p. 255-60. Cybulski, N., P. Polak, J. Auwerx, M. A. Ruegg, and M. N. Hall, 2009, mTOR complex 2 in adipose tissue negatively controls whole-body growth: Proc Natl Acad Sci U S A, v. 106, p. 9902-7. Dammak, M., S. Tounsi, D. B. Hamadou, L. Abdelkafi, P. Schultz, and S. Jaoua, 2009, Restoration of the crystallization of altered delta-endotoxins Cry1Ac, by the promotion of their in vivo integration into the Bacillus thuringiensis native crystals: FEMS Microbiol Lett, v. 292, p. 268-73. Daujat, S., T. Weiss, F. Mohn, U. C. Lange, C. Ziegler-Birling, U. Zeissler, M. Lappe, D. Schubeler, M. E.Torres-Padilla, and R. Schneider, 2009, H3K64 trimethylation marks heterochromatin and is dynamically remodeled during developmental reprogramming: Nat Struct Mol Biol, v. 16, p. 777-81. Delfosse, V., E. Girard, C. Birck, M. Delmarcelle, M. Delarue, O. Poch, P. Schultz, and C. Mayer, 2009, Structure of the archaeal pab87 peptidase reveals a novel self-compartmentalizing protease family: PLoS ONE, v. 4, p. e4712. Didiot, M. C., M. Subramanian, E. Flatter, J. L. Mandel, and H. Moine, 2009, Cells lacking the fragile X mental retardation protein (FMRP) have normal RISC activity but exhibit altered stress granule assembly: Mol Biol Cell, v. 20, p. 428-37. Dieterich K, Zouari R, Harbuz R, Vialard F, Martinez D, Bellayou H, Prisant N, Zoghmar A, Guichaoua MR, Koscinski I, Kharouf M, Noruzinia M, Nadifi S, Sefiani A, Lornage J, Zahi M, Viville S, Sèle B, Jouk PS, Jacob MC, Escalier D, Nikas Y, Hennebicq S, Lunardi J, Ray PF. The Aurora Kinase C c.144delC mutation causes meiosis I arrest in men and is frequent in the North African population. Hum Mol Genet. 2009 Apr 1;18(7):1301-9. Epub 2009 Jan 15. El Ramy, R., N. Magroun, N. Messadecq, L. R. Gauthier, F. D. Boussin, U. KolthurSeetharam, V. Schreiber, M. W. McBurney, P. Sassone-Corsi, and F. Dantzer, 2009, Functional interplay between Parp-1 and SirT1 in genome integrity and chromatinbased processes: Cell Mol Life Sci, v. 66, p. 3219-34. Fauny, J. D., B.Thisse, and C.Thisse, 2009, The entire zebrafish blastula-gastrula margin acts as an organizer dependent on the ratio of Nodal to BMP activity: Development, v. 136, p. 3811-9. Fechter, P., C. Chevalier, G.Yusupova, M. Yusupov, P. Romby, and S. Marzi, 2009, Ribosomal initiation complexes probed by toeprinting and effect of trans-acting translational regulators in bacteria: Methods Mol Biol, v. 540, p. 247-63. Ferry, C., M. Gianni, S. Lalevee, N. Bruck, J. L. Plassat, I. Raska, Jr., E. Garattini, and C. Rochette-Egly, 2009, SUG-1 Plays Proteolytic and Non-proteolytic Roles in the Control of Retinoic Acid Target Genes via Its Interaction with SRC-3: J Biol Chem, v. 284, p. 8127-35. Fischer, M., P. M. Pereira, B. Holtmann, C. M. Simon, A. Hanauer, M. Heisenberg, and M. Sendtner, 2009, P90 Ribosomal s6 kinase 2 negatively regulates axon growth in motoneurons: Mol Cell Neurosci, v. 42, p. 134-41. Floquet, S., S. Brun, J. F. Lemonnier, M. Henry, M. A. Delsuc, Y. Prigent, E. Cadot, and F. Taulelle, 2009, Molecular weights of cyclic and hollow clusters measured by DOSY NMR spectroscopy: J Am Chem Soc, v. 131, p. 17254-9. Fournier, B., E. Bendeif el, B. Guillot, A. Podjarny, C. Lecomte, and C. Jelsch, 2009, Charge density and electrostatic interactions of fidarestat, an inhibitor of human aldose reductase: J Am Chem Soc, v. 131, p. 10929-41. Fridlich, R., F. Delalande, C. Jaillard, J. Lu, L. Poidevin, T. Cronin, L. Perrocheau, G. Millet-Puel, M. L. Niepon, O. Poch, A. Holmgren, A. Van Dorsselaer, J. A. Sahel, and T. Leveillard, 2009, The Thioredoxin-like Protein Rod-derived Cone Viability Factor (RdCVFL) Interacts with TAU and Inhibits Its Phosphorylation in the Retina: Mol Cell Proteomics, v. 8, p. 1206-18. Gaillard, T., A. Dejaegere, and R. H. Stote, 2009, Dynamics of beta3 integrin I-like and hybrid domains: insight from simulations on the mechanism of transition between open and closed forms: Proteins, v. 76, p. 977-94. Gallien, S., E. Perrodou, C. Carapito, C. Deshayes, J. M. Reyrat, A. Van Dorsselaer, O. Poch, C. Schaeffer, and O. Lecompte, 2009, Ortho-proteogenomics: multiple proteomes investigation through orthology and a new MS-based protocol: Genome Res, v. 19, p. 128-35. Gally, C., F. Wissler, H. Zahreddine, S. Quintin, F. Landmann, and M. Labouesse, 2009, Myosin II regulation during C. elegans embryonic elongation: LET-502/ROCK, MRCK-1 and PAK-1, three kinases with different roles: Development, v. 136, p. 3109-19. Ganguli-Indra, G., C. Wasylyk, X. Liang, R. Millon, M. Leid, B. Wasylyk, J. Abecassis, and A. Indra, 2009, CTIP2 expression in human head and neck squamous cell carcinoma is linked to poorly differentiated tumor status: PLoS ONE, v. 4, p. e5367. Ganguli-Indra, G., C. Wasylyk, X. Liang, R. Millon, M. Leid, B. Wasylyk, J. Abecassis, and A. Indra, 2009, CTIP2 expression in human head and neck squamous cell carcinoma is linked to poorly differentiated tumor status: PLoS ONE, v. 4, p. e5367. Galluzzi, L., S. A. Aaronson, J. Abrams, E. S. Alnemri, D. W. Andrews, E. H. Baehrecke, N. G. Bazan, M. V. Blagosklonny, K. Blomgren, C. Borner, D. E. Bredesen, C. Brenner, M. Castedo, J. A. Cidlowski, A. Ciechanover, G. M. Cohen, V. De Laurenzi, R. De