Abstracts - International Mammalian Genome Society
Transcription
Abstracts - International Mammalian Genome Society
Aegean Conferences Series-Vol. 54 AEGEAN CONFERENCES Linking the international scientific community Bringing the humanity scholars together together with the INTERNATIONAL MAMMALIAN GENOME SOCIETY 24th International Mammalian Genome Conference October 17-21, 2010 Aldemar: Knossos Royal Village Conference Center Hersonissos, Crete, Greece 24th International Mammalian Genome Conference Aldemar Knossos Royal Village October 17-21, 2010 Hersonissos, Crete, Greece COMMITTEES International Mammalian Genome Society Secretariat Maja Bucan, Past President (2010) Karen Steel, President (2010) David Threadgill, Vice-President (President 2011) Kent Hunter (2010) David Beier (2011) Ian Jackson (2011) Nancy Jenkins (2011) Simon Foote (2012) Xavier Montagutelli (2012) Darren Logan (2011) VM Chapman award winner Nomination and Election Committee Kathryn Hentges (2010) Bruce Herron (2010) Amy Moser (2010) Rosemary Elliott (2011) Beverly Mock (2011) Thomas Saunders (2011) Committee on the Standardized Genetic Nomenclature for Mice Janan Eppig, Chair Ruth Arkell Piero Carninci Sally Cross Deanna Church Jonathan Flint Simon Foote Jiri Forejt Teresa Gunn David Largespada Lois Maltais 3 Amy Moser Jean-Jacques Panthier David Threadgill Wolfgang Wurst Aamir Zuberi Local Organizing Committee Joe Nadeau Director of Research and Academic Affairs Institute for Systems Biology 1441 North 34th Street Seattle, WA 98103-8904 Tel: 1-206-732-1207 E-mail: jnadeau@systemsbiology.org Rudi Balling Director Luxembourg Centre for Systems Biomedicine Universite' du Luxembourg Campus Limpertsberg 162a Rue de la Faiencerie L-1511 Luxembourg E-mail: rudi.balling@uni.lu David J. Galas Professor & Senior Vice President, Institute for Systems Biology 1441 N. 34th Street, Seattle, WA 98103 E-mail: dgalas@systemsbiology.org George Kollias President & Scientific Director B.S.R.C. "Alexander Fleming" P.O. Box 74145 16602, Varkiza, Greece E-mail: kollias@fleming.gr John D. Lambris Dr. Ralph and Sallie Weaver Professor of Research Medicine Founder & Executive Director of AegeanConferences University of Pennsylvania 422 Curie Boulevard Philadelphia, PA 19104 E-mail:lambris@upenn.edu www.lambris.net 4 Official Journal of the International Mammalian Genome Society Sponsored by the International Committee for Standardized Genetic Nomenclature Editors Joseph H. Nadeau Director of Research and Academic Affairs Institute for Systems Biology 1441 North 34th Street Seattle, WA 98103-8904 USA Steve D.M. Brown Medical Research Council Mammalian Genetics Unit Harwell, Didcot Oxfordshire OX11 ORD United Kingdom Informatics Editors Janan T. Eppig, Bar Harbor, Maine USA Reviews Editor Maja Bucan, Philadelphia, Pennsylvania, USA Managing Editor Louise Tinsley, Oxfordshire, UK IMGS Business Manager Darla Miller, Chapel Hill, NC, USA Editors D. Adams, Cambridge, United Kingdom M.D. Adams, Cleveland, Ohio, USA T. Aitman, London, United Kingdom L. Andersson, Uppsala, Sweden R. Arkell, Canberra, Australia P. Avner, Paris, France E. Bailey, Lexington, Kentucky, USA R. Balling, Braunschweig, Germany D.R. Beier, Boston, Massachusetts, USA B. Beutler, La Jolla, California, USA S. Camper, Ann Arbor, Michigan, USA G. Churchill, Bar Harbor, Maine, USA N.G. Copeland, Singapore E. Fisher, London, United Kingdom S. Foote, Hobart, Tasmania, Australia W. Frankel, Bar Harbor, Maine, USA D. Gauguier, Oxford, United Kingdom M. Georges, Liege, Belgium C. Goodnow, Canberra, Australia K.A. Gould, Omaha, Nebraska, USA E.D. Green, Bethesda, Maryland, USA P. Gros, Montreal, Quebec, Canada Y. Herault, Orleans, France M. Hrabe´ de Angelis, Neuherberg, Germany D.A. Hume, Edinburgh, UK I.J. Jackson, Edinburgh, United Kingdom N. Jenkins, Singapore M. Justice, Houston, Texas, USA G. Kelsey, Cambridge, UK H. Khatib, Madison, Wisconsin, USA E. Lander, Cambridge, Massachusetts, USA M. Lyon, Oxfordshire, United Kingdom T. Magnuson, Chapel Hill, NC, USA K. Murphy, College Station, Texas, USA D. Norris, Oxfordshire, United Kingdom E.A. Ostrander, Bethesda, Maryland, USA W.J. Pavan, Bethesda, Maryland, USA D. Pomp, Chapel Hill, North Carolina, USA L.C. Schalkwyk, London, United Kingdom T. Shiroishi, Shizuoka-ken, Japan K. Steel, Cambridge, United Kingdom C.K. Tuggle, Ames, Iowa, USA E. Whitelaw, Brisbane, Australia L. Wicker, Cambridge, United Kingdom J. Womack, College Station, Texas, USA J.G. Zhang, Ann Arbor, Michigan, USA 5 TABLE OF CONTENTS General Information 8 Program Outline 10 Program 13 Abstracts 35 Bio Informatics Workshop Program 187 List of Participants 190 Sponsors 195 Upcoming Events 196 Conference Map 197 Hotel Map 198 6 AWARDS The Secretariat, members of The International Mammalian Genome Society, and Aegean Conferences are proud of the students and postdoctoral fellows who present oral or poster presentations at the annual meeting. The outstanding contributions of these young scientists are recognized through several awards. The first of these awards is the Verne Chapman Young Scientist Award, which is given to the most outstanding oral presentation from a postdoctoral fellow or student. This is a monetary award of $500 that reflects Dr. Chapman’s dedication to mentoring junior scientists. This award also includes a two year term on the secretariat. Two second place monetary awards of $250 each are given this year by The Genetics Society of America. A large group of publishing companies sponsor (see page 195) this year’s presentation prizes, which are a one year subscription to their journal. Genesis is donating books. These awards are given to the most outstanding poster presentations by graduate students and postdoctoral fellows, and are chosen by members of the IMGS Secretariat during the course of the meeting. 7 GENERAL INFORMATION Accommodations The conference participants will be staying at the Aldemar: Knossos Royal Village (A' class), which is located in Hersonissos, 25 km from the airport. Arrival and Check-in It is expected that attendees will arrive/check-in on Sunday after 2:00 PM. Anyone arriving after 10:30 pm on Sunday must make alternative dinner arrangements. Conference Venue The Aldemar: Knossos Royal Village Conference Center is located at the hotel. (#29 on Hotel Map) Handicapped Accessibility The hotel and conference facilities are handicapped accessible. Posters Posters should be mounted Monday, October 18, on the designated boards in Conference Hall II and removed at the closing of the conference. The dimensions of the boards are 90 cm wide x 120 cm high. Adhesive tape for mounting the posters on the boards will be available at the poster area. Odd number posters will be viewed at session 1. Even number posters will be viewed at session 2. Posters will be on display for the entire duration of the meeting. (#29 on Hotel Map) Oral Presentations Speakers are asked to bring their USB flash drive or CD to the conference reception desk at least 30 minutes prior to the session. The allocated time for all presentations, unless otherwise indicated, includes five minutes Q&A. Tour to Knossos and Museum Buses for the city & archaeological tour will be leaving from the hotel on Tuesday, October 19 at 2:00p.m. Participants will be picked up at the hotel front lobby at that time. Badges are required for participation. Breakfast Breakfast for registered participants and registered accompanying persons will be served at the Aldemar Knossos Royal Village hotel main restaurant during the hours scheduled in this program. (#2 on Hotel Map) 8 Lunches/Informal Discussions Lunches for registered participants and registered accompanying persons will be served during the times indicated in this program at the Main Restaurant. Badges are required for admission. (#2 on Hotel Map) Dinners/Informal Discussions Three of the dinners/informal discussion will be served during the times indicated in the program. Vegetarian entrees are available only upon request in advance of the meeting. Please contact info@aegeanconferences.org. Internet, Phone, & Computer Wireless Internet access is located throughout the conference center free of charge during meeting hours. There are computers available in the hotel’s business center for the use of the attendees. Phone service is available for an additional charge from your hotel room. Other Information Animals are not permitted on site. Badges are required for admission to all events. All Aegean Conferences events are non-smoking. No Photographs and Recording Devices Participants are not allowed to photograph and/or record using cameras, mobile telephones and other recording devices during the slide and poster presentations. Tour and Travel Information A desk operated by the personnel of our official travel agency Conferex LLC, P.O. Box 12, Wynnewood, PA 19096, USA Tel.: 1-484-685-0558, FAX: 1-610-771-4224 E-mail: info@conferex.com, www.conferex.com ERA Ltd., 17, Asklipiou Str., 106 80 Athens-Greece Tel.: 30-210-363-4944, FAX: 30-210-3631690, E-Mail: info@era.gr will be located at the conference center throughout the meeting. 9 PROGRAM OUTLINE 2:00 PM – 6:00 PM 7:00 AM-8:00 AM 8:00 AM-9:00 AM 9:00 AM-12:30 PM Saturday, October 16, 2010 Secretariat Meeting Theater Room Hotel Lobby Sunday, October 17, 2010 Breakfast Registration: Bioinformatics Workshop Lobby I Bioinformatics Workshop (see appendix 1) Tutorial 1 Room Orpheas Tutorial 2 Room Apollon Tutorial 3 Room Hermes 12:30 PM-1:00 PM Coffee Break 11:00 AM Registration: Student Satellite Symposium Lobby I Student Satellite Symposium Session 1 Conference Hall I 1:00 PM-2:45 PM 2:45 PM-3:30 PM Coffee Break 3:30 PM-5:30 PM Student Satellite Symposium Session 2 Conference Hall I 4:00 PM – 7:00 PM 24th IMGS Registration Open Lobby I 7:00 PM-10:30 PM Welcome Reception and Dinner 7:00 AM-8:20 AM Monday, October 18, 2010 Breakfast 8:00 AM-5:30 PM 24th IMGS Registration Open Lobby I 8:20 AM-8:30 AM Official 24th IMGS Opening 10 8:30 AM-10:30 AM Session 1: Infection and Immunity Conference Hall I 10:30 AM–12:30 PM Coffee Break/ Exhibition and Poster Session 1 Odd Numbered Posters Conference Hall II 12:00 PM-1:30 PM Editorial Board Meeting with lunch Theater Room Hotel Lobby 12:30-1:30 PM Lunch 1:30 PM-3:30 PM Session 2: Modeling Disease-Development, Metabolism and Physiology Conference Hall I 3:30 PM-4:00 PM Coffee Break 4:00 PM-5:00 PM IMGS Business meeting (all welcome) Conference Hall I 5:00 PM-6:00 PM Verne Chapman Lecture Conference Hall I 6:30 PM-9:30 PM Dinner 7:00 AM-8:30 AM Tuesday, October 19, 2010 Breakfast and Mentor Breakfast 8:30 AM-10:00 AM Session 3: Neuroscience, Behavior, and Sensory Systems I Conference Hall I 10:00 AM Session 4: Large Scale and Genome-wide Resources Conference Hall I 10:30AM Coffee Break 11:00 AM Session 4 continued: Large Scale and Genome-wide Resources 1:00 PM-2:00 PM Lunch 11 2:00 PM-5:00 PM Guided Tour of the Knossos Archeological Site Buses depart hotel lobby at 2:00 PM 5:00 PM-6:30 PM SPECIAL SESSION Human Disease and Mouse Models Conference Hall I 6:30 PM-9:30 PM Dinner 7:00 AM-8:30 AM Wednesday, October 20, 2010 Breakfast 8:30 AM-10:30AM Session 5: Epigenetics, Neoplasia and Aging I Sponsored by The Ellison Medical Foundation Conference Hall I 10:30 AM-12:30 PM Coffee Break/Exhibition and Poster Session 2 Even Numbered Posters Conference Hall II 12:30 PM-1:30 PM 12:00 PM-1:30 PM Lunch Secretariat meeting with lunch Theater Room Hotel Lobby 1:45 PM-3:15 PM Session 6: Epigenetics, Neoplasia and Aging II Sponsored by The Ellison Medical Foundation Conference Hall I 3:15 PM-3:45 PM Coffee Break 3:45 PM-4:30 PM Session 7: Neuroscience, Behavior and Sensory Systems II Conference Hall I 7:00 PM Farewell Dinner 12 PROGRAM 13 PROGRAM 2:00 PM – 6:00 PM 7:00 AM-9:00 AM Saturday, October 16, 2010 Secretariat Meeting Theater Room Hotel Lobby Sunday, October 17, 2010 Breakfast 8:00 AM-9:00 AM Registration: Bioinformatics Workshop Lobby I 9:00 AM-12:30 PM Bioinformatics Workshop (see appendix 1) Tutorial 1 Room Orpheas Tutorial 2 Room Apollon Tutorial 3 Room Hermes 12:30 PM-1:00 PM Coffee Break 11:00 AM Registration: Student Satellite Symposium Lobby 1 Student Satellite Symposium Session 1 Chairs: Karen Steel, Maja Bucan Conference Hall I 1:00 PM 1 1:15 PM 2 1:30 PM 3 1:45 PM 4 2:00 PM 5 The F2 population for dissecting host susceptibility to periodontitis Ariel Shusterman, Yael Houri-Haddad, Ervin I Weiss, and Fuad A Iraqi Mouse genetic model of cardiovirulent coxsackievirus B3 infection Sean A. Wiltshire and Silvia M. Vidal Association between spatial proximity and functional similarity in human genome Ekaterina Khraneeva, Andrey Mironov, and Mikhail Gelfand Olfactory receptor gene family evolution in marsupials Amir Mohammadi, Margaret L. Delbridge, and Jennifer A. Marshal Graves Advancing paternal age is associated with deficits in social and exploratory behaviours in the offspring Rebecca G. Smith, Rachel L. Kember, Leonard C. 15 2:15 PM 6 2:30 PM 7 2:45 PM Schalkwyk, Joseph D Buxbaum, Abraham Reichenberg, Cathy Fernandes, and Jonathan Mill Genome-wide differential methylation patterns in intersubspecific hybrid mice John D. Calaway, Hyuna Yang, Elena de la CasaEsperon, Megan E. Hanson, Ezequiel C. Cambranis, Maria A. Perez-Lamigueiro, David L. Aylor, Leonard McMillan, Gary A. Churchill, and Fernando PardoManuel de Villena Optimization of a genome-wide, allele-specific DNA methylation assay for the mouse John P. Didion, John D. Calaway, Isa Kemal Pakatci, Leonard McMillan, and Fernando Pardo-Manuel de Villena Coffee Break Student Satellite Symposium Session 2 Chairs: David Beier, David Threadgill Conference Hall I 3:30 PM 8 3:45 PM 9 4:00 PM 10 4:15 PM 11 4:30 PM 12 Characterisation of laterality and cardiac developmental defects in the K27 mutant mouse Louise Stephen, Karen Mitchell, and Kathryn Hentges Kyoto Epileptic Rat (KER): A novel rat model of Kcna1 mutation showing spontaneous seizures and muscle involuntry contractions Saeko Ishida, Tomoji Mashimo, Takeshi Nishio, Yu Sakamoto, Shuji Kaneko, and Tadao Serikawa Identifying modifier genes of mpnsts in the Nf1;p53cis mouse model of neurofibromatosis type 1 Jessica A Van Schaick, Keiko Akagi, Sandra Burkett, Christina DiFabio, Robert Tuskan, Jessica Walrath, and Karlyne Reilly A novel genetic model of osteoporosis by overexpression of human RANKL in transgenic mice Alexandra Niti, R Dacquin, V Rinotas, P Jurdic, and E Douni Positive selection might contribute to functional divergence of NAIPs following gene duplication in mammals during the evolution Zhaoliang Su, Chenglin Zhou, Jianguo Chen, Shengjun Wang, and Huaxi Xu 16 4:45 PM 13 5:00 PM 14 5:15 PM 15 A forward genetics approach identifies a novel member of the Dnajc family that causes neuromuscular disease in mice Fotios Ioakeimidis, Vagelis Rinotas, Eleni Makrinou, George Kollias, and Eleni Douni Gene expression studies of ncRNAs within Odz4 locus Tyler Buit, Chiao-Ling Lo, Feichen Shen, and Amy C. Lossie Early life stress is associated with altered behavior and epigenetic alterations to Nr3c1, Avp and Nr4a1 Rachel L. Kember, Emma Dempster, Leonard C. Schalkwyk, Jonathan Mill and Cathy Fernandes 4:00 PM -7:00 PM 24th IMGS Registration Open Lobby I 7:00 PM-10:30 PM Welcome Reception and Dinner 7:00 AM-8:20 AM Monday, October 18, 2010 Breakfast 8:00 AM-5:30 PM 24th IMGS Registration Open Lobby I 8:20 AM Official 24th IMGS Opening Session 1: Infection and Immunity Chairs: Simon Foote, Karen Svenson Conference Hall I 8:30 AM 16 Stress mechanisms, inflammation, infection and host resistance Katia P. Karalis 9:15 AM 17 Physiologic consequences of constitutive MTOR inhibition: alterations in cell/organ size and immune cell development Shuling Zhang, Julie A. Readinger, Wendy DuBois, Mirkka Janka-Junttila, Richard Robinson, Magaret Pruitt, Val Bliskovsky, Julie Z. Wu, Kaori Sakakibara, Jyoti Patel, Carole A. Parent, Lino Tessarollo, Pamela L. Schwartzberg, and Beverly A. Mock 9:30 AM 18 Severe Combined Immunodeficiency (SCID) rats generated by zinc finger nuclease technology Tomoji Mashimo, Akiko Takizawa, Birger Voigt, 17 9:45 AM 19 10:00 AM 10:15 AM Kazuto Yoshimi, Hiroshi Hiai, Takashi Kuramoto, and Tadao Serikawa A recombination hotspot leads to sequence variability within a novel gene and contributes to complex disease susceptibility Iris KL Tan, Leanne Mackin, Nancy Wang, Anthony T Papenfuss, Colleen M Elso, Michelle P Ashton, Belinda Phipson, Melanie Bahlo, Terrence P Speed, Gordon K Smyth, Grant Morahan, and Thomas C Brodnicki Selected Student Talk Selected Student Talk 10:30 AM Coffee Break/ Exhibition and Poster Session 1 Odd Numbered Posters Conference Hall II 12:30 PM-1:30 PM Lunch 12:00PM-1:30 PM Mammalian Genome Editorial Board Meeting with Lunch Theater Room Hotel Lobby Session 2: Modeling Disease: Development, Metabolism and Physiology Chairs: Nancy Jenkins, Gail Herman Conference Hall I 1:30 PM 20 1:45 PM 21 2:00 PM 22 2:15 PM 23 A sensitized suppressor screen to identify modifier genes for factor V leiden-dependent thrombosis in the mouse Randal Westrick, Goujing Zhu, Sara Manning, Angela Yang, David Siemieniak, and David Ginsburg Genetic and dietary controls of alternative exon use and mRNA levels in the systems architecture of dietinduced metabolic diseases Sabrina H. Spiezio, Annie Hill-Baskin, Karen Fitch, Keith Jones, Michael H. Shapero, and Joseph H. Nadeau A gene driven ENU mutagenesis screen reveals a novel method of ZIC protein nuclear localization R Ali, H Bellchambers, N Warr, D Quwailid, P Denny and Ruth Arkell Non-muscle myosin IIB is required for formation of the coronary vasculature Karen Mitchell, C Clowes, Louise Stephen, and Kathryn 18 2:30 PM 24 2:45 PM 25 3:00 PM 26 3:15 PM 27 E Hentges Short circuit, a homozygous lethal ENU mutation that affects lung development Michael J. Parsons, Laura Yates, Jessica Edwards, Lauren Chessum, Charlotte H. Dean, and Patrick M. Nolan Identification of the Danforth’s short tail mutation using next generation sequencing Christopher N. Vlangos, Amanda N. Siuniak, Dan Robinson, Arul M. Chinnaiyan, James Cavalcoli, Robert H. Lyons, and Catherine E. Keegan A novel role for Atmin, controlling ciliogenesis through modulation of dynein light chain expression Paraskevi Goggolidou, Jonathan Stevens, Gabrielle Wheway, Antonella Di Paolo, Rosario Romero, James Briscoe, Colin Johnson, and Dominic Norris Genetic analysis of complex traits in the emerging collaborative cross David L. Aylor, William Valdar,, Wendy FouldsMathes, Ryan J. Buus, Ricardo A. Verdugo,, Ralph S. Baric, Martin T. Ferris, Jeffrey A. Frelinger, Mark Heise, Matt B. Frieman, Lisa E. Gralinski, Timothy A. Bell, John P. Didion, Kunjie Hua, Derrick L. Nehrenberg, Christine L. Powell, Jill Steigerwalt, Yuying Xie, Samir N.P. Kelada, Francis S. Collins, Ivana V. Yang, David A. Schwartz, Lisa A. Branstetter, Elissa J. Chesler, Darla R. Miller, Jason Spence, Eric Yi Liu, Leonard McMillan, Abhishek Sarkar, Jeremy Wang, Wei Wang, Qi Zhang, Karl W. Broman, Ron Korstanje, Caroline Durrant, Richard Mott, Fuad A. Iraqi, Daniel Pomp, David Threadgill, Fernando PardoManuel de Villena, and Gary A. Churchill 3:30 PM-4:00 PM Coffee Break 4:00 PM-5:00 PM IMGS Business meeting (all welcome) Conference Hall I 5:00 PM – 6:00 PM Verne Chapman Lecture Realising the potential of mouse disease models – from pathway to therapy Steve D.M. Brown Conference Hall I 28 19 6:30 PM-9:30 PM Dinner 7:00 AM-8:30 AM Tuesday, October 19, 2010 Breakfast and Mentor Breakfast Session 3: Neuroscience, Behavior, and Sensory Systems I Chairs: Ian Jackson, Elizabeth Bryda Conference Hall I 8:30 AM 29 8:45 AM 30 9:00 AM 31 9:15 AM 32 9:30 AM 33 9:45 AM Importance of Cadm1 and cell adhesion in depressive behavior C Santos, B Miller, M Pletcher, A Su, L Tarantino, and Tim Wiltshire An ENU sensitization screen to determine the physiological function of α - synuclein Deborah E Cabin, M Casey, and D Zou Genomic resources in Peromyscus – mapping the audiogenic seizure sensitivity locus Gabor Szalai, Janet Crossland, Jay Coleman, and Michael Felder Mutations in the PDZ domain containing protein Gipc3 cause progressive sensorineural degeneration (Ahl5 and jams1) in mice and recessive hearing impairment in humans (DFNB95) Nikoletta Charizopoulou, Andrea Lelli, Margit Schraders, Kausik Ray, Ronald J.C. Admiraal, Harold R. Neely, Joseph R. Latoche, John K. Northup, Hannie Kremer, Jeffrey R. Holt, and Konrad Noben-Trauth A mutation in the gene encoding mitochondrial Mg2+ channel MRS2 results in demyelination in rats Takashi Kuramoto, Mitsuru Kuwamura, Satoko Tokuda, Takeshi Izawa, Yoshifumi Nakane, Kazuhiro Kitada, Masaharu Akao, Jean-Louis Guénet, and Tadao Serikawa Selected Student Talk Session 4: Large Scale and Genome-wide Resources Chairs: David Beier, Joe Nadeau Conference Hall I 10:00 AM 34 10:15 AM 35 Annotation of long non-coding RNA transcripts Mark Thomas and Jennifer Harrow The JAX Cre Repository: Improving the utility of Cre 20 driver strains Stephen A. Murray, Caleb Heffner, Michael Sasner, Cathleen Lutz, Brandon Grossman, Stephen Rockwood, Yashoda Sharma, and Leah Rae Donahue 10:30 AM Coffee Break 11:00 AM 36 11:15 AM 37 11:30 AM 38 11:45 AM 39 12:00 Noon 40 12:15 PM 41 12:30 PM 42 12:45 PM 43 Mouse genomes project: map of SNPS and short indels Petr Danecek, Gan Xiangchao, Thomas M Keane, James Stalker, Binnaz Yalcin, Martin Goodson, Sendu Balasubramaniam, Kim Wong, Guy Slater, Andreas Heger, Eleazar Eskin, Nick Furlotte, Chris Ponting, Jonathan Flint, and David J Adams Use of PiggyBac-mediated transient transgenic RNAi expression for rapid characterization of gene function during embryonic development David R. Beier, Yuko Fujiwara, Shannon W. Davis, Haiyan Qiu, Thomas L. Saunders, Stuart Orkin, Sally A. Camper, and Bryan C. Bjork The Sanger Institute Mouse Genetics Programme Ramiro Ramirez-Solis, J White, E Ryder, R Houghton, and J Bottomley Next level systemic phenotyping of mice Martin Hrabé de Angelis and the German Mouse Clinic consortium Portal to large-scale mouse knockout resources William Skarnes Tooling up MGI for a deluge in mouse phenotype and disease model data Anna Anagnostopoulos, Janan Eppig, and Mouse Genome Informatics Group New approaches to conditionality Aris N. Economides, David Frendewey, Peter Yang, David M. Valenzuela, Andrew J. Murphy, and George D. Yancopoulos Mouse metabolic phenotyping centers: National consortium focusing on diabetic complications Renee LeBoeuf 1:00 PM-2:00 PM Lunch 2:00 PM-5:00 PM Guided Tour of the Knossos Archeological Site Buses depart hotel lobby at 2:00 PM 21 5:00 PM-6:30 PM 5:00 PM 44 5:45 PM 45 SPECIAL SESSION Human Disease and Mouse Models Conference Hall I Lupus Edward Wakeland Cancerous microRNAs and regulatory RNA binding proteins Reuven Agami 6:30 PM Dinner 7:00 AM-8:30 AM Wednesday, October 20, 2010 Breakfast Session 5: Epigenetics, Neoplasia and Aging I Sponsored by The Ellison Medical Foundation Chairs: Teresa Gunn, Kent Hunter Conference Hall I 8:30 AM 46 Genomic imprinting: Insights from the Gnas cluster Jo Peters 9:15 AM 47 The battle of the signals: Non-coding RNAs vs. enhancers in the transcriptional regulation of the Kcnq1 imprinted region Anjali Raval, Lisa Korostowski, and Nora Engel 9:30 AM 48 An identity crisis: Abnormal male germ cell development and the initiation of testicular cancer Jason Heaney, Jean Kawasoe, Megan Michelson, and Joseph Nadeau 9:45 AM 49 Age-associated change of energy metabolism are genetically dissected by mouse inter-subspecific consomic strains Toyoyuki Takada, Akihiko Mita, Shigeharu Wakana, Kazuo Moriwaki, Hiromichi Yonekawa, and Toshihiko Shiroishi 10:00 AM 50 Genetic regulation of aging related phenotypes and lifespan in mice Rong Yuan, S W Tsaih, Q Meng, K Flurkey, J Nautiya, SB Petkova, MA Bogue, KD Mills, L L Peters, CJ Bult, CJ Rosen, JP Sundberg, M Parker, DE Harrison, GA Churchill, and B Paigen 10:15 AM 51 Subspecific origin and haplotype diversity in the laboratory mouse Hyuna Yang, Jeremy R Wang, John P Didion, Ryan J 22 Buus, Timothy A Bell, Catherine E Welsh2, François Bonhomme, Alex Hon-Tsen Yu, Michael W Nachman, Jaroslav Pialek, Priscilla Tucker, Pierre Boursot, Leonard McMillan, Gary A Churchill, and Fernando Pardo-Manuel de Villena 10:30 AM Coffee Break/Exhibition and Poster Session 2 Even Numbered Posters Conference Hall II 12:30 PM-1:30 PM Lunch 12:00 PM-1:30 PM Secretariat meeting with lunch Theater Room Hotel Lobby Session 6: Epigenetics, Neoplasia and Aging II Chairs: Beverley Mock, Xavier Montagutelli Conference Hall I Sponsored by The Ellison Medical Foundation 1:45 PM 52 2:30 PM 53 2:45 PM 54 3:00 PM 55 3:15 PM TBA David Galas Genetic architecture of hybrid sterility: Matching the puzzle pieces together Jiri Forejt, Maria Dzur-Gejdosova, Vaclav John, Tanmoy Bhattacharyya, Petr Simecek, Sona Gregorova, Ondrej Mihola, Petr Flachs, and Zdenek Trachtulec Molecular interactions of dead end (Dnd1) Angabin Matin, Zhu Rui, KangLi Luo, Chitralekha Bhattacharya, Michelina Iacovino, Elisabeth Mahen, Michael Kyba, and Sita Aggarwal Genetic dissection of metatasis susceptibility converges on a common mechanism J Alsarraj, S Winter, N Goldberger, K Mattaini, M William, L Lukes, R Walker, and Kent W Hunter Coffee Break Session 7: Neuroscience, Behaviour and Sensory Systems II Conference Hall I 3:45 PM 56 Icst is a dominant negative mutation of Lmx1b Sally H. Cross, Lisa Mckie, Margaret Keighren, Dan 23 4:00 PM 57 4:15 PM 58 7:00 PM Macalinao, Alison L. Kearney, Simon W. John, and Ian J. Jackson Elucidating the role of Nsdhl and cholesterol metabolism in CNS development using a conditional knockout allele Gail E Herman, N Bir, L Binkley, K McLarren, C Boerkoel, and D Cunningham Genetic, stem cell, and systems analyses of neurodegenerative diseases George A Carlson, R Bennett, ME Orr, I Lee, H Yoo, JH Cho, D Hwang, and LE Hood Farewell Dinner 24 Posters 59 60 61 62 63 64 65 66 67 68 69 Stage and cell subtype-specific epigenetic regulation of mammary gland development Deanna Acosta, Melissa J Fazzari, John M Greally, and Cristina Montagna Toward an unified measure of intraspecific selective pressure Roberto Amato, Gennaro Miele, Michele Pinelli, and Sergio Cocozza A new targeted mutation, Caspa, in the Gnas complex shows hyperactivity and ataxia Simon Ball, Sally Eaton, Christine Williamson, Charlotte Tibbit, and Jo Peters Rat resource and research center Beth A. Bauer, EC Bryda, CL Franklin, LK Riley, and JK Critser Loci on chromosome 2 are associated with naïve airway hyperresponsiveness in CD-1 outbred mice David R Beier, Andrew Kirby, Mary Prysak, Mark Daly, and Emily Cozzi High-resolution map and candidate gene analysis for ébouriffé (ebo), a hair mutation on mouse Chromosome 2 associated with infertility Fernando Benavides, J Jaubert, CJ Perez, J-L Guénet, J Barrera, B Cole, and CJ Conti Molecular genetic analysis of the NADPH oxidase of the vestibular system Catrina A Spruce, John P Flaherty, Heather E Fairfield and David E Bergstrom Mouse models of Costello syndrome Heather E Fairfield, John P Flaherty, Catrina A Spruce, Leah Rae Donahue and David E Bergstrom The application of high-throughput sequencing technologies for mutation discovery in the Jackson Laboratory’s mouse mutant resource Laura G Reinholdt, David E Bergstrom, Muriel T Davisson-Fahey, Cathleen M Lutz, Michael Sasner, Steven A Murray, Jeff Lake, Steven Rockwood, Leah Rae Donahue and the MMR team The role of CAML in the inner ear Elizabeth C Bryda, Nathan T Johnson, Cynthia L Besch-Williford, Kevin K Ohlemiller, and Richard J Bram Interesting phenotypes found as part of the infection challenge in the Wellcome Trust Sanger Institute’s Mouse Genetics Programme Simon Clare, Leanne Kane, Lynda Mottram, Jacqui White, Ramiro 25 70 71 72 73 74 75 76 77 78 79 80 81 Ramirez – Solis, and Gordon Dougan The expression of SEPTIN 9 in human breast cancer Diana Connolly, Zhixia Yang, Elias T. Spiliotis, Maria Castaldi, Nichelle Simmons, Pascal Verdier-Pinard, and Cristina Montagna Genetic contribution to liver fibrosis David DeSantis, Michelle Pritchard, Laura Nagy, Joseph Nadeau, and Colleen M Croniger Diet, obesity, and susceptibility to colon polyps Stephanie K. Doerner and Joseph H. Nadeau Genetic regulation of fracture risk in inbred mice Leah Rae Donahue GA Churchill, S-W Tsaih, and WG Beamer Imprinted XLAS: A new player in bone and adipocyte metabolism Sally Eaton, Simon Ball, Colin Beechey, Christine Williamson, and Jo Peters The Sanger Mouse Genetics Programme: High throughput recessive lethality screen Jeanne Estabel, Elizabeth Tuck, Damian Carragher, Jennifer Salisbury, and Jacqueline K. White on behalf of the Mouse Genetics Programme Multiple forebrain cis-regulatory elements at the Dlx1/2 locus: redundant function or complex control mechanisms? Marc Ekker, Luc Poitras, Man Yu, Noel Ghanem, and Lisa Tran Novel mutation discovery on ENU mouse mutagenesis by nextgeneration sequencer Ryutaro Fukumura, Hayato Kotaki, Yuichi Ishitsuka, Takuya Murata, Shigeru Makino, Yuji Nakai, and Yoichi Gondo The Sanger Mouse Genetics Programme: High throughput characterisation of knockout mice for genes involved in glucose metabolism Anna-Karin Gerdin, L Roberson, Jacqueline K White on behalf of the Mouse Genetics Programme EuroPhenome: Large dataset visualisation, statistical analysis and data exploratory tool for mouse phenotyping data Ahmad Hassan, Hugh Morgan, Andrew Blake, Simon Greenaway, The EUMODIC Consortium, John M. Hancock, and Ann-Marie Mallon The intragene resource centre, a national centre for mouse genetics, distribution, archiving and functional imaging Cecile Fremond, Stephanie Lerondel, Christelle Martin, and Yann Herault The Down syndrome as a complex trait resulting from the interaction of dosage sensitive genes unravelled in the mouse model V Brault, A Duchon, E Dalloneau, P Lopes, I Abizanda, S Luiller, JC Bizot, C Borel, M Raveau, D Marechal, J Lignon, S Pothion, F Trovero, V Tybulewicz, EM Fisher, S Antonarakis, M Dierssen, and 26 82 83 84 85 86 87 88 89 90 91 92 Yann Herault The European Mouse Mutant Archive (EMMA) Michael Hagn, G Tocchini-Valentini, Y Hérault, S Brown, U Lendahl, J Demengeot, M Hrabé de Angelis (EMMA Director), E Birney, R Ramirez-Solis, JL Mandel, and L Montoliu Broad based phenotyping approaches for the characterisation of mutant mouse lines in EUMODIC and the Harwell aging screen Heena Vanmalibhai Lad, KA Lee, A Blake, H Morgan, A Hassan, S Greenaway, AM Mallon, HJ Gates, SDM Brown, and The EUMODIC Consortium FBXO11 regulates TGF beta signaling through TRP53 Hilda Tateossian, Susan Morse, and Steve DM Brown The development of the integrated database of mammals in RIKEN Hiroshi Masuya, Yuko Makita, Koro Nishikata, Yuko Yoshida, Terue Takatsuki, Kasunori Waki, Nobuhiko Tanaka, Yoshiki Mochizuki, Norio Kobayashi, Riichiro Mizoguchi, Teiichi Furuichi, Hideya Kawaji, Daub Carsten, Yoshihide Hayashizaki, Shigeharu Wakana, Atsushi Yoshiki, Kaoru Fukami-Kobayashi, and Tetsuro Toyoda Combining clinical and expression QTLs in the context of infection: new insights into susceptibility to influenza Gregory A. Boivin, Julien Pothlichet, Emil Skamene, Earl G. Brown, Robert Sladek, and Silvia M. Vidal A nature of the blastomer DNA methylation determination Andrey A. Ivanov Not all wild-derived Mus spretus mice do resist virulent Yersinia pestis Jean Jaubert, Charlotte Leblanc, Charlène Blanchet, Garcia Sylvie, Elisabeth Carniel, Christian Demeure, Jean-Jacques Panthier, and Xavier Montagutelli Evidence of hypothalamic degeneration in the anorectic anx/anx mouse IAK Nilsson, S Thams, C Lindfors, A Bergstrand, S Cullheim, T Hökfelt, and Jeanette E Johansen Identification of a novel Slc25 member that causes autosomal recessive ataxia in ENU-mutagenized mice A Karakostas, F Ioakeimidis, V Rinotas, G Kollias, and Eleni Douni Consomic analysis of genetic factors related to temporal difference of home-cage activity between B6 and MSM Ayako Ishii, Akinori Nishi, Toshihiko Shiroishi, Aki Takahashi, and Tsuyoshi Koide Genetic determinants for intramuscular fat content and water holding capacity in mice selected for high muscle mass Stefan Kärst and Gudrun A. Brockman 27 93 94 95 96 97 98 99 100 101 102 Discovering novel regulatory pathways of imflammation by ENU mutagenesis Philip L Kong, Emma Timms, Andrew Coulson, Kristina BlagoevaHubbard, Paul Potter, and Marc Feldmann Epigenetic changes in Friedreich ataxia pathogenesis and therapy S Al-Mahdawi, C Sandi, R Mouro-Pinto, V Ezzatizadeh, and Mark A Pook A mouse mutant exhibiting a unique pattern of cochlear inner hair cell degeneration Morag A. Lewis, Tracy J. Bussoli, Amy Taylor, Michael A. Cheong, Cordelia F. Langford, and Karen P. Steel A comprehensive whole-genome map of endogenous retroviral elements and their functional effects across 17 laboratory mouse strains Thomas M Keane, K Wong, J Flint, W Frankel, David J Adams Phenotyping of knockout mice using bacterial pathogens as part of the Wellcome Trust Sanger Institute’s Mouse Genetics Programme Leanne Kane, Lynda Mottram, Simon Clare, Jacqui White, Ramiro Ramirez – Solis, and Gordon Dougan Imprinted methylation vs. imprinted expression – are they necessarily associated? John D. Calaway, Hyuna Yang, Megan E. Hanson, Ezequiel C. Cambranis, Maria A. Perez-Lamigueiro, David L. Aylor, Leonard McMillan, Gary A. Churchill, Fernando Pardo-Manuel de Villena, and Elena de la Casa-Esperon Dual activity on a single locus: Existence of a novel RNA control mechanism for microRNA-650 and immunoglobulin lambda light chain variable genes in primates Sabyasachi Das, Jianxu Li, Masayuki Hirano, Christopher L. Haga, Murali Gururajan, Götz R.A. Ehrhardt, and Max D. Cooper Identification of positional candidates for Chr15 obesity quantitative trait loci using microarray and RT-PCR analysis of F2 congenic crosses in mice K Cirnski, M Stanonik, R Keber, P Juvan, R Košir, D Rozman, and Simon Horvat Crezoo - the european virtual repository for cre driver strains Christina Chandras, Michael Zouberakis, Damian Smedley, Nadia Rosenthal, and Vassilis Aidinis Host immune responses and genetic factors modulating resistance to Salmonella enterica serovar typhimurium in the inbred mouse strain SPRET/Ei: a role for neutrophils Lien Dejager, Iris Pinheiro, Pieter Bogaert, Liesbeth Huys, and Claude Libert 28 103 104 105 106 107 108 109 110 111 112 113 114 115 LPS-resistance of the mouse strain SPRET/Ei: an important role of the X-chromosome Iris Pinheiro, Lien Dejager, Tina Mahieu, Marnik Vuylsteke, and Claude Libert Complete knockout of the adrenocortical dysplasia gene encoding the shelterin protein TPP1 is associated with telomere dysfunction and early embryonic lethality Gail A Osawa, C Harris, T Kibe, T de Lange, S Kalantry, and CE Keegan The FaceBase Cre driver project: creating new mouse tool strains for clefting research Stephen A. Murray, Jocelyn Sharp, John Flaherty, Thomas Gridley, and Leah Rae Donahue Epigenome sequencing comes of age in development, differentiation and immune regulation mechanisms of mammalian research Ning Li and Jun Wang Generation of customized and read-to-use genetically engineered mice at the institute Clinique de la souris – ICS (The French Mouse Clinical Institute) Marie-Christine Birling, Andrée Dierich, Karim Essabri, Lydie Venteo, Guillaume Pavlovic, Tania Sorg, and Yann Hérault Pudding – a novel model of glomerularnephritis Paul K Potter, L Wisby, FW Tam, S Wells, T Hough, HT Cook, M Cheeseman, and SDM Brown The Harwell ageing screen Paul K Potter Gelsolin plays a role in the actin polymerization complex of hair cell stereocilia Philomena Mburu, MR Romero, Helen Hilton, Andrew Parker, Stuart Townsend, Yoshiaki Kikkawa, and SDM Brown New sterile model mouse from RIKEN ENU-based gene-driven mutagenesis Takuya Murata, Shigeru Makino, Ryutaro Fukumura, and Yoichi Gondo Genetic diversity among C57BL/6 substrains based on SNPs Kazuyuki Mekada, Ikuo Miura, Takehide Murata, Atsushi Toyoda, Mayu Hirose, Shigeharu Wakana, Kuniya Abe, and Atsushi Yoshiki Novel ENU screens for immune-related phenotypes Vera M Ripoll, Marie Hutchison, Paul Potter, and Roger Cox Grb10 modifies MPNST tumorigenesis in a mouse model of NF1 Karlyne M. Reilly, Jessica Van Schaick, Keiko Akagi, Christina DiFabio, Robert Tuskan, Sandra Burkett, and Jessica Walrath Homology mapping: development of the mouse hindlimb 29 116 117 118 119 120 121 122 123 124 musculoskeletal model using the human musculoskeletal model and Scleraxis (Scx)-GFP mouse Satoshi Oota, Nobunori Kakusho, Yosuke Ikegami, Kazuyuki Mekada, Koh Ayusawa, Hirotaka Imagawa, Yuichi Obata, Ryutaro Himeno, Hideo Yokota, Yoshihiko Nakamura, and Atsushi Yoshiki Mutation identification in mice by exome sequencing Laura Reinholdt, David Bergstrom, Steve Murray, Michelle Curtain, Carol Bult, Joel Richardson, Lucy Rowe, Mary Barter, Daniel J. Gerhardt, Mark D'Ascenzo, Todd Richmond, Tom J. Albert, Jeffrey A. Jeddeloh, Benjamin Kile, Ivo Gut, Jay Shendure, and Leah Rae Donahue Status of the US lines of the Collaborative Cross: Completed lines Darla R Miller, Ryan J Buus, Jennifer N Shockley, Stephanie D Hansen, Ginger D Shaw, Terry J Gooch, Jason S Spence, Timothy A Bell, Catherine E Welsh, Leonard McMillan, Wei Wang, Jeremy Wang, Eric Yi Liu, Kenneth F Manly, Gary A Churchill, David W Threadgill, and Fernando Pardo-Manuel de Villena Whole genome resequencing of Estonian dairy cow (Estonian Holstein) Rutt Lilleoja, Ü Jaakma, and S Kõks Hypothalamic mitochondrial dysfunction, a possible cause of anorexia Charlotte Lindfors, Ida A.K. Nilsson, Pablo M. Garcia-Roves, Aamir R. Zuberi, Mohsen Karimi, Lea Rae Donahue, Derry C. Roopenian, Jan Mulder, Mathias Uhlén, Tomas J. Ekström, Muriel T. Davisson, Tomas Hökfelt, Martin Schalling, and Jeanette E. Johansen High-throughput genotyping and quality control of gene-targeted mutations in the mouse Edward Ryder, Diane Gleeson, Debarati Bhattacharjee, Ross Cook and the Mouse Genetics Programme The Sanger Mouse Genetics Programme: High throughput characterisation of immunological phenotypes in knockout mice Mark Lucas, C Podrini, E Cambridge, S Clare, L Mottram, JK White on behalf of the Mouse Genetics Programme The Mouse Reference GENOME assembly: updates and resources Valerie A Schneider and DM Church on behalf of the Genome Reference Consortium and NCBI Genome Annotation Team dbVar and dbSNP: NCBI Databases of Simple and Structural Variations Valerie A Schneider, Tim Hefferon, John Garner, Azat Mardanov, Ming Ward, Aleksey Vinokurov, Melvin Quintos, Mike Kholodov, David Shao, John Lopez, Steve Sherry, and Deanna Church Current progress of phenotypic analysis in Japan mouse clinic 30 125 126 127 128 129 130 131 132 133 134 Tomohiro Suzuki, Hideki Kaneda, Kimio Kobayashi, Ikuo Miura, Tamio Furuse, Ikuko Yamada, Osamu Minowa, Hideaki Toki, Hiromi Motegi, Maki Inoue, Tetsuo Noda, Hiroshi Maduya, and Sigeharu Wakana Hyperactive mice show a cluster of associated metabolic features Jacqueline K White, Anna-Karin Gerdin, Christine Podrini on behalf of the Mouse Genetics Programme, and Karen P Steel Multiplicity of phenotypes in early generations of the mouse diversity outcross population Karen L Svenson and GA Churchill SDOP-DB: a database for international sharing and standardization of mouse phenotyping protocols Nobuhiko Tanaka, Kazunori Waki, Hideki Kaneda, Tomohiro Suzuki, Ikuko Yamada, Tamio Furuse, Kimio Kobayashi, Hiromi Motegi, Hideaki Toki, Maki Inoue, Osamu Minowa, Tetsuo Noda, Shigeharu Wakana, and Hiroshi Masuya Modeling human chromosome 21-associated mental retardation disorders in mice Tao Yu, Zhongyou Li, Steven J Clapcote, Zhengping Jia, Chunhong Liu, Annie Pao, Sei-ichi Matsui, Norma J Nowak, John C Roder, Chu Chen, Allan Bradley, and Y. Eugene Yu Taiwan Mouse Clinic – an experiment for mouse phenotyping service Jeffrey Jong-Young Yen, Yen-Hui Chen, and Ya-Wen Hsaio Initial assembly of 17 mouse strains from the mouse genomes project Guy Slater, Thomas M Keane, Jared Simpson, Aylwyn Scally, Xiangchao Gan, Richard Mott, Jonathan Flint, and David J Adams Clustered protocadherin genes: Genetic codes for generating the complex brain Takeshi Yagi, Shunsuke Toyoda, Yasushi Itoga, Keizo Hirano, Atsushi Okayama, Shinichi Yokota, Shota Katori, Teruyoshi Hirayama, and Takahiro Hirabayashi Comprehensive phenotyping of mouse models Tania Sorg, Marie-France Champy, Elodie Bedu, Roy Combe, Hamid Meziane, and Yann Herault Targeted and whole exome resequencing of mouse monogenic mutants, quantitative trait loci and cancer models David R. Beier, Jennifer L. Moran, Evan Mauceli, Snaevar Sigurdsson, Tim Fennell, Lauren Ambrogio, Miriam H. Meisler, John C. Schimenti, Karen Cichowski, Laura Reinholdt, Jane Wilkinson, Stacey B. Gabriel, Federica di Palma, and Kerstin Lindblad-Toh Dynamic transcriptomes during neural differentiation of human 31 135 136 137 138 139 140 141 142 143 144 embryonic stem cells revealed by short, long, and paired-end sequencing Jia Qian Wu, Lukas Habegger, Parinya Noisa, Anna Szekely, Caihong Qiu, Stephen Hutchison, Debasish Raha, Michael Egholm, Haifan Lin, Sherman Weissman, Wei Cui, Mark Gerstein, and Michael Snyder Improving mouse genome annotation through new technologies Laurens Wilming, Gary Saunders, Markus Brosch, Gavin Laird, Adam Frankish, and Jennifer Harrow Genomic alterations in tumors of the Mcm4Chaos3 breast cancer mouse model Marsha D. Wallace, Ethan Cerami, Stefan Stefanov, Lishuang Shen, and John C. Schimenti A missense mutation in the novel WD-repeat-containing protein, WDR81, causes Purkinje cell and photoreceptor cell death in the ENU-induced neurological mouse mutant nur5 Maria Traka, Kathleen J. Millen, and Brian Popko Using whole genome next-generation sequencing as a tool to find ENU-induced mutations Michelle Simon, S Greenaway, P Denny, A-M Mallon, and JM Hancock The Sanger Mouse Genetics Programme; High throughput morphological analysis of the eye Valerie E. Vancollie, J Estabel, D Sunter, VB Mahajan, SH Tsang, MC Naumann, M Mahajan, JK White, on behalf of the Mouse Genetics Programme A Cellular Genetics Platform to identify pharmacogenetic toxicity pathways O Suzuki, N Butz, M Pletcher, A Su, B Steffy, D Scoville, A Frick, J Trask, R Thomas, and Tim Wiltshire Lessons from phenotype of KO mouse, The role of Snur4 on adipogenesis and insulin sensitivity Jae Hoon Shin, Il Yong Kim, Yo Na Kim, Ji Won Choi, Kyung Jin Roh, Mi Ra Sohn, Cheol soo Choi, Yun Soo Bae, and Je Kyung Seon Epiblast-ablation of Sox2 expression leads to neurocristopathies in mouse embryos Nikolaos Mandalos, Marannia Saridaki, Jessica Lea Harper, Anastasia Kotsoni, Aris N. Economides, and Eumorphia Remboutsika A comprehensive catalogue of structural variation from! ! 17! ! inbred mouse strains Kim Wong, B Yalcin, T Keane, M Goodson, A Agam, C Nellaker, J Stalker, J Flint, and D Adams Collection and distribution of CRE/FLP-drivers at the Riken Bioresource Center 32 145 146 147 Atsushi Yoshiki, Kazuyuki Mekada, Hatsumi Nakata, Ayumi Murakami, Masayo Kadota, Fumio Ike, Noriko Hiraiwa, Kaoru Fukami-Kobayashi, Yuichi Obata Analysis of the expression profile of CRH-POMC system genes in vitiligo skin biopsies Ene Reimann,, K Kingo, M Karelson, T Salum, E Aunin, P Reemann, K Abram, E Vasar, H Silm, and S Kõks Effects of a high spontaneous mutation rate in mammalian germline by using mutator mice modified replicative DNA polymerase delta Arikuni Uchimura, Yuko Hidaka, Ikuo Miura, Shigeharu Wakana, and Takeshi Yagi Tooling up MGI for a deluge in mouse phenotype and disease model data Anna Anagnostopoulos, Janan Eppig, and Mouse Genome Informatics Group 33 ABSTRACTS 35 ABSTRACT 1 THE F2 POPULATION FOR DISSECTING HOST SUSCEPTIBILITY TO PERIODONTITIS Ariel Shusterman1, Yael Houri-Haddad1, Ervin I Weiss1, and Fuad A Iraqi2 1 Department of Prosthodontics Hadassah Medical Center, Israel 2 Deptartment of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Israel Introduction: Epidemiological studies in human have suggested that susceptibility to periodontal disease is controlled by host genetic factors. Recently, we have shown quantitatively by using microCT that inbred mouse strains respond differently to the infection. Based on these results, we have developed 408 mice of A/JxBALB/cJ F2 resource population, knowing that A/J and BALB/cJ are the resistant and susceptible founder strains for the infection, respectively. Objective: 1) To determine the phenotypic response; i.e. alveolar bone loss, of all progenies of the F2 population. 2) To perform genome wide search for quantitative trait loci (QTL) associated with host susceptibility to periodontitis using selective genotyping approach. Materials and Methods: 408 mice of the F2 population were generated by a controlled mating scheme of the two parental inbred founders. All mice were orally infected with Porphyromonas gingivalis and Fusobacterium nucleatum. The infection was repeated three times at 2 day intervals. Six weeks following the final infection, the maxillary jaws were harvested and alveolar bone loss was quantified using microCT. Results: The mean of phenotypic response of F2 population (0.0083mm3) was intermediate between the two parental founders, 0.0103mm3 and 0.0062mm3 for A/J and BABLB/cJ, respectively. The results suggest no genomic imprinting (mitochondrial) effect as well as no significant sex effect. Conclusions: The phenotypic variety of F2 population suggests more than one quantitative trait loci (QTL) affecting host susceptibility to periodontitis. This is the first study which uses mixed infection as experimental periodontitis that: (i) compares different F2 groups (ii) analyze quantitatively a high number of mice by microCT. Currently, we are finalizing the genotyping of 38% of the phenotypic extremes of F2 resource population with 1500 single nucleotide polymorphic (SNP) markers, covering the whole genome which allows subsequently performing QTL mapping analysis. Mapping the QTL will point candidate gene involved in periodontitis. 37 ABSTRACT 2 MOUSE GENETIC MODEL OF CARDIOVIRULENT COXSACKIEVIRUS B3 INFECTION Sean A. Wiltshire1,2 and Silvia M. Vidal1,2 1 Department of Human Genetics, McGill University 2 McGill Centre for the Study of Host Resistance, McGill University In North America, up to 50% of Myocarditis can be attributed to Coxsackie virus B3 (CVB3) infection. Host genetics, viral factors and the environment of their interaction determine variable susceptibility to pathogenesis within exposed human populations. Virally induced pathology in the mouse model mimics the progression of disease in humans. Inbred mouse strains respond differently to CVB3 infection including susceptible strains with A/J background and resistant strains with C57BL/10 background. Through the analysis of a segregating (A/JXB10.A)F2 cross we have recently detected three loci linked to susceptibility to CVB3 induced pathology: Vms1, Vms2 and Vms3 (viral myocarditis susceptibility). We have subsequently confirmed the existence of Vms1 through the use of the consomic substitution strain CSS3 and the localization of Vms1 through the analysis of a segregating (CSS3 x B6)F2 cross. In order to identify causal genes we have undertaken a congenic and subcongenic approach to dissecting Vms1. Ongoing analysis of potential cell autonomous mechanisms in isolated adult ventricular cardiomycytes may reveal novel immune mechanisms which control infection in the heart. 38 ABSTRACT 3 ASSOCIATION BETWEEN SPATIAL PROXIMITY AND FUNCTIONAL SIMILARITY IN HUMAN GENOME Ekaterina Khraneeva, Andrey Mironov, and Mikhail Gelfand Faculty of Bioengineering and Bioinformatics, M.V.Lomonosov Moscow State University, Russia A.A.Kharkevich Institute for Information Transmission Problems, RAS, Moscow, Russia Recent progress in determination of 3D structure of nuclear chromatin allows one to study correlations between spatial proximity of genome domains and their functional state. We combined the 3D data from (Lieberman-Aiden et al. 2009) with the results of several high-throughput studies of the chromatin functional state. All pairs of regions from different chromosomes were divided into groups according to their proximity, and the distribution of various chromatin marks was calculated within these groups and then compared between the groups. The results show that, indeed, gene regions that are spatially close tend to have similar patterns of histone modifications (Encode 2004), methylation state (Encode 2004), DNAse hypersensitivity (Encode 2004), and expression level (Lieberman-Aiden et al. 2009). We also analyzed chimeric transcripts as determined by genome mapping of paired-read RNA-Seq data (Xu et al. 2010 and Berger et al. 2010) and observed that the frequency of pairs mapping to two different genome loci is higher among spatially proximal regions. This could be caused by (at least) two reasons: trans-splicing and genome rearrangement compared to the reference genome (either somatic recombination or allelic forms). To test these possibilities we analyzed the data on genome trans-chromosomal rearrangements (Kidd et al. 2008) and demonstrated that again the number of chimeric pairs is higher among spatially close regions, although the effect is weaker than the one observed in the transcriptomic data. Hence, there remains a possibility that the additional chimeras indeed originate from trans-splicing. Finally, we analyzed GO annotations of genes in spatially closest pairs of genome regions. The respective sets were enriched for GO categories related to signal transduction and regulation of transcription. In addition, pairs of spatially close regions had more common GO terms than control random pairs. Taken together, these results seem to demonstrate the presence of co-regulated genome domains formed by regions of different chromosomes. 39 ABSTRACT 4 OLFACTORY RECEPTOR GENE FAMILY EVOLUTION IN MARSUPIALS Amir Mohammadi, Margaret L. Delbridge, and Jennifer A. Marshal Graves ARC Centre of Excellence for Kangaroo Genomics and Research School of Biology, The Australian National University, Canberra, Australia The ability to smell is critical to most mammals, so it is not surprising that the olfactory receptor genes are the largest gene family in genomes of eutherian (placental) mammals. Pouch-bearing mammals (marsupials) diverged from eutherian mammals about 148 million years ago (MYA), so we wished to compare the size and diversity of this gene family between the two mammalian infracalsses, as well as between an American and an Australian marsupial. The complete genomes of two marsupials have now been sequenced, allowing us to look at the conservation of large gene families in this group for the first time. We therefore examined the composition of the olfactory receptor (OR) gene family in the Brazilian short-tailed opossum (Monodelphis domestica) and the tammar wallaby (Macropus eugenii), which diverged about 70 MYA. We isolated all olfactory receptor gene sequences from the first assembly of the Australian model marsupial, the tammar wallaby, classified them by phylogenetic analysis and compared the distribution with the OR repertoire of the opossum. Genomic location of most of ORG clusters was also identified in tammar wallaby by using fluorescence in situ hybridization. Sequence homology, and the genomic distribution of OR genes is well conserved between these two distantly related marsupials. This high level of conservation contrasts with the divergence of ORG families between eutherian mammals with similar divergence times. Our results suggest a significant role for adaptive evolution, rather than genomic drift, in the evolution of chemosensory genes in vertebrates. 40 ABSTRACT 5 ADVANCING PATERNAL AGE IS ASSOCIATED WITH DEFICITS IN SOCIAL AND EXPLORATORY BEHAVIOURS IN THE OFFSPRING Rebecca G. Smith1, Rachel L. Kember1, Leonard C. Schalkwyk1, Joseph D Buxbaum2, Abraham Reichenberg1, Cathy Fernandes3*, and Jonathan Mill1* 1 Medical Research Council Social Genetic and Developmental Psychiatry Centre, King's College London, London, United Kingdom 2 Department of Psychiatry, Mount Sinai School of Medicine, New York, New York, United States of America 3 Department of Psychosis, King's College London, London, United Kingdom * Contributed equally to this work Many epidemiological studies have demonstrated an association between advanced paternal age and risk for several psychiatric disorders including autism. In this study we used an animal model to investigate the effects of advanced paternal age on behaviour in the offspring. C57BL/6J offspring were bred from fathers of two different ages, two months old (to represent young fathers) and 10 months old (to represent older fathers) using mothers aged two months in both groups. The offspring underwent open field, holeboard and social recognition tasks to explore differences in their behaviours. We observed that the offspring of older fathers were found to engage in significantly less social and exploratory behaviours than the offspring of younger fathers without any reduction in their motor activity. Brain and peripheral tissues were obtained from both parents and offspring and used to investigate de novo genomic changes associated with increased paternal age using Nimblegen 720K CGH microarrays. Spermatozoa undergo multiple divisions throughout the male lifespan, potentially leading to a higher incidence of de novo copy number variation (CNV) in any resulting offspring. Given recent human data reporting an increased burden of CNVs in autism and other neurodevelopmental disorders, we speculated that such genomic changes could be mediating the behavioural changes we observed. Our study provides the strongest evidence for deleterious effects of advancing paternal age on social and exploratory behaviour and suggests that de-novo chromosomal changes are plausible explanatory factors. 41 ABSTRACT 6 GENOME-WIDE DIFFERENTIAL METHYLATION PATTERNS IN INTERSUBSPECIFIC HYBRID MICE John D. Calaway1,2,3,4, Hyuna Yang5, Elena de la Casa-Esperon7, Megan E. Hanson6, Ezequiel C. Cambranis6, Maria A. Perez-Lamigueiro6, David L. Aylor2, Leonard McMillan8, Gary A. Churchill5, and Fernando Pardo-Manuel de Villena1,2,3,4 1 Curriculum in Genetics and Molecular Biology, University of North Carolina University of North Carolina, Chapel Hill, NC 2 Department of Genetics, University of North Carolina, NC, USA 3 Lineberger Comprehensive Cancer Center, University of North Carolina, NC, USA 4 Carolina Center for Genome Sciences, University of North Carolina, NC, USA 5 The Jackson Laboratory, Bar Harbor, ME, USA 6 Department of Biology, University of Texas Arlington, Arlington, TX, USA 7 Regional Center for Biomedical Research (C.R.I.B.) Albacete Science and Technology Park/ University of Castilla-La Mancha, Spain 8 Department of Computer Science, University of North Carolina, NC, USA Here we report the first genome-wide survey of the mouse differential methylome. Using the high-density Mouse Diversity genotyping array, we exploited the whole-genome amplification step of the Affymetrix genotyping protocol by adding a pre-digestion step with either methylation-sensitive HpaII or methylation-insensitive MspI endonucleases. Reported previously as methylation-sensitive single nucleotide polymorphism analysis (MSNP), this technique compares genotyping calls between undigested and HpaII samples to determine allele-specific methylation. We expanded the analysis by directly analyzing probe intensity data to classify methylation patterns. By doing so, we identified stably inherited epigenetic states attributed to parent-of-origin or strain-of-origin. In addition, global methylation analysis suggests that female mice exhibit a higher level of autosomal methylation when compared to males. This approach identified three novel differential methylated regions (DMR). We chose one located in the 3’ UTR of Actn1 for method validation using two independent molecular assays. Our findings suggest that the mouse Actn1 DMR has moderate tissue and strain specificity and a comparison of the orthologous regions in rat and human reveal a possible rodent-specific DMR. Lastly, we demonstrate that the Actn1 DMR influences gene expression based on Collaborative Cross mice. 42 ABSTRACT 7 OPTIMIZATION OF A GENOME-WIDE, ALLELE-SPECIFIC DNA METHYLATION ASSAY FOR THE MOUSE John P. Didion1, John D. Calaway1, Isa Kemal Pakatci2, Leonard McMillan2, and Fernando Pardo-Manuel de Villena1 1 Department of Genetics, Lineberger Comprehensive Cancer Center, Carolina Center for Genome Science, University of North Carolina Chapel Hill, NC, USA 2 Department of Computer Science, University of North Carolina Chapel Hill, NC, USA Cytosine (CpG) methylation is involved in many critical cellular functions, including genomic imprinting, Xchromosome inactivation and silencing of retrotransposons and disease-related genes. Genomic studies increasingly call for consideration of genome-wide methylation patterns, however the methods available for assaying methylation in the mouse are currently limited either by expense or the number and distribution of CpGs interrogated. This later shortcoming is the result of most methylation studies focusing on CpG islands regions of high CpG concentration whose methylation status tends to be correlated with the activity of nearby genes. However, recent studies have shown that CpGs proximal to islands (as much as 4kb distant) are highly correlated with allele-specific gene expression and thus of great interest to researchers. We have optimized an existing method based on genotyping arrays to assay allele-specific CpG methylation using the Mouse Diversity array and a combination of methylation-sensitive and -insensitive restriction endonucleases. Our method provides fully informative coverage of more than 100,000 CpGs, and partially informative coverage of an additional 90,000 CpGs, most with at least one technical replicate, and roughly even genomic distribution. We present here a general bioinformatic approach for optimization of the Mouse Diversity array to address a range of biological parameters. In addition, we have applied our method to study parent-of-origin and strain-specific effects on methylation in the Collaborative Cross, and have thus generated an optimized data set of untreated controls that we provide as a community resource. Our findings will reduce the expense and challenge of incorporating genome-wide methylation analysis in genomic studies. 43 ABSTRACT 8 CHARACTERISATION OF LATERALITY AND CARDIAC DEVELOPMENTAL DEFECTS IN THE K27 MUTANT MOUSE Louise Stephen, K Mitchell, and K Hentges University of Manchester, UK Congenital heart defects account for 3% of all infant deaths and many more prenatal. Cardiac development is a highly regulated process, reliant on the cooperation of a vast number of genes. Whilst advances have been made in defining the physiological processes occurring during cardiac development, the genetic regulation is less well understood. Random mutagenesis screens offer a phenotype-driven approach to producing novel mammalian models of human congenital birth defects. We have applied this approach to the study of cardiac development and are currently characterising the K27 mutant mouse, identified from a balancer chromosome chemical mutagenesis screen. The K27 mutant exhibits a recessive embryonic lethal phenotype, apparent from embryonic day (E)9.5, with death occurring by E12. The K27 mutant exhibits a developmental delay and abnormal cardiac morphology. Approximately 50% of mutants demonstrate a reversal of cardiac looping. Analysis of Nkx2-5 and Fgf8 has revealed that both the primary and secondary heart fields are correctly specified in mutants. Likewise K27 mutants express Mef2c in a pattern indistinguishable from control littermates, indicating that cardiac muscle is present in mutants. Laterality defects suggested by the reversal in looping were confirmed by investigation of the expression of genes required for left-right asymmetry, including down-regulation of the Sonic hedgehog-regulated Gli1 gene in mutants. Meiotic mapping has refined the K27 candidate region to a 5 Mb interval on mouse Chromosome 11. This region contains over 100 genes, none of which are currently known to have an established role in cardiac development. Further work will include using high-throughput sequencing to identify the K27 gene. Continued characterisation of the K27 mutants will define the role the K27 gene plays in cardiac development and left-right asymmetry. Combined these studies will identify a novel gene that contributes to our understanding of cardiac congenital birth defects and of left-right axis formation. 44 ABSTRACT 9 KYOTO EPILEPTIC RAT (KER): A NOVEL RAT MODEL OF KCNA1 MUTATION SHOWING SPONTANEOUS SEIZURES AND MUSCLE INVOLUNTRY CONTRACTIONS Saeko Ishida1, Tomoji Mashimo1, Takeshi Nishio2, Yu Sakamoto3, Shuji Kaneko 3, and Tadao Serikawa1 1 Institute of Laboratory animals,and 2Department of Integrative Brain Science, Graduate School of Medicine, 3 Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Japan Epilepsy is a neurological disorder characterized by unprovoked, recurring seizures with a wide range of manifestations. We have so far performed phenotype-driven ENU mutagenesis in rats to establish animal models of epilepsy. Here we identified a rat exhibiting involuntary muscle movements and convulsive seizures, named Kyoto Epileptic Rat (KER). We characterized KER pathologically and physiologically and identified the responsible gene (Ker) for involuntary muscle movements by positional cloning. KER was backcrossed by more than 10 generations against F344/NSlc background strain to remove latent ENU-induced mutations in other chromosomal regions. In KER, generalized convulsive seizures were observed from 10 weeks of age, and most of the rats died until 7 month of age. By electroencephalogram (EEG) recording, polyspikes and polyspike-and-wave discharges were typically observed during the seizures. By electromyogram (EMG) analysis of the anterior limb muscle, 7 Hz rhythmic spikes were detected during the interictal phase. Using 97 (KER x BN/SsNSlc) x BN backcross progeny, Ker was mapped within the 10Mb region of rat Chromosome 4. Within the region, a missense mutation T925A (S309T) in voltage-gated potassium channel shaker-related subfamily member 1 (Kcna1) gene was identified by sequencing analysis. The mutant homotetrameric Kv1.1 channels were non-functional in Xenopus oocytes. In humans, mutations of KCNA1 cause autosomal dominant episodic ataxia type 1 (EA1) characterized by brief episodes of ataxia associated with continuous myokymia. In addition, the patients with EA1 are ten times more likely to develop epilepsy than normal individuals. In mice, Kcna1 knockout mutation shows a recessive seizure disorders (Smart et al., Neuron 1998), while V408A missense mutation shows no seizures, but exhibits stress-induced loss of motor coordination (Herson et al., Nat Neurosci. 2003). Since KER shows muscular symptoms and spontaneous seizures as an autosomal dominant manner, which are different from the phenotype of the mouse models, KER can be a novel animal model of EA1 to understand the underling mechanism of the KCNA1 diseases. 45 ABSTRACT 10 IDENTIFYING MODIFIER GENES OF MPNSTS IN THE NF1;P53CIS MOUSE MODEL OF NEUROFIBROMATOSIS TYPE 1. Jessica A Van Schaick1,2, Keiko Akagi3, Sandra Burkett1, Christina DiFabio1, Robert Tuskan1, Jessica Walrath1, and Karlyne Reilly1 1 National Cancer Institute, Frederick, MD, USA 2 The George Washington University, Washington, DC, USA 3 The Ohio State University, Columbus, OH, USA The current study aimed to identify modifier genes of malignant peripheral nerve sheath tumors (MPNSTs) in the Nf1;p53cis (NPcis) mouse model of NF1. Previous studies have shown that the incidence of MPNST development in the NPcis mouse model is affected by the parental transmission of the mutant Chromosome 11. In this study microarray analysis was used to examine gene expression differences between MPNST primary tumors derived from NPcis mice varying in inheritance of the NPcis chromosome from the mother (NPcis maternal) or father (NPcis paternal). Grb10 was found to be more highly expressed in NPcis maternal MPNSTs. Zrsr1 was found to be more highly expressed in NPcis paternal MPNSTs. qPCR was used to validate both gene expression differences. We chose to focus first on Grb10 due to its role as a cytoplasmic signaling adapter protein. Fluorescence in situ hybridization was used to examine the presence of Grb10 on Chromosome 11. Grb10 was found to be lost more frequently in NPcis paternal MPNST cell lines, potentially contributing to the decrease in Grb10 gene expression seen in these tumors. Grb10 is paternally imprinted in the periphery of the mouse, therefore we examined Grb10 isoform expression and found paternal and maternal isoforms expressed in the MPNSTs. Due to these results we are examining whether loss of imprinting is contributing to tumorigenesis. Finally we have generated NPcis;Grb10cis mutant mice. Preliminary studies indicate that NPcis;Grb10cis maternal mice have an increase in PNST incidence and a decrease in survival. Our studies indicate that Grb10 is a candidate modifier gene of MPNSTs, acting in a tumor suppressive manner. 46 ABSTRACT 11 A NOVEL GENETIC MODEL OF OSTEOPOROSIS BY OVEREXPRESSION OF HUMAN RANKL IN TRANSGENIC MICE Alexandra Niti1, R Dacquin2, V Rinotas1, P Jurdic2 and Eleni Douni1 1 Institute of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece 2 Institut de Génomique Fonctionnelle de Lyon, UMR5242 CNRS/INRA/UCBL/ENS Ecole Normale Supérieure de Lyon, France Receptor Activator of Nuclear Factor-κB Ligand (RANKL, official symbol TNFSF11) is a central regulator of bone remodelling by mediating osteoclastinduced bone resorption, whereas specific inhibition of RANKL by a fully human monoclonal antibody effectively reduces the incidence of fractures in postmenopausal women. We have recently generated transgenic mice overexpressing human RANKL (TghuRANKL) in order to model human RANKL-mediated pathologies. To achieve a correct pattern of human RANKL expression in the mouse, a transgenic 200kb genomic fragment containing the whole human RANKL gene has been introduced into the mouse genome. Among the five independent transgenic lines obtained, a striking skeletal phenotype was observed in the highest copy number TghuRANKL founder characterized by multiple bone fractures, increased osteoclast numbers, severe trabecular bone loss and cortical porosity. A similar but milder bone phenotype was identified by another high copy TghuRANKL line that developed both trabecular bone loss and cortical bone porosity by the age of 3 months. Additionally, a low copy number TghuRANKL line developed exclusively trabecular bone loss by the age of 3 months. The observed phenotypes in various TghuRANKL lines developed in both sexes, whereas the levels of human RANKL expression were correlated with disease severity. Notably, the osteopetrotic phenotype of mutant mice expressing an inactive form of the endogenous RANKL protein was rescued upon crossing these mice with TghuRANKL mice, showing that the transgenic human RANKL protein is fully active in the mouse. These novel human RANKL transgenic mice represent a unique tool for understanding the pathogenic mechanisms that cause bone resorption and for the evaluation of novel therapeutic approaches targeting RANKL-mediated pathologies such as osteoporosis. 47 ABSTRACT 12 POSITIVE SELECTION MIGHT CONTRIBUTE TO FUNCTIONAL DIVERGENCE OF NAIPS FOLLOWING GENE DUPLICATION IN MAMMALS DURING THE EVOLUTION Zhaoliang Su1, Chenglin Zhou4, Jianguo Chen2*, Shengjun Wang1*, and Huaxi Xu1* 1 Department of Immunology & Laboratory Immunology, Jiangsu University, Zhenjiang, China 2 The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China 3 Zhenjiang Entry-exit Inspection and Quarantine Bureau, Jiangsu, China The neuronal apoptosis inhibitor proteins (NAIPs) are members of NLRs (Nodlike receptors) protein family. Recent researches have shown that some NAIP genes were strongly associated with both mammalian innate immunity and many diseases, whereas some NAIP genes had been pseudogenization. So the present works were to clarify whether the functional divergence had appeared, as well as whether natural selection played an important role during their evolution. Our results showed that NAIP genes had produced functional divergence and appeared genes duplicated before the divergence; during evolution, lineagespecific duplications of NAIP genes were gradually replaced by pseudogenization and NAIP genes were driven by the positive selection; and the selective force changed the functional sites of NAIP, affected the tertiary structure of NAIP, and then produced the functional divergence. Our results also indicated that the positive selection and functional divergence of NAIP might provide mammals advantages to adapt evolution of geographical environment. 