Maria, M. Deshmukh, B. D. Dynlacht, W. S. El-Deiry, R. A. Flavell, S. Fulda, C. Garrido, P. Golstein, M. L. Gougeon, D. R. Green, H. Gronemeyer, G. Hajnoczky, J. M. Hardwick, M. O. Hengartner, H. Ichijo, M. Jaattela, O. Kepp, A. Kimchi, D. J. Klionsky, R. A. Knight, S. Kornbluth, S. Kumar, B. Levine, S. A. Lipton, E. Lugli, F. Madeo, W. Malomi, J. C. Marine, S. J. Martin, J. P. Medema, P. Mehlen, G. Melino, U. M. Moll, E. Morselli, S. Nagata, D. W. Nicholson, P. Nicotera, G. Nunez, M. Oren, J. Penninger, S. Pervaiz, M. E. Peter, M. Piacentini, J. H. Prehn, H. Puthalakath, G. A. Rabinovich, R. Rizzuto, C. M. Rodrigues, D. C. Rubinsztein, T. Rudel, L. Scorrano, H. U. Simon, H. Steller, J. Tschopp, Y. Tsujimoto, P. Vandenabeele, I. Vitale, K. H. Vousden, R. J. Youle, J. Yuan, B. Zhivotovsky, and G. Kroemer, 2009, Guidelines for the use and interpretation of assays for monitoring cell death in higher eukaryotes: Cell Death Differ, v. 16, p. 1093-107. Garcel, A., J. Perino, J. M. Crance, R. Drillien, D. Garin, and A. L. Favier, 2009, Phenotypic and genetic diversity of the traditional Lister smallpox vaccine: Vaccine, v. 27, p. 708-17. Garcia, J., H. Khanwalkar, R. Pereira, C. Erb, J. J. Voegel, P. Collette, P. Mauvais, W. Bourguet, H. Gronemeyer, and A. R. de Lera, 2009, Pyrazine Arotinoids with Inverse Agonist Activities on the Retinoid and Rexinoid Receptors: Chembiochem, v. 10, p. 1252-1259. Gas, M.-E. and Séraphin, B. Twins take the job. EMBO J. (2010) 29, in press (Editorial) Gazdag, E., A. Santenard, C. ZieglerBirling, G. Altobelli, O. Poch, L.Tora, and M. E.Torres-Padilla, 2009, TBP2 is essential for germ cell development by regulating transcription and chromatin condensation in the oocyte: Genes Dev, v. 23, p. 2210-23. Gazit, K., S. Moshonov, R. Elfakess, M. Sharon, G. Mengus, I. Davidson, and R. Dikstein, 2009, TAF4/4b x TAF12 displays a unique mode of DNA binding and is required for core promoter function of a subset of genes: J Biol Chem, v. 284, p. 26286-96. Gazulla, J., I. Benavente, I. P. Lopez-Fraile, P. Modrego, and M. Koenig, 2009, Sensorimotor neuronopathy in ataxia with oculomotor apraxia type 2: Muscle Nerve, v. 40, p. 481-5. Germain, P., C. Gaudon, V. Pogenberg, S. Sanglier, A. Van Dorsselaer, C. A. Royer, M. A. Lazar, W. Bourguet, and H. Gronemeyer, 2009, Differential action on coregulator interaction defines inverse retinoid agonists and neutral antagonists: Chem Biol, v. 16, p. 479-89. B. K. Bay, M. K. Gross, C. Kioussi, and M. Leid, 2009, Ctip2/Bcl11b controls ameloblast formation during mammalian odontogenesis: Proc Natl Acad Sci U S A, v. 106, p. 4278-83. Ho, M. S., H. Chen, M. Chen, C. Jacques, A. Giangrande, and C. T. Chien, 2009, Gcm protein degradation suppresses proliferation of glial progenitors: Proc Natl Acad Sci U S A, v. 106, p. 6778-83. Gianni, M., A. Boldetti, V. Guarnaccia, A. Rambaldi, E. Parrella, I. Raska, Jr., C. Rochette-Egly, G. Del Sal, A. Rustighi, M. Terao, and E. Garattini, 2009, Inhibition of the peptidyl-prolyl-isomerase Pin1 enhances the responses of acute myeloid leukemia cells to retinoic acid via stabilization of RARalpha and PML-RARalpha: Cancer Res, v. 69, p. 1016-26. Hoque, M. M., S. Shimizu, E. C. Juan, Y. Sato, M. T. Hossain, T. Yamamoto, S. Imamura, K. Suzuki, H. Amano, T. Sekiguchi, M. Tsunoda, and A. Takenaka, 2009, Structure of D-3-hydroxybutyrate dehydrogenase prepared in the presence of the substrate D-3-hydroxybutyrate and NAD+: Acta Crystallogr Sect F Struct Biol Cryst Commun, v. 65, p. 331-5. Goula, A. V., B. R. Berquist, D. M. Wilson, 3rd, V. C. Wheeler, Y.Trottier, and K. Merienne, 2009, Stoichiometry of base excision repair proteins correlates with increased somatic CAG instability in striatum over cerebellum In Huntington’s disease transgenic mice: PLoS Genet, v. 5, p. e1000749. Iwema,T., A. Chaumot, R. A. Studer, M. Robinson-Rechavi, I. M. Billas, D. Moras, V. Laudet, and F. Bonneton, 2009, Structural and evolutionary innovation of the heterodimerization interface between USP and the ecdysone receptor ECR in insects: Mol Biol Evol, v. 26, p. 753-68. Goeldner, C., D. Reiss, J. Wichmann, B. L. Kieffer, and A. M. Ouagazzal, 2009, Activation of nociceptin opioid peptide (NOP) receptor impairs contextual fear learning in mice through glutamatergic mechanisms: Neurobiol Learn Mem, v. 91, p. 393-401. O. Golonzhka, D. Metzger, J.-M. Bornert, B. K. Bay, M. K. Gross, C. Kioussi, and M. Leid. 2009,Ctip2/Bcl11b controls ameloblast formation during mammalian odontogenesis. Proc. Natl. Acad. Sci. (USA) 106 : 4278-4283. Golonzhka, O., D. Metzger, J. M. Bornert, Golonzhka, O., X. Liang, N. Messaddeq, J. M. Bornert, A. L. Campbell, D. Metzger, P. Chambon, G. Ganguli-Indra, M. Leid, and A. K. Indra, 2009, Dual role of COUP-TFinteracting protein 2 in epidermal homeostasis and permeability barrier formation: J Invest Dermatol, v. 129, p. 1459-70. Gronemeyer, H., and A. Zelent, 2009, Fingering modulators of retinoic acid signaling identifies new prognostic marker for neuroblastoma: Cancer Cell, v. 15, p. 249-51. Gronemeyer, H., and W. Bourguet, 2009, Allosteric effects govern nuclear receptor action: DNA appears as a player: Sci Signal, v. 2, p. pe34. Guillon, B., A. L. Bulteau, M. WattenhoferDonze, S. Schmucker, B. Friguet, H. Puccio, J. C. Drapier, and C. Bouton, 2009, Frataxin deficiency causes upregulation of mitochondrial Lon and ClpP proteases and severe loss of mitochondrial Fe-S proteins: Febs J, v. 276, p. 1036-47. Hajjar, E., A. Dejaegere, and N. Reuter, 2009, Challenges in pKa predictions for proteins: the case of Asp213 in human proteinase 3: J Phys Chem A, v. 113, p. 11783-92. Hashimoto, S., and J. M. Egly, 2009, Trichothiodystrophy view from the molecular basis of DNA repair/transcription factor TFIIH: Hum Mol Genet, v. 18, p. R224-30. Heikkinen, S., C. Argmann, J. N. Feige, H. Koutnikova, M. F. Champy, N. Dali-Youcef, E. E. Schadt, M. Laakso, and J. Auwerx, 2009, The Pro12Ala PPARgamma2 