48 ABSTRACT 13 A FORWARD GENETICS APPROACH IDENTIFIES A NOVEL MEMBER OF THE DNAJC FAMILY THAT CAUSES NEUROMUSCULAR DISEASE IN MICE Fotios Ioakeimidis, Vagelis Rinotas, Eleni Makrinou, George Kollias, and Eleni Douni Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece Neuromuscular diseases encompass a wide range of clinical conditions remaining incurable while the genetic and molecular basis of most of these conditions remains unknown. Using mouse ENU mutagenesis we have identified a novel autosomal recessive neuromuscular phenotype. This phenotype manifests with hind limb weakness, progressing with muscle atrophy, reduced body weight, generalized paralysis and death within 30 days after birth. Genetic linkage analysis mapped the causal mutation within a 2Mbp interval containing 24 genes. Sequencing revealed an intronic T>A transversion within a member of the DnaJ homolog, subfamily C (DNAJC) of heat shock proteins (Hsp40s). This is a novel gene with completely unknown function, whose protein is suggested to be located in the mitochondrion, suggestion that is supported by our own confocal imaging data. The intronic mutation generates a novel splicing acceptor site resulting in the insertion of 109 bp in the mature transcript, causing a frameshift of the last 50 aminoacids of the protein that are replaced by 44 different aminoacids. This DNAJC family member shares 96% identity with its human ortholog, highlighting the functional importance of the protein. Our expression analysis data show constitutive and high expression of the normal gene in all tissues studied, neuronal and non neuronal. To genetically prove the causality of this mutation in our phenotype, we have generated BAC transgenic mice for the human DNAJC gene in order to rescue the neuromuscular phenotype. Our current studies focus on the histological and immunohistochemical characterization of the CNS in our mutant mice, and analysis of the expression pattern and function of the normal and the mutated protein. The identification of a novel DNAJC protein involved in neuromuscular disease and future functional characterization of this protein can shed light into new pathogenetic pathways involved in neuromuscular diseases. 49 ABSTRACT 14 GENE EXPRESSION STUDIES OF ncRNAS WITHIN THE ODZ4 LOCUS Tyler Buit, Chiao-Ling Lo, Feichen Shen, and Amy C. Lossie Department of Animal Sciences, Purdue University, West Lafayette, IN, USA The Odz4 allelic series is comprised of five phenotypically distinct embryonic lethal mouse mutants, which are characterized by abnormal embryonic and extraembryonic mesoderm development, irregular somite formation, imperfect patterning of the skeleton and defective maternal blood flow to the embryo. Odz4 gene regulation is complicated and not well understood. Whole transcriptome and cross-species comparison studies suggest that the 735 kb locus produces over 20 alternatively spliced transcripts, contains over 800 CAGE tags and includes 93 highly conserved non-coding elements (HCEs). These HCEs are >200bp long and exhibit ≥70% identity among five mammalian species. Similar highly-conserved DNA sequences can contain mRNA-like noncoding RNAs that control developmental gene expression. We hypothesize that several of these HCEs will encode cis-acting ncRNAs that direct alternative splicing at the Odz4 locus. Our long-term goal is to determine the role of these HCEs in directing Odz4 transcription during development. Using RT-PCR, we demonstrated that 92 of the 93 HCEs are expressed in the adult and/or at various embryonic stages in normal embryos. Each transcript exhibited a unique developmental expression profile. One HCE was detected at the blastocyst stage, which is before the primary Odz4 transcripts are produced. Six HCEs were detected in E6.5 embryos, an important stage in ODZ4 signaling. Northern and RT-PCR analyses indicate that although several of the ncRNAs are new Odz4 exons, at least 60 of these HCEs are expressed sequences of unknown function, all of which lie within 5 kb of an Odz4 CAGE tag. In situ hybridization studies at later time points indicate that these six HCEs demonstrate dynamic expression profiles. For example, at E9.5, Hce39 is restricted to punctate regions in the limbbud, brain and first brachial arch. By E12.5, we detect broad expression across the neural ectoderm, with high-levels at the midbrain-hindbrain junction. RT-PCR and in situ hybridization studies of the HCEs in Odz4 mutant embryos is ongoing, and future studies are aimed at determining the effects of these ncRNAs in specific Odz4 mutants. 50 ABSTRACT 15 EARLY LIFE STRESS IS ASSOCIATED WITH ALTERED BEHAVIOUR AND EPIGENETIC ALTERATIONS TO NR3C1, AVP AND NR4A1 Rachel L. Kember, Emma Dempster, Leonard C. Schalkwyk, Jonathan Mill, and Cathy Fernandes Institute of Psychiatry, Kings College London, UK Stressful early life environments have consistently been implicated in the development of adverse behavioural phenotypes. Environmental effects can be mediated by epigenetic processes, whereby changes in epigenetic marks due to environment can produce long lasting changes to phenotype. A mouse model is an ideal way to disentangle the contributions of environment, genetics and epigenetics towards phenotypic outcome, as it allows both the genetic and the environmental contribution to be controlled. The current study investigates the behavioural effects of a model of early life stress (maternal separation) in mouse using two inbred strains (C57BL/6J and DBA/2J). A battery of behavioural tests demonstrates that the maternal separation group exhibit phenotypic differences when compared to controls, including variation in anxiety levels. Corticosterone levels, measured at baseline and post-forced swim test, show increased hormone levels in C57BL/6J maternally separated animals after the forced swim. We identified three candidate genes that have previously shown associations with early life stress. Epigenetic changes in the glucocorticoid receptor (Nr3c1) and arginine vasopressin (Avp) genes have both been associated with poor maternal behaviour and maternal separation, respectively. Additionally, nerve growth factor IB (Nr4a1) is rapidly induced by various stress responses as an early response mechanism. I will present findings that show significant methylation differences in the hippocampus of maternally separated animals compared to controls at CpG sites in the promoter regions of these genes. Furthermore, preliminary data from a genomewide methylation assay will be presented. Studies such as these will allow further disentanglement of environment, genotype and epigenetic mechanisms in the contribution towards adverse phenotypes. 51 ABSTRACT 16 STRESS MECHANISMS, INFLAMMATION, INFECTION AND HOST RESISTANCE Katia P. Karalis Biomedical Research Foundation of the Academy of Athens (BRFAA), Greece and Children’s Hospital, Harvard Medical School, Boston, USA Stress response, a process well-preserved in mammals, is mediated by the activated hypothalamic-pituitary-adrenal (HPA) axis. Immune system challenges such as inflammatory and infectious stimuli activate stress response leading to release of glucocorticoid. The latter is a very potent immunosuppressive and anti-inflammatory agent when administered exogenously. Glucocorticoid acts through binding to glucocorticoid receptor (GR), two types α and β have been identified in humans with antagonistic effects in some cases. In addition to the GRs abundance, the local effects of glucocorticoid are regulated by the expression of 11betaHSD, an enzyme that modulates the availability of biologically active levels of glucocorticoid, in a tissue-specific manner. The hypothalamic mediator of the stress response, Corticotropin Releasing Hormone or Factor (CRH/CRF), is also expressed in various peripheral tissues and exerts potent immunomodulatory effects. This factor belongs to a family of peptides acting, although with different affinity, via two G-protein coupled receptors (CRF receptor 1 and 2) expressed in several sites in the nervous system and in a plethora of peripheral tissues. Studies from our and other laboratories have demonstrated the bidirectional, direct and indirect, effects of CRF in inflammatory processes and have started to elucidate the mechanisms mediating these actions of CRF. In this presentation we will review the evidence that supports the critical role of the, so-called, stress hormones in the resistance to inflammatory challenges. Next, we will present our findings from mouse models demonstrating a dual role of the stress system in host resistance and the molecular pathways implicated. Finally, we will discuss the possibilities for novel therapeutic applications for inflammatory conditions, given the challenges provided by the complex regulation of the genes involved in the stress response. 52 ABSTRACT 17 PHYSIOLOGIC CONSEQUENCES OF CONSTITUTIVE MTOR INHIBITION: ALTERATIONS IN CELL/ORGAN SIZE AND IMMUNE CELL DEVELOPMENT Shuling Zhang1, Julie A. Readinger2, Wendy DuBois1, Mirkka Janka-Junttila3, Richard Robinson1, Magaret Pruitt1, Val Bliskovsky1, Julie Z. Wu2, Kaori Sakakibara1, Jyoti Patel1, Carole A. Parent3, Lino Tessarollo4, Pamela L. Schwartzberg2, and Beverly A. Mock1 1 Laboratory of Cancer Biology and Genetics, CCR/NCI, 2Genetic Disease Research Branch, NHGRI, and 3Laboratory of Cellular and Molecular Biology, CCR/NCI, 4Mouse Cancer Genetics Program, CCR/NCI Mammalian TOR (MTOR) regulates cell growth, proliferation and migration. Because Mtor knock-outs are embryonic lethal, we generated a hypomorphic mouse with a neo-insertion that partially disrupts MTOR transcription and reduces its expression, thus creating a physiologic model of TORC1/TORC2 inhibition. Homozygous knock-in mice exhibited reductions in body, organ and cell size. Although reductions in most organ sizes were proportional to decreased body weight, spleens were disproportionately smaller. Decreases in the total number of T cells, particularly memory cells, and reduced responses to chemokines suggested alterations in T cell homing/homeostasis. TCRstimulated T cells also proliferated less, produced lower cytokine levels, and induced an increased number of regulatory T cells (iTreg). Surprisingly, B cell developmental effects were more pronounced, with a partial block in B cell development in bone marrow, altered splenic populations, decreased migration to chemokines, and decreased proliferation. Moreover, AKT phosphorylation was increased in activated B cells, reminiscent of cancers treated with rapamycin. Furthermore, decreased neutrophil numbers were observed in the spleen, despite normal development and migration in the bone marrow. Thus, MTOR is required for the maturation and differentiation of multiple immune cell lineages. These mice provide a novel platform for studying consequences of constitutively reduced TORC1/TORC2 activity. 53 ABSTRACT 18 SEVERE COMBINED IMMUNODEFICIENCY (SCID) RATS GENERATED BY ZINC FINGER NUCLEASE TECHNOLOGY Tomoji Mashimo, Akiko Takizawa, Birger Voigt, Kazuto Yoshimi, Hiroshi Hiai, Takashi Kuramoto, and Tadao Serikawa Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, Japan Although the laboratory rat has been widely used as animal models across many fields in biomedical sciences, the inability to utilize germline-competent embryonic stem (ES) cells was a major drawback for research activities that aimed to elucidate gene functions. Recently, Zinc-Finger Nucleases (ZFNs) have been used successfully to create site-specific DNA double-strand breaks, and thereby stimulate targeted gene mutations in a wide variety of organisms including plants, xenopus, drosophila, zebrafish, and rats. Here we demonstrate ZFN-stimulated gene-targeting at an endogenous rat gene, for which human and mouse mutations are known to cause a severe combined immunodeficiency (SCID). Co-injection of mRNAs encoding the custom-designed ZFNs into pronucleus of fertilized oocytes yielded 25% gene-modified offspring, including a wide variety of deletion or insertion mutations. ZFN-modified founders faithfully transmit these genetic changes to the next generation with the SCID phenotypes. The high frequency of gene-targeting and the rapid creation of gene knockouts indicate that ZFN technology can provide a new strategy in rats for creating animal models of human diseases. 54 ABSTRACT 19 A RECOMBINATION HOTSPOT LEADS TO SEQUENCE VARIABILITY WITHIN A NOVEL GENE AND CONTRIBUTES TO COMPLEX DISEASE SUSCEPTIBILITY Iris KL Tan, Leanne Mackin1, Nancy Wang1,2, Anthony T Papenfuss3, Colleen M Elso1, Michelle P Ashton, Belinda Phipson2,3, Melanie Bahlo3, Terrence P Speed3, Gordon K Smyth3, Grant Morahan4, and Thomas C Brodnicki1 1 St Vincent’s Institute of Medical Research, Fitzroy, Victoria, Australia 2 The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia 3 The Western Australian Institute of Medical Research, Perth, Western Australia, Australia Type 1 diabetes (T1D) is a multigenic autoimmune disease in which lymphocytes mediate destruction of insulin-producing pancreatic beta cells. Despite the recent success of human genome-wide association studies, genetic heterogeneity and tissue availability still hinder the identification of causative alleles, as well as their effects upon gene function and T1D pathogenesis. A synergistic approach to ongoing human studies is the use of the nonobese diabetic (NOD) mouse strain, which has provided key insights into T1D. We have recently positionally cloned a T1D susceptibility locus, termed Idd11, located on mouse Chromosome 4. Sequence analysis across a critical 6.9 kb interval in a series of congenic NOD mouse strains and in 25 other inbred strains identified several haplotypes, including a unique NOD haplotype, associated with varying levels of T1D susceptibility. Haplotype diversity within this interval between congenic NOD mouse strains was due to a recombination hotspot that generated four crossover breakpoints, including one with a complex conversion tract. The Idd11 haplotype and recombination hotspot are located within a predicted gene of unknown function. This gene, temporarily named Latet, exhibits deficient splicing and decreased expression in relevant immunological tissues of NOD mice. Notably, it was the recombination hotspot that aided our mapping of Idd11 and confirms that recombination hotspots can create genetic variation representing ‘private mutations’ and affecting a common polygenic disease. This finding has implications for studies of complex genetic disease, which may be affected by the ~33,000 estimated recombination hotspots in the human genome. 55 ABSTRACT 20 A SENSITIZED SUPPRESSOR SCREEN TO IDENTIFY MODIFIER GENES FOR FACTOR V LEIDEN-DEPENDENT THROMBOSIS IN THE MOUSE Randal Westrick, Goujing Zhu, Sara Manning, Angela Yang, David Siemieniak, and David Ginsburg University of Michigan, Ann Arbor MI USA Venous thrombosis affects ~300,000 individuals/year in the USA. A factor V polymorphism, Factor V Leiden, (FVL) is the most common genetic risk factor for venous thrombosis, though it displays only 10% penetrance. We previously demonstrated synthetic lethality between homozygosity for FVL (FVQ/Q) and heterozygous tissue factor pathway inhibitor deficiency (TFPI+/-) in mice. To identify modifier genes contributing to FVL penetrance, we used this lethal phenotype for a sensitized ENU mutagenesis screen. As proof of concept, we tested the loss of one tissue factor (TF+/-) allele to suppress the lethal FVQ/Q TFPI+/- phenotype. Analysis of 257 offspring from a FVQ/Q X FVQ/+ TFPI+/TF+/- cross demonstrated that haploinsufficiency for TF suppressed FVQ/Q TFPI+/-. Thus, suppressor mutations in the TF gene should emerge from our screen. We next performed a genome-wide mutagenesis screen for suppressors of FVQ/Q TFPI+/-. Male FVQ/Q mice were exposed to ENU and bred to FVQ/+ TFPI+/- females. Surviving G1 offspring were analyzed to identify mice with the otherwise lethal FVQ/Q TFPI+/- genotype. Analysis of 7,128 G1offspring (~2X genome coverage) identified 98 FVQ/Q TFPI+/- mice that survived to weaning. 13 FVQ/Q TFPI+/- G1 mice exhibited successful transmission of a putative suppressor mutation to two or more FVQ/Q TFPI+/- G2 offspring. 11 of these 13 putative suppressors are being subjected to positional cloning to identify the corresponding mutations. Analysis of 20 offspring from one of the lines resulted in the identification of a Chromosome 3 region encompassing the TF gene (LOD=4.93). Sequencing of this region is in progress to identify the causative mutation. Mapping and genome sequencing is underway for the remaining 10 lines. Identification of these mutations should provide novel insights into hemostatic regulation and suggest candidate modifier genes for Factor V Leiden and other human hemostatic disorders. 56 ABSTRACT 21 GENETIC AND DIETARY CONTROLS OF ALTERNATIVE EXON USE AND MRNA LEVELS IN THE SYSTEMS ARCHITECTURE OF DIETINDUCED METABOLIC DISEASES Sabrina H. Spiezio1, Annie Hill-Baskin1, Karen Fitch2, Keith Jones2, Michael H. Shapero2, and Joseph H. Nadeau1 1 Department of Genetics, Case Western Reserve University, Cleveland, Ohio, USA 2 Affymetrix Inc., Santa Clara, CA, USA Because of their rapidly increasing incidence worldwide, the need is urgent to characterize the genetics and systems biology of obesity and related metabolic conditions such as metabolic syndrome, type 2 diabetes, and non-alcoholic fatty liver disease (NAFLD). Two mouse inbred strains, C57BL/6J (B6) and A/J, show contrasting metabolic responses to high fat vs low fat diets. Using a complete panel of chromosome substitution strains (CSSs) that we made to study genetically complex traits such as these, we discovered 100s of complex trait genes that have unexpectedly large and non-additive phenotypic effects. Moreover these networks of interacting genes buffer systems properties from genetic perturbations. To examine molecular features of these systems, we compared mRNA and alternative exon profiles in three metabolically relevant tissues, for two diets, and several CSSs and B6 control mice that differ in their response to high fat vs low fat diet. The timepoint was midway through the study period, after 50 days on the test or control diet, but before significant weight gain was evident. Key findings include 103 more mRNAs showing dietinduced differential expression and 104 more diet-induced alternative exon usages in CSSs than B6, cis-effects that were more common than trans-effects on the low fat vs. high fat diet, remarkable heterogeneity in trans-responses to different chromosome substitutions, and finally a modest correlation between changes in mRNA expression and use of alternative exons. Together, these results suggest that changes in alternative exon use are much more common than changes in mRNA levels, that B6 responded in a markedly different molecular manner than CSSs to dietary perturbations, and that alternative exon use may be an important but generally neglected source of functional heterogeneity for modulating molecular and physiological responses to genetic and dietary perturbations. 57 ABSTRACT 22 A GENE DRIVEN ENU MUTAGENESIS SCREEN REVEALS A NOVEL METHOD OF ZIC PROTEIN NUCLEAR LOCALIZATION R Ali1, H Bellchambers1, N Warr2, D Quwailid2, P Denny2 and Ruth Arkell1,2 1 Early Mammalian Development, Research School of Biological Sciences, The Australian National University, Canberra, ACT, Australia 2 Mammalian Genetics Unit, MRC Harwell, Oxfordshire, UK The Zic genes are an important family of transcription factors about which little is known. Germ line mutation of Zic genes leads to a variety of congenital defects and somatic mutation of ZICs is implicated in a wide range of cancers. The Zic genes contain a zinc finger domain with supposed DNA binding, protein binding and nuclear localization roles. To learn more about the specific molecular functions of the zinc finger domain we undertook a search for relevant point mutations within the Harwell archive of mutagenised mouse DNAs. The Zic5 gene screen lead to the isolation of 2 missense alleles, one with a conservative amino acid change and one with a non-conservative change. In cell based transcription assays both mutant proteins exhibit a partial loss-offunction and mouse strains harbouring theses mutations have a milder phenotype than a targeted null allele of Zic5. Together this demonstrates that both mutations have generated hypomorphic alleles of Zic5. We have further investigated how a conservative change causes a partial loss-of function. The missense mutation alters a lysine residue and the lysine is a predicted substrate for SUMOylation. In vivo studies indicate that Zic5 is normally SUMOylated at this and other lysines and that this modification is required for localization to the nucleus. Other ZIC proteins have been shown to be trafficked to the nucleus via the importin pathway. Our data therefore point to a previously unrecognized mode of transport for the ZIC proteins. SUMOylation is a post-translational modification that is frequently associated with transcription factors. Previous work has demonstrated that the absence of all SUMOylation is incompatible with development of an organism, however, the effect on individual proteins has been harder to demonstrate. This work provides the first in vivo evidence that SUMOylation is required for the function of an individual protein. 58 ABSTRACT 23 NON-MUSCLE MYOSIN IIB IS REQUIRED FOR FORMATION OF THE CORONARY VASCULATURE Karen Mitchell, C Clowes, Louise Stephen, and Kathryn E Hentges Faculty of Life Sciences, University of Manchester, Manchester UK The coronary vasculature is an essential network providing the blood supply to the heart. Disruptions in the coronary circulation deprive the working myocardium of oxygen, leading to irreversible damage to cardiac muscle and myocardial infarction. Cardiovascular disease is a major contributor to mortality worldwide. The generation of treatments for cardiovascular disease will be aided by a deeper understanding of the developmental processes underlying the formation of the coronary vessels. We have identified a requirement for Non-Muscle Myosin IIB (NMIIB) in the development of the coronary vasculature. We isolated an embryonic lethal mouse mutant with Embryonic Hydrocephalus and Cardiac defects (EHC) from a balancer chromosome mutagenesis screen. Positional cloning revealed that the EHC mutant phenotype is caused by a splice donor mutation in Myh10, which encodes NMIIB. EHC mutants produce an aberrant transcript lacking exon 18, disrupting the reading frame of NMIIB and introducing a premature stop codon. RT-PCR analysis demonstrates that the mutation destabilises the Myh10 transcript, such that heterozygous embryos preferentially produce wild type Myh10. EHC mutant embryos display cardiac morphological defects such as double-outlet right ventricle. Additionally mutants do not form coronary vessels, evident from gross inspection of hearts and a lack of vascular endothelial PECAM staining. The failure to form the coronary vasculature is suggestive of epicardial defects. Accordingly, EHC mutant epicardial cells fail to form an epithelial layer covering the myocardium, and have disruptions in the localisation of extracellular matrix between the myocardium and epicardium. NMIIB therefore may facilitate coronary vessel formation by supporting adhesive interactions between the epicardium and myocardium mediated via the extracellular matrix. Future work will determine the mechanisms by which Myh10 deficiency contributes to coronary vascular failure. 59 ABSTRACT 24 SHORT CIRCUIT, A HOMOZYGOUS LETHAL ENU MUTATION THAT AFFECTS LUNG DEVELOPMENT Michael J. Parsons, Laura Yates, Jessica Edwards, Lauren Chessum, Charlotte H. Dean, and Patrick M. Nolan Mammalian Genetics Unit, MRC Harwell, Oxfordshire, United Kingdom The use of N-ethyl-N-nitrosourea (ENU) mutagenesis screening is a powerful tool for discovering novel genes associated with numerous biological pathways and phenotypes. The mutant called short circuit (Sci) was found in a dominant enu mutagenesis screen. This mutant, first characterized in a screen for circadian phenotypes, was found to be homozygous lethal with Sci homozygous animals dying between embryonic day 14.5 to post-natal day one. The gene containing the Sci mutation encodes for a protein that is a transcription factor with multiple zinc fingers. This gene has been implicated in various functions including neuronal differentiation and development. In mouse, this gene is highly expressed in developing brain, lung and heart, with its expression peaking at embryonic day 13.5. These expression patterns combined with the timing of the lethality in homozygous animals led us to explore the possibility of the Sci mutation interfering with normal lung development, thus leading to the observed homozygous lethality. Upon conducting a time series of lung development, we found significant decreases in distal airway volume in Sci homozygous animals, as compared to wildtype animals, at embryonic day 18.5 (p<0.05); morphological changes in distal airways were observed as early as embryonic day 15.5. Adult heterozygous Sci animals showed impaired lung function as measured by full body plethysmography following metacholine challenge. Specifically, heterozygous Sci animals had significant decreases in baseline PenH (a measure of lung resistance), peak inspiratory flow and peak expiratory flow (p<0.05). We are currently conducting immunohistochemical analysis using lung differentiation and proliferation markers, to further characterize the effects of this gene on lung development. Taken together these data suggest that this gene may play an important role in lung development. 60 ABSTRACT 25 IDENTIFICATION OF THE DANFORTH’S SHORT TAIL MUTATION USING NEXT GENERATION SEQUENCING Christopher N. Vlangos, Amanda N. Siuniak, Dan Robinson, Arul M. Chinnaiyan, James Cavalcoli, Robert H. Lyons, and Catherine E. Keegan University of Michigan, Ann Arbor, MI USA The Danforth’s short tail (SdSd) mouse first appeared as a semi-dominant spontaneous mutation in an inbred colony at Stanford University in the 1920s. The phenotype of heterozygous (SdSd/+) animals includes unilateral kidney agenesis, vertebral anomalies, and a shortened and kinked tail. Homozygous (SdSd/Sd) mice are more severely affected with bilateral renal agenesis, lack of tail, vertebral anomalies, spina bifida, lack of urogenital and anal openings, and persistence of the cloaca. Homozygous mice are born live but die within 24 hours of birth. Though genetically mapped to a 1.0cM (1.5Mb) region of mouse Chromosome 2qA3, the Sd mutation has not yet been identified. Using bioinformatic analysis of the mouse DNA physical map we identified 9 annotated genes with a total of 86 coding exons spanning the corresponding 1.5Mb critical region. Direct sequencing of the exonic DNA and intron/exon boundaries did not reveal any causative mutations. Since direct sequencing of the exonic DNA only provided ~1% coverage of the Sd critical region we performed next generation sequencing (NGS) of the entire 1.5Mb region. Prior to NGS we performed an enrichment capture using an Agilent oligo DNA chip designed to cover all unique (non-repeat masked) DNA of the Sd critical region. After successful locus specific DNA enrichment, 36bp paired end NGS on an Illumina Genome Analyzer IIx was completed, generating 1.85Gb of sequence. 93% of reads were of proper size and mapped to the enriched region in the proper orientation resulting in an average of 200X coverage. Analysis of the mapped reads using standard computational techniques did not reveal any causative mutations. We then turned to interrogation of reads where only a single end of the paired end sequencing mapped correctly to the Sd locus. By using this novel technique, we were able to identify the presence of an endogenous retroviral-like (ERV) insertion of the MusD family at the Sd locus. The ERV insertion was confirmed via Southern analysis, and is not present in additional inbred mouse lines tested. The phenotypic characteristics of homozygous SdSd/Sd mice are comparable to those seen in human patients with caudal regression syndrome, cloacal exstrophy, and VACTERL association. Thus, the Sd mouse is an excellent model for identification of novel genes responsible for human disorders. 61 ABSTRACT 26 A NOVEL ROLE FOR ATMIN, CONTROLLING CILIOGENESIS THROUGH MODULATION OF DYNEIN LIGHT CHAIN EXPRESSION Paraskevi Goggolidou1, Jonathan Stevens1, Gabrielle Wheway3, Antonella Di Paolo, Rosario Romero1, James Briscoe2, Colin Johnson3, and Dominic P Norris 1 Mammalian Genetics Unit, Medical Research Council Harwell, Oxfordshire OX11 0RD, UK 2 National Institute for Medical Research, London, UK, 3 Section of Ophthalmology and Neurosciences, Wellcome Trust Brenner Building, Leeds Institute of Molecular Medicine, St James's University Hospital, Beckett Street, Leeds, U.K. Through a phenotype driven genetic screen, we identified the DNA damage response locus Atmin as encoding a zinc finger factor required for normal ciliogenesis. Point mutants destroying Atmin zinc finger structure result in embryonic death around 13.5 days of development. Homozygous mutants exhibit a complex developmental syndrome indicative of a ciliopathy, including exencephaly, coloboma, cardiac outflow tract defects, pulmonary hypoplasia and left-right (L-R) patterning defects. Indeed, impaired ciliogenesis is evident from a decreased number of ciliated cells and shortened cilia in the embryonic node. Notably, these morphologically abnormal cilia retain motility, although LR patterning is abnormal. Cilial structure is known to be important for SHH signaling and for correct establishment of dorso-ventral (D-V) patterning in the neural tube. A defect in SHH signalling is evident in Atmin mutants, as assessed by GLI3 processing. However, no obvious defects in neural tube D-V patterning were detected, suggesting a specific role of Atmin in GLI3 but not GLI2 processing. We have further identified a dynein light chain as acting downstream of Atmin; mRNA expression analysis shows a ~16-fold downregulation in Atmin mutant embryos. Using si-RNA mediated knock down of Atmin and the dynein, we show that Atmin mediates ciliogenesis through regulation of expression of this specific dynein subunit. Sub-cellular localisation places this dynein subunit in the primary cilium in both cultured cells and the embryo, consistent with it directly influencing retrograde intraflagellar transport. 62 ABSTRACT 27 GENETIC ANALYSIS OF COMPLEX TRAITS IN THE EMERGING COLLABORATIVE CROSS David L. Aylor1, William Valdar1,#, Wendy Foulds-Mathes1,#, Ryan J. Buus1,#, Ricardo A. Verdugo2,#, Ralph S. Baric3,4, Martin T. Ferris1, Jeffrey A. Frelinger4, Mark Heise1, Matt B. Frieman4, Lisa E. Gralinski3, Timothy A. Bell1, John P. Didion1, Kunjie Hua1, Derrick L. Nehrenberg1, Christine L. Powell1, Jill Steigerwalt5, Yuying Xie1, Samir N.P. Kelada6, Francis S. Collins6, Ivana V. Yang7, David A. Schwartz7, Lisa A. Branstetter8, Elissa J. Chesler2, Darla R. Miller1, Jason Spence1, Eric Yi Liu9, Leonard McMillan9, Abhishek Sarkar9, Jeremy Wang9, Wei Wang9, Qi Zhang9, Karl W. Broman10, Ron Korstanje2, Caroline Durrant11, Richard Mott11, Fuad A. Iraqi12, Daniel Pomp1,*, David Threadgill5,*, Fernando Pardo-Manuel de Villena1,* and Gary A. Churchill2,* 1 Department of Genetics, University of North-Carolina-Chapel Hill, Chapel Hill, North Carolina, USA 2 The Jackson Laboratory, Bar Harbor, Maine, USA 3 Department of Epidemiology, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, USA 4 Department of Microbiology and Immunology, University of North Carolina– Chapel Hill, Chapel Hill, North Carolina, USA 5 Department of Genetics, North Carolina State University, Raleigh, North Carolina, USA 6 National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, US 7 National Jewish Health, Denver, CO, USA 8 Oak Ridge National Laboratory, Oak Ridge, TN, USA 9 Department of Computer Science, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina, USA 10 Department of Biostatistics and Medical Informatics, University of WisconsinMadison, Madison, Wisconsin, USA 11 Welcome Trust Centre for Human Genetics, Oxford University, Oxford, UK 12 Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel The Collaborative Cross (CC) is an eight-way mouse recombinant inbred strain panel that is being developed as a resource for mammalian system genetics. Here we describe an experiment using partially inbred CC lines to evaluate the genetic structure, mapping power and resolution of the CC. Genome-wide analysis of high density SNP data in the incipient strains reveals high genetic diversity, balanced allele frequencies, and dense, evenly distributed recombination sites – all ideal qualities for a genetic mapping and systems genetics resource. We mapped white head spotting, a discrete coat color trait; 63 body weight, a highly polygenic complex trait; and more than 11,000 liver gene expression traits. We demonstrate that analysis based on inferred haplotypes in the eight-way cross population improves power and provides additional information about candidate genes not available from standard crosses or strain surveys. We were able to identify Kitl as a candidate gene for white head spotting in the WSB/EiJ strain and Asph as a novel candidate gene for body weight. Genetic mapping of gene expression data demonstrates that resolution in this experiment is on the order of 1Mb, and this will improve in the finished CC panel. The number of eQTL discovered here exceeds all previous efforts at eQTL mapping in mouse strain panels and crosses. The notable absence of trans- eQTL clustering suggests that multiple functional genetic variants are evenly distributed throughout the genome of CC mice. We have demonstrated that the unprecedented genetic diversity of the CC, which derives from random mixing of an estimated 45 million SNPs, results in high phenotypic diversity and enhances our ability to map the causative loci underlying complex diseaserelated traits 64 ABSTRACT 28 REALISING THE POTENTIAL OF MOUSE DISEASE MODELS – FROM PATHWAY TO THERAPY Steve D.M. Brown MRC Mammalian Genetics Unit, MRC Harwell, Harwell Science and Innovation Campus, OX11 0RD, UK Classical genetic approaches utilising random mutagenesis coupled with focused phenotyping continues to remain an important tool for discovering novelty in biological systems. Moreover, the development of a comprehensive mutant resource for every gene in the mouse genome, followed by large-scale phenotyping, is also set to transform the available models for dissecting disease states. The discovery of novelty is the key basis for a cycle of discovery comprising genetics, molecular, cellular and physiological studies all of which provide a basis for developing new therapeutic approaches. These paradigms provide a powerful platform for elucidating the genetic pathways underlying disease and bringing new insights to the therapeutic strategies that might be investigated. This is no more important for common diseases for which there is little genetic understanding and for which current treatments are ineffective. One such disease is recurrent (ROM) and chronic (COME) forms of otitis media. ROM and COME are known to have a strong genetic component, but nothing is known of the underlying genes involved in the human population. Otitis media with effusion (OME) is the most common cause of hearing impairment in children in the Western world, potentially causing language delays, learning and behavioural disruption. The high prevalence of the disease, coupled with its recurrent and chronic nature, accounts for the large number of ventilation tubes inserted into the tympanic membrane of affected children. Placement of ventilation tubes is the most common operation in the UK however the mechanism by which they work remains uncertain. We have identified two novel dominant mutants, Jeff and Junbo, which develop a conductive deafness due to a chronic suppurative OM. The Jeff mutant carries a mutation in an F-box gene, Fbxo11 (Hum. Mol. Genet. 15, 1-7 2006). Junbo carries a mutation in the Evi1 transcription factor (PLoS Genetics 2: e149 2006). Evi1 represses the TGFb signalling pathway by the binding of Smad3. Jeff homozygotes die shortly after birth displaying developmental abnormalities including cleft palate, eyes open at birth and impaired lung function. TGF-b signalling is involved in all these epithelial developmental processes and we find that pSmad2 is significantly upregulated in epithelia of Jeff homozygotes. Mice heterozygous for both Jeff and Smad2 mutations recapitulate the Jeff homozygous phenotype. Fbxo11 is known to neddylate p53, a co-factor of pSmad2, and we found that p53 levels are substantially reduced in Jeff suggesting that Fbxo11 plays a role in stabilizing p53. Our findings support a model whereby Fbxo11, via 65 stabilization of p53, is required to limit the accumulation of pSmad2 in the nucleus of epithelial cells. Hypoxia is a common feature of inflamed microenvironments and the impact of hypoxic mechanisms in OM has received little attention. Responses to hypoxia are mediated via Hypoxia Inducible Factor (HIF) and there is considerable cross-talk between TGF-b and HIF-1a pathways that mediate hypoxic responses. We surmise that dysregulation of TGF-b signalling in the Jeff and Junbo mutations is having an affect on the response of the middle ear, including neutrophil and macrophage function, to the hypoxic environment, leading to the chronic unresolved inflammatory condition. The role of chronic inflammatory hypoxia and hypoxia-inducible factor (HIF) mediated responses were investigated in Junbo and Jeff mouse models. Mice labeled in vivo with Pimonidazole showed cellular hypoxia in middle ear mucosa and inflammatory cells in the middle ear lumen and there was upregulation of Il-1b and Tnf-a that modulate HIF. Hif-1a gene expression was elevated in ear fluid white blood cells and there was upregulation of Vegf pathway genes including Vegfa gene and protein expression. VEGFR signaling inhibitors PTK787/ZK 222584, SU-11248 and BAY 43-9006 reduced hearing loss and modulated inflammatory changes in middle ear mucosa. The effectiveness of VEGFR signaling inhibitors implies HIF mediated VEGF plays a pivotal role in otitis media pathogenesis and targeting molecules in HIF-VEGF signaling pathways has therapeutic potential in the treatment of chronic otitis media. 66 ABSTRACT 29 IMPORTANCE OF CADM1 AND CELL ADHESION IN DEPRESSIVE BEHAVIOR C Santos1, B Miller2, M Pletcher3, A Su4, L Tarantino5, and Tim Wiltshire1 1 University of North Carolina at Chapel Hill, Division of Pharmacotherapy and Experimental Therapeutics, NC, USA 2 The Scripps Research Institute Jupiter, Florida, USA 3 Pfizer Incorporated Groton, Connecticut, USA 4 Genomics Institute of Novartis Research Foundation at La Jolla, California, USA 5 University of North Carolina at Chapel Hill, Department of Psychiatry, NC, USA Despite evidence for genetic vulnerability to depressive behavior, genes that can predict risk or response to anti-depressants have not been identified. Factors including disease heterogeneity, environmental effects, and complex neurobiological processes complicate studies in mood disorder. In order to elucidate genetic mechanisms underlying depression, we have performed a series of experiments using a panel of genetically diverse mouse inbred strains. We collected genome-wide gene expression data as well as levels of forty biochemical molecules including neurotransmitters, neurotrophic factors, neuropeptides, and neuroamine regulators and receptors under control, fluoxetine, and treatment-naïve conditions from brains of inbred mice. Furthermore, we have recorded seven baseline anxiety and depressive behaviors as well as depressive-like responses after administration of fluoxetine or water across 30-35 mouse inbred strains. Since mice within a strain are isogenic, inter-strain genotypic and phenotypic differences are analyzed using haplotype-association mapping algorithm to identify genomic regions associated with the phenotypes of interest known as quantitative trait loci (QTLs). Candidate regions were then prioritized based on convergence of evidence from gene expression, behavior, and biochemical data. A biochemical QTL on Chromosome 9 overlaps with a depressive QTL (which suggests that genes within this region are associated with behavior as well as biological changes associated with differential levels of glutamate decarboxylase 67 (GAD67, official symbol GAD1). There are two genes within this region including cell adhesion molecule 1 (Cadm1) and 2900052N01Rik. Analyses reveal moderate correlation between expression of Cadm1 and anxiety-like and depressive-like behaviors in addition to levels of several biochemical molecules. Furthermore, genes within candidate haplotype-depressive QTL peaks (-logP >3.5) were found to be enriched for genes involved in cell adhesion. Our findings suggest Cadm1 and other cell adhesion molecules may mediate molecular and cellular processes that can result in depressive behavior. Studies are underway to elucidate how cellular adhesive properties modulate biological changes that lead to depression. 67 ABSTRACT 30 AN ENU SENSITIZATION SCREEN TO DETERMINE PHYSIOLOGICAL FUNCTION OF α- SYNUCLEIN Deborah E Cabin, M Casey, and D Zou McLaughlin Research Institute, Great Falls, MT USA THE α−synuclein is a small presynaptic and nuclear neuronal protein that is linked to Parkinson's disease (PD) by its presence in Lewy bodies, the intracellular inclusions characteristic of sporadic PD, and by mutations responsible for rare familial forms of that disease. The physiological function of α-synuclein is not well understood, though several lines of mice that lack the protein have been studied. Whether or not α-synuclein's normal function is a factor in PD cannot be assessed until that normal function is better defined. We have taken a genetic approach to determine α-synuclein's function, a sensitized ENU mutagenesis screen using mice null for the protein. One hundred twenty pedigrees have been screened for neurological phenodeviants; 45 lines have produced non-sensitized phenotypes, 18 lines that transmit phenotypes are being tested for sensitization, and one confirmed sensitized mutation has been identified. Among the neurological phenotypes still being tested for sensitization are 2 circling mutants, hyperactivity with odd gait, shaky and poor grip, slow righting and poor grip, lack of exploration, and no response to a clickbox. The confirmed sensitized mutation is in Atp7a the X-linked transGolgi copper transporter mutated in human Menkes disease. While all males carrying the Atp7a I610S mutation die regardless of α-synuclein status, Atp7a mutant females that lack α-synuclein die prior to 35 days at a significantly higher rate than Atp7a mutant females that express α-synuclein (p= 0.00069, Χ2). While Atp7a mutations affect many systems in the body, the protective effect of α-synuclein must be neuronal based on its expression pattern. Menkes disease has a severe neurological component that can progress to decerebration. In the absence of α-synuclein, brain steady-state levels of both wild type and mutant ATP7A protein are highly variable compared to ATP7A levels in mice that express α-synuclein. The absence of α-synuclein also appears to cause mislocalization of mutant ATP7A in brain; mislocalization might further impair the function of a hypomorphic protein. Thus α−synuclein appears to have a neuroprotective function; in Parkinson's disease, sequestration of the protein in Lewy bodies may prevent it from performing that function, which may be particularly important under stress conditions. 68 ABSTRACT 31 GENOMIC RESOURCES IN PEROMYSCUS - MAPPING THE AUDIOGENIC SEIZURE SENSITIVITY LOCUS Gabor Szalai, Janet Crossland, Jay Coleman, and Michael Felder Peromyscus Genetic Stock Center, University of South Carolina, Columbia, SC,USA A Mice of the genus Peromyscus are among the most abundant mammals in North America. They range from Alaska to Central America and occur in many natural habitats. Though superficially resembling laboratory mice (Mus musculus) and rats (Rattus norvegicus), deer mice are not closely related to either of these species. One of the major advantages of research with Peromyscus is their ready adaptability to colony conditions. Research on this genus has been widespread across so many disciplines that the genus has aptly been referred to as “The Drosophila of North American Mammalogy”. In order to make Peromyscus as a viable alternative to Mus, we have been working with collaborators on developing comparable genomic resources such as an intermediate resolution genetic map (500 markers to date), synteny map, EST libraries (50 000 EST clones), BAC libraries and whole genome sequences (to be completed in December, 2010). These resources are used to map the genetic locus controlling the audiogenic seizure sensitivity in deer mice. Epilepsy is a debilitating disease that can arise from either acquired brain lesions or from an inherited susceptibility to cortical hyperexcitability. At least 40-50% of epilepsies have a presumed genetic basis. Although few human epilepsy syndromes are inherited in a simple Mendelian manner, single gene animal models offer valuable opportunities to isolate gene mutations, to identify underlying molecular mechanisms and to explore strategies for therapy. In 1935 a spontaneous recessive mutation appeared among laboratory stocks of Peromyscus maniculatus artemisiae, which has been maintained as a separate stock since. Using homozygousity mapping analyzing a panel 250 F2 and 100 N2 animals we have identified 4 candidate markers. A comparative cytogenetic map between Peromyscus maniculatus and Mus musculus reveal that the M. musculus homologs of the candidate markers on P.m. chromosome 1 and chromosome 23 would be on M.m. chromosomes 7 and 5, respectively. The two mapped mouse audiogenic seizure susceptibility genes, mass1 and jams1 are localized on M. m. chromosomes 13 and 10, respectively. Taken together, our preliminary data suggests that the genetic mutation underlying seizure sensitivity in deer mice resides in a novel gene and is not a homolog of a previously identified susceptibility locus. 69 ABSTRACT 32 MUTATIONS IN THE PDZ DOMAIN CONTAINING PROTEIN GIPC3 CAUSE PROGRESSIVE SENSORINEURAL DEGENERATION (AHL5 AND JAMS1) IN MICE AND RECESSIVE HEARING IMPAIRMENT IN HUMANS (DFNB95) Nikoletta Charizopoulou1, Andrea Lelli2, Margit Schraders3,4, Kausik Ray5, Ronald J.C. Admiraal4, Harold R. Neely1, Joseph R. Latoche1, John K. Northup5, Hannie Kremer3,4,6, Jeffrey R. Holt2, and Konrad Noben-Trauth1 1 Section on Neurogenetics, 5Section on Signal Transduction, Laboratory of Molecular Biology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, MD, USA 2 Department of Neuroscience and Otolaryngology, University of Virginia School of Medicine, Charlottesville, Virginia, USA 3 Department of Otorhinolaryngology, 6Department of Human Genetics, 4 Nijmegen Centre of Molecular Life Sciences and Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands Progressive sensorineural hearing loss affects the quality of life and communication of millions of people but the underlying molecular mechanisms remain elusive. Recently, the age-related hearing loss 5 (Ahl5) and juvenile audiogenic monogenic seizure 1 (jams1) loci were shown to underlie progressive hearing loss and audiogenic seizures in Black Swiss mice, respectively. Here, we identify Ahl5 and jams1 as a 343G>A transition in Gipc3 changing the conserved Gly115 to Arg in its PDZ domain and causing a significant reduction in protein levels. GIPC3 localizes to inner ear sensory hair cells and spiral ganglia and we show that the mutation disrupts the structure of the stereocilia hair bundle affecting mechano-transduction currents and longterm survival of spiral neurons. We demonstrate an adverse effect of the Gipc3343A allele on wave I amplitudes of afferent neurons, which we correlate with susceptibility and resistance of audiogenic seizures. A Gipc3 transgene rescues both hearing loss and audiogenic seizures. Lastly, we identify a truncating mutation in human GIPC3 in a family segregating autosomal recessive hearing loss DFNB95. Our study reveals a novel and pivotal role of GIPC3 for the function of the stereocilia hair bundle and for synaptic transmission. 70 ABSTRACT 33 A MUTATION IN THE GENE ENCODING MITOCHONDRIAL MG2+ CHANNEL MRS2 RESULTS IN DEMYELINATION IN RATS Takashi Kuramoto1, Mitsuru Kuwamura2, Satoko Tokuda1,2, Takeshi Izawa2, Yoshifumi Nakane1, Kazuhiro Kitada1,3, Masaharu Akao4, Jean-Louis Guénet5, and Tadao Serikawa1 1 Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Japan 2 Laboratory of Veterinary Pathology, Osaka Prefecture University, Japan 3 Laboratory of Mammalian Genetics, Genome Dynamics Research Center, Graduate School of Science, Hokkaido University, Japan 4 Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan 5 Département de Biologie du Développement, Institut Pasteur, France The rat demyelination (dmy) mutation serves as a unique model system to investigate the maintenance of myelin, because it provokes severe myelin breakdown in the central nervous system (CNS) after normal postnatal completion of myelination. Here, we identified the dmy mutation and characterized dmy/dmy rats to clarify the pathomechanisms underlying demyelination. By positional cloning, we found that a G-to-A transition, 177 bp downstream of exon 3 of the Mrs2 gene, generated a novel splice acceptor site which resulted in functional inactivation of the mutant allele. Transgenic rescue with wild-type Mrs2-cDNA validated our finding. Mrs2 encodes an essential component of the major Mg2+ influx system in mitochondria of yeast as well as human cells. We found an increased number of mitochondria in the swollen cytoplasm of oligodendrocytes and elevated serum lactic acid concentration in mutant rats, indicating that mitochondria were indeed functionally defective. MRS2-GFP recombinant BAC transgenic rats showed that MRS2 was dominantly expressed in neurons rather than oligodendrocytes, and ultrastructurally observed in the inner membrane of mitochondria. These findings clearly demonstrate that dmy is a loss-of-function mutation of Mrs2 and suggest that dmy/dmy rats suffer from mitochondrial disease. Our findings imply that the maintenance and turnover of myelin are genetically independent from its initial production, and Mg2+ homeostasis in CNS mitochondria is essential for the maintenance of myelin. 71 ABSTRACT 34 ANNOTATION OF LONG NON-CODING RNA TRANSCRIPTS Mark Thomas and Jennifer Harrow HAVANA Group, Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom With the publication of the human genome 10 years ago, came the realisation that developmental complexity is not related to gene number. Instead, it would seem that the increased complexity of higher organisms is achieved through greater regulation of more diverse transcripts. Recent studies would suggest that a significant proportion of our genome is transcribed, however only 1-2% of the genome is associated with protein coding loci. A large number of transcripts are therefore non-coding with no known function and are the subject of much debate as to whether they are indeed functional or represent transcriptional noise. It is the remit of the HAVANA group at the Wellcome Trust Sanger Institute to annotate all transcriptional elements in the human and mouse genomes. As part of this ongoing effort and in collaboration with the Mattick laboratory and HGNC, we are proposing a nomenclature system for long non-coding RNA transcripts (lncRNAs). In the absence of any functional characterisation data for most transcripts, the proposed schema will ascribe a name for each transcript based on its genomic context relative to the nearest protein coding locus. We are also constantly updating our methods to make the most of recent advances in sequencing technology (eg. RNAseq) and structure prediction methods (eg. lincRNAs). This has led to the introduction of new non-coding transcript biotypes, such as antisense, lincRNA, ncRNA-hostgene and 3’overlapping ncRNA to further define this rapidly emerging class of genes. Furthermore, analysis of lincRNAs combined with existing data from the Rfam, miRBase and piRNABank databases have identified potentially new hostgenes for these small ncRNAs. 72 ABSTRACT 35 THE JAX CRE REPOSITORY: IMPROVING THE UTILITY OF CRE DRIVER STRAINS Stephen A. Murray, Caleb Heffner, Michael Sasner, Cathleen Lutz, Brandon Grossman, Stephen Rockwood, Yashoda Sharma, and Leah Rae Donahue The Jackson Laboratory, Bar Harbor, ME, USA Capitalizing on mouse gene targeting projects of the International Knockout Mouse Consortium (IKMC) will require that a large, diverse set of wellcharacterized Cre driver lines. To fill this need, The Jackson Laboratory (JAX) has committed to increasing the number of Cre lines available to the scientific community. The JAX Cre Repository currently houses and distributes the single largest collection of Cre driver strains totaling more than 200 lines, including 156 that are currently distributed as live colonies. The JAX Cre Repository has embarked on an ambitious project to add value to these strains by comprehensively characterizing Repository Cre lines. Despite the best efforts of those developing new Cre lines, the fidelity of Cre activity is not always ideal. Many difficulties have been reported in various Cre lines, including mosaic or incomplete deletion in a target tissue/cell type, inconsistent activity, expression in non-target tissues, and/or Cre-related toxicity. In many cases, however, this data is not reported or available to the potential user. We have embarked on an ambitious project to add value to these strains by comprehensively characterizing all of the Cre lines distributed. We have developed a comprehensive pipeline for the characterization of Cre driver strains using a LacZ reporter strain in a wide range of tissues and at multiple time points, including both target and non-target tissues. In addition, we are examining possible instances of Cre-induced toxicity, effect of genetic background on excision fidelity, and functional differences that result from maternal versus paternal inheritance of the Cre allele. Thus far we have completed the characterization of 26 strains, and an additional 37 are underway. Our results indicate the vast majority of Cre driver strains exhibit unexpected recombinase activity in a number of tissue types, highlighting the need for extended analysis. We have standardized our data annotation scheme to include 11 broad organ systems, 30 individual organs/structures and 89 substructures, all of which are consistent with the mouse Anatomical Dictionary. Slide-scanned images and associated annotations are published on a dedicated website and submitted to Creportal.org. This information will allow users to make informed judgments about the suitability of a particular line for their experiments, and enhance the power of large-scale mouse gene targeting projects. 73 ABSTRACT 36 MOUSE GENOMES PROJECT: MAP OF SNPS AND SHORT INDELs Petr Danecek, Gan Xiangchao2, Thomas M Keane1, James Stalker1, Binnaz Yalcin2, Martin Goodson2, Sendu Balasubramaniam1, Kim Wong1, Guy Slater1, Andreas Heger2, Eleazar Eskin3, Nick Furlotte3, Chris Ponting, Jonathan Flint2, and David J Adams1 1 Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK 2 Wellcome Trust Centre for Human Genetics, Oxford, UK 3 University of California, Los Angeles, USA The Mouse Genomes Project has sequenced the genomes of 17 inbred mouse strains to between 20-35x coverage, using paired-end Illumina sequencing. One of the major goals of this project is to generate a comprehensive catalogue of all SNPs across these strains. Our strategy for calling SNPs was to employ several next-generation sequencing SNP calling programs and then merge the calls to produce the final callset. The final set consists of more than 65 millions of SNPs across the strains. We find extremely high concordance with the Perlegen SNP set and using 10Mbp of manually finished sequence for NOD/ShiLtJ find a false positive rate to be 1.4% and false negative rate of 1.7% thus indicating the high quality of our calls. The final call set consists of 24 thousand coding SNPs of which 700 were found to be potentially truncating coding mutations. The wild derived strains such as CAST/EiJ, PWK/PhJ, Spretus/EiJ show an order of magnitude more variation than the classical strains. From the sequenced based SNP calls, we have also created an imputed set of SNPs across a further 94 strains. The full set of SNPs has been uploaded to dbSNP and can be queried on our website which also gives the consequences per SNP, tables of the SNP patterns across the 17 strains, and enables users to visually inspect the raw sequence data surrounding individual SNPs. http://www.sanger.ac.uk/mousegenomes 74 ABSTRACT 37 USE OF PIGGYBAC-MEDIATED TRANSIENT TRANSGENIC RNAI EXPRESSION FOR RAPID CHARACTERIZATION OF GENE FUNCTION DURING EMBRYONIC DEVELOPMENT David R. Beier1, Yuko Fujiwara2, Shannon W. Davis3, Haiyan Qiu1, Thomas L. Saunders3, Stuart Orkin2, Sally A. Camper3, and Bryan C. Bjork1 1 Genetics Division, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA 2 Division of Hematology and Oncology, Children’s Hospital, Harvard Medical School/Howard Hughes Medical Institute, Boston, MA, USA 3 Departments of Human Genetics and Internal Medicine, University of Michigan, Ann Arbor, MI, USA The production of targeted mutations in mice remains the gold standard for the analysis of loss-of-function of specific genes in mammals. However, even with the emergence of large-scale knockout mouse resources, generation of such mutants using embryonic stem cells may still require substantial time and resources. In particular, this approach is difficult to pursue for high throughput applications. For instance, linkage and association studies for mutations or strain-specific traits may yield genomic intervals of only moderate resolution, containing a large number of positional candidate genes. Similarly, microarray analyses typically result in lists of differentially expressed genes, with little indication regarding which ones may be key regulators. An efficient methodology to rapidly screen genes in vivo would enhance the functional analysis of outputs from high throughput screening. RNA interference (RNAi) is a powerful strategy for studying the phenotypic consequences of reduced gene expression. To develop a method for the rapid characterization of the developmental consequences of gene dysregulation, we tested the use of RNAi for “transient transgenic” knockdown of mRNA in mouse embryos. These methods included lentiviral infection as well as transposition using the Sleeping Beauty (SB) and PiggyBac (PB) transposable element systems. Of the three methodologies tested, the PB transposon system produced high numbers of transgenic embryos with the expected phenotype, demonstrating its utility as a screening method. This approach can be useful for phenotypic validation of putative mutant loci, as we demonstrate by confirming that knockdown of Prdm16 phenocopies the ENU-induced cleft palate mutant, csp1. This strategy is attractive as an alternative to gene targeting in embryonic stem cells, as it is simple and yields phenotypic information in a matter of weeks. Furthermore, we found the efficiency of PB for transgenesis was routinely over 65%, suggesting this method has more widespread applications, and we have adapted it for multigene knock-down, for expression of affinity-tagged proteins, and for analysis of presumptive transcriptional regulatory sequences. 75 ABSTRACT 38 THE SANGER INSTITUTE MOUSE GENETICS PROGRAMME Ramiro Ramirez-Solis, J White, E Ryder, R Houghton, and J Bottomley Mouse Genetics Programme, Wellcome Trust Sanger Institute, Hinxton, UK The Sanger Institute Mouse Genetics Programme (MGP) capitalizes on the mutant ES cell resources generated by the EUCOMM and KOMP projects by generating and phenotyping mutant mouse lines at a large scale. The program makes the mutant strains and the phenotypic data, available to the scientific community to galvanize deeper analysis aimed at uncovering the molecular mechanisms involved in the phenotypic alterations resulting from the mutant alleles. The MGP has produced over 400 mutant strains, of which more than 200 hundred have finished the phenotypic screen. The phenotypic data can be obtained by visiting the Sanger Mouse Portal (http://www.sanger.ac.uk/mouseportal/). The website offers the opportunity to download a weekly updated summary heat map that includes all the strains being examined. Scientists are also encouraged to sign up for a phenotypic alert email list to receive early warnings on interesting phenotypes. When two (2) heterozygote mice are genotyped for a particular allele, the strain is advertised to the community at the International Knockout Mouse Consortium (IKMC) website (http://www.knockoutmouse.org/about). Although the specific mice to be distributed may not be available immediately, the MGP seeks to receive notifications of interest, and while the mice are on the shelf, a reasonable effort is made to distribute them to interested parties with minimum delay. Although a majority of strains is currently selected because of prior interest on the gene, we expect that increasingly strains will be selected because of the phenotypic data generated from the MGP. The selection of phenotypic tests included in the program is strongly influenced by an active interaction with experts from the community, and is aimed at exploring a biological space of medical relevance. We will present an update on the program activities and highlight some interesting novel phenotypic findings. 76 ABSTRACT 39 NEXT LEVEL SYSTEMIC PHENOTYPING OF MICE Martin Hrabé de Angelis and the German Mouse Clinic consortium Helmholtz Zentrum München and Technical University Munich; Institute of Experimental Genetics, Munich, Germany Next challenges in functional annotation of mammalian genomes are yet of a much larger scope than previous genomics initiatives. Mouse mutant resources must be phenotyped systematically (one after the other) and systemically (assessing all organ systems). In addition, for the next generation of mouse models the “envirotypes”, that humans are exposed to need to be modeled. We established the German Mouse Clinic (GMC) as the first mouse phenotyping platform worldwide with the logistics of systemic, standardized phenotypic analysis and interpretation, with open access for the scientific community on a collaborative basis. The German Mouse Clinic is well integrated into EUMODIC and Infrafrontier, pan European projects for functional annotation of mouse models. A new worldwide effort - the International Mouse Phenotyping Consortium – has been established and will move systemic mouse model phenotyping to a new scale. To explore the complex relationship between environmental changes and genetic factors, we have been setting up standardized challenge platforms for mouse phenotyping. By simulating specific environmental exposures or life styles we mimic envirotypes that have a strong impact on human health. Five platforms have been implemented in the areas of nutrition, exercise, air, infection and stress. Goal is to decipher their effects on disease etiology and progression, uncovering the physiological and molecular mechanisms of genome-environment interactions. In addition, identification and non-invasive screening of biomarkers and risk factors for distinct diseases will be a future goal in the GMC using a new metabolomic platform and a new breath-gas analysis set up. First results will be presented. 77 ABSTRACT 41 TOOLING UP MGI FOR A DELUGE IN MOUSE PHENOTYPE AND DISEASE MODEL DATA Anna Anagnostopoulos, Janan Eppig, and Mouse Genome Informatics Group The Jackson Laboratory, Bar Harbor, ME, USA Global efforts to functionally annotate the mouse genome through large-scale mutagenesis and phenotyping consortia have catapulted the size and diversity of new mouse resources aimed at building predictive models of human disease. The Mouse Genome Informatics resource (MGI, www.informatics.jax.org) for the genetics, genomics and biology of the laboratory mouse grants free access to current integrated biological knowledge spanning from sequence to phenotype and disease model information to unified data on worldwide mouse resource holdings via IMSR. MGI curates aberrant mouse phenotypes in the context of mutations, strain variations, QTLs and complex traits shaped by intricate epigenome-environment networks. To support the data deluge fueled by ongoing translational research, MGI has devised automated processes to gradually incorporate all multipurpose alleles generated by various IKMC partners, and built a Recombinase Data Portal (www.creportal.org) to provide expression and specificity data for each cre-containing transgene and knock-in allele, aiming to integrate phenotype characterization of newly generated mutant mouse lines as available. MGI users can access mouse phenotypes, alleles and disease models of preclinical value using a suite of tools, including an enhanced Quick Search Tool, Mouse GBrowse, web-based vocabulary browsers and data-specific query forms. Robust search inputs include standardized terms from the Mammalian Phenotype Ontology, a widely adopted ontological model that enables phenotype annotations to background-specific allelic genotypes at varying degrees of granularity. Use of human disease (OMIM) terms serves to harness associations between phenodeviant mouse features and orthologous human gene mutations or disease syndromes for which defined mouse genotypes model the human condition. We will review MGI phenotype viewing options, customized retrieval of complex phenogenomic datasets and disease model mining from various perspectives. Supported by NIH grant HG000330 78 ABSTRACT 42 NEW APPROACHES TO CONDITIONALITY Aris N. Economides, David Frendewey, Peter Yang, David M. Valenzuela, Andrew J. Murphy, and George D. Yancopoulos Regeneron Pharmaceuticals, Inc. Tarrytown, NY 10591, USA Conditional mutagenesis is rapidly becoming the method of choice for the study of gene function, but the types of alleles that can be engineered are limited both by vector design and target gene structure. We therefore developed a new technology — Conditional-by-Inversion (COIN) — that utilizes an optimized invertible gene trap-like cassette, the COIN module. The COIN module is placed in the antisense strand of the target gene, where it is effectively inert until activated by a recombinase-mediated inversion event that flips the COIN module into the sense strand, thereby disrupting transcription of downstream exons while simultaneously providing a reporter for tracking the mutation. The COIN module can either be inserted into one of the target gene’s natural introns (an intronic COIN) or directly into a coding exon as part of an artificial intron (an exonic COIN), greatly increasing allele design flexibility over existing conditional knockout approaches. The unique artificial intron feature enables the introduction of the COIN module (or other elements) in nearly any location within the target gene, freeing up design choices and increasing the types of alleles that can be designed. We have constructed COIN alleles for a large number of genes and performed detailed phenotypic analysis on ES cells and mice before and after activation of the COIN modules. Beyond establishing the robustness, reliability, and broad applicability irrespective of exon-intron structure of this method, our study uncovered rare cases of post-inversion hypomorphic alleles, observed only with intronic COINs, that were caused by ‘skipping’ of the inverted COIN module, resulting in expression of a mixture of both wild type and COIN-encoding mRNA. We showed that re-engineering the intronic COIN allele as an exonic COIN by placing the COIN module into the immediate neighboring exon could rectify this problem. These results not only inform future design decisions, but may explain the hypomorphism occasionally observed with traditional gene trap alleles, which are by nature intronic. Finally, the exon-splitting and reporter features of COINs open up new engineering modalities for the generation of multifunctional alleles that go beyond conditional-nulls, ranging from simple splitting of exons to introduce novel intronic elements to complex multifunctional alleles with more than two functionalities. Examples of these novel allele designs will be presented. 79 ABSTRACT 43 MOUSE METABOLIC PHENOTYPING CENTERS: NATIONAL CONSORTIUM FOCUSING ON DIABETIC COMPLICATIONS Renee LeBoeuf University of Washington, Seattle, WA, USA The Mouse Metabolic Phenotyping Center consortium (www.mmpc.org) is funded by the National Institutes of Health and has the mission to advance research by investigators using mice primarily for diabetes, diabetes complications and metabolic diseases. There are six science and one bioinformatics centers located across the United States of America which provide unique or well standardized phenotyping tests. We provide in depth consultations for planning experiments and for data analysis and biological interpretations. We are also compiling a data base for mouse models studied within our consortium which is open to NIH and industry-contributing researchers. We support courses including glucose clamping and tracer usage in metabolic research, and have two grant awarding mechanisms. The Animal Models of Diabetic Complications Consortium, made up of individual investigators who obtain special strains from The Jackson Laboratory, is another group using our services. Examples of data and MMPC usage will be given. 