Variant Determines Metabolism at the Gene-Environment Interface: Cell Metab, v. 9, p. 88-98. Henri, J., Rispal, D., Bayart, E., van Tilbeurgh, H., Séraphin, B.and Graille, M. Structural and functional insights into S. cerevisiae Tpa1, a putative prolyl hydroxylase influencing translation termination and transcription. J. Biol. Chem. (2010) 285, in press. Jacques, C., L. Soustelle, I. Nagy, C. Diebold, and A. Giangrande, 2009, A novel role of the glial fate determinant glial cells missing in hematopoiesis: Int J Dev Biol, v. 53, p. 1013-22. Jobert, L., M. Argentini, and L.Tora, 2009, PRMT1 mediated methylation of TAF15 is required for its positive gene regulatory function: Exp Cell Res, v. 315, p. 1273-86. Jobert, L., N. Pinzon, E. Van Herreweghe, B. E. Jady, A. Guialis, T. Kiss, and L.Tora, 2009, Human U1 snRNA forms a new chromatin-associated snRNP with TAF15: EMBO Rep, v. 10, p. 494-500. Jungbluth H, Wallgren-Pettersson C, Laporte JF, Centronuclear (Myotubular) Myopathy Consortium. 164th ENMC International workshop: 6th workshop on centronuclear (myotubular) myopathies, 16-18th January 2009, Naarden, The Netherlands.Neuromuscul Disord. 2009 Oct;19(10):721-9. Kadkhodaei, B., T. Ito, E. Joodmardi, B. Mattsson, C. Rouillard, M. Carta, S. Muramatsu, C. Sumi-Ichinose, T. Nomura, D. Metzger, P. Chambon, E. Lindqvist, N. G. Larsson, L. Olson, A. Bjorklund, H. Ichinose, and T. Perlmann, 2009, Nurr1 is required for maintenance of maturing and adult midbrain dopamine neurons: J Neurosci, v. 29, p. 15923-32. Kang, S., S. Elf, S. Dong, T. Hitosugi, K. Lythgoe, A. Guo, H. Ruan, S. Lonial, H. J. Khoury, I. R. Williams, B. H. Lee, J. L. Roesel, G. Karsenty, A. Hanauer, J. Taunton, T. J. Boggon, T. L. Gu, and J. Chen, 2009, Fibroblast growth factor receptor 3 associates with and tyrosine phosphorylates p90 RSK2, leading to RSK2 activation that mediates hematopoietic transformation: Mol Cell Biol, v. 29, p. 2105-17. Kang-Park, M. H., B. L. Kieffer, A. J. Roberts, M. Roberto, S. G. Madamba, G. R. Siggins, and S. D. Moore, 2009, {micro}Opioid Receptors Selectively Regulate Basal Inhibitory Transmission in the Central Amygdala: Lack of Ethanol Interactions: J Pharmacol Exp Ther, v. 328, p. 284-93. Khiar, N., M. P. Leal, R. Baati, C. Ruhlmann, C. Mioskowski, P. Schultz, and I. Fernandez, 2009, Tailoring carbon nanotube surfaces with glyconanorings: new bionanomaterials with specific lectin affinity: Chem Commun (Camb), p. 4121-3. Kieffer, B. L., and C. J. Evans, 2009, Opioid receptors: from binding sites to visible molecules in vivo: Neuropharmacology, v. 56 Suppl 1, p. 205-12. Kim, H., S. Hayashi, P. Chambon, H. Watanabe, T. Iguchi, and T. Sato, 2009, Effects of diethylstilbestrol on ovarian follicle development in neonatal mice: Reprod Toxicol, v. 27, p. 55-62. 121 publications 2009 Kirigaya, A., H. Kim, S. Hayashi, P. Chambon, H. Watanabe, T. Lguchi, and T. Sato, 2009, Involvement of estrogen receptor Beta in the induction of polyovular follicles in mouse ovaries exposed neonatally to diethylstilbestrol: Zoolog Sci, v. 26, p. 704-12. Liu, Y., C. F. Bourgeois, S. Pang, M. Kudla, N. Dreumont, L. Kister, Y. H. Sun, J. Stevenin, and D. J. Elliott, 2009, The germ cell nuclear proteins hnRNP G-T and RBMY activate a testis-specific exon: PLoS Genet, v. 5, p. e1000707. Kolotuev, I., A. Apaydin, and M. Labouesse, 2009, Secretion of Hedgehog-related peptides and WNT during Caenorhabditis elegans development: Traffic, v. 10, p. 803-10. Maclean, G., P. Dolle, and M. Petkovich, 2009, Genetic disruption of CYP26B1 severely affects development of neural crest derived head structures, but does not compromise hindbrain patterning: Dev Dyn, v. 238, p. 732-45. Kolthur-Seetharam, U., K.Teerds, D. G. Rooij, O. Wendling, M. McBurney, P. Sassone-Corsi, and I. Davidson, 2009, The Histone Deacetylase SIRT1 Controls Male Fertility in Mice Through Regulation of Hypothalamic-Pituitary Gonadotropin Signaling: Biol Reprod, v. 80, p. 384-91. Madan, B., V. Madan, O. Weber, P. Tropel, C. Blum, E. Kieffer, S. Viville, and H. J. Fehling, 2009, The pluripotency-associated gene Dppa4 is dispensable for embryonic stem cell identity and germ cell development but essential for embryogenesis: Mol Cell Biol, v. 29, p. 3186-203. Krebs, A., and L.Tora, 2009, Keys to open chromatin for transcription activation: FACT and Asf1: Mol Cell, v. 34, p. 397-9. Madan, V., B. Madan, U. Brykczynska, F. Zilbermann, K. Hogeveen, K. Dohner, H. Dohner, O. Weber, C. Blum, H. R. Rodewald, P. Sassone-Corsi, A. H. Peters, and H. J. Fehling, 2009, Impaired function of primitive hematopoietic cells in mice lacking the Mixed-Lineage-Leukemia homolog MLL5: Blood, v. 113, p. 1444-54. Labouesse, M., and L. Solnica-Krezel, 2009, Shaping embryos in Barcelona: Nat Cell Biol, v. 11, p. 3-6. Lamour, V., L. F. Westblade, E. A. Campbell, and S. A. Darst, 2009, Crystal structure of the in vivo-assembled Bacillus subtilis Spx/RNA polymerase alpha subunit C-terminal domain complex: J Struct Biol, v. 168, p. 352-6. Le Merrer, J., J. A. Becker, K. Befort, and B. L. Kieffer, 2009, Reward processing by the opioid system in the brain: Physiol Rev, v. 89, p. 1379-412. Leclerc, E. A., A. Huchenq, N. R. Mattiuzzo, D. Metzger, P. Chambon, N. B. Ghyselinck, G. Serre, N. Jonca, and M. Guerrin, 2009, Corneodesmosin gene ablation induces lethal skin-barrier disruption and hair-follicle degeneration related to desmosome dysfunction: J Cell Sci, v. 122, p. 2699-709. Lee, S., D. W. Kang, S. Hudgins-Spivey, A. Krust, E. Y. Lee, Y. Koo, Y. Cheon, M. C. Gye, P. Chambon, and C. Ko, 2009, Theca-specific