80 ABSTRACT 46 GENOMIC IMPRINTING: INSIGHTS FROM THE GNAS CLUSTER Jo Peters MRC Harwell, Mammalian Genetics Unit, Oxfordshire, UK Genomic imprinting results in gene expression according to parental origin. Thus imprinted genes are functionally haploid and so genomic imprinting must confer significant advantage to offset the cost associated with haploidy. Over one hundred and thirty well established imprinted genes are known in the mouse and they tend to occur in clusters. The Gnas cluster contains two protein coding genes with major phenotypic effects. These are Gnas itself that is maternally expressed in some tissues and Gnasxl that is exclusively paternally expressed. Gnas determines the stimulatory G-protein subunit Gsα and Gnasxl encodes a variant G-protein subunit XLΑS. Both GSA and XLΑS are signal transduction molecules but they have opposite and potentially antagonistic functions in the regulation of metabolism and behavior, and each has major effects on preweaning viability. We used a gene targeting approach to investigate how the imprinted expression of Gnas and Gnasxl is regulated. Imprinted expression within the cluster is under the overall control of an imprinting control region (ICR). This region is DNA-methylated on the maternal allele but unmethylated on the paternal allele. The unmethylated ICR contains an active promoter for a macro noncoding RNA, Nespas, that we have shown is a cis-acting regulator for the paternal allele. This finding adds to other evidence that noncoding RNAs are major elements for regulating imprinted gene expression, although the mechanisms are not well understood. Nespas runs antisense to Nesp, a paternally repressed protein coding transcript and our results show that Nespas is required to silence Nesp so that Gnas can be silenced in turn on the paternal allele. In addition we have shown that Nespas is required for methylation and therefore inactivation of the Nesp promoter. Furthermore, a low level of Nespas can downregulate Nesp in cis through chromatin modification at the Nesp promoter in the absence of DNA methylation. Our results indicate that a complex set of RNA and epigenetic interactions have evolved to regulate imprinted gene expression. 81 ABSTRACT 47 THE BATTLE OF THE SIGNALS: NON-CODING RNAS VS. ENHANCERS IN THE TRANSCRIPTIONAL REGULATION OF THE KCNQ1 IMPRINTED REGION Anjali Raval, Lisa Korostowski, and Nora Engel Fels Institute, Temple University School of Medicine, Philadelphia, PA, USA Long non-coding RNAs (lncRNAs) are a growing category of RNAs with regulatory functions, few of which have been functionally characterized. The Kcnq1 imprinted domain is involved in Beckwith-Wiedemann syndrome. The highly conserved mouse cluster is regulated by a paternally expressed 90kb lncRNA, Kcnq1ot1, that silences most of its neighboring genes, acting strictly in cis. Kcnq1ot1 is transcribed from an intron of Kcnq1 in antisense direction. Intriguingly, the paternal Kcnq1 is released from silencing in a tissue-specific manner, suggesting that strong enhancers can override silencing by the ncRNA. To study the in vivo spatial organization of the domain and provide insight into the complex expression patterns, we carried out chromatin conformation capture (3C) assays. Three-dimensional chromatin topology plays a vital role in bringing wide-spread regulatory elements into close proximity. We anchored the assays at the Kcnq1 and Kcnq1ot1 promoters and tested embryos and neonatal tissues of wild-type mice and mice with a truncation at Kcnq1ot1. Results showed that interaction frequencies are developmentally regulated and exhibit tissuespecificity. In fact, physical contact between the Kcnq1 promoter and specific DNA sequences allowed us to identify novel enhancers. Furthermore, we found major differences in interaction profiles between wild-type and mutant mice. Transcription of the lncRNA physically restricts the regions that the Kcnq1 promoter can interact with in the wild-type mice, whereas mutant mice producing a severely truncated ncRNA exhibit promiscuous contacts and deregulated expression of Kcnq1. These results suggest a unique model whereby the silencing function of Kcnq1ot1 is exerted by restricting the access of promoters to specific enhancers by modifying the flexibility of the chromatin fiber. Nevertheless, strong tissue-specific enhancers can override this effect and establish the three-dimensional contacts required to promote transcription. 82 ABSTRACT 48 AN IDENTITY CRISIS: ABNORMAL MALE GERM CELL DEVELOPMENT AND THE INITIATION OF TESTICULAR CANCER Jason Heaney, Jean Kawasoe, Megan Michelson, and Joseph Nadeau Case Western Reserve University, Department of Genetics, OH, USA Testicular germ cell tumors (TGCTs) are the most common cancer in young men. TGCTs result from anomalies in the development of primordial germ cells, totipotent embryonic stem cells that normally differentiate into mature gametes. Spontaneous TGCTs occur at an appreciable frequency only in the 129 family of inbred strains. TGCTs in mice initiate around embryonic day 13.5 (E13.5) during the same developmental time-point at which germ cells commit to meiosis or mitotic arrest (the mitotic:meiotic switch). In females, embryonic oocytes pluripotent gene expression decreases and Stra8 expression is induced to initiates entry into meiosis. In males, pluripotent gene expression decreases, Stra8 is not induced and gonocytes, the embryonic precursors of adult male germ cells, become quiescent until after birth when they differentiate to form spermatogonia. Given the overlap in the timing of TGCT initiation and the decision to enter meiosis or mitotic arrest, we tested whether a defect in the mitotic:meiotic switch contributes to TGCT development. We discovered that TGCT susceptible gonocytes prematurely express markers of adult spermatogonia and embryonic oocytes between embryonic days E13.5 and E16.5. However, the similarities in differentiation stop at a stage resembling Ngn3-positive, Kit-negative, undifferentiated A spermatogonia. A sub-population of TGCT susceptible gonocytes express Ccnd2 but not Kit. Because expression of both genes occurs at and contributes to male germ cell differentiation to meiotically-commitment, differentiated A spermatogonia, the ultimate fate of TGCT susceptible gonocytes may be altered from meiotic to tumorigenic. To test whether prematurely differentiating, TGCT susceptible gonocytes commit to meiosis, we immunolabeled chromosome spreads for components of the synaptonemal complex (SCP3 and SCP1). Interestingly, axial elements form but synapsis does not occur in TGCT susceptible gonocytes. These results suggest that prophase I of meiosis is initiated but aborted in TGCT susceptible gonocytes. We recently discovered that suppression of pluripotent gene expression is delayed in TGCT susceptible gonocytes. Interestingly, in vitro stimulation of pluripotent gene expression in undifferentiated A spermatogonia induces the formation of pluripotent stem cells with teratoma forming capacity. Similarly, TGCT formation in vivo may result from the establishment of A spermatogonia-like cells with pluripotent stem cell properties. Ongoing studies are testing the influence of retinoic acid on TGCT susceptibility and the contributions of Stra8 expression and meiotic initiation to TGCT formation. 83 ABSTRACT 49 AGE-ASSOCIATED CHANGE OF ENERGY METABOLISM ARE GENETICALLY DISSECTED BY MOUSE INTER-SUBSPECIFIC CONSOMIC STRAINS Toyoyuki Takada1, Akihiko Mita1, Shigeharu Wakana2, Kazuo Moriwaki2, Hiromichi Yonekawa3, and Toshihiko Shiroishi1 1 National Institute of Genetics, Shizuoka, Japan 2 RIKEN BRC, Ibaraki, Japan 3 Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan Energy metabolism-related traits including fat deposition are governed by environmental and complex genetic factors, which are also largely influenced by aging with concomitant change in basal metabolic rate. In this study, we intend to uncover the genetic determinants that control the age-associated changes of energy metabolism by functional genomics with mouse inter-subspecific consomic strains. We have established a full set of consomic (chromosome substitution) strains, B6-ChrNMSM, with each of C57BL/6 (B6) chromosomes replaced by its counterpart of MSM/Ms, which was derived from M. m. molossinus. Using the panel of consomic strains, we conducted systematic phenotype screening of body growth and physiological traits by 10 weeks of age, and successfully detected several hundred of energy metabolism-related QTLs (http://molossinus.lab.nig.ac.jp/). Currently, we extend the period of observation of the same traits to 25 weeks of age. Comparing phenotype of each strain at the two different observation periods, we systematically explored strains that show significant change in measurement values. We found that many strains drastically changed the values of energy metabolism-related traits when compared with change of the consomic background strain B6. For examples, consomic strains of chromosome 11 and 15 showed extremely low rate in increase of fat deposition. This suggests that although wild-derived MSM/Ms strain has thrifty-type metabolic pathway as a whole, it also carries genetic factors leading to age-associated anti-obesity. Thus, this study shows that the inter-subspecific consomic strains would provide powerful tools to dissect ageassociated change of energy metabolic-related traits. 84 ABSTRACT 50 GENETIC REGULATION OF AGING RELATED PHENOTYPES AND LIFESPAN IN MICE Rong Yuan1, SW Tsaih1, Q Meng1, K Flurkey1, J Nautiya2, SB Petkova1, MA Bogue1, KD Mills1, LL Peters1, CJ Bult1, CJ Rosen1, JP Sundberg1, M Parker2, DE Harrison1, GA Churchill1, and B Paigen1 1 The Jackson Laboratory, Maine, USA 2 Imperial College London, London, UK To better characterize aging in mice, the Jackson Aging Center carried out a lifespan study of 31 inbred strains. Clinical assessments were carried out every 6 months, measuring multiple age-related phenotypes including development traits, organ functions, body composition, hematology, hormonal levels, and immune system parameters. In a concurrent cross-sectional study of the same 31 strains at 6, 12, and 20 months, more invasive measurements were carried out followed by necropsy to assess apoptosis, DNA repair, chromosome fragility, and histopathology. Survival curves varied dramatically among strains. The median lifespans ranged from 251 to 964 days. By conducting correlation analyses, we found some phenotypes correlated with lifespan. Particularly, plasma IGF1 levels showed an inverse correlation with a median lifespan at 6 months (R =-0.33, P = 0.01). This correlation became stronger if the short-lived strains with a median lifespan < 600 days were removed (R = 0.53, P < 0.01). IGF1 levels at 6 months also significantly correlated with the ages of vaginal patency (AVP), a measurement for sexual maturation (R=-0.44, P=0.01). AVP positively correlated with median lifespan, however it is not significant (P=0.22) until the short-lived strains were excluded (R=0.38, P<0.05). Haplotype association mapping identified three loci — Vpq1, 2, 3 — on Chromosomes 4 and 16 that significantly associated with the variation in AVP. At these loci, all four wild-derived strains — WSB/EiJ, CAST/EiJ, PWD/PhJ, MOLF/EiJ — each of which represents a different sub-species of the mouse family, have the same haplotypes, which differ from those of almost all domesticated inbred strains. A consomic strain, which carries chromosome 16 of PWD/PhJ on the C57BL/6J background, had significantly delayed AVP. Bioinformatic analysis suggested that nuclear-receptor-interacting protein 1 (Nrip1) is a candidate gene for Vpq3. AVP was delayed in the Nrip1 knock out mice compared to controls, demonstrating that Nrip1 can influence the age of sexual maturation. These results suggest common genetic mechanisms may exist for regulating IGF1 levels, AVP and lifespan. Using aging related phenotypes as entrées may facilitate the identification of genes that regulate aging and longevity. 85 ABSTRACT 51 SUBSPECIFIC ORIGIN AND HAPLOTYPE DIVERSITY IN THE LABORATORY MOUSE Hyuna Yang1, Jeremy R Wang2, John P Didion3, Ryan J Buus3, Timothy A Bell3, Catherine E Welsh2, François Bonhomme4, Alex Hon-Tsen Yu5, Michael W Nachman6, Jaroslav Pialek7, Priscilla Tucker8, Pierre Boursot4, Leonard McMillan2, Gary A Churchill1, and Fernando Pardo-Manuel de Villena3 1 The Jackson Laboratory, Bar Harbor, ME 2Department of Computer Science; 3, Department of Genetics, Lineberger Comprehensive Cancer Center, Carolina Center for Genome Science, University of North Carolina Chapel Hill, NC, USA 4 Université Montpellier, Institut des Sciences de l'Evolution, Montpellier, France 5 Institute of Zoology and Department of Life Science, National Taiwan University, Taipei Taiwan ROC 10617 6 Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ, USA 7 Department of Population Biology, Institute of Vertebrate Biology, Studenec, Czech Republic 8 Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, MI, USA In this study we provide the first genome-wide, high-resolution map of the phylogenetic origin of the genome of the majority of extant laboratory mouse inbred strains. Our analysis is based on the genotypes of wild caught mice from three distinct subspecies of Mus musculus. We demonstrate tha classical laboratory strains are derived from a small pool of fancy mice with limited haplotype diversity. Their genomes are overwhelmingly M. m. domesticus in origin and the remainder is mostly of Japanese (M. m. molossinus) origin. We have generated genome-wide maps of haplotype sharing in classical inbred strains based on identity by descent from fancy mice and demonstrate that, despite broad phenoptypic diversity, classical inbred strains have limited and non-randomly distributed genetic diversity. In contrast, wild-derived laboratory strains represent a broad sampling of diversity within the species M. musculus. However, intersubspecific introgression is pervasive in these strains and contamination by laboratory stocks has played an important role in this process. The subspecific origin, haplotype diversity and identity by descent map in laboratory strains can be visualized and searched at www.cgd.jax.org. This work provides a detailed characterization of the evolutionary origins of this widely used model for biomedical research. 86 ABSTRACT 53 GENETIC ARCHITECTURE OF HYBRID STERILITY: MATCHING THE PUZZLE PIECES TOGETHER Jiri Forejt, Maria Dzur-Gejdosova, Vaclav John, Tanmoy Bhattacharyya, Petr Simecek, Sona Gregorova, Ondrej Mihola, Petr Flachs, and Zdenek Trachtulec Institute of Molecular Genetics and Center for Applied Genomics, Academy of Sciences of the Czech Republic,Videnska, Czech Republic Hybrid sterility genes are involved in reproductive isolation and consequently in speciation by restricting gene flow between related taxa. The house mouse Mus m. domesticus and Mus m. musculus, two subspecies in statu nascendi, represent unique mammalian models to study speciation because of their recent evolutionary divergence, and because of the availability of variety of genetic tools and sequence information. We are using C57BL/6J (B6) inbred strain as a model of Mus m. domesticus (cca 95% of its genome is of domesticus origin) and PWD/Ph wild-derived inbred strain of musculus origin. All F1 hybrid males from crosses of PWD females and B6 males are sterile with no sperm. By substituting B6.PWD-Chr# consomics for B6 parent in PWD x B6 hybrid cross, we tested, separately for each chromosome, the necessity of PWD/B6 heterozygosity for male sterility. Using these crosses we found that PWD/PWD homozygosity of Chr 17 or Chr 19, and B6 form of Chr X rescue hybrid sterility while PWD/PWD genotype of all other autosomes did not interfere with sterility of hybrid males. In independent experiments, we mapped hybrid sterility genes as QTLs using several genetic backcrosses, F2 intercrosses and testcrosses. At least 3 hybrid sterility loci were identified, in agreement with the consomic mapping. On Chr 17 we identified Hybrid sterility 1 locus with Prdm9 gene and, more recently, we localized the Hstx2 gene on Chr X. Genetic mapping of the gene(s) responsible for hybrid sterility rescue on Chr 19 is in progress. Detailed analysis of intrameiotic block characteristic for (PWD x B6) hybrid male sterility revealed failure of meiotic pairing of variable number of autosomes. The unpaired autosomes, differentially decorated by SYCP1 and SYCP3 antibody, displayed failure of DSB repair at pachynema marked by the persistence of ATR, RAD51 and γH2AX. The X chromosome showed lack of di-methylation of histone H3K9, and transcription profiling of prepubertal testes and FACS- sorted primary spermatocytes indicated improper inactivation of the X chromosome. Thus the interference with meiotic male sex chromosome inactivation (MSCI) could represent the major surveillance mechanism of hybrid sterility that could be under the control of at least three hybrid sterility genes. We want to propose that the interference with the MSCI is the major molecular mechanism explaining the Haldane’s rule in gametogenesis of interspecific hybrids. 87 ABSTRACT 54 MOLECULAR INTERACTIONS OF DEAD END (DND1) Angabin Matin1, Zhu Rui1, KangLi Luo1, Chitralekha Bhattacharya1, Michelina Iacovino2, Elisabeth Mahen2, Michael Kyba2 and Sita Aggarwal3 1 University of Texas, MD Anderson Cancer Center, Houston, Texas, USA, 2 University of Minnesota, Minneapolis, USA 3 Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA Inactivation of the Dead End (Dnd1) gene in mice results in sterility or development of testicular germ cell tumors in a strain specific manner. Dnd1 encoded protein, DND1, possesses canonical RNA recognition motifs. It has been shown that DND1 binds to the 3’-untranslated regions (UTRs) of mRNAs to block microRNA (miRNA) mediated inhibition of translation. As DND1 helps maintain translation from specific mRNAs, it is expected that inactivation of DND1 will result in loss of cell-type specific protein expression and cell death. DND1 is expressed in germ cells as well as in embryonic stem (ES) cells. Early germ cells and ES cells share gene expression patterns, markers and miRNAs in common. For example, both cell types express markers such as POU5F1, VASA, NANOG and FRAGILIS. They express similar miRNA families (example, miR 209-295, miR 302-367 and miR 17-92) and pluripotency factors such as POU5F1, SOX2 and NANOG. Indeed, there is speculation that ES cells may be derived from germ cells of the early embryo. Because ES cells express DND1, it is expected that they also express the mRNAs and miRNAs whose activity is modulated by DND1. Therefore, we generated a stable, modified ES cell line that expresses tagged DND1 in a regulatable manner. We used this line to perform ribonucleo immunoprecipitation (RIP) assays followed by RT-PCR. We found that the mRNA targets of DND1 in ES cells include pluripotency factors, apoptotic factors and tumor suppressors. We will present our RIP results of candidate DND1 mRNA targets from ES cells. Because protein levels of many factors are precisely regulated in ES cells, our results suggest that DND1 likely imposes another level of translational regulation of critical factors in ES cells. Second, we examined the effect of DND1 interaction with the cytidine deaminase, APOBEC3. We had previously demonstrated that APOBEC3 interacts with DND1. Using luciferase reporter assays we found that APOBEC3 can block DND1 activity and restore miRNA mediated translation repression of p27. Based on our results we hypothesize that the APOBEC3DND1 interaction may be involved in providing specificity of DND1 function. In summary, our results indicate that DND1 likely imposes another level of translational regulation to control expression of critical factors in ES cells. In addition, interaction of DND1 with other proteins such as APOBEC3 may provide further specificity of DND1 function. These studies are beginning to shed light on the role of DND1 in cell death and transformation. 88 ABSTRACT 55 GENETIC DISSECTION OF METATASIS SUSCEPTIBILITY CONVERGES ON A COMMON MECHANISM J Alsarraj, S Winter, N Goldberger, K Mattaini, M William, L Lukes, R Walker, and Kent W Hunter Laboratory of Cancer Biology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA Metastasis, or dissemination and growth of tumors at secondary sites, remains a major unsolved problem that accounts for the majority of cancer related mortality. The metastatic process is a complex cascade of events that includes escape from the primary tumor, invasion through surrounding tissue, penetration of the vasculature, resistance to anchor-independence induced apoptosis, arrest in the secondary site, exiting from the vasculature and finally growth in a novel microenvironment. Acquisition of these abilities has traditionally been thought to be due to the accumulation of somatic mutations in a small subset of cells during tumor evolution. More recently however, our laboratory has demonstrated that there is a significant inherited susceptibility for breast cancer metastasis in both mouse and human populations. Using a quantitative trait strategy we have identified and validated a number of metastasis susceptibility genes, including the genes Sipa1, Brd4, and Rrp1b. Applications of additional genome-wide analytical tools have revealed additional candidate metastasis susceptibility genes from our genetic mapping crosses. Unexpectedly, many of the genes identified appear to encode proteins that operate within the same transcriptional control and chromatin modification pathway, physically interact, and oppose each others function. These data suggest that inherited metastatic susceptibility is due to modification of a complex mechanism regulating basal transcriptional control of breast epithelial cells. 89 ABSTRACT 56 Icst IS A DOMINANT NEGATIVE MUTATION OF Lmx1b Sally H. Cross1, Lisa Mckie1, Margaret Keighren1, Dan Macalinao2, Alison L. Kearney2, Simon W. John2,3, and Ian J. Jackson1 1 MRC Human Genetics Unit, Edinburgh, UK 2 The Jackson Laboratory, Bar Harbor, ME USA 3 Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME USA The ENU-induced mutation iris-corneal strands (Icst) causes raised intraocular pressure, bulging eyes and anterior segment scarring when heterozygous and is recessive lethal. Homozygotes have limb and skull abnormalities and absent cerebellum. Icst is a missense mutation of the LIM-homeodomain transcription factor Lmx1b that prevents the binding of the mutant protein to its target DNA sequence. Mutations in LMX1B underlie the dominant human disorder nailpatella syndrome (NPS). This is a pleiotrophic disorder characterised by nail and joint dysplasia that is sometimes accompanied by kidney defects and glaucoma. It is thought to be caused by haploinsufficiency, although there is great variability in disease aetiology both within and between families. Lmx1b has been knocked-out in the mouse and although the reported homozygous phenotype is very similar to that of Icst, heterozygous knock-out mice are reported to be normal, in contrast to the strongly penetrant eye phenotype of Icst heterozygotes. Both mutant models are on C57BL/6 suggesting strain background is unlikely to account for the observed phenotypic differences. We hypothesised that Icst mutant LMX1B protein has a dominant negative mode of action and that the relative level of wild-type to mutant protein is crucial. We used recombineering to introduce the Icst mutation into an Lmx1b-containing BAC and made transgenic lines with both the wild-type and the mutant BAC. Both express the Lmx1b transgene at 65-70% the level of the endogenous gene. One copy of the wild-type BAC rescues the dominant eye phenotype and two copies rescue the homozygous lethality but not the limb phenotype. The limbs of transgenic rescued homozygotes are symmetrically ventralised, indicating that although the absolute level of wild-type LMX1B protein is greater than in Icst heterozygotes, the excess mutant protein prevents normal development. Furthermore, when we introduce the mutant Lmx1b transgene into Icst heterozygotes we see embryonic lethality, demonstrating that a normally sufficient, heterozygous, wild-type gene is insufficient when there is an increased dose of mutant protein. It has previously been suggested in humans that mutant LMX1B alleles do not influence the wild-type allele. However, we show that the gene can have dominant negative mutant forms, and some cases of NPS could be due this mechanism. 90 ABSTRACT 57 ELUCIDATING THE ROLE OF NSDHL AND CHOLESTEROL METABOLISM IN CNS DEVELOPMENT USING A CONDITIONAL KNOCKOUT ALLELE Gail E Herman1, N Bir1, L Binkley1, K McLarren2, C Boerkoel2, and D Cunningham1 1 The Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA 2 Department of Medical Genetics, University of British Columbia, Vancouver, BC, USA NSDHL (NADH steroid dehydrogenase-like) is a 3β-sterol dehydrogenase involved in the removal of C-4 methyl groups in one of the later steps of cholesterol biosynthesis. Mutations in the murine gene are associated with the X-linked, male-lethal mouse mutations bare patches (Bpa) and striated (Str). Intellectual developmental disabilities (IDD) and developmental CNS malformations are prominent features of human disorders of cholesterol synthesis. In addition, abnormalities of cholesterol metabolism have been implicated in more common CNS disorders ranging from autism to Alzheimer’s. Recently, two families with X-linked IDD in males have been found to have hypomorphic human NSDHL mutations. In one family, female carriers exhibited significant callousness (P=0.002), linking cholesterol and behavior. To begin to understand the role of cholesterol and its intermediates in CNS development and behavior, we generated a conditional Nsdhl targeted allele (Nsdhlflx5). The “floxed” animals are fertile and without any obvious phenotype. Nsdhlflx5/+ females mated to Sox2-Cre males exhibit an identical phenotype to that observed in surviving Bpa1H/+ females carrying a null allele. There was also no detectable NSDHL protein in Sox2-cre deleted E9.5 or E10.5 male embryos. Nsdhlflx /Nsdhlflx5 females were mated to nestin, GFAP, or Thy1 cre males that have cre expression in neural precursors starting at E10.5; radial glia at E13.5; and selected cortical neurons at P15, respectively. No liveborn deleted NsdhlnesΔ5/Y have been recovered, while Thy1 cre deleted mice have no overt phenotypes at >100 days of age. Hemizygous GFAP cre deleted males develop early postnatal ataxia (P10-12), wasting, and cerebellar degeneration with massive apoptosis and death by P15-P20. While early cerebellar differentiation appears normal, cells of the external granule layer do not appear to exit the cell cycle and migrate properly. In addition, although GFAP cre is not expressed in Purkinje cells, these neurons also die, suggesting that they require the continued presence of supporting glia or are exposed to toxic metabolites. The hippocampus similarly exhibits neuronal cell loss beginning in the late prenatal period. Possible mechanisms, including altered cell signaling and accumulation of toxic intermediates, will be discussed. 91 ABSTRACT 58 GENETIC, STEM CELL, AND SYSTEMS ANALYSES OF NEURODEGENERATIVE DISEASES George A Carlson1, R Bennett1, ME Orr1, I Lee2, H Yoo2, J-H Cho2, D Hwang3, and LE Hood2 1 McLaughlin Research Institute, Great Falls, Montana, USA 2 Institute for Systems Biology, Seattle, Washington, USA 3 POSTECH, Pohang, Republic of Korea Prions cause transmissible neurodegenerative diseases and replicate by conformational conversion of benign forms of prion protein (PrPC) to diseasecausing PrPSc isoforms. Tracking global gene expression changes in the brains of 8 distinct mouse strain-prion strain combinations that differed widely in disease incubation time enabled capture of the effects of prion strain, host genetics, and PrP concentration. A core of 333 differentially expressed genes (DEGs) appear central to prion pathogenesis. Most of these core DEGs appear before clinical disease but are related to pathological processes. Core DEGs also change in PrP-overexpressing transgenic mice with very short incubation times, but the magnitude of differential expression is smaller and closer in time to clinical illness than in other mouse strain-prion strain combinations. The most significant DEGs in these transgenic mice change early and are not seen in the other combinations; we hypothesize that these DEGs may reflect the earliest responses to prion infection and that their gene products may be involved in prion replication. To test this, CNS stem cell-containing neurosphere cultures were infected with mouse prions. Prior to induction of differentiation, no obvious pathogenic effects of prion replication on neurospheres were observed; thus, DEGs found in prion-infected neurospheres likely reflect processes related to prion replication. This systems approach is enabling identification of intersecting networks of genes crucial for establishing and maintaining prion infection and will help define the link between replication and pathogenesis. Similar approaches are now being applied to a mouse model for frontotemporal dementia with the ultimate goal of identifying gene and protein expression signatures that can discriminate among diverse neurodegenerative diseases. 92 ABSTRACT 59 STAGE AND CELL SUBTYPE-SPECIFIC EPIGENETIC REGULATION OF MAMMARY GLAND DEVELOPMENT Deanna Acosta1, Melissa J Fazzari1, John M Greally1, and Cristina Montagna1 1 Albert Einstein College of Medicine, Bronx, NY, USA Postnatal development of the mammary gland is a complex and highly specialized process characterized by cell proliferation and tissue remodeling. Many genes involved in mammary gland development have been identified and their expression extensively investigated at all major developmental stages. However, the means by which these genes are regulated have not yet been established. Despite advances in understanding the role of epigenetics in gene regulation, little is known about the relationship between cell subtype-specific cytosine methylation and gene expression in the developing mammary gland. Based on previous reports that cytosine methylation is dynamic during development and that cell type-specific methylation affects gene expression, we hypothesize that alterations of DNA methylation levels occur during postnatal mammary gland development in a stage- and cell subtype-specific manner. We propose that these changes act as one of the mechanisms regulating the expression of transcription factors that drive development and differentiation. Using a known gene in breast development (Serpinb5) we determined if cytosine methylation changes occur during mouse mammary gland development and if these changes correlate with expression. Serpinb5 is regulated by alterations in cytosine methylation in breast cancer cells. Our preliminary studies suggest that the Serpinb5 promoter region is differentially methylated through the developmental stages and that its methylation status correlates with changes in expression. To identify novel candidate loci, we performed a genome-wide analysis of cytosine methylation levels during mammary gland development. Our study indicates that conserved, non-coding regions around the transcription start sites of a subset of genes undergo changes in methylation during development. Of these, we selected Serpinb5, Gata3, Tnfsf11 (Rankl) Cebpb, and Cebpd because they have been shown to play critical roles in mammary gland development whereas Gata2, a novel candidate gene, is involved in the development of other cell types. To approach the issue of mammary gland tissue heterogeneity, we sorted luminal and myoepithelial cells from different developmental stages. With these cell subtypes, we aim to establish a genome-wide methylation profile and validate the role of cytosine methylation in the regulation of our candidate genes during mammary gland development. This study would not only uncover the role of cell subtypespecific cytosine methylation in tissue morphogenesis, but it will help pinpoint genes that could be altered at early stages of breast tumorigenesis. 93 ABSTRACT 60 TOWARD AN UNIFIED MEASURE OF INTRASPECIFIC SELECTIVE PRESSURE Roberto Amato1,2, Gennaro Miele1,2, Michele Pinelli1,3, and Sergio Cocozza1,3 1 Gruppo Interdipartimentale di Bioinformatica e Biologia Computazionale, Università di Napoli "Federico II" & Università di Salerno, Italy 2 Dipartimento di Scienze Fisiche, Università di Napoli "Federico II" - INFN Sezione di Napoli, Naples, Italy 3 Dipartimento di Biologia e Patologia Cellulare e Molecolare "L. Califano", Università di Napoli "Federico II", Naples, Italy During recent decades the study of human evolution has been of increasing interest, also due to the large amount of data now available. In the mean time, new applications of evolutionary biology to medical problems are being discovered at an accelerating rate. Several estimators for the selective pressure have been proposed. Being introduced to face different aspects of selective pressure, each measure has its own pros and cons. We focused on the most widely used intraspecific estimators of selective pressure. In particular, to cope with both inter- and intra-group phenomena, we analyzed the fixation index (FST) and some measures based on the extended haplotype homozygosity (namely REHH, iHS, XP-EHH). We assessed the performances of each measure on simulated data produced using Fregene, a tool developed and calibrated to reproduce, in a biologically sound manner, our evolutionary history, also taking into account for complex demographic, selection and recombination scenarios. By using GRID facilities to produce a large enough amount of data, we were able to exhaustively assess the performances of each estimator with regard to several biological parameters of the sites under selection (e.g. selection coefficient, dominance model, age of selection). Our analysis highlighted a complementary behavior of these estimators suggesting a possible strategy of merging them in an unified and versatile measure of selective pressure. 94 ABSTRACT 61 A NEW TARGETED MUTATION, CASPA, IN THE GNAS COMPLEX SHOW S HYPERACTIVITY AND ATAXIA Simon Ball1, Sally Eaton1, Christine Williamson1, Charlotte Tibbit1,2, and Jo Peters1 1 MRC Harwell, Mammalian Genetics Unit, Oxon, UK 2 Current address: MRC Functional Genomics Unit, Oxford, UK Imprinted genes, by definition, are preferentially expressed from either the maternally or paternally derived allele. They are known to affect growth, survival and behaviour. We report here a new targeted mutation, Caspa, within the Gnas imprinted gene complex. On maternal inheritance, Caspa/+ mice appear chunky have a tail kink or bend and most die before weaning. Notably they are hyperactive, crawling by the day after birth and running by 7-8 days, several days before their wild-type sibs. Ataxia has been seen in some several days after birth, but this is not associated with cerebellar defects. The hyperactivity is reminiscent of that seen in mice with paternal uniparental partial disomy for distal chromosome 2 (PatDp.dist2). In both Caspa/+ and PatDp.dist2, Gnasxl, a paternally expressed transcript in the Gnas complex, is expressed at twice the normal level. We suggest that over expression of Gnasxl, that encodes a variant alpha subunit XLαs of the signalling Gs protein, is the cause of the hyperactivity. 95 ABSTRACT 62 RAT RESOURCE AND RESEARCH CENTER Beth A. Bauer, EC Bryda, CL Franklin, LK Riley, and JK Critser Department of Veterinary Pathobiology, Research Animal Laboratory, University of Missouri, MO, USA Diagnostic The Rat Resource and Research Center (RRRC) was established in 2001 with funding from the National Institutes of Health (NIH) National Center for Research Resources (NCRR). The goals of the RRRC are to (1) provide the biomedical community with a repository and distribution center for valuable rat strains, and (2) to shift the burden for maintaining and distributing unique rat models from investigators to a National Resource Center. Currently, the RRRC has more than 160 rat lines received through active recruitment of valuable rat models and donations from investigators who have created models. Upon importation into the RRRC, gametes and embryos are cryopreserved to insure against future loss of the model. Recent additions to the repository include a collection of ENU and transposon-generated mutants from the Medical College of Wisconsin. Also, newly developed rat embryonic stem cell lines have been imported into the RRRC and are available for distribution to the research community. Models in the RRRC are available for distribution as live animals, tissues, or cryopreserved gametes and embryos. In addition to repository and distribution functions, the RRRC also conducts research. To date, research efforts by the RRRC have significantly advanced cryobiology and assisted reproductive technologies (ART) for the rat. Due to high success rates with intracytoplasmic sperm injection (ICSI), the RRRC can use sperm cryopreservation as a cost effective method for banking large collections of single gene mutations and ensuring reliable recovery when models are requested. The RRRC (http://www.rrrrc.us) is a valuable resource to the community of rat users as it continues to expand the number of accessible rat models and services available to the biomedical community. 96 ABSTRACT 63 LOCI ON CHROMOSOME 2 ARE ASSOCIATED WITH NAÏVE AIRWAY HYPERRESPONSIVENESS IN CD-1 OUTBRED MICE David R Beier1, Andrew Kirby2, Mary Prysak1, Mark Daly2, and Emily Cozzi1 1 Department of Medicine, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston MA, USA 2 Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA Asthma is a complex syndrome characterized by episodic and reversible airway obstruction, airway inflammation, and airway hyperresponsiveness. The genetic mechanisms predisposing individuals to asthma and in particular airway hyperresponsiveness (AHR) are largely unknown. Quantitative trait linkage analyses (QTL) have been performed to identify loci associated with naive AHR in a limited number of inbred murine populations. The goal of this study was to uncover loci associated with AHR in a genetically heterogeneous outbred murine population. Outbred CD1 mice were intercrossed to generate F1 progeny from four male/female breeding pairs (N=88). All mice were phenotyped by determining methacholine-induced airway resistance with the invasive flexiVent system. CD1 F1 mice were genotyped using an Illumina medium density 1440 SNP panel and linkage analysis was carried out using Merlin and GeneHunter software. The CD1 F1 progeny had a highly variable AHR phenotype (2.2-21.5 cmH2O.s/mL). Linkage analysis of this population was done by inferring parental haplotypes; this identified loci on chromosome 2 as being significantly associated with the AHR phenotype in 2 of the 4 families (additive LOD score = 3). Remarkably, this data is consistent with what we have previously discovered for the A/J and AKR/J inbred hyperresponsive strains. In order to obtain high resolution of the loci identified on chromosome 2, CD1 F1 mice were genotyped on the JAX 600K SNP Diversity Array panel. Analysis of this data may facilitate the identification of specific haplotypes associated with the AHR trait. In summary, we have found a robust distribution and segregation of AHR as a trait in outbred CD1 F1 mice, and identified loci on chromosome 2 that are significantly associated with AHR. Overall, these data highlight that common AHR loci may exist between hyperresponsive murine strains. 97 ABSTRACT 64 HIGH-RESOLUTION MAP AND CANDIDATE GENE ANALYSIS FOR ÉBOURIFFÉ (EBO), A HAIR MUTATION ON MOUSE CHROMOSOME 2 ASSOCIATED WITH INFERTILITY Fernando Benavides1, J Jaubert2, CJ Perez1, J-L Guénet2, J Barrera1, B Cole1, and CJ Conti1 1 Department of Carcinogenesis, M.D. Anderson Cancer Center, Science ParkResearch Division, Smithville, TX 2 Unité de Génétique Fonctionnelle de la Souris, Institut Pasteur, Paris, France Forward genetics is a powerful approach to discover genes with pleiotropic effects in the mouse. We previously described ébouriffé (ebo), a mouse mutation with a hair phenotype and infertility (male and female). This autosomal recessive mutation arose spontaneously in a substrain of BALB/c and was localized on proximal chromosome 2 using an interspecific backcross. Affected ebo/ebo mice show curly vibrissae and wavy hair from day P7, and males have defective spermatogenesis with structural abnormalities of the acrosome that lead to abnormal head shapes and isolated flagellum (Biology of Reproduction 55: 355-363, 1996). New mapping crosses allowed us to narrow down the segment of Chromosome 2 containing ebo to a ~1 Mb interval, between markers D2Mit153 and D2Mit64 (29.27 - 31.20 Mb), a region of homology with human chromosome 9q34. Here, we will summarize the status of our ongoing candidate gene analysis for ebo. Positional candidate genes include Rapgef1, Slc27a4, Coq4, Gle1, Spna2, Zdhhc12, Zer1, Tbc1d13, Ccbl1, and Nup188. 98 ABSTRACT 65 MOLECULAR GENETIC ANALYSIS OF THE NADPH OXIDASE OF THE VESTIBULAR SYSTEM Catrina A Spruce1, John P Flaherty2, Heather E Fairfield1, and David E Bergstrom1 1 The Jackson Laboratory, Bar Harbor, ME USA 2 New York College of Osteopathic Medicine of New York Institute of Technology, Old Westbury, NY USA Age-related vestibular decline is a prevalent and increasingly common ailment within the American population. Much of the clinical spectrum is due to the loss and/or displacement of otoconia within the inner ear. Otoconia are proteinaceous particles containing crystalline inorganic calcite. Affixed to underlying stereocilia within the vestibular portion of the inner ear, otoconia act as inertial masses that shift in response to an organism’s linear movements and to the force of gravity. To better understand the molecular genetics of otoconial deposition and maintenance, we have previously explored the molecular genetic basis for three mouse vestibular mutants named “neuroscience mutagenesis facility 333” (nmf333), “head tilt” (het), and “head slant” (hslt), each of which displays the classic phenotype of congenital otoconial agenesis. Together these studies identified Cyba, Nox3, and Noxo1, respectively, as the dysfunction-causative genes. Cyba, in addition to its role in the inner ear, is also a known component of the well-characterized and canonical NADPH oxidase of phagocytic cells. Moreover, Nox3 and Noxo1 are paralogs of the canonical NADPH oxidase components, Cybb and Ncf1. Thus, the emerging picture is that of a Cyba, Nox3 and Noxo1-containing inner ear NADPH oxidase with structural parallels to the well-known phagocytic complex. The critical roll of the inner ear oxidase in otoconiogenesis and proper vestibular function suggests Cyba, Nox3, and Noxo1 as candidate genes potentially involved in human disease- and age-related otoconial loss. Moreover, the Nox3-based NADPH oxidase, as a source of potentially harmful reactive oxygen species, is now under investigation as a possible factor in drug-, age- and noise-related hearing loss. Our presentation will describe recent gene trap and gene targeting studies aimed at elucidating the roles of other NADPH oxidase components in the inner ear. 99 ABSTRACT 66 MOUSE MODELS OF COSTELLO SYNDROME Heather E Fairfield1, John P Flaherty2, Catrina A Spruce1, Leah R Donahue1, and David E Bergstrom1 1 The Jackson Laboratory, Bar Harbor, ME USA 2 New York College of Osteopathic Medicine of New York Institute of Technology, Old Westbury, NY USA Costello Syndrome (CS) is a rare, complex, developmental disorder characterized by a number of features including— failure to thrive, characteristic facies, delay in intellectual development, hypertrophic cardiomyopathy, arrhythmia, and predisposition to both benign and malignant tumors. Past studies identifying gain-of-function mutations in the HRAS gene as the basis of human CS, and strong conservation with the mouse ortholog Hras1, have led to the development of a Gly12Val (G12V) mouse model of CS. To extend animal modeling studies to include various allelic forms of the disease, we are developing five additional mouse models of CS. Using a recombineeringbased “knock-in” approach, we have completed construction of five vectors, each containing a loxP-flanked, neomycin resistance (Neor) selection cassette in intron 1 of Hras1 and site-directed mutations encoding each of the following five pathogenic alleles — G12A encoded by GCA, G12A encoded by GCC, G12S encoded by AGC, G12V encoded by GTA and G12V encoded by GTT. Four of the five strains have been completed creating animal models for the G12AGCA, G12AGCC, G12VGTA and G12VGTT forms of the disease. Suppression of each CS-causative allele by the oppositely transcribed Neor transcript can be relieved by breeding affected mice to inner cell mass- or germline-specific “deleter” Cre lines such as Meox2-Cre or Zp3-Cre, respectively. Craniofacial abnormalities like those seen in human patients are clearly evident. Preliminary phenotypic assessment also suggests possible neoplasia as well as cardiac and skin abnormalities. Each strain will be deposited into The Jackson Laboratory Repository. Together, these five models hold the potential for uncovering allelic and codon preference influences on development and neoplasia in CS, identifying CS modifying genes, and dissecting tissue-specific facets of CS using spatially-controlled Cre driver lines. 100 ABSTRACT 67 THE APPLICATION OF HIGH-THROUGHPUT SEQUENCING TECHNOLOGIES FOR MUTATION DISCOVERY IN THE JACKSON LABORATORY’S MOUSE MUTANT RESOURCE Laura G Reinholdt, David E Bergstrom, Muriel T Davisson-Fahey, Cathleen M Lutz, Michael Sasner, Steven A Murray, Jeff Lake, Steven Rockwood, Leah Rae Donahue and the MMR team The Jackson Laboratory, Bar Harbor, ME USA The application of high-throughput sequencing technologies is revolutionizing the process of mutation detection. By incorporating these technologies, we in The Jackson Laboratory’s Mouse Mutant Resource (JAX MMR) are extending our characterization of new spontaneous mutant strains to the point of causative gene identification. For over fifty years, the mission of the MMR (and its predecessors) has been to provide mouse models of human genetic illness to the scientific community. Through our Mouse Phenodeviant Search process, atypical mice are identified from among the millions of inbred mice produced yearly at JAX and isolated. This not only ensures genetic stability within the originating strain; but also, provides a pool of potential spontaneous mutant mice for further characterization. After colonies are established, heritability is proven, and the modes of inheritance are determined; subchromosomal locations for each mutant locus are established by backcross or intercross in conjunction with SSLP or SNP genotyping. In place of Sanger-based sequencing of promising candidate genes from select mutant strains, interval-specific array capture and Illumina GAIIx-based high-throughput sequencing are now being incorporated to broaden the scope of mutation detection and dramatically shorten the time to causative gene identification. A general phenotypic assessment, cryopreservation of embryos or gametes, and archival of mutant DNA completes the characterization. Findings for each mutant strain are disseminated through the scientific literature or on the MMR website at http://mousemutant.jax.org/. Additional projects are underway to incorporate whole-exome approaches to spontaneous/induced mutation discovery, reducing the need for genetic mapping. The presentation will enumerate the many advantages of studying spontaneous mutant mice and summarize current resource offerings. 101 ABSTRACT 68 THE ROLE OF CAML IN THE INNER EAR Elizabeth C Bryda1, Nathan T Johnson1, Cynthia L Besch-Williford1, Kevin K Ohlemiller2, and Richard J Bram3 1 Department of Veterinary Pathobiology, Research Animal Diagnostic Laboratory, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA 2 Central Institute for the Deaf at Washington University, Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, USA 3 Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA Calcium modulating cyclophilin ligand (CAML) is a ubiquitously expressed cytoplasmic protein which appears to play a role in embryonic and thymocyte development, EGFR and LCK signaling, and chromosomal stability. In previous studies, we demonstrated that there is a protein –protein interaction between CAML and the cytoplasmic region of Cadherin23. This led us to speculate that CAML might be important in the inner ear and play a role in the development and/or function of hair cells. To enable spatially and temporally controlled Caml gene expression, a mouse line was generated that allows knockout of Caml expression specifically in the inner ear upon administration of tamoxifen. Using this mouse model, tamoxifen was administered immediately after birth to neonates or daily to pregnant females from day E10.5 to E15 to assess the effect of loss of CAML during postnatal or embryonic development respectively. Hearing in treated animals was tested by auditory brainstem response (ABR) analysis. The presence of hair cells was evaluated via histological analysis of the inner ears of treated mice. To date, there is evidence that lack of Caml expression in the inner ear leads to deafness. Elucidating a role for CAML in the inner ear will aid our understanding of the molecular pathways important for auditory development and function. 102 ABSTRACT 69 INTERESTING PHENOTYPES FOUND AS PART OF THE INFECTION CHALLENGE IN THE WELLCOME TRUST SANGER INSTITUTE’S MOUSE GENETICS PROGRAMME Simon Clare, Leanne Kane, Lynda Mottram, Jacqui White, Ramiro Ramirez – Solis, and Gordon Dougan All the Mouse genome project team at the Sanger Institute Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridgeshire, UK As part of the Wellcome Trust Sanger Institute’s Mouse Genetics Programme all mutant mouse lines generated in this high throughput programme are challenged with Salmonella Typhimurium an intracellular pathogen which induces a systemic disease and Citrobacter rodentium a natural mouse pathogen which forms attaching and effacing lesions on the surface of the gastrointestinal lumen (details of the challenges can be found on the poster “Phenotyping of knockout mice using bacterial pathogens as part of the Wellcome Trust Sanger Institute’s Mouse Genetics Programme”). To date we have identified 18 genes which contribute to controlling the susceptibility to bacterial infection out over 250 knockout mice lines phenotyped so far. These phenotypes include hits in novel genes as well as gene of known function and range from severe phenotypes, were the mice succumb to infection rapidly, too more subtle phenotypes. Here we would like to show a selection of the data generated as part of this programme and details of how to freely access all the data and resources from the programme as a whole. 103 ABSTRACT 70 THE EXPRESSION OF SEPTIN 9 IN HUMAN BREAST CANCER Diana Connolly1, Zhixia Yang1, Elias T. Spiliotis2, Maria Castaldi3, Nichelle Simmons3, Pascal Verdier-Pinard4, and Cristina Montagna 1 Department of Genetics, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY, USA 2 Department of Biology, Drexel University, Philadelphia, PA, USA 3 Department of Surgery, Jacobi Medical Center, Bronx, NY, USA 4 INSERM UMR 911 CR02, Aix-Marseille Université, Marseille, France Septins are a family of GTPases known for their involvement in various cellular processes and interactions with the cytoskeleton. Septin 9 (SEPT9) in particular, is one of the fourteen members of this family that is of particular interest because of its contribution to breast tumorigenesis. Our group identified SEPT9 as a novel oncogene that is amplified and over-expressed in breast cancer. Alteration of SEPT9 expression has also been implicated in ovarian carcinoma and results in cell cycle arrest when down-regulated. The study of SEPT9 is complex due to the potential ability of this gene to encode 18 different isoform transcripts and at least 5 different polypeptides. Studies show that SEPT9 isoforms are differentially expressed between normal and tumor breast cells, but their relation to stages of breast cancer development and breast cancer subtypes is still unclear. Our group has approached this question by analyzing isoform expression at the DNA, RNA and protein levels. Using mouse models for breast cancer progression, and human tumor and matching adjacent normal breast tissue, we have detected gene amplification and expression changes of the SEPT9 isoforms among the tissue types. For the first time we could show that pre-malignant cells have a SEPT9 expression profile that distinguishes them from both normal breast and advanced adenocarcinomas. Moreover, we found that the differential expression of at least one isoform (SEPT9_v3) was due to epigenetic regulation via the methylation status of an alternative promoter region. Our data suggest that other alternative promoters within this gene could also be regulated by DNA methylation and thus dramatically affect the expression of SEPT9 isoforms in tumorigenesis. These findings are supported by the establishment of a diagnostic SEPT9 DNA methylation assay for detection of colorectal cancer in peripheral blood, and furthermore indicate that the amplification and differential expression of SEPT9 isoforms may represent a potential molecular signature with functional and diagnostic significance for breast tumor progression and early detection. 104 ABSTRACT 71 GENETIC CONTRIBUTION TO LIVER FIBROSIS David DeSantis1, Michelle Pritchard3, Laura Nagy3, Joseph Nadeau2, and Colleen M Croniger1 1 Departments of Nutrition, 2Department of Genetics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA 3 Department of Gastroenterology, Department of Pathobiology, Cleveland Clinic, Cleveland, Ohio, USA Liver disease is one of the serious complications associated with either chronic excessive alcohol consumption or calorie consumption. Complications can range from steatosis to hepatitis and eventually fibrosis and cirrhosis. Pathological evolution of alcoholic steatohepatitis (ASH) and non-alcoholic steatohepatitis (NASH) are very similar yet only 20-50% of these patients develop cirrhosis. The mechanism for progression to fibrosis in these patients is presently unknown. Current nutritional and transgenic models of hepatic steatosis replicate preconditions for ASH and NASH, but fail to represent the multifactorial pathogenesis of these diseases. To identify susceptibility genes to fibrosis, we have analyzed the response to high fat high sucrose diet (HFHS) in chromosome substitution strains (CSS) developed by Dr. Joseph Nadeau (Case Western Reserve University). CSS strains have one chromosome from the A/J inbred strain that has been substituted for the corresponding chromosome on the B6 genetic background. CSS-17 which contains A/J Chromosome 17 was resistant to obesity and steatosis. Analysis of congenic strains derived from CSS-17 identified a very small segment of A/J chromosome that conferred resistance to obesity and steatosis. In collaboration with Dr. David Adams (The Wellcome Trust Sanger Institute), A/J Chromosome 17 has been deep short-read sequenced to 22X coverage. Using comparative analysis of the sequence from A/J and B6 Chromosome 17, we have identified genes in the small region of A/J chromosmone that have single nucleotide polymorphisms (SNPs) in the proximal region or coding region. We hypothesize that these genes play a critical role in development of hepatic fibrosis. Our preliminary data support this hypothesis. To induce fibrosis we have used CCl4 administration in A/J mice, CSS17 and congenic strains from CSS17. A/J mice remained resistant to CCl4induced injury while CSS-17 and B6 mice developed CCl4- induced injury and fibrosis. Most importantly the congenic strain containing only 27.8 Mb of A/J chromosome had CCl4- induced fibrosis at significantly higher levels compared to B6 mice. A region more distal was protected from CCl4-induced fibrosis. Thus Chromosome 17 contains genes that are contributing to susceptibility to CCl4 liver injury. 105 ABSTRACT 72 DIET, OBESITY, AND SUSCEPTIBILITY TO COLON POLYPS Stephanie K. Doerner 1,3 and Joseph H. Nadeau 1,2,3 1Department of Genetics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA 2Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA 3Case Center for Transdisciplinary Research on Energetics and Cancer Colon cancer is the third most common cancer worldwide and is the second leading cause of cancer related deaths in the United States. Both genetic and environmental factors such as diet, obesity, microbial profile, inflammatory diseases and heritability contribute to susceptibility. However, assessing the contributions of obesity and diet on the development and progression of colon cancer is difficult because they are strongly correlated in humans. Recent work from our laboratory suggests that the amount of dietary saturated fat plays a critical role in the development of intestinal polyps in the B6.ApcMin/+ mouse model. Using Congenic-Consomic strains, we show that a high-fat diet, regardless of susceptibility to diet-induced obesity, affects polyp number. We show that the type of fat (saturated versus omega-6 polyunsaturated) significantly affects polyp number after 60 days. We also show that polyp numbers stabilize in mice after 30 days on the high-fat and low-fat diets. Preliminary data suggests that pro-inflammatory cytokines are elevated in intestinal polyps of mice fed the high-fat diet compared to the low-fat control diet and that circulating inflammatory factors are also elevated in mice fed a high-fat diet. Together, our diet studies demonstrate that the quantity and kind of dietary fat influences polyp number, independent of obesity. Although much has been learned about the effects of diet on cancer severity, the mechanism remains unknown. The B6.ApcMin/+ mouse model for colon cancer will be used to test the effects of alternative fat compositions on cancer severity, the influence of inflammation in carcinogenesis, and the interaction between diet and WNT signaling. 106 ABSTRACT 73 GENETIC REGULATION OF FRACTURE RISK IN INBRED MICE Leah Rae Donahue, GA Churchill, S-W Tsaih, and WG Beamer The Jackson Laboratory, Bar Harbor, Maine, USA The measurement often used to predict bone strength is bone mineral density measured by x-ray absorptiometry. Other bone traits - size, shape, architecture and material properties, as well as muscle mass - are important qualitative contributors to bone strength and should be considered when predicting fracture risk. There is intense interest in genetic determinants of fracture risk based on these indices of bone strength. Interpretation of human studies is complicated by heterogeneity and environmental differences, indicating that genetic regulation of bone strength is complex and that animal models are critical to progress in this field. Our goal was to determine which muscle and bone phenotypes predict femoral bone strength in mice and to discover genetic models for further analyses of factors that contribute to fracture risk. We collected skeletal geometry, muscle mass, volumetric BMD (vBMD), and bone strength data from female mice of 9 inbred strains. pQCT (SA Plus,Stratec) was used to measure vBMD, periosteal perimeter (Ps.pm), and muscle area at the mid diaphysis; digital calipers were used to measure femur length and medio-lateral diameter (M/L) at mid diaphysis; peak load, stiffness, and energy to break (EB) were measured by three point bending (MTS). EB is often used as a measure of the amount of energy required to cause bone to fracture. We found that peak load and stiffness were both predicted by femur length (p=.002;.001), M/L diameter (p=.009;.029), muscle area (p=.001;.011), and vBMD (p=006;.002). EB was predicted by Ps.pm (p=.044), M/L (p=.037), and muscle area (p=.009), but was independent of vBMD. A strategy to genetically decompose the EB phenotype could be based on crosses between a low EB (low bone size/muscle area) strain and a high EB (plus high bone size/muscle area) strain. Such a combination is present in SWR/J and FVB/NJ, or in 129/SvImJ crossed to C57BL/6J. The F2 generation progeny from either cross would provide mice that could be used to analyze the genetic regulation of EB. These data show that bone geometry, density, and muscle mass are important determinants of femoral strength, and illustrate that bone strength is a complex trait with genetic regulation. We propose a mouse genetic model to discover the genetic regulation bone geometry, muscle mass, and BMD, and to determine the relative contribution of each to overall bone strength and fracture risk. 107 ABSTRACT 74 IMPRINTED XLAS: A NEW PLAYER IN BONE AND ADIPOCYTE METABOLISM Sally Eaton, Simon Ball, Colin Beechey, Christine Williamson, and Jo Peters MRC Harwell, Mammalian Genetics Unit, Harwell Science & Innovation Campus, Oxfordshire, OX110RD, United Kingdom It is recently understood that bone and energy metabolism are tightly coregulated through the central and sympathetic nervous systems. Now that this mechanism of the control of bone metabolism has been described we are better placed to understand how new entrants regulate both bone and adipocyte metabolism. We describe here a new mouse mutation which presents with a severe growth retardation, a reduction in fat mass, an increased metabolic rate, a lowered bone mineral density, an increase in Ucp1 and a reduction in leptin. We further describe that this phenotype can be attributed to truncation of the paternally expressed transcript, Gnasxl resulting in loss of the protein, XLAS. Gnasxl arises from the imprinted Gnas cluster and is expressed exclusively from the paternally derived allele. Gnasxl encodes for a protein that acts as the alpha subunit of the heterotrimeric Gs protein and regulates receptor-stimulated cAMP production. This is to our knowledge the first report describing a role of XLAS in bone metabolism and we propose that XLAS is involved in the co-regulation of bone and adipocyte metabolism at the level of the hypothalamus through its actions on the sympathetic nervous system. 108 ABSTRACT 75 THE SANGER MOUSE GENETICS PROGRAMME: HIGH THROUGHPUT RECESSIVE LETHALITY SCREEN Jeanne Estabel, Elizabeth Tuck, Damian Carragher, Jennifer Salisbury, and Jacqueline K. White on behalf of the Mouse Genetics Programme The Sanger M.G.P., Wellcome Trust Sanger Institute, Genone Campus, Hinxton, Cambridgeshire, United Kingdom The Sanger Institute Mouse Genetics Programme aims to make a significant contribution to the understanding of the function of genes and their role in disease by generating, characterising and archiving 200 lines of knockout mice per year. In addition to a standardised battery of primary phenotypic testing relevant to key disease areas including diabetes, obesity, hearing and vision disorders, immune disorders, pain and motor function, one key aspect of understanding the function of genes is to identify their expression profile. The presence of the β-Galactosidase reporter gene, expressed under the control of the endogenous promoter of the target gene, makes wholemount expression profiling in the adult and at various developmental stages possible. Furthermore, ~31% of the knockout lines of mice assessed to date display a recessive lethality phenotype. To explore the underlying developmental defects, we are collecting and assessing embryos from heterozygous intercrosses at E14.5. Here we report the strategy that we have implemented to facilitate the efficient, high throughput, large scale expression profile and recessive lethality analysis of knockout lines, including a tiered approach that we have to document and interrogate the data. This systematic process involves husbandry management, embryo malformation assessment and record keeping. We present a summary of recessive lethality data available to date, and examples of novel findings for a subset of interesting mutant lines. 109 ABSTRACT 76 MULTIPLE FOREBRAIN CIS-REGULATORY ELEMENTS AT THE DLX1/2 LOCUS: REDUNDANT FUNCTION OR COMPLEX CONTROL MECHANISMS? Marc Ekker, Luc Poitras, Man Yu, Noel Ghanem, and Lisa Tran Center for Advanced Research in Environmental Genomics, Department of Biology, University of Ottawa, Canada The Dlx homeobox genes play an important role in the development of the vertebrate forebrain. DLX function is essential for the migration and differentiation of GABAergic interneuron precursors. Of the three Dlx bigene clusters, the Dlx1/Dlx2 and Dlx5/Dlx6 genes are expressed in the forebrain with patterns suggesting concerted control mechanisms. We have identified four cisacting regulatory elements (CRE) with forebrain enhancer activity. Of the four CREs, URE2, and I12b are found at the Dlx1/Dlx2 locus whereas I56i and I56ii are e located in the Dlx5/Dlx6 intergenic region. The CREs are highly conserved amongst distant vertebrates, structurally and functionally. However, the four enhancers diverge markedly in sequence although they target gene expression with partially overlapping patterns. Immunohistochemical analyses revealed that the mouse URE2 and I12b/I56i are active in distinct populations of progenitor cells that will be give rise to different subtypes of adult GABAergic interneurons. We produced mice with targeted deletion of the I12b CRE. Mice homozygous for the mutation show partial reductions in Dlx1 and Dlx2 expression in the E13.5 forebrain as well as small increases in the levels of Mash1 transcripts. They did not display overt developmental abnormalities and newborn pups were viable. These results suggest partial functional redundancy of this Dlx CRE at least in subset of GABAergic interneurons and their precursors. Supported by CIHR MOP14460 110 ABSTRACT 77 NOVEL MUTATION DISCOVERY ON ENU MOUSE MUTAGENESIS BY NEXT-GENERATION SEQUENCER Ryutaro Fukumura, Hayato Kotaki, Yuichi Ishitsuka, Takuya Murata, Shigeru Makino, Yuji Nakai, and Yoichi Gondo Mutagenesis and Genomics Team, RIKEN BioResource Center, Japan We developed a reverse genetics tool to provide allelic series of point mutations in any mouse genes. We have produced about 10,000 Generation-1 (G1) male mice derived from ENU-treated C57BL/6J males. We have preserved them as frozen sperm and extracted each genomic DNA for the mutation screening of target genes. Thus, the dual archives of frozen sperm and genomic DNA are considered to be the Mutant Mouse Library. We have called this system the RIKEN Gene-Driven Mutagenesis System (RGDMS). By using various methods to detect novel point mutations, we have estimated the ENU-induced mutation rate is ~1/1,000,000bp in a G1 genomic DNA sample. It implies that one G1 mouse has ~3,000 mutations. Therefore, a total of 30,000,000 mutations are reserved in the RIKEN Mutant Mouse Library. We have made the RIKEN Library open to research community and have been finding ~100 ENU-induced mutations per year in various target genes based upon the requests. To significantly enhance the mutation detection efficiency, we have started to resequence the G1 mouse genomic DNAs in the library by the next-generation sequencers. The shotgun sequencing of the whole mouse genome by the available next-generation sequencers has not been cost effective yet, thus we target protein-coding sequences to find the mutation. First, we selected 10 chromosomal regions and extracted about 4Mb exon-sequences. We analyzed them by illmumina GAIIx (two G1 genomes; 1675 and 381) and by AB SOLiD3plus (three G1 genomes; 1675, 381 and 15373). We estimated the mutation detection efficiency with known 57 SNPs on Chr 8 between DBA/2J and C57BL/6J as positive controls. The mutation detection efficiency was more than 90% by both systems. We have so far found 20 and 7 novel ENU-induced mutations from targeted exon-sequences and non-targeted regions on three G1 genomes, respectively. Eight and one mutations were missense and nonsense mutations, respectively. 111 ABSTRACT 78 THE SANGER MOUSE GENETICS PROGRAMME: HIGH THROUGHPUT CHARACTERISATION OF KNOCKOUT MICE FOR GENES INVOLVED IN GLUCOSE METABOLISM Anna-Karin Gerdin, L Roberson, Jacqueline K White on behalf of the Mouse Genetics Programme The Sanger Mouse Genetics Programme, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, United Kingdom The Sanger Mouse Genetics Programme aims to make a significant impact on our understanding of the function of genes and their role in disease, by generating, characterising and archiving 200 knockout lines per year. To date knockout mice for more than 200 genes have been characterised using a battery of primary phenotyping assays relevant to key disease areas including diabetes, obesity, hearing and vision disorders, immune disorders, pain and motor function. Expression profiling via the lacZ reporter gene is performed for each mutant line in adults and at E14.5. We will describe the phenotypic characterisation of seven knockout lines for genes involved in glucose metabolism. The genes we will focus on are Gys2, Gsk3a, Akt2, Pfkl, Tpi1, Ldha and Ide. We will explore how the data generated by the primary screen might be used to further the understanding of the interplay of genes and disease, and how it provides insight into the underlying biological pathways. Our data provides support for some previously published studies but also adds significant novel information to the scientific community. The tests used to investigate the metabolism of these mice include high fat diet exposure, body weight curve, indirect calorimetry, glucose tolerance test, body composition examination, core body temperature and clinical chemistry as well as expression profiling and tissue collection to our Biobank. 