estrogen receptor-alpha knockout mice lose fertility prematurely: Endocrinology, v. 150, p. 3855-62. Lehraiki, A., C. Racine, A. Krust, R. Habert, and C. Levacher, 2009, Phthalates impair germ cell number in the mouse fetal testis by an androgen- and estrogen-independent mechanism: Toxicol Sci, v. 111, p. 372-82. Levenga, J., R. A. Buijsen, M. Rife, H. Moine, D. L. Nelson, B. A. Oostra, R. Willemsen, and F. M. de Vrij, 2009, Ultrastructural analysis of the functional domains in FMRP using primary hippocampal mouse neurons: Neurobiol Dis, v. 35, p. 241-50. Levy, N., M. Oehlmann, F. Delalande, H. P. Nasheuer, A. Van Dorsselaer, V. Schreiber, G. de Murcia, J. Menissier-de Murcia, D. Maiorano, and A. Bresson, 2009, XRCC1 interacts with the p58 subunit of DNA Pol alpha-primase and may coordinate DNA repair and replication during S phase: Nucleic Acids Res, v. 37, p. 3177-88. Li, M., P. Hener, Z. Zhang, K. P. Ganti, D. Metzger, and P. Chambon, 2009, Induction of thymic stromal lymphopoietin expression in keratinocytes is necessary for generating an atopic dermatitis upon application of the active vitamin D3 analogue MC903 on mouse skin: J Invest Dermatol, v. 129, p. 498-502. Lippert, W. P., C. Burschka, K. Gotz, M. Kaupp, D. Ivanova, C. Gaudon, Y. Sato, P. Antony, N. Rochel, D. Moras, H. Gronemeyer, and R. Tacke, 2009, Silicon analogues of the RXR-selective retinoid agonist SR11237 (BMS649): chemistry and biology: ChemMedChem, v. 4, p. 1143-52. Manzo, F., A. Nebbioso, M. Miceli, M. Conte, F. De Bellis, V. Carafa, G. Franci, F. P. Tambaro, and L. Altucci, 2009, TNF-related apoptosis-inducing ligand: signalling of a ‘smart’ molecule: Int J Biochem Cell Biol, v. 41, p. 460-6. Marion, V., C. Stoetzel, D. Schlicht, N. Messaddeq, M. Koch, E. Flori, J. M. Danse, J. L. Mandel, and H. Dollfus, 2009, Transient ciliogenesis involving Bardet-Biedl syndrome proteins is a fundamental characteristic of adipogenic differentiation: Proc Natl Acad Sci U S A, v. 106, p. 1820-5. Mark, M., N. B. Ghyselinck, and P. Chambon, 2009, Function of retinoic acid receptors during embryonic development: Nucl Recept Signal, v. 7, p. e002. Mark, M., Ghyselinck, N.B. Effects of vitamin A on spermatogenesis (2009). mt Méd. Reprod., 11 (3), pp. 215-226. Marquis, R., C. Greco, P. Schultz, S. Meunier, and C. Mioskowski, 2009, Isothermal microcalorimetry, a tool for probing SWNT bundles: J Nanosci Nanotechnol, v. 9, p. 6777-82. Mascrez, B., N. B. Ghyselinck, P. Chambon, and M. Mark, 2009, A transcriptionally silent RXRalpha supports early embryonic morphogenesis and heart development: Proc Natl Acad Sci U S A, v. 106, p. 4272-7. Masiero, E., L. Agatea, C. Mammucari, B. Blaauw, E. Loro, M. Komatsu, D. Metzger, C. Reggiani, S. Schiaffino, and M. Sandri, 2009, Autophagy is required to maintain muscle mass: Cell Metab, v. 10, p. 507-15. McCrea, H. J., C. Kretz, J. Laporte, and L. R. Ment, 2009, Dementia in a child with myotubular myopathy: Pediatr Neurol, v. 40, p. 483-5. Merienne, K., and Y.Trottier, 2009, SCA8 CAG/CTG expansions, a tale of two TOXICities: a unique or common case?: PLoS Genet, v. 5, p. e1000593. Michel, F., C. Crucifix, F. Granger, S. Eiler, J. F. Mouscadet, S. Korolev, J. Agapkina, R. Ziganshin, M. Gottikh, A. Nazabal, S. Emiliani, R. Benarous, D. Moras, P. Schultz, and M. Ruff, 2009, Structural basis for HIV1 DNA integration in the human genome, role of the LEDGF/P75 cofactor: Embo J, v. 28, p. 980-91. Minoux, M., G. S. Antonarakis, M. Kmita, D. Duboule, and F. M. Rijli, 2009, Rostral and caudal pharyngeal arches share a common neural crest ground pattern: Development, v. 136, p. 637-45. Misteli, T., and Soutoglou E., 2009, The emerging role of nuclear architecture in DNA repair and genome maintenance: Nat Rev Mol Cell Biol, v. 10, p. 243-54. Modder, U. I., D. G. Monroe, D. G. Fraser, T. C. Spelsberg, C. J. Rosen, M. Gehin, P. Chambon, B. W. O’Malley, and S. Khosla, 2009, Skeletal consequences of deletion of steroid receptor coactivator-2/transcription intermediary factor-2: J Biol Chem, v. 284, p. 18767-77. Moras, D., 2009, Structure of full-length PPARgamma-RXRalpha: a snapshot of a functional complex?: Cell Metab, v. 9, p. 8-10. Moroy, G., E. Martin, A. Dejaegere, and R. H. Stote, 2009, Molecular basis for Bcl-2 homology 3 domain recognition in the Bcl2 protein family: identification of conserved hot spot interactions: J Biol Chem, v. 284, p. 17499-511. Müller F. and Tora L. (2009) TBP2: a specialized general transcription factor in female germ cells. Journal of Biology, 8:97, 1-4, Minireview. Myasnikov, A. G., A. Simonetti, S. Marzi, and B. P. Klaholz, 2009, Structure-function insights into prokaryotic and eukaryotic translation initiation: Curr Opin Struct Biol, v. 19, p. 300-9. Nagy, Z., A. Riss, C. Romier, X. le Guezennec, A. R. Dongre, M. Orpinell, J. Han, H. Stunnenberg, and L.Tora, 2009, The Human SPT20-Containing SAGA Complex Plays a Direct Role in the Regulation of Endoplasmic Reticulum Stress-Induced Genes: Mol Cell Biol, v. 29, p. 1649-60. Nagy, Z., and E. Soutoglou, 2009, DNA repair: easy to visualize, difficult to elucidate: Trends Cell Biol, v. 19, p. 617-29. Nakade, K., J. Pan, T. Yamasaki, T. Murata, B. Wasylyk, and K. K. Yokoyama, 2009, JDP2 (Jun Dimerization Protein 2)-deficient Mouse Embryonic Fibroblasts Are Resistant to Replicative Senescence: J Biol Chem, v. 284, p. 10808-10817. Nebbioso, A., F. Manzo, M. Miceli, M. Conte, L. Manente, A. Baldi, A. De Luca, D. Rotili, S. Valente, A. Mai, A. Usiello, H. Gronemeyer, and L. Altucci, 2009, Selective class II HDAC inhibitors impair