112 ABSTRACT 79 EUROPHENOME: LARGE DATASET VISUALISATION, STATISTICAL ANALYSIS AND DATA EXPLORATORY TOOL FOR MOUSE PHENOTYPING DATA Ahmad Hassan, Hugh Morgan, Andrew Blake, Simon Greenaway, The EUMODIC Consortium, John M. Hancock, and Ann-Marie Mallon Medical Research Council, Mammalian Genetics Unit, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, UK A major aspect of medical research is to utilise results to understand disease and improve human health. In recent years, a lot of effort has been made in medical science in linking the genotype data with phenotype data. The correlation of genotype with phenotype information enables a better understanding of human diseases and phenotype alterations that results from genetic changes. The EuroPhenome project (http://www.EuroPhenome.org) is a mouse phenotyping data repository that hosts the raw and annotated high-throughput phenotyping data arising from different projects such as European Mouse Disease Clinic (http://eumodic.org). EUMODIC is gathering data from the EMPReSSslim pipeline (http://www.empress.har.mrc.ac.uk) which is performed on inbred mouse strains and knock-out lines arising from the EUCOMM project. Europhenome provides the tools to visualise mouse phenotype and genotype data in an integrated manner. In addition, it enables the phenotyping data to be viewed in variety of ways i.e. Graphical representation of phenotyping data for inbred and mutant strain, graphical representation of statistically significant phenovariant data, mouse phenotype (MP) ontology tree and access to the data via web services. The raw phenotyping data captured in EuroPhenome is annotated by an annotation pipeline which automatically identifies statistically different mutants from the appropriate baseline and assigns ontology terms for that specific test. The mutant phenotypes can be viewed using two EuroPhenome tools: PhenoMap, a graphical representation of statistically relevant phenotypes, and mining for a mutant using ontology terms. To assist with data definition and cross-database comparisons, phenotype data is annotated using combinations of terms from biological ontologies. 113 ABSTRACT 80 THE INTRAGENE RESSOURCE CENTRE, A NATIONAL CENTRE FOR MOUSE GENETICS, DISTRIBUTION, ARCHIVING AND FUNCTIONAL IMAGING Cecile Fremond, Stephanie Lerondel, Christelle Martin, and Yann Herault Transgenesis, Archiving of Animal Models, CNRS, TAAM, UPS44, Institut de Transgenose, 3B rue de la Ferollerie, 45071 Orleans cedex2, France The INTRAGENE Ressource Centre provides the scientific community with all the technical facilities essential for creating, maintaining, distributing, conserving and analyzing transgenic murine models, either for the study of human diseases, or for fundamental modelling of genetic analysis in physiology, development, immunology, neurobiology, etc... Its main activities are associated to the Transgenesis, Archiving of Animal Models service unit from the CNRS based in Orleans and Villejuif. The Resource Center creates new transgenic models, mainly on the Villejuif site in the SErvice for Transgenic Animals (SEAT) while on its Orleans site, it hosts the Centre for Distribution, Transgenesis and Archiving (CDTA), a "mouse repository", which represents a large collection of mutant and transgenic mouse strains available on request from, or in agreement with, the EMMA network (www.emmanet.org). Furthermore the CDTA offers a large set of zootechnical services to the scientific community including rederivation, sanitary and genetic controls and gnotoxenisation. Finally, the CIPA department is devoted to small animal imaging with a large set of complementary imaging techniques. The operational phenotyping analysis and different services fulfill the recommendations set out by the EUMORPHIA program and also to ensure appropriate animal ethics and welfare. The Resource Center has been recognized since 2001, by way of an accreditation French Infrastructure in Biology, Health and Agronomy, and since then has been an integral part of the TGIR department of the Life Science Ministry: CELPHEDIA, and as a partner of the Infrafrontier European funded consortium. It is moving to the large scale infrastructure for Life Science and Biotechnology to answer further to the needs of the scientific community in mouse models for fundamental research and human diseases. 114 ABSTRACT 81 THE DOWN SYNDROME AS A COMPLEX TRAIT RESULTING FROM THE INTERACTION OF DOSAGE SENSITIVE GENES UNRAVELLED IN THE MOUSE MODEL V Brault1, A Duchon1, E Dalloneau1, P Lopes1, I Abizanda2, S Luiller3, JC Bizot3, C Borel4, M Raveau1, D Marechal1, J Lignon1, S Pothion2, F Trovero3, V Tybulewicz5, EM Fisher5, S Antonarakis4, M Dierssen2, and Yann Herault1,7,8 1 IGBMC, CNRS, INSERM, UdS, UMR7104, U964, Illkirch, France 2 Genes and Disease Program, Center for Genomic Regulation, Dr Aiguader 88, 08003 Barcelona, Spain and CIBER de Enfermedades Raras CIBERER, Barcelona, Spain 3 Key-Obs S.A., Allée du Titane, Orléans, France 4 Department of Genetic Medicine and Development, University of Geneva Medical School, 1 Rue Michel-Servet, 1211 Geneva, Switzerland 5 MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London UK 6 Department of Neurodegenerative Disease Institute of Neurology, London, UK 7 CNRS UPS44 TAAM, Orleans, France 8 ICS, ILLKIRCH, France More than 50 Years after the discovery of the Trisomy 21, the underlying genetic basic of the Down syndrome (DS), is still a challenge. Human genetic studies clearly demonstrated the complex genotypes - phenotypes relationship in DS with several loci participating to the DS traits. The current hypothesis is based on interactions between “dosage sensitive” genes or regions spread along the Hsa21 that are responsible for the complex feature of the pathology and could explain the penetrance and the variability of the traits. Currently a little of this loci has been identified. In order to go further and to decipher the gene interactions generating the DS phenotype, we created new partial trisomies and monosomies in the mouse for the different regions homologous to Hsa21 by using chromosomal engineering. Orthologuous genes on Hsa21 are located on mouse Chromosomes (Mmu) 16, 17 and 10. The Ts65Dn well-known mouse models for DS that is trisomic for part of the Mmu16 homologous region and exhibits many of the characteristics of DS has been a key model for the better understanding of the disease and the development of pharmacological intervention (Fernandez et al. Nat Neurosci 2007, 10, 411-413). Interestingly several new reports for larger trisomies, including the Hsa21 transchromosomic mice, Tc1 (O'Doherty et al., Science 2005, 309, 2033-2037) and the complete trisomic model developed by Yu and colleagues (Yu, T. et al.., Hum Mol Genet 2010, 19, 2780-2791) demonstrate that increasing the gene number in trisomic model could also result in the masking of specific phenotypes. Using our series of mouse trisomic and monosomic models, we deciphered the contribution of 115 several regions in the induction of DS phenotypes and identified several dosage sensitive genes controlling behavior, learning and memory, cardiovascular, craniofacial morphology and respiratory function. Now we are looking at the pathways that are affected, with the hope that this would lead to development of new therapeutic approaches. We will report here the phenotypic analysis of the new models and the results obtained on several traits including cognition, learning and memory and additional phenotypes. Our data support the current hypothesis that the DS phenotype is a complex trait with several dosage sensitive loci found on Hsa21, impacting the central nervous system and other organs, that interact together. A few of those loci and the pathways that are modified in DS will be discussed. 116 ABSTRACT 82 THE EUROPEAN MOUSE MUTANT ARCHIVE (EMMA) Michael Hagn6, G Tocchini-Valentini1, Y Hérault2, S Brown3, U Lendahl4, J Demengeot5, M Hrabé de Angelis (EMMA Director)6, E Birney7, R RamirezSolis8, JL Mandel9, and L Montoliu10 1 Consiglio Nazionale delle Ricerche, Istituto di Biologia Cellulare (CNR-IBC), Monterotondo Scalo, Italy 2 Centre National de la Recherche Scientifique, Transgénèse et Archivage d’Animaux Modèles (CNRS-TAAM), Orleans, France 3 Medical Research Council, Mammalian Genetics Unit (MRC-MGU), Harwell, UK 4 Karolinska Institutet, Department of Cell and Molecular Biology (KI-CMB), Stockholm, Sweden 5 Fundação Calouste Gulbenkian, Instituto Gulbenkian de Ciência, Oeiras, Portugal 6 Helmholtz Zentrum München, Institute of Experimental Genetics (HMGUIEG), Neuherberg, Germany 7 European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK 8 Genome Research Limited, Wellcome Trust Sanger Institute (WTSI), Hinxton, UK 9 GIE-Centre Européen de Recherche en Biologie et en Médecine, Institut Clinique de la Souris (GIE-CERBM-ICS), Illkirch, France 10 Consejo Superior de Investigaciones Scientificas, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain The European Mouse Mutant Archive (EMMA) offers the worldwide scientific community a free archiving service for its mutant mouse lines and access to a wide range of disease models and other research tools. A full description of these services can be viewed on the EMMA website at http://www.emmanet.org. The EMMA network is comprised of ten partners who operate as the primary mouse repository in Europe and is funded by the participating institutes and the European Commission FP7 Capacities Specific Program. EMMA’s primary objectives are to establish and manage a unified repository for maintaining mouse mutations and to make them available to the scientific community. In addition to these core services, the consortium can generate germ-free (axenic) mice for its customers and also hosts courses in cryopreservation. All applications for archiving and requests for mutant mouse strains are submitted through the EMMA website. Mouse strains submitted for archiving are evaluated by EMMA’s external scientific committee. Once approval has been granted depositors are asked to send mice of breeding age to one of the EMMA partners for embryo or spermatozoa cryopreservation. Strains 117 held under the EMMA umbrella can be provided as frozen materials or rederived and shipped as live mice depending on the customer’s needs. However, certain strains that are in high demand are maintained as breeding colonies to facilitate their rapid delivery. All animals supplied by EMMA are classified as SPF in accordance with the FELASA recommendations. EMMA is a founding member of FIMRe (International Federation of Mouse Resources) and actively cooperates with other leading repositories like TJL and the MMRRC in the US and BRC RIKEN from Japan. 118 ABSTRACT 83 BROAD BASED PHENOTYPING APPROACHES FOR THE CHARACTERISATION OF MUTANT MOUSE LINES IN EUMODIC AND THE HARWELL AGING SCREEN Heena Vanmalibhai Lad, KA Lee, A Blake, H Morgan, A Hassan, S Greenaway, AM Mallon, HJ Gates, SDM Brown, and The EUMODIC Consortium MRC Harwell, Harwell Science and Innovation Site, Oxfordshire, UK Central to successfully characterising the function of every gene in the mouse genome is the use of broad based phenotyping screens that will capture a range of phenotypes across mutant mouse lines. In the EUMODIC consortium, in order to capture robust and reliable phenotype information we have adopted and developed validated Standard Operating Procedures (SOPs), each which has the potential to reveal a variety of phenotypic perturbations underlying the genetic variant of interest. The SOPs are combined into the EMPReSSslim phenotyping platform. EMPReSSslim comprises two main strands of phenotyping that form ‘Pipeline 1’ and ‘Pipeline 2’, each designed to minimise the impact that preceding and successive protocols may have on the phenotypic outcome. Data combined from these pipelines reveals aspects of: dysmorphology; metabolism and physiology; bone structure and density; behaviour; sensory systems; and immunology. Complementary to the rigorous screening of mutant lines is the cataloguing of the phenotypes in a standardised format that will allow these data to be linked to ontological terms. In EUMODIC, standardisation of data capture has been achieved through devising a set of defined parameter lists for each SOP. Phenotypic ontologies are used to identify phenodeviants where a parameter for the mutant is statistically different to the control value. These are displayed in a heatmap (www.europhenome.org/ontologybrowser/heatmap.php), called ‘PhenoMap’, for each of the mutant lines. Adjusting the significance level and the effect size determines the colour displayed and thereby its related ontologies. For the data display and mark-up process to be operational, each of the components contributing at the successive levels is precisely mined to index the information for each mutant by pipeline or phenotype(s) of interest. Using a similar approach and many of the tests included within the EUMODIC pipelines, a large aging screen is underway at Harwell on mutant lines generated from the ENU programme. The main objective of the undertaking will be to identify the severity of the phenotype(s) and its associated genetic pathways with respect to late onset and/or age-related diseases. 119 ABSTRACT 84 FBXO11 REGULATES TGF BETA SIGNALING THROUGH TRP53 Hilda Tateossian, Susan Morse, and Steve DM Brown MRC Mammalian Genetics Unit, Harwell, Oxfordshire, UK The Jeff mouse mutant develops deafness due to chronic proliferative otitis media. The gene mutated in Jeff was identified as Fbxo11, a member of the Fbox family (Hardisty et al. 2006, Hum. Mol. Genet. 15: 3273). We previously reported that Fbxo11 affects TGF beta signaling by regulating the levels of phospho-SMAD2 in the epithelial cells of palatal shelves, eyelids and airways of the lungs (Tateossian et al. 2009, PathoGenetics, 2:5, 1-14). FBXO11 has recently been identified as a NEDD8 ligase for the tumor suppressor protein p53 in human colorectal carcinoma cells. It has been also reported that FBXO11 can inhibit TP53 activity without affecting its stability (Abida et al. 2007, J. Biochem Chem., 282, 1797-1804). Given the cross-talk between TGF beta signalling and p53, and the reported interaction between FBXO11 and TP53, we proceeded to examine the genetic and biochemical interactions of Fbxo11 and Trp53 in the developing mouse. To study the genetic interaction we crossed mice heterozygous for Fbxo11 to homozygous Trp53 knock-outs. The adult double heterozygote mice are smaller than Trp53 heterozygotes and their palates are not properly fused. The double homozygote new born mice also appear underdeveloped compared to their littermates. The phenotype of the double mutants indicates a genetic interaction of Fbxo11 and Trp53. To confirm the physical interaction between FBXO11 and TRP53 in developing tissues we used an immunoprecipitation strategy. Total protein from E15.5 lungs was immunoprecipitated with FBXO11 antibody and the Western blot analysis revealed that the antibody precipitated TRP53 as well as FBXO11. Furthermore TRP53 appears NEDD8-conjugated when complexed with FBXO11. Immunoprecipitation with TRP53 also confirmed that the two proteins are in a complex. Moreover, we have shown that only one isoform of FBXO11 interacts with TRP53. It is known that TRP53 is a partner of SMAD2 in the activation of multiple TGF beta target genes in mammalian cells and Xenopus embryos (Cordenonsi et al. 2003, Cell, 113, 301-314). We are currently investigating if the FBXO11-SMAD2-TRP53 complex in the developing mouse affects expression of genes under the joint control of TRP53 and TGF beta. Further studies into the interacting partners of FBXO11 and the signaling pathways it impacts will improve our understanding of the molecular mechanisms of middle ear inflammatory disease. 120 ABSTRACT 85 THE DEVELOPMENT OF THE INTEGRATED DATABASE OF MAMMALS IN RIKEN Hiroshi Masuya1, Yuko Makita2, Koro Nishikata2, Yuko Yoshida2, Terue Takatsuki1, Kasunori Waki1, Nobuhiko Tanaka1, Yoshiki Mochizuki2, Norio Kobayashi2, Riichiro Mizoguchi, Teiichi Furuichi4, Hideya Kawaji5, Daub Carsten5, Yoshihide Hayashizaki5, Shigeharu Wakana1, Atsushi Yoshiki1, Kaoru Fukami-Kobayashi1, and Tetsuro Toyoda 1 RIKEN BioResource Center, Tsukuba, Japan 2 RIKEN BASE, Yokohama, Japan 3 Department of Knowledge Systems, ISIR, Osaka University, Ibaraki, Japan 4 Laboratory of Molecular Neurogenesis, RIKEN BSI, Wako, Japan 5 RIKEN OSC, Yokohama, Japan To secure the sustainability, accessibility, utility and publicity of the data produced from multiple large-scale programs, RIKEN has developed an official integrated database for mammal data. The database integrates not only RIKEN’s original databases, such as FANTOM, ENU mutagenesis program and RIKEN Cerebellar Development Transcriptome Database (CDT-DB) and BRC Mouse Web Catalog, but also imported public databases such as Ensembl, MGI and OBO ontologies. Our integrated database has been constructed on RIKEN’s semantic-web-based cloud system termed SciNeS or Scientists’ Networking System, where the data and metadata are structured as a semantic web and are downloadable in various standardized formats. The outstanding feature of this database is the integrated implementation of the general knowledge (i.e. “allele is a variant of a genetic marker”) represented in the top-level ontology, Yet Another More Advanced Top-level Ontology (YAMATO) and public ontologies, and the instantiated knowledge in databases (i.e. “a datum for an ENU induced allele has a informational relationship to a gene recode in the MGI database”). This will provides 1) the dissemination of RIKEN’s data to be coordinated (or annotated) with public meta-information to increase its accessibility and 2) comprehensive, sustainable and cost-effective data management and maintenance across multiple databases. The Sustainability of database becomes a major issue on the bioinformatics field. Therefore, by this database, we would like to propose one of the generally useful and cost-effective ways to solve this problem. 121 ABSTRACT 86 COMBINING CLINICAL AND EXPRESSION QTLS IN THE CONTEXT OF INFECTION: NEW INSIGHTS INTO SUSCEPTIBILITY TO INFLUENZA Gregory A. Boivin1,2, Julien Pothlichet2, Emil Skamene2, Earl G. Brown 3, Robert Sladek1,4,5, and Silvia M. Vidal 1,2,6 1 Department of Human Genetics, McGill University, Canada 2 McGill Centre for the Study of Host Resistance, McGill University, Canada 3 Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada 4 Genome Quebec Innovation Center, Montreal, Canada 5 Department of Medicine, Experimental Medicine Division, McGill University, Canada 6 Department of Microbiology and Immunology, McGill University, Canada Seasonal influenza infections cause millions of severe cases and over 250,000 deaths annually worldwide. Influenza virus also causes recurrent pandemics in humans (e.g., 1918 [over 20 million deaths], 1957 [~2 million deaths], 1968 [~1 million deaths]). The large collection of influenza viruses circulating in animal species (including 16 “H” subtypes and 9 “N” subtypes as classified by surface cell receptors) makes the prediction of causal strains of future pandemics and epidemics difficult. Further, the high mutation rate inherent in influenza virus replication makes the timeline of future influenza outbreaks hard to predict. By studying immunological responses against influenza in the host species, it may be possible to discover common mechanisms of susceptibility in response to many influenza strains. This will ideally lead to quickly implementable therapies with wide clinical relevance, reducing economic and health burdens of future influenza epidemics and pandemics. We have created a mouse model of highly pathogenic influenza (hp-flu), which reproduces the hallmark inflammatory response and over-expression of cytokines associated with susceptibility to hpflu in humans by using a mouse adapted strain of influenza (A/HK/1/68-MA20 [H3N2]). By studying a panel of 34 closely related mouse strains created from susceptible A/J and resistant C57BL/6 mice (the AcB/BcA set of recombinant strains) we identified four loci strongly associated with altered influenza susceptibility, which range in size from 6 to 30 Mb. To more efficiently identify causative genes underlying these loci, we incorporated expression profiles obtained from uninfected lung tissue of 54 AcB/BcA mice. Functional characterization was performed for a subset of genes showing evidence of cisregulation. These co-localized clinical and expression QTLs have confirmed a previously reported influenza susceptibility locus and have identified a novel mechanism of susceptibility to influenza virus infection related to lipid biology. This study represents a potential road map for future studies attempting 122 to incorporate expression QTLs with traditional forward genetics towards efficient gene discovery in the context of infection. 123 ABSTRACT 87 A NATURE OF THE BLASTOMER DNA METHYLATION DETERMINATION Andrey A. Ivanov Vernadsky Institute of Geochemistry and Analytical chemistry RAS, Moscow, Russia All known biochemical processes involves the fractionation of isotopes [1]. In a separate series of the molecular biology experiments devoted to studies and simulation modeling on the metabolism environment isotopy, a phenomenon of a non-equal methylation of the isotopically different DNA copies has been found. It seems that the nature of this phenomenon somehow relates to a change in the DNA double helix conformational status [2]. This phenomenon is no doubt significant for making perfectly clear the very nature of the blastomers functional determination. This indicates to the zygote-specific, universal for zygote as well as for a whole biodiversity range, fundamental regularity. A regularity that consists in a non-random, truly regular, distribution of the isotopically different nucleotide forms among the blastomer DNA polynucleotide pairs. Noteworthy, the free nucleotides pool in any zygote originally contain approx. 10% of the isotopically heavy (Carbon-13 enriched) nucleotides. But in a course of closed, self-isolated, zygote cleavage process – there is no regeneration (no “refilling”) of the free nucleotides pool occurred. The isotopically light nucleotides are the most mobile ones, so they are about to get involved into a DNA synthesis more actively compared to the isotopically heavy ones. That’s why the zygote cleavage kinetic - from one stage to the next one - steadily changes the ratio of isotopically different nucleotide forms to a favor of the heavy ones. Owing to this, the DNA isotopy patterns should be different at different zygote cleavage stages. Particularly, this difference comes up to its maximal level by the zygote cleavage terminal stages. Then, due to depletion of the zygote free nucleotides pool, a C-13 isotopic self-organization takes place within the DNA double helix forming polynucleotide chains. This, in turn, leads to the fact that every system of DNA double helix should have an individual, unique, conformational status in all blastomers. This would also mean an individual methylation pattern. References 1. Galimov E.M. Biological Fractionation of Isotopes. N.Y. etc.: Academic Press Inc., 1985. 2. Ivanov A.A. Does the conformation of DNA depend on the differences in the isotope composition of it’s threads? Russian Journal of Physical Chemistry. B. 2007. vol. 2. 6: 649-652. 124 ABSTRACT 88 EVIDENCE OF HYPOTHALAMIC DEGENERATION IN THE ANX/ANX MOUSE IAK Nilsson1, S Thams2, C Lindfors1, A Bergstrand2, S Cullheim2, T Hökfelt2, and Jeanette E Johansen1 1 Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden 2 Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden The anx/anx mouse is an interesting model for studies concerning different conditions of disturbed feeding behavior, such as anorexia nervosa, cachexia and failure to thrive. Mice homozygous for the anorexia (anx) mutation are characterized by poor feeding and die around 3-5 weeks after birth. Histochemical studies of anx/anx mice have shown aberrant appearance of transmitter and neuropeptidergic systems originating in the arcuate nucleus (Arc) of the hypothalamus, important for the regulation of food intake and energy metabolism. For example, by P21 a lower density of hypothalamic appetite regulating neuropeptides, including Agouti gene-related protein (AGRP) and neuropeptide y (NPY) has been detected in these mice. The AGRP/NPY-system of the anx/anx mice develops normally until P12, when the normal increase in fiber density ceases, in some areas even distinctly decreases. This overlaps both in time and space with activation of microglia, indicating an inflammatory and/or degenerative process. To further evaluate this process we studied the expression of major histocompatibility complex (MHC) class Irelated molecules and markers for cellular reactivity in the hypothalamus of anx/anx mice. MHC class I transcript and -related proteins were found in Arc, both in neurons and glia. Using TUNEL labeling, a significantly higher number of apoptotic cells were found in anx/anx hypothalamus compared to +/+ mice, and active caspase 6 immunoreactivity was detected in degenerating NPY-fibers as well as signs of ‘microglia-associated cell death’. In addition, Y1 receptorlabeled processes and soma of pro-opiomelanocortin (POMC) neurons, were markedly decreased, but not until P21. These results support the hypothesis of degeneration of hypothalamic arcuate neuron populations in the anx/anx mice, where the AGRP/NPY system may be affected first and the changes in the POMC system seem to be secondary. 125 ABSTRACT 89 EVIDENCE OF HYPOTHALAMIC DEGENERATION IN THE ANORECTIC ANX/ANX MOUSE IAK Nilsson1, S Thams2, C Lindfors1, A Bergstrand2, S Cullheim2, T Hökfelt2, and Jeanette E Johansen1 1 Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden 2 Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden The anx/anx mouse is an interesting model for studies concerning different conditions of disturbed feeding behavior, such as anorexia nervosa, cachexia and failure to thrive. Mice homozygous for the anorexia (anx) mutation are characterized by poor feeding and die around 3-5 weeks after birth. Histochemical studies of anx/anx mice have shown aberrant appearance of transmitter and neuropeptidergic systems originating in the arcuate nucleus (Arc) of the hypothalamus, important for the regulation of food intake and energy metabolism. For example, by P21 a lower density of hypothalamic appetite regulating neuropeptides, including Agouti gene-related protein (AGRP) and neuropeptide y (NPY) has been detected in these mice. The AGRP/NPY-system of the anx/anx mice develops normally until P12, when the normal increase in fiber density ceases, in some areas even distinctly decreases. This overlaps both in time and space with activation of microglia, indicating an inflammatory and/or degenerative process. To further evaluate this process we studied the expression of major histocompatibility complex (MHC) class Irelated molecules and markers for cellular reactivity in the hypothalamus of anx/anx mice. MHC class I transcript and -related proteins were found in Arc, both in neurons and glia. Using TUNEL labeling, a significantly higher number of apoptotic cells were found in anx/anx hypothalamus compared to +/+ mice, and active caspase 6 immunoreactivity was detected in degenerating NPY-fibers as well as signs of ‘microglia-associated cell death’. In addition, Y1 receptorlabeled processes and soma of pro-opiomelanocortin (POMC) neurons, were markedly decreased, but not until P21. These results support the hypothesis of degeneration of hypothalamic arcuate neuron populations in the anx/anx mice, where the AGRP/NPY system may be affected first and the changes in the POMC system seem to be secondary. 126 ABSTRACT 90 IDENTIFICATION OF A NOVEL SLC25 MEMBER THAT CAUSES AUTOSOMAL RECESSIVE ATAXIA IN ENU-MUTAGENIZED MICE A Karakostas, F Ioakeimidis, V Rinotas, G Kollias, and Eleni Douni Institute of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece We have recently generated by random mutagenesis using the mutagen N-ethylN-nitrosourea (ENU), a novel mouse model of severe autosomal recessive neurological disease characterized by ataxia, unsteady locomotion, episodic crises, and growth retardation. The symptoms start at 3 weeks of age with severe disease progression that leads to lethality of the majority of the mice within 7 weeks after birth. Using genome-wide linkage analysis we localized the mutation in a 2.9Mb genomic interval containing 26 genes. Upon sequencing of the candidate genes we identified a nonsense point mutation (C to T) in the coding region of a novel gene member of the Solute Carrier Family 25 (SLC25) that introduces a premature stop codon and results in a loss-of-function protein. All SLC25 members are nuclear-coded proteins that are imported into the inner mitochondrial membrane where they shuttle a variety of metabolites across it. Until now, almost 50 SLC25 members have been identified whereas most of them remain uncharacterized. Mutations in SLC25 genes impair mitochondria functions and result in at least 10 various human diseases, by affecting either the synthesis of ATP through oxidative phosphorylation, or the selective transport of solutes in and out of the mitochondrial matrix. This novel SLC25 member is highly conserved among various species, but its function remains completely unknown. Our ongoing studies are focused on a) the identification of the primary site of lesion in order to align the brain pathology of the mouse model with that of similar human neurodegenerative diseases, b) the identification of mitochondrial dysfunctions, and c) the analysis of the expression profile and the function of this novel SLC25 protein which constitutes a novel pathogenic target in neurological diseases such as ataxia. 127 ABSTRACT 91 CONSOMIC ANALYSIS OF GENETIC FACTORS RELATED TO TEMPORAL DIFFERENCE OF HOME-CAGE ACTIVITY BETWEEN B6 AND MSM Ayako Ishii1,2, Akinori Nishi1,2, Toshihiko Shiroishi1,3, Aki Takahashi1,2, and Tsuyoshi Koide1,2 1 Mouse Genomics Resource Laboratory, NIG, 2 SOKENDAI, 3 Mammalian Genetics Laboratory, NIG, Japan We have been studying genetic basis of behavioral diversity using a variety of mouse strains including wild-derived strains. Recent studies have shown that genetic factors have no small effect on home-cage activity. The wild-derived mouse strain MSM/Ms (MSM) exhibits higher activity in the home-cage than C57BL/6 (B6), a commonly used laboratory strain. In addition, there is a clear strain difference in the temporal pattern of within-day activity. We have analyzed consomic strains derived from B6 and MSM to elucidate genetic mechanism responsible for strain differences in the temporal regulation of home-cage activity. The results showed that many genetic factors are related to home-cage activity with a complex manner and different genetic factors related to different temporal component of the activity. In order to identify a causative gene related to different level of home-cage activity, we focused on one of the loci on telomeric region of Chromosome 6. One of the sub-consomic strains which has only a short segment (~5Mbp) of candidate locus from MSM exhibited different activity suggesting existence of the causative gene in this region. We are currently conducting comparative analyses of DNA sequences and gene-expression levels of candidate genes using the sub-consomic strain and B6 to find the causative gene in this region. 128 ABSTRACT 92 GENETIC DETERMINANTS FOR INTRAMUSCULAR FAT CONTENT AND WATER HOLDING CAPACITY IN MICE SELECTED FOR HIGH MUSCLE MASS Stefan Kärst and Gudrun A. Brockman Humboldt Universität zu Berlin, Berlin, Germany Intramuscular fat content and water holding capacity are important traits in livestock as they influence meat quality, nutritive value of the muscle and animal health. The intramuscular fat content may influence the animals' metabolic stability via blood glucose levels and the taste of meat products, whereas low water holding capacity leads to impaired meat processing properties and does not meet consumers' requirements. This study was performed to identify genomic regions affecting intramuscular fat content and water holding capacity. As a model for livestock, we used the two inbred mouse strains of the Berlin Muscle Mouse population. The inbred Berlin Muscle Mouse strains 806 (BMMI806) and 816 (BMMI816) have been long term selected for high muscle mass. Although both strains have increased lean mass, the strains differ in intramuscular fat content. The intramuscular fat content of the Musculus longissimus of BMMI806 mice is approximately 1.4 fold higher than in BMMI816 mice. Additionally, lower water holding capacity was observed in BMMI816 mice compared to BFMI806. In a reciprocal intercross between the two strains, a F3 population with 345 mice was generated. All animals were phenotyped and genotyped at 164 SNP markers. Linkage analyses revealed three quantitative trait loci for intramuscular fat content on Chromosomes 7, 12 and 15 and two loci for water holding capacity on Chromosomes 2 and 17. These loci explained 10% and 6% of the observed phenotypic variance in intramuscular fat content and water holding capacity in the F3 population, respectively. In part, the quantitative trait loci mapped for these traits in mice coincide with homologue regions in pigs, in which quantitative trait loci for the same traits were found. Therefore, the mouse strains and this F3 population are interesting genetic resources to identify responsible genes, which could also contribute to quantitative trait loci effects in other species. 