myogenesis by modulating the stability and activity of HDAC-MEF2 complexes: EMBO Rep, v. 10, p. 776-82. Nicolet, C., E. Guerin, A. Neuville, J. P. Kerckaert, N. Wicker, E. Bergmann, C. Brigand, M. Kedinger, M. P. Gaub, and D. Guenot, 2009, Evidence for various 20q status using allelotyping, CGH arrays, and quantitative PCR in distal CIN colon cancers: Cancer Lett, v. 282, p. 195-204. Oda, H., I. Okamoto, N. Murphy, J. Chu, S. M. Price, M. M. Shen, M. E.TorresPadilla, E. Heard, and D. Reinberg, 2009, Monomethylation of histone H4-lysine 20 is involved in chromosome structure and stability and is essential for mouse development: Mol Cell Biol, v. 29, p. 2278-95. Ogata, T., J. Laporte, and M. Fukami, 2009, MAMLD1 (CXorf6): A New Gene Involved in Hypospadias: Horm Res, v. 71, p. 245-252. Ohe, K., K.T.Tamai, M. Parvinen, and P. Sassone-Corsi, 2009, DAX-1 and SOX6 molecular interplay results in an antagonistic effect in pre-mRNA splicing: Dev Dyn, v. 238, p. 1595-1604. Oksenych, V., B. B. de Jesus, A. Zhovmer, J. M. Egly, and F. Coin, 2009, Molecular insights into the recruitment of TFIIH to sites of DNA damage: Embo J, v. 28, p. 2971-80. publications 2009 Olieric, V., P. Wolff, A. Takeuchi, G. Bec, C. Birck, M. Vitorino, B. Kieffer, A. Beniaminov, G. Cavigiolio, E. Theil, C. Allmang, A. Krol, and P. Dumas, 2009, SECIS-binding protein 2, a key player in selenoprotein synthesis, is an intrinsically disordered protein: Biochimie, v. 91, p. 1003-9. Olmstead, M. C., A. M. Ouagazzal, and B. L. Kieffer, 2009, Mu and delta opioid receptors oppositely regulate motor impulsivity in the signaled nose poke task: PLoS ONE, v. 4, p. e4410. Pankotai,T., A. S. Hoffbeck, C. Boumendil, and E. Soutoglou, 2009, DNA damage response in the absence of DNA lesions continued: Cell Cycle, v. 8, p. 4025-6 Pannier, S., V. Couloigner, N. Messaddeq, M. Elmaleh-Berges, A. Munnich, R. Romand, and L. Legeai-Mallet, 2009, Activating Fgfr3 Y367C mutation causes hearing loss and inner ear defect in a mouse model of chondrodysplasia: Biochim Biophys Acta, v. 1792, p. 140-7. Papai, G., M. K. Tripathi, C. Ruhlmann, S. Werten, C. Crucifix, P. A. Weil, and P. Schultz, 2009, Mapping the Initiator Binding Taf2 Subunit in the Structure of Hydrated Yeast TFIID: Structure, v. 17, p. 363-73. Parry, D. A., A. J. Mighell, W. El-Sayed, R. C. Shore, I. K. Jalili, H. Dollfus, A. Bloch-Zupan, R. Carlos, I. M. Carr, L. M. Downey, K. M. Blain, D. C. Mansfield, M. Shahrabi, M. Heidari, P. Aref, M. Abbasi, M. Michaelides, A. T. Moore, J. Kirkham, and C. F. Inglehearn, 2009, Mutations in CNNM4 cause Jalili syndrome, consisting of autosomal-recessive cone-rod dystrophy and amelogenesis imperfecta: Am J Hum Genet, v. 84, p. 266-73. Pascal, C., F. Pate, V. Cheynier, and M. A. Delsuc, 2009, Study of the interactions between a proline-rich protein and a flavan-3-ol by NMR: Residual structures in the natively unfolded protein provides anchorage points for the ligands: Biopolymers, v. 91, p. 745-56. Patti, M. E., S. M. Houten, A. C. Bianco, R. Bernier, P. R. Larsen, J. J. Holst, M. K. Badman, E. Maratos-Flier, E. C. Mun, J. Pihlajamaki, J. Auwerx, and A. B. Goldfine, 2009, Serum bile acids are higher in humans with prior gastric bypass: potential contribution to improved glucose and lipid metabolism: Obesity (Silver Spring), v. 17, p. 1671-7. Pellicciari, R., A. Gioiello, A. Macchiarulo, C. Thomas, E. Rosatelli, B. Natalini, R. Sardella, M. Pruzanski, A. Roda, E. Pastorini, K. Schoonjans, and J. Auwerx, 2009, Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity: J Med Chem, v. 52, p. 7958-61. Percival, T., S. E. Aylett, F. Pool, A. Bloch-Zupan, G. J. Roberts, and V. S. Lucas, 2009, Oral health of children with intractable epilepsy attending the UK National Centre for Young People with Epilepsy: Eur Arch Paediatr Dent, v. 10, p. 19-24. Perez Santin, E., P. Germain, F. Quillard, H. Khanwalkar, F. Rodriguez-Barrios, H. Gronemeyer, A. R. de Lera, and W. Bourguet, 2009, Modulating retinoid X receptor with a series of (E)-3-[4-hydroxy3-(3-alkoxy-5,5,8,8-tetramethyl-5,6,7,8tetrahydronaphtha len-2-yl)phenyl]acrylic acids and their 4-alkoxy isomers: J Med Chem, v. 52, p. 3150-8. Petit-Haertlein, I., M. P. Blakeley, E. Howard, I. Hazemann, A. Mitschler, M. Haertlein, and A. Podjarny, 2009, Perdeuteration, purification, crystallization and preliminary neutron diffraction of an ocean pout type III antifreeze protein: Acta Crystallogr Sect F Struct Biol Cryst Commun, v. 65, p. 406-9. Petrova, T., V. Y. Lunin, S. Ginell, I. Hazemann, K. Lazarski, A. Mitschler, A. Podjarny, and A. Joachimiak, 2009, X-ray-radiation-induced cooperative atomic movements in protein: J Mol Biol, v. 387, p. 1092-105. Robert, I., F. Dantzer, and B. Reina-SanMartin, 2009, Parp1 facilitates alternative NHEJ, whereas Parp2 suppresses IgH/cmyc translocations during immunoglobulin class switch recombination: J Exp Med, v. 206, p. 1047-56. Pignon, J. C., B. Koopmansch, G. Nolens, L. Delacroix, D. Waltregny, and R. Winkler, 2009, Androgen receptor controls EGFR and ERBB2 gene expression at different levels in prostate cancer cell lines: Cancer Res, v. 69, p. 2941-9. Rochette-Egly, C., and P. Germain, 2009, Dynamic and combinatorial control of gene expression by nuclear retinoic acid receptors (RARs): Nucl Recept Signal, v. 7, p. e005. Poitras, P., and C.Tomasetto, 2009, The potential of ghrelin as a prokinetic: Regul Pept, v. 155, p. 24-7. Poulain, S., S. Lacomme, S. F. BattagliaHsu, S. du Manoir, L. Brochin, J. M. Vignaud, and N. Martinet, 2009, Signalling with retinoids in the human lung: validation of new tools for the expression study of retinoid receptors: BMC Cancer, v. 9, p. 423. Pradhan, A. A., J. A. Becker, G. Scherrer, P.Tryoen-Toth, D. Filliol, A. Matifas, D. Massotte, C. Gaveriaux-Ruff, and B. L. Kieffer, 2009, In vivo delta opioid receptor internalization controls behavioral effects of agonists: PLoS ONE, v. 4, p. e5425. Prosdocimi, F., B. Chisham, E. Pontelli, J. D. Thompson, and A. Stoltzfus, 2009, Initial implementation of a comparative data analysis ontology: Evol Bioinform Online, v. 5, p. 47-66. Puccio, H., 2009, Multicellular models of Friedreich ataxia: J Neurol, v. 256 Suppl 1, p. 18-24. Quenet, D., M. Mark, J. Govin, A. van Dorsselear, V. Schreiber, S. Khochbin, and F. Dantzer, 2009, Parp2 is required for the differentiation of post-meiotic germ cells: identification of a spermatid-specific complex containing Parp1, Parp2, TP2 and HSPA2: Exp Cell Res, v. 315, p. 2824-34. Rajan, P., C. Dalgliesh, C. F. Bourgeois, M. Heiner, K. Emami, E. L. Clark, A. Bindereif, J. Stevenin, C. N. Robson, H. Y. Leung, and D. J. Elliott, 2009, Proteomic identification of heterogeneous nuclear ribonucleoprotein L as a novel component of SLM/Sam68 Nuclear Bodies: BMC Cell Biol, v. 10, p. 82. Reibel, A., M. C. Maniere, F. Clauss, D. Droz, Y. Alembik, E. Mornet, and A. Bloch-Zupan, 2009, Orodental phenotype and genotype findings in all subtypes of hypophosphatasia: Orphanet J Rare Dis, v. 4, p. 6. Renaud, J. P., and M. A. Delsuc, 2009, Biophysical techniques for ligand screening and drug design: Curr Opin Pharmacol, v. 9, p. 622-8. Rhinn, M., K. Lun, R. Ahrendt, M. Geffarth, and M. Brand, 2009, Zebrafish gbx1 refines the midbrain-hindbrain boundary border and mediates the Wnt8 posteriorization signal: Neural Dev, v. 4, p. 12. Ribes, V., I. Le Roux, M. Rhinn, B. Schuhbaur, and P. Dolle, 2009, Early mouse caudal development relies on crosstalk between retinoic acid, Shh and Fgf signalling pathways: Development, v. 136, p. 665-76. Riclet, R., M. Chendeb, J. L. Vonesch, D. Koczan, H. J. Thiesen, R. Losson, and F. Cammas, 2009, Disruption of the Interaction between Transcriptional Intermediary Factor 1{beta} and Heterochromatin Protein 1 Leads to a Switch from DNA Hyper- to Hypomethylation and H3K9 to H3K27 Trimethylation on the MEST Promoter Correlating with Gene Reactivation: Mol Biol Cell, v. 20, p. 296-305. Rohde, H. M., H. Tronchere, B. Payrastre, and J. Laporte, 2009, Detection of myotubularin phosphatases activity on phosphoinositides in vitro and ex vivo: Methods Mol Biol, v. 462, p. 265-78. C. Romier, J.M. Wurtz, J.P. Renaud, J. Cavarelli. Structural biology of epigenetic targets in “Epigenetics Targets in Drug Discovery”,Editors W. Sippl and M. Jung, (2009), Wiley-VCH, 23-56. Ruano-Rubio, V., O. Poch, and J. D. Thompson, 2009, Comparison of eukaryotic phylogenetic profiling approaches using species tree aware methods: BMC Bioinformatics, v. 10, p. 383. Saliou, J. M., C. F. Bourgeois, L. A. Mena, D. Ropers, S. Jacquenet, V. Marchand, J. Stevenin, and C. Branlant, 2009, Role of RNA structure and protein factors in the control of HIV-1 splicing: Front Biosci, v. 14, p. 2714-29. Santenard, A., and M. E.Torres-Padilla, 2009, Epigenetic reprogramming in mammalian reproduction: contribution from histone variants: Epigenetics, v. 4, p. 80-4. Santin, E. P., H. Khanwalkar, J. Voegel, P. Collette, P. Mauvais, H. Gronemeyer, and A. R. de Lera, 2009, Highly Potent Naphthofuran-Based Retinoic Acid Receptor Agonists: ChemMedChem, v. 4, p. 780-791. Sarrazin, S., N. Mossadegh-Keller, T. Fukao, A. Aziz, F. Mourcin, L. Vanhille, L. Kelly Modis, P. Kastner, S. Chan, E. Duprez, C. Otto, and M. H. Sieweke, 2009, MafB restricts M-CSF-dependent myeloid commitment divisions of hematopoietic stem cells: Cell, v. 138, p. 300-13. Sayama K, Kajiya K, Sugawara K, Sato S, Hirakawa S, Shirakata Y, Hanakawa Y, Dai X, Ishimatsu-Tsuji Y, Metzger D, Chambon P, Akira S, Paus R, Kishimoto J, Hashimoto K. Inflammatory mediator TAK1 regulates hair follicle morphogenesis and anagen induction shown by using keratinocytespecific TAK1-deficient mice. PLoS One. 2010 Jun 23;5(6). Scherrer, G., N. Imamachi, Y. Q. Cao, C. Contet, F. Mennicken, D. O’Donnell, B. L. Kieffer, and A. I. Basbaum, 2009, Dissociation of the opioid receptor mechanisms that control mechanical and heat pain: Cell, v. 137, p. 1148-59. Schultz, P., C. Crucifix, and L. Lebeau, 2009, Two-dimensional crystallisation of soluble protein complexes: Methods Mol Biol, v. 543, p. 353-67. Sellars, M., B. Reina-San-Martin, P. Kastner, and S. Chan, 2009, Ikaros controls isotype selection during immunoglobulin class switch recombination: J Exp Med, v. 206, p. 1073-87. Sermon, K. D., C. Simon, P. Braude, S. Viville, J. Borstlap, and A. Veiga, 2009, Creation of a registry for human embryonic stem cells carrying an inherited defect: joint collaboration between ESHRE and hESCreg: Hum Reprod, v. 24, p. 1556-60. Shankaranarayanan, P., A. Rossin, H. Khanwalkar, S. Alvarez, R. Alvarez, A. Jacobson, A. Nebbioso, A. R. de Lera, L. Altucci, and H. Gronemeyer, 2009, Growth factor-antagonized rexinoid apoptosis involves permissive PPARgamma/ RXR heterodimers to activate the intrinsic death pathway by NO: Cancer Cell, v. 16, p. 220-31. Tocchini-Valentini, G. D., N. Rochel, H. Escriva, P. Germain, C. Peluso-Iltis, M. Paris, S. Sanglier-Cianferani, A. Van Dorsselaer, D. Moras, and V. Laudet, 2009, Structural and Functional Insights into the Ligand-binding Domain of a Nonduplicated Retinoid X Nuclear Receptor from the Invertebrate Chordate Amphioxus: J Biol Chem, v. 284, p. 1938-1948. Shimizu, S., E. C. Juan, Y. Sato, Y. Miyashita, M. M. Hoque, K. Suzuki, T. Sagara, M. Tsunoda, T. Sekiguchi, A. C. Dock-Bregeon, D. Moras, and A. Takenaka, 2009, Two complementary enzymes for threonylation of tRNA in crenarchaeota: crystal structure of Aeropyrum pernix threonyl-tRNA synthetase lacking a cis-editing domain: J Mol Biol, v. 394, p. 286-96. Toutain, C. E., L. Brouchet, I. RaymondLetron, P. Vicendo, H. Berges, J. Favre, M. J. Fouque, A. Krust, A. M. Schmitt, P. Chambon, P. Gourdy, J. F. Arnal, and F. Lenfant, 2009, Prevention of skin flap necrosis by estradiol involves reperfusion of a protected vascular network: Circ Res, v. 104, p. 245-54. Simonetti, A., S. Marzi, L. Jenner, A. Myasnikov, P. Romby, G.Yusupova, B. P. Klaholz, and M.Yusupov, 2009, A structural view of translation initiation in bacteria: Cell Mol Life Sci, v. 66, p. 423-36. Singh, J., W. Zeller, N. Zhou, G. Hategen, R. Mishra, A. Polozov, P. Yu, E. Onua, J. Zhang, D. Zembower, A. Kiselyov, J. L. Ramirez, G. Sigthorsson, J. M. Bjornsson, M. Thorsteinsdottir, T. Andresson, M. Bjarnadottir, O. Magnusson, J. E. Fabre, K. Stefansson, and M. E. Gurney, 2009, Antagonists of the EP(3) Receptor for Prostaglandin E(2) Are Novel Antiplatelet Agents That Do Not Prolong Bleeding: ACS Chem Biol, v. 4, p. 115-26. C. Stoetzel, S. Riehm, V. Bennouna Greene, V. Pelletier, J. Vigneron, B. Leheup, V. Marion, S. Helle, J. M. Danse, C. Thibault, L. Moulinier, F. Veillon, and H. Dollfus, 2009, Confirmation of TFAP2A gene involvement in branchio-oculo-facial syndrome (BOFS) and report of temporal bone anomalies: Am J Med Genet A, v. 149A, p. 2141-6. Tacke, R., V. Muller, M. W. Buttner, W. P. Lippert, R. Bertermann, J. O. Daiss, H. Khanwalkar, A. Furst, C. Gaudon, and H. Gronemeyer, 2009, Synthesis and pharmacological characterization of Disila-AM80 (Disila-tamibarotene) and Disila-AM580, silicon analogues of the RARalpha-selective retinoid agonists AM80 (Tamibarotene) and AM580: ChemMedChem, v. 4, p. 1797-802. Tazir, M., L. Ali-Pacha, A. M’Zahem, J. P. Delaunoy, M. Fritsch, S. Nouioua, T. Benhassine, S. Assami, D. Grid, J. M. Vallat, A. Hamri, and M. Koenig, 2009, Ataxia with oculomotor apraxia type 2: A clinical and genetic study of 19 patients: J Neurol Sci, v. 278, p. 77-81. Terrand, J., V. Bruban, L. Zhou, W. Gong, Z. El Asmar, P. May, K. Zurhove, P. Haffner, C. Philippe, E. Woldt, R. L. Matz, C. Gracia, D. Metzger, J. Auwerx, J. Herz, and P. Boucher, 2009, LRP1 Controls Intracellular Cholesterol Storage and Fatty Acid Synthesis through Modulation of Wnt Signaling: J Biol Chem, v. 284, p. 381-8. Thomas, C., A. Gioiello, L. Noriega, A. Strehle, J. Oury, G. Rizzo, A. Macchiarulo, H. Yamamoto, C. Mataki, M. Pruzanski, R. Pellicciari, J. Auwerx, and K. Schoonjans, 2009, TGR5-mediated bile acid sensing controls glucose homeostasis: Cell Metab, v. 10, p. 167-77. Thuaud F, Bernard Y, Türkeri G, Dirr R, Aubert G, Cresteil T, Baguet A, Tomasetto C, Svitkin Y, Sonenberg N, Nebigil CG, Désaubry L. Synthetic analogue of rocaglaol displays a potent and selective cytotoxicity in cancer cells: involvement of apoptosis inducing factor and caspase-12. J Med Chem. 2009 Aug 27;52(16):5176-87. Trompouki, E., A. Tsagaratou, S. K. Kosmidis, P. Dolle, J. Qian, D. L. Kontoyiannis, W. V. Cardoso, and G. Mosialos, 2009, Truncation of the catalytic domain of the cylindromatosis tumor suppressor impairs lung maturation: Neoplasia, v. 11, p. 469-76. Ueda,T., E. Compe, P. Catez, K. H. Kraemer, and J. M. Egly, 2009, Both XPD alleles contribute to the phenotype of compound heterozygote xeroderma pigmentosum patients: J Exp Med, v. 206, p. 3031-46. Urayama, K., D. B. Dedeoglu, C. Guilini, S. Frantz, G. Ertl, N. Messaddeq, and C. G. Nebigil, 2009, Transgenic myocardial overexpression of prokineticin receptor-2 (GPR73b) induces hypertrophy and capillary vessel leakage: Cardiovasc Res, v. 81, p. 28-37. Urzhumtsev, A., P. V. Afonine, and P. D. Adams, 2009, On the use of logarithmic scales for analysis of diffraction data: Acta Crystallogr D Biol Crystallogr, v. 65, p. 1283-91. Urzhumtseva, L., P. V. Afonine, P. D. Adams, and A. Urzhumtsev, 2009, Crystallographic model quality at a glance: Acta Crystallogr D Biol Crystallogr, v. 65, p. 297-300. Van Zandt, M. C., B. Doan, D. R. Sawicki, J. Sredy, and A. D. Podjarny, 2009, Discovery of [3-(4,5,7-trifluoro-benzothiazol2-ylmethyl)-pyrrolo[2,3-b]pyridin-1-yl]ac etic acids as highly potent and selective inhibitors of aldose reductase for treatment of chronic diabetic complications: Bioorg Med Chem Lett, v. 19, p. 2006-8. Végran F, Boidot R, Coudert B, Fumoleau P, Arnould L, Garnier J, Causeret S, Fraise J, Dembélé D, Lizard-Nacol S. Gene expression profile and response to trastuzumab-docetaxel-based treatment in breast carcinoma. Br J Cancer. 2009 Oct 20;101(8):1357-64. Vergne, I., E. Roberts, R. A. Elmaoued, V. Tosch, M. A. Delgado, T. Proikas-Cezanne, J. Laporte, and V. Deretic, 2009, Control of autophagy initiation by phosphoinositide 3-phosphatase jumpy: Embo J, v. 28, p. 2244-2258. Vetter, G., A. Le Bechec, J. Muller, A. Muller, M. Moes, M. Yatskou, Z. Al Tanoury, O. Poch, L. Vallar, and E. Friederich, 2009, Time-resolved analysis of transcriptional events during SNAI1-triggered epithelial to mesenchymal transition: Biochem Biophys Res Commun, v. 385, p. 485-91. Vincent, D. F., K. P. Yan, I. Treilleux, F. Gay, V. Arfi, B. Kaniewsky, J. C. Marie, F. Lepinasse, S. Martel, S. Goddard-Leon, J. L. Iovanna, P. Dubus, S. Garcia, A. Puisieux, R. Rimokh, N. Bardeesy, J. Y. Scoazec, R. Losson, and L. Bartholin, 2009, Inactivation of TIF1gamma cooperates with Kras to induce cystic tumors of the pancreas: PLoS Genet, v. 5, p. e1000575. 123 publications 2009 Notes Vlastos, F., S. Lacomme, P. Wild, S. Poulain, J. Siat, G. Grosdidier, S. du Manoir, B. Monga, G. Hillas, R. Varsovie, F. Claudot, B. Marie, J. M. Vignaud, and N. Szymanski, 2009, Do evolving practices improve survival in operated lung cancer patients? A biobank may answer: J Thorac Oncol, v. 4, p. 505-11. Volle, D. H., M. Decourteix, E. Garo, J. McNeilly, P. Fenichel, J. Auwerx, A. S. McNeilly, K. Schoonjans, and M. Benahmed, 2009, The orphan nuclear receptor small heterodimer partner mediates male infertility induced by diethylstilbestrol in mice: J Clin Invest, v. 119, p. 3752-64. Wang, X., C. G. Sansam, C. S. Thom, D. Metzger, J. A. Evans, P. T. Nguyen, and C. W. Roberts, 2009, Oncogenesis caused by loss of the SNF5 tumor suppressor is dependent on activity of BRG1, the ATPase of the SWI/SNF chromatin remodeling complex: Cancer Res, v. 69, p. 8094-101. Wendling, O., J. M. Bornert, P. Chambon, and D. Metzger, 2009, Efficient temporallycontrolled targeted mutagenesis in smooth muscle cells of the adult mouse: Genesis, v. 47, p. 14-18. Williams, J. A., N. Kondo, T. Okabe, N. Takeshita, D. M. Pilchak, E. Koyama, T. Ochiai, D. Jensen, M. L. Chu, M. A. Kane, J. L. Napoli, M. Enomoto-Iwamoto, N. Ghyselinck, P. Chambon, M. Pacifici, and M. Iwamoto, 2009, Retinoic acid receptors are required for skeletal growth, matrix homeostasis and growth plate function in postnatal mouse: Dev Biol, v. 328, p. 315-27. Wolf, A. I., D. Buehler, S. E. Hensley, L. L. Cavanagh, E. J. Wherry, P. Kastner, S. Chan, and W. Weninger, 2009, Plasmacytoid dendritic cells are dispensable during primary influenza virus infection: J Immunol, v. 182, p. 871-9. Wu, C., F. Ivars, P. Anderson, R. Hallmann, D. Vestweber, P. Nilsson, H. Robenek, K. Tryggvason, J. Song, E. Korpos, K. Loser, S. Beissert, E. Georges-Labouesse, and L. M. Sorokin, 2009, Endothelial basement membrane laminin alpha5 selectively inhibits T lymphocyte extravasation into the brain: Nat Med, v. 15, p. 519-27. Yu, J., and J. Auwerx, 2009, The role of sirtuins in the control of metabolic homeostasis: Ann N Y Acad Sci, v. 1173 Suppl 1, p. E10-9. Zeitlin SG, Baker NM, Chapados BR, Soutoglou E, Wang JY, Berns MW, Cleveland DW. Double-strand DNA breaks recruit the centromeric histone CENP-A. Zhang, Z., P. Hener, N. Frossard, S. Kato, D. Metzger, M. Li, and P. Chambon, 2009, Thymic stromal lymphopoietin overproduced by keratinocytes in mouse skin aggravates experimental asthma: Proc Natl Acad Sci U S A, v. 106, p. 1536-41. Zhao, Z., C. Park, M. A. McDevitt, C. Glidewell-Kenney, P. Chambon, J. Weiss, J. L. Jameson, and J. E. Levine, 2009, p21-Activated kinase mediates rapid estradiol-negative feedback actions in the reproductive axis: Proc Natl Acad Sci U S A, v. 106, p. 7221-6. Ziegler-Birling, C., A. Helmrich, L.Tora, and M. E.Torres-Padilla, 2009, Distribution of p53 binding protein 1 (53BP1) and phosphorylated H2A.X during mouse preimplantation development in the absence of DNA damage: Int J Dev Biol, v. 53, p. 1003-11. Zsindely, N., T. Pankotai, Z. Ujfaludi, D. Lakatos, O. Komonyi, L. Bodai, L.Tora, and I. M. Boros, 2009, The loss of histone H3 lysine 9 acetylation due to dSAGAspecific dAda2b mutation influences the expression of only a small subset of genes: Nucleic Acids Res, v. 37, p. 6665-80. 125 NOTES Notes 127 where we are olm eC te d rou Highway exit N°5 Illkirch-Nord Baggersee ar A35 PARIS NANCY METZ STRASBOURG STRASBOURG CITY CENTER Tram A Direction Illkirch Lixenbuhl Hy p erm ark et N Direction Parc d’Innovation N NW NE E W E S SW d Kast Alfre Faculté de Pharmacie ler MULHOUSE COLMAR Aéroport Strabourg-Enztheim Highway exit N°5 Illkirch-Graffenstaden Strasbourg-Meinau S Rue A35 SE O Tram A Station Campus Illkirch Tram A Direction Hautepierre Maillon Pôle API Bld Sé Bra bastien ndt Parc d’Innovation From Strasbourg airport (Entzheim): Take the shuttle (train, TER every 15 mins) that stops at the Strasbourg railway station: Journey time is 10 minutes. From Strasbourg railway station: Underground trams leave every 5 minutes. Take Tram A (direction Illkirch Lixenbuhl). Alight at the stop Campus d’Illkirch. Journey time is 25 minutes. From the Tram station Campus d’Illkirch: Take the Bus 63 (direction Plobsheim Rhin) and alight at the stop Parc d’Innovation. Follow Boulevard Sebastien Brant. At the roundabout, follow the second exit, Boulevard Gonthier d’Andernach and turn left on to Rue Laurent Fries. The IGBMC will be on your right. Or walking: Cross the tracks and passing by the Velo Parc, continue on to the roundabout. Take Boulevard Sebastien Brant (third exit). At the second roundabout take the first exit, Boulevard Gonthier d’Andernach and turn left in to Rue Laurent Fries. The IGBMC will be on your right. By car: Take the 5th Highway exit and then follow the direction Parc d’innovation. At the first roundabout take the fourth exit, Boulevard Sebastien Brant, then at the second take the second exit and turn left in to Rue Laurent Fries. The IGBMC will be on your right. © Copyright 2010 Produced by: IGBMC Texts: IGBMC Teams, Technical Platforms, Core Facilities, Students & Post-docs Board Print: Valblor Conception, Design, and Layout: Office of Communication IGBMC, Photography: Lola Velasquez, Patrice Latron, Office of Communication IGBMC A big thank you goes out to all ! Institut de Génétique et de Biologie Moléculaire et Cellulaire 1 rue Laurent Fries / 67404 Illkirch CEDEX / France igbmc@igbmc.fr