129 ABSTRACT 93 DISCOVERING NOVEL REGULATORY PATHWAYS OF IMFLAMMATION BY ENU MUTAGENESIS Philip L Kong1, Emma Timms1, Andrew Coulson1, Kristina Blagoeva-Hubbard1, Paul Potter1,2, and Marc Feldmann1 1 Kennedy Institute of Rheumatology, Imperial College London, United Kingdom 2 MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, United Kingdom Inflammation is a complex process whose outcome underpins many physiological functions. Likewise, its dysregulation is the key driver in many pathological conditions, including autoimmune diseases such as rheumatoid arthritis. Over the years a multitude of regulatory mechanisms of inflammation have been defined, including those driven by interleukin 10 (IL10) and glucocorticoids. However despite intensive research significant gaps still exist in our knowledge of their mechanisms of action, hampering our efforts to understand the diseases in which these molecules play a part, and to design therapies. In this screen of ENU-mutagenised mice, we aim to uncover novel genes and pathways that are involved in the action of these anti-inflammatory molecules. The phenotype we screen is the inhibition of cytokine production by IL10 and dexamethasone in macrophages, a key cell type in effecting inflammation in the joint. Bone marrow-derived macrophages are stimulated with LPS to drive the production of pro-inflammatory cytokines, in the presence or absence of a predetermined amount of IL10 or dexamethasone. Cytokine production is determined by the multiplexing Luminex platform. Animals that exhibit reduced responsiveness to IL10 or dexamethasone are selected for follow-up studies using cells archived in liquid nitrogen. Some of the data from the primary screen and the follow-ups are presented here. Pedigrees that harbor significant and interesting phenotypes are then chosen for mapping studies in order to identify the underlying mutation responsible for the phenotype. 130 ABSTRACT 94 EPIGENETIC CHANGES IN FRIEDREICH ATAXIA PATHOGENESIS AND THERAPY S Al-Mahdawi, C Sandi, R Mouro-Pinto, V Ezzatizadeh, and Mark A Pook Brunel University, Uxbridge, UK Friedreich ataxia (FRDA) is a lethal inherited neurodegenerative disorder caused by a homozygous GAA repeat expansion mutation within intron 1 of the FXN gene, which leads to reduced expression of the essential mitochondrial protein, frataxin. Evidence indicates that the GAA mutation induces epigenetic changes and heterochromatin formation, thereby impeding FXN gene transcription elongation. In particular, studies using FRDA patient blood and lymphoblastoid cell lines have detected increased DNA methylation of specific CpG sites upstream of the GAA repeat, together with histone post-translational modifications in regions flanking the GAA repeat. Our own studies have revealed similar epigenetic changes in FRDA patient autopsy brain and heart tissues, the primary systems that are affected in this disorder. Furthermore, we have established GAA repeat expansion-containing human FXN YAC transgenic mice that exhibit comparable epigenetic changes in brain and heart tissues to those detected in FRDA patient tissues. We have thus developed a mouse model that is a valuable resource for pre-clinical testing of epigenetic-acting compounds that may target the FXN gene to increase frataxin expression. We have established fibroblast and neural stem cell lines from our FRDA transgenic mice, and several DNA methylation inhibitors, histone deacetylase inhibitors and GAA-interacting compounds have been tested on these cultured cells, resulting in the identification of good candidates to take forward to further preclinical and clinical studies. 131 ABSTRACT 95 A MOUSE MUTANT EXHIBITING A UNIQUE PATTERN OF COCHLEAR INNER HAIR CELL DEGENERATION Morag A. Lewis, Tracy J. Bussoli, Amy Taylor, Michael A. Cheong, Cordelia F. Langford, and Karen P. Steel Wellcome Trust Sanger Institute, Cambridge, UK Deafness is the most common sensory deficit in the human population, but the genetic basis for nonsyndromic hearing loss is still largely unknown. It is thought that hundreds of genes may be required for development and function of the ear, many of which remain undiscovered. Deaf mouse mutants are a powerful tool for discovering genes which affect hearing and characterising the pathways which, when perturbed, result in dysfunction of the ear. Bronx waltzer is a spontaneous autosomal recessive mutant which has been previously described. Homozygotes demonstrate rapid degeneration of vestibular hair cells and cochlear inner hair cells at a late embryonic stage, resulting in deafness and vestibular dysfunction, but outer hair cells remain intact. The mutation has been mapped to a 2.7Mb region on Chromosome 5, but exon resequencing has failed to find a potential causative mutation. We have investigated the mutation using numerous methods including Illumina resequencing of the entire critical region, analyses of which detected multiple mutations ranging from large deletions to single nucleotide polymorphisms. We are currently studying several candidate genes which are expressed in the organ of Corti. The pattern of degeneration of bronx waltzer hair cells is unique, so characterisation of the gene responsible for the phenotype will enhance understanding of the development of the ear and potentially lead to more effective therapies for hearing loss. 132 ABSTRACT 96 A COMPREHENSIVE WHOLE-GENOME MAP OF ENDOGENOUS RETROVIRAL ELEMENTS AND THEIR FUNCTIONAL EFFECTS ACROSS 17 LABORATORY MOUSE STRAINS Thomas M Keane1, K Wong1, J Flint2, W Frankel3, and David J Adams1 1 Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom 2 Wellcome Trust Centre for Human Genetics, Oxford University, Oxford, Oxford, United Kingdom 3 The Jackson Laboratory, The Jackson Laboratory, Bar Harbor, ME, USA It has been estimated that endogenous retroviral elements (ERVs) are a significant source (~10%) of spontaneous germline mutations among laboratory mouse strains. Two high-copy families of ERVs in particular, IAP and ETns, have been found to be responsible for the vast majority of these mutations. As part of the Mouse Genomes Project (http://www.sanger.ac.uk/mousegenomes), we have recently completed deep paired-end illumina sequencing of 17 key mouse laboratory strains to between 20-35x depth. In this study, we report the first use of new sequencing technologies data to catalogue the full repertoire of the major classes of ERVs (IAP, ETn, MLV, LINE, and SINE) across these strains. Our strategy utilises paired-end information by finding clusters of matepairs where either only one end maps well to the reference genome and the other to a set of ERV reference sequences or incorrectly mapped pairs where one end maps to a canonical copy in the reference. From the initial set of 1-2kb call regions, we then further refine the calls by using the read depth information to identify the exact breakpoints. We assess the accuracy of our calls by comparing the set of calls we make in the C57B6NJ strain sequenced by us compared to the insertions annotated in the C57B6J reference genome. We find extremely high concordance between these two sets with just a few differing insertions. We have also compared our findings to a large set of manually PCR validated insertions falling into introns across five strains and find that our illumina predictions show extremely high confirmation rates. In our analysis, we have focused on young ERVs that occur in just a single strain and compared this set of ERVs found across the strains to existing expression data finding several examples of gene expression being potentially affected by intronic or proximal ERVs and use this information to infer potential phenotypic traits amongst the strains. 133 ABSTRACT 97 PHENOTYPING OF KNOCKOUT MICE USING BACTERIAL PATHOGENS AS PART OF THE WELLCOME TRUST SANGER INSTITUTE’S MOUSE GENETICS PROGRAMME Leanne Kane, Lynda Mottram, Simon Clare, Jacqui White, Ramiro Ramirez – Solis, and Gordon Dougan All the Mouse genome project team at the Sanger Institute Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridgeshire, UK The Wellcome Trust Sanger Institute’s Mouse Genetics Programme is running an extensive sequence/phenotype driven mutagenesis programme as part of international efforts aimed at mutating over 95 percent of known mouse genes in embryonic stem cells. The knockout mice, resources and data generated in this programme are then freely available to the scientific community. All mutant mouse lines generated in this high throughput programme are examined using a common battery of phenotyping tools. At the Sanger Institute a component of this phenotyping programme involves challenging the knockout mice with selected pathogens. We are currently using two standard models of infection Salmonella Typhimurium an intracellular pathogen which induces a systemic disease in mice similar to that of human typhoid fever and Citrobacter rodentium a natural mouse pathogen which forms attaching and effacing lesions on the surface of the lumen to colonise the host gastrointestinal tract in a similar way to enterohaemorrhagic E.coli (EPEC). We have screened over 250 targeted mouse knockout lines and have found 15 phenotypes ranging in severity from subtle antibody differences to mice succumbing rapidly to the infection. 134 ABSTRACT 98 IMPRINTED METHYLATION VS. IMPRINTED EXPRESSION -ARE THEY NECESSARILY ASSOCIATED? John D. Calaway1, Hyuna Yang2, Megan E. Hanson3, Ezequiel C. Cambranis3, Maria A. Perez-Lamigueiro3, David L. Aylor1, Leonard McMillan1, Gary A. Churchill2, Fernando Pardo-Manuel de Villena1, and Elena de la CasaEsperon3,4 1 University of North Carolina, Chapel Hill, NC, USA 2 The Jackson Laboratory, Bar Harbor, ME, USA 3 University of Texas Arlington, Arlington, TX, USA 4 Regional Center for Biomedical Research. University of Castilla-La Mancha/Albacete Science and Technology Park. Spain Imprinted genes are preferentially expressed from either the maternal or the paternal copy, and their pattern of expression has been correlated with the methylation status of nearby CpG sites. At these sites, only one of the two alleles is methylated and alterations of these differentially methylated regions (DMRs) can disrupt imprinted expression. While currently known DMRs have been identified due to their proximity to known imprinted genes, we have adopted the opposite strategy: to search for novel DMRs in order to uncover nearby imprinted genes. Our genome-wide survey of the mouse methylome has allowed us to identify several DMRs. Most of them are associated to known imprinted genes, but not all: such is the case of a DMR located in the 3' of the Actn1 gene, which has never been tested for imprinting. Consequently, we have analyzed the expression of this gene in heterozygous mice resulting from reciprocal crosses, both male and female, in diverse tissues. In spite of finding widespread differential methylation, none of the tissues studied show any indication of imprinted expression. Our results suggest that imprinted expression of Actn1 might be restricted to a specific cell type and/or developmental stage, while imprinted methylation of Actn1 is maintained in most cells. Alternatively, our observations may support our hypothesis that parental origin-dependent differences in methylation have been selected in sexually reproducing organisms for reasons other than gene expression regulation. 135 ABSTRACT 99 DUAL ACTIVITY ON A SINGLE LOCUS: EXISTENCE OF A NOVEL RNA CONTROL MECHANISM FOR MICRORNA-650 AND IMMUNOGLOBULIN LAMBDA LIGHT CHAIN VARIABLE GENES IN PRIMATES Sabyasachi Das, Jianxu Li, Masayuki Hirano, Christopher L. Haga, Murali Gururajan, Götz R.A. Ehrhardt, and Max D. Cooper Department of Pathology and Laboratory Medicine, Emory Vaccine Center, School of Medicine, Emory University, Atlanta, GA, USA MicroRNAs (miRNAs) are small noncoding RNA molecules that regulate gene expression post-transcriptionally in a wide spectrum of biological systems. A survey of the genomic context of human microRNA (miRNA) genes reveals that the miR-650 gene overlaps with the protein-coding region of the immunoglobulin lambda light chain variable (IGVL) gene in the same transcriptional orientation. This genomic organization offers a unique opportunity to explore the evolutionary and functional consequences of overlapping miRNA and protein coding genes. The miR-650-bearing IGVL genes are present in multiple copies and belong to a specific phylogenetic clan (clan II). Sequence comparison and structural prediction indicate that the accumulation of nucleotide changes in the leader exons of clan II IGVL genes has led to the generation of functionally stable miRNA hairpins. In contrast to the expectation that expression of miRNAs overlaps with other genes in the same transcriptional orientation, our analysis of ten different cell lines demonstrates that, using the same promoter, the miR-650 gene is transcribed independently of the IGVL gene. This suggests the existence of a novel RNA control mechanism by which the same locus is expressed as either IGVL (VJrearrangement dependent) or miRNA (VJ-rearrangement independent) in different cells. Our study on ten mammalian genome sequences suggests that the control mechanism for mutually exclusive expression of miR-650 and IGVL genes evolved in the primate lineage. 136 ABSTRACT 100 IDENTIFICATION OF POSITIONAL CANDIDATES FOR CHR15 OBESITY QUANTITATIVE TRAIT LOCI USING MICROARRAY AND RT-PCR ANALYSES OF F2 CONGENIC CROSSES IN MICE K Cirnski1, M Stanonik1, R Keber1, P Juvan2, R Košir2, D Rozman2, and Simon Horvat1,3 1 Biotechnical Faculty, Dept. Anim. Sci. University of Ljubljana, Domžale, Slovenia 2 Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Slovenia 3 National Institute of Chemistry, Ljubljana, Slovenia Obesity is a risk factor for a number of chronic diseases including diabetes, cardiovascular diseases and cancer and is controlled by a complex interplay between genetic and environmental factors. Additional genes from the common polygenic form of obesity need to be identified to elucidate the mechanisms of susceptibility or resistance to obesity development. We previously identified a Chr 15 quantitative trait locus (QTL) Fob3b in lines of mice selected on high (Fat line) and low (Lean line) body fat content that represent a unique model of polygenic obesity. Follow up studies revealed that Fob3b consists of at least two separate linked QTLs Fob3b1 and Fob3b2. The objective of the study presented here was to identify differentially expressed candidates for Fob3b1 and Fob3b2 using microarray and quantitative RT-PCR analyses. Brain, gonadal fat and liver RNA of homozygotes for alternative alleles from an F2 cross between the congenic line containing Fob3b1 and Fob3b2 QTL and Fat line was analyzed. Affymetrix GeneChip 1.0 ST arrays were used and analyzed using modified ttest (R/Bioconductor/Limma) of individual genes, gene enrichment analysis (R/Bioconductor/PGSEA) of gene sets from GO terms and KEGG pathways, and Venn analysis of differentially expressed genes/gene sets. A focused analysis on Chromosome 15 revealed 1 candidate in brain, 2 in gonadal fat and 9 in liver. Most of these genes were found to be involved in obesity-related processes such as biosynthesis of fatty acids, steroid hormone metabolism, glycerophospholipid metabolism, PPAR signaling pathway and Wnt signaling pathway. A comprehensive quantitative RT-PCR analysis to validate the microarray candidates in a larger set of F2 animals is under way and will be presented. This study should enable us to narrow down the list of candidates for the obesity Fob3b1 and Fob3b2 QTL loci. Identifying new obesity loci should help to uncover important inherited risk factors and provide novel diagnostics and therapeutic approaches. 137 ABSTRACT 101 CREZOO - THE EUROPEAN VIRTUAL REPOSITORY FOR CRE DRIVER STRAINS Christina Chandras1, Michael Zouberakis1, Damian Smedley2, Nadia Rosenthal3, and Vassilis Aidinis1 1 B.S.R.C. Al. Fleming, Athens Greece 2 European Bioinformatics Institute, EMBL, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK 3 EMBL-Monterotondo Outstation, Italy CreZOO (www.fleming.gr/crezoo) is the European database of genetically modified mice expressing cre recombinase and other conditional systems under the control of cell-specific promoters/enhancers. Its aim is to promote European collaboration towards unified mouse databases and deposition of existing cre mouse strains into EMMA and to share and publicize information on mouse characteristics and availability. Transgenic mice carry detailed information on promoter(s), alleles and genetic background (DNA origin, targeted, host and backcrossing background), in addition to recombination efficiency, specificity, integration site and copy number where available. Each promoter/gene index includes IDs and direct links to MGI, Ensembl and NCBI’s Entrez Gene database. The site of transgene expression is described using the Mouse adult gross anatomy (MA) as developed by the Jackson Laboratories, while embryonic development is standardised based on EMAP’s Mouse gross anatomy and development ontology. Availability (e.g. live mice, cryopreserved embryos, sperm, ES cells) is clearly indicated, along with handling and genotyping details (in the form of documents or hyperlinks) and all relevant contact information (including EMMA and Jax/IMSR hyperlinks where available). CreZOO is the front-end of a normalized PostgreSQL database, built around EJBs technology and is deployed on a Glassfish application server. CreZOO also enables programmatic access via web services. Its design is based on the users’ convenience and is thus an easy to use database, offering a userfriendly query interface and providing instant access to the list of transgenic mice, transgenes and alleles. Finally, handling and genotyping instructions as well as the primary and additional references are also available in the final tab for each mouse. CreZOO is still under construction and is created in the context of the CREATE consortium, a core of major European and international mouse database holders and research groups involved in conditional mutagenesis, primarily to develop a strategy for integration and dissemination of cre driver strains for modeling aspects of complex human diseases in the mouse. CREATE aims integrate new and existing information on cre driver strains, through a virtual international repository of genetically modified mice expressing cre recombinase, the cre8 portal (http://www.creline.org/search_cre_mice). 138 Participating databases (CreZOO/EU, MGI's Recombinase/USA and Cre-XMice/Canada) will exchange/contribute seven agreed datafields to the cre8 portal thus fulfilling the urgent need on the production and expansion of an international creZOO to complement the conditional mouse alleles currently being generated worldwide. 139 ABSTRACT 102 HOST IMMUNE RESPONSES AND GENETIC FACTORS MODULATING RESISTANCE TO SALMONELLA ENTERICA SEROVAR TYPHIMURIUM IN THE INBRED MOUSE STRAIN SPRET/EI: A ROLE FOR NEUTROPHILS Lien Dejager, Iris Pinheiro, Pieter Bogaert, Liesbeth Huys, and Claude Libert Molecular Mouse Genetics Unit, VIB and Ghent University, Ghent, Belgium Infection with Salmonella enterica serovar typhimurium is a complex disease in which the host-bacteria interactions are strongly influenced by genetic factors of the host. We demonstrate that SPRET/Ei, an inbred strain derived from Mus spretus, is resistant to S. typhimurium infections. The kinetics of bacterial proliferation as well as histological examinations of tissue sections suggest that SPRET/Ei mice can control bacterial multiplication and spreading despite significant attenuation of the cytokine response. The resistance of SPRET/Ei mice to S. typhimurium infection is associated with increased leukocyte counts in circulation and enhanced neutrophil influx into the peritoneum during the course of infection. A critical role of neutrophils was confirmed by neutrophil depletion: neutropenic SPRET/Ei mice were sensitive to infection with S. typhimurium and showed much higher bacterial loads. To identify genes that modulate the natural resistance of SPRET/Ei mice to S. typhimurium infection, we performed a genome-wide study using an interspecific backcross between C3H/HeN and SPRET/Ei mice. This analysis demonstrates that at least two loci, located on chromosomes 6 and 11, affect survival following lethal infection with S. typhimurium. These two loci contain several interesting candidate genes, which may have important implications for the search for genetic factors controlling Salmonella infections in humans and for our understanding of the complex host-pathogen interactions in general. 140 ABSTRACT 103 LPS-RESISTANCE OF THE MOUSE STRAIN SPRET/EI: AN IMPORTANT ROLE OF THE X-CHROMOSOME Iris Pinheiro, Lien Dejager, Tina Mahieu, Marnik Vuylsteke, and Claude Libert Molecular Mouse Genetics Unit, VIB and Ghent University, Ghent, Belgium Mus spretus diverged from the Mus musculus complex of house mouse subspecies about 1.5 million years ago. Several inbred strains, as SPRET/Ei (S), have been derived from this species. Recently, we have described that this strain shows a remarkable resistance in models of acute inflammation, e.g. triggered by cytokines or by bacterial components, as lipopolysaccharides (LPS). Objectives: Study the genetic basis for X-located LPS-resistance observed in SPRET/Ei mice. Methods: Linkage analysis, microarrays analysis, in vitro models (bone marrow and ES-cells derived macrophages), in vivo models, tetraploid aggregation. Results: The nature of S LPS-resistance is complex, and multiple genetic factors seem to be involved. It is a dominant trait, and two major protective loci, namely on chromosomes 2 and X and two minor protective loci on chromosomes 10 and 13 were identified by linkage analysis. Moreover, F1 females resulting from the cross between C57BL/6 (B) females and S males, and therefore carrying the S X-chromosome, display higher resistance than F1 males. Both in vivo and in vitro results indicate that induction of IL6 after LPS challenge is lower in (BxS)F1 females. Furthermore, (SxB)F1 males resulting from the reverse cross, i.e. between S females and B males, do also show an increased resistance to LPS when compared to (BxS)F1 males, a fact confirming the contribution of the S X-chromosome to this trait. A genomewide expression profile on both parental strains and F1 offsprings of both sexes was performed by using Affymetrix microarrays. A few X-located candidate genes were found to be differentially regulated by LPS in F1 females and males bone marrow derived macrophages. The contribution of these genes to LPSresistance of both F1 females and S mice will be further investigated by the use of in vitro models as well as by the generation of (BxS)F1 male mutants by tetraploid aggregation. Conclusion: The use of SPRET/Ei as a mouse model can be of valuable use as a tool to disclose the molecular mechanism which is responsible for the observed LPS-resistance. Ultimately we hope to contribute to the development of a new therapeutic approach in the treatment of endotoxemia and sepsis. 141 ABSTRACT 104 COMPLETE KNOCKOUT OF THE ADRENOCORTICAL DYSPLASIA GENE ENCODING THE SHELTERIN PROTEIN TPP1 IS ASSOCIATED WITH TELOMERE DYSFUNCTION AND EARLY EMBRYONIC LETHALITY Gail A Osawa1, C Harris1, T Kibe2, T de Lange2, S Kalantry1, and CE Keegan1 1 University of Michigan, Ann Arbor, MI. USA 2 Rockefeller University, New York, NY, USA Adrenocortical dysplasia (Acdacd) is a spontaneous autosomal recessive mouse mutation that originated on the DW/J strain. Homozygous acd mutant mice exhibit a strain-dependent pleiotropic phenotype; on the DW/J strain, the phenotype is lethal shortly following birth and includes caudal truncation, vertebral segmentation, and limb patterning defects. In contrast, acd mutant mice on a mixed DW/J X CAST/Ei background are growth retarded, hyperpigmented, and infertile but can survive into adulthood. We previously characterized the acd mutation as a splicing defect in the Acd gene, which encodes TPP1, a component of the shelterin complex that functions to maintain telomere integrity. Because of the critical role of Tpp1 in the shelterin complex, we suspected that acd is a hypomorphic mutation. In addition, previous studies of acd mutant MEF cells showed worsening of the cellular phenotype following shRNA knockdown of Acd expression. To examine the phenotype of a complete knockout, we created a null allele by deleting exons 3 through 8 of the Acd gene. Acdnull MEF cells exhibit evidence of significant telomere dysfunction similar to double Pot1a/Pot1b knockout cells, including telomere dysfunction-induced foci, chromosomal fusions, and endoreduplication. In addition, Acdnull cells show loss of POT1 proteins from the telomeres. These data indicate that the main function of TPP1 is to allow POT1A and POT1B to protect chromosome ends. To examine the mutant phenotype in early stage embryos, we performed timed pregnancies with heterozygous Acdnull mice, and we found no homozygous mutant embryos as early as E8.5. We are currently analyzing blastocyst stage embryos to determine whether homozygous mutants are present at these early timepoints. These studies demonstrate that the acd mutation is hypomorphic and that Acd/TPP1 function is critical for telomere integrity and is required at very early stages of embryonic development. 142 ABSTRACT 105 THE FACEBASE CRE DRIVER PROJECT: CREATING NEW MOUSE TOOL STRAINS FOR CLEFTING RESEARCH Stephen A. Murray, Jocelyn Sharp, John Flaherty, Thomas Gridley, and Leah Rae Donahue The Jackson Laboratory, Bar Harbor, ME, USA Orofacial clefting is one of the most common birth defects in humans, affecting approximately 1 in 700 live births. This frequency highlights the complexity of craniofacial morphogenesis, which requires precise regulation of gene expression changes, alterations in cell physiology and morphogenic movements. The mouse has played an instrumental role in advancing our understanding of the mechanisms that govern mid-face and palate development. Future progress, however, will require an increasingly sophisticated set of genetic models and tools. As part of the NIDCR-funded FaceBase Consortium, we will generate 15 Cre driver strains to facilitate orofacial clefting research in the mouse. These driver strains will comprise both inducible and non-inducible approaches, and include a fluorescent marker for simultaneous visualization of Cre expression patterns. The individual driver promoters have been chosen to encompass both early midface and primary/secondary palate development and fusion. These tools are designed to complement existing lines, allowing users to define gene function in new cell types and regions. A combination of approaches will be employed to produce the lines, including BAC transgenic, knock-in and targeting to the ROSA26 locus. In addition to detailed characterization of Cre functionality in the midface and palate, we will use our existing comprehensive characterization pipeline to ensure specificity and carefully define any nontarget activity. All of this data will be made available as part of the FaceBase effort, and lines will be distributed through the JAX FaceBase Repository, which includes both Cre lines and other mouse models and tools useful in clefting research. Development and rapid distribution of these lines will greatly enhance the genetic “toolbox” available to the scientific community for understanding the mechanisms that govern orofacial clefting. 143 ABSTRACT 106 EPIGENOME SEQUENCING COMES OF AGE IN DEVELOPMENT, DIFFERENTIATION AND IMMUNE REGULATION MECHANISMS OF MAMMALIAN RESEARCH Ning Li and Jun Wang Beijing Genomics Institute, Shenzhen, China Epigenetics is a new field, defined as heritable changes in gene expression that are not accompanied by changes in DNA sequence, which could open up many possibilities for future development process control and disease treatment. DNA methylation plays a key role in epigenome. There are numerous approaches to decipher a whole genome DNA methylation profile (“methylome”), each varying in cost, throughput and resolution. Bisulfite-sequencing is the golden standard of these methods. Compared with it, methylated DNA immunoprecipitation (MeDIP-Seq) and methyl-binding protein (MBD-Seq) are complementary strategies. MeDIP-Seq is more sensitive to highly methylated, high-CpG densities and MBD-Seq is more sensitive to highly methylated, moderate-CpG densities. Bisulfite-Seq has high reliability and accuracy to determine each cytosine methylation state. In our study, we have generated a single-base-resolution human DNA methylation profile in peripheral blood mononuclear cells (PBMC), using Bisulfite-Seq, which was determined by 20X coverage genome-wide resequencing after bisulfite treatment. In this research, essentially complete (92.62%) methylome of PBMC was reported, and only <0.2% of non-CpG sites were methylated, demonstrating that non-CpG cytosine methylation is negligible in human PBMC. Integration of this methylome data with the previously determined genome sequence of the same Asian individual analysed here, enabled a first assessment of allele-specific methylation (ASM) differences between the two haploid methylomes of any individual. We estimate that about 30% of the haploid differentially methylated regions (hDMR) may be attributed to imprinting and about 70% to other mechanisms such as sequencedependent ASM, suggesting that imprinting may be more common than previously thought. The study performed herein not only provides a comprehensive method for detailed understanding of epigenetic regulation mechanisms, but also serves as a catalyst for future studies of the epigenetic mechanisms that regulates development, differentiation and immune regulation mechanisms of mammalian research. 144 ABSTRACT 107 GENERATION OF CUSTOMIZED AND READ-TO-USE GENETICALLY ENGINEERED MICE AT THE INSTITUT CLINIQUE DE LA SOURIS – ICS (THE FRENCH MOUSE CLINICAL INSTITUTE) Marie-Christine Birling, Andrée Dierich, Karim Essabri, Lydie Venteo, Guillaume Pavlovic, Tania Sorg, and Yann Hérault Institut Clinique de la Souris (ICS) and IGBMC, Illkirch, France, http://www.ics-mci.fr/ The Institut Clinique de la Souris - ICS is a research infrastructure that provides extensive services ranging from the development of mouse models to comprehensive phenotyping. The Mutagenesis, Transgenesis and Molecular Phenotyping Department is dedicated to the development and molecular validation of new mouse models. Four main services are offered through the Department: 1) The Genetic Engineering service provides expert advises, proposes strategies and takes care of the design of new targeting constructs. All kind of constructs can be developed in the service: a. Constructs for random insertion of a transgene (traditional or BAC construct), b. Constructs targeted insertion of a transgene, or for targeted transgenesis (Constitutive knock out, Tissue specific knock out, Point mutation, Humanization, Flex models, etc) The Genetic Engineering service is also in charge of the screening and validation of the ES cell clones. Long range PCR analysis, sequencing of the inserted fragment, as well as Southern blot analysis on the genomic DNA of embryonic stem (ES) cells are routinely performed. 2) The ES cell service is dedicated to the electroporations, selections, picking, amplification and karyotyping of the ES cell clones. New ES cell lines from various genetic backgrounds have been derived. Notably, since 2009, the standard electroporations are performed in an in house derived C57Bl6/N ES cell line. This line has been shown to be quite efficient for homologous recombination and germ line transmission. 3) The Genotyping service confirms the insertion of the transgene when the mice are born. A PCR genotyping strategy is provided to the client at the delivery of the model. 4) The Molecular Phenotyping service analyzes, and most often validate, the functionality of the genetic modifications introduced in vivo. The techniques choosen for these analyzes are dependent on the mouse model (qPCR, RTqPCR, Southern blot, Western blots, in situ Hybridization). The relevance of this molecular analysis before the generation of phenotyping cohorts is very high. 145 In addition to the service activities of the department, internal research and development programs (eg. development of new mouse tools) in concertation with European funded project such as the Create consortium (http://www.creline.org/) are carried out. Two main resources as freely available the community: -The CreERT2 zoo. More than 50 cell or tissue specific promoter driven CreERT2 transgenic lines are available to the whole scientific community (http://www.ics-mci.fr/crezoo.html). Most of these lines have been partially validated and show at least specificity in the mRNA expression. -The NR zoo: most of the nuclear receptors and their co-factors have been developed as mouse conditional KO models (http://www.ics-mci.fr/nrzoo.html) 146 ABSTRACT 108 PUDDING - A NOVEL MODEL OF GLOMERULARNEPHRITIS Paul K Potter1, L Wisby1, FW Tam2, S Wells1, T Hough1, HT Cook2, M Cheeseman1, and SDM Brown1 1 MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, UK 2 Renal Unit, Division of Medicine, Hammersmith Campus, Imperial College, London, UK As part of the ongoing phenotype driven ENU mutagenesis programme at MRC Harwell we identified mice from a recessive pedigree that exhibited normal post-natal growth and behavior but at around 90 days of age rapidly deteriorated, exhibiting clear signs of a debilitating disease. Further breeding was carried out confirming this was an inherited recessive trait. Initial histological analysis of kidneys in affected individuals indicated the mice had a glomerulopathy. To further characterize renal function in these mice we carried out clinical chemistry analysis of serum and urine. The mice developed proteinuria and azotemia (serum creatinine 50-112 umol/L vs 12.8-13.5 umol/L and urea >32 mmol/L vs 9 mmol/L in affected and aged matched controls respectively). This is associated with reduced serum albumin concentration and raised amylase. Further detailed histological analysis showed that these mice had crescentic glomerulonephritis. The disease results in the rapid deterioration of renal function around 90 days of age with only minimal changes in serum creatinine levels at 50 days of age. Preliminary mapping studies indicate that the causative mutation lies within a region spanning 80.1-93.2 Mb on chromosome 1. These initial studies indicate that this is a novel model of spontaneous glomerulonephritis and we are carrying out further investigations to identify the underlying mutation and further define the pathogenesis of disease in this line. 147 ABSTRACT 109 THE HARWELL AGEING SCREEN Paul K Potter MRC Harwell, Harwell Science and Innovation Site, Oxfordshire, UK Diseases associated with ageing pose an increasing social and financial burden on society and represent a vital imperative for research in the biomedical sciences. Despite the complications of genetic background in human populations and the confounds of environment there has been considerable advances, particularly through Genome-wide association studies (GWAS) in identifying loci involved in diseases of aging. Nevertheless, this is only a first step towards a more fundamental understanding of the genetic pathways involved. Animal models are required both to test our understanding of these pathways, as well as to provide the tools for developing and assessing therapeutics. We are undertaking the first large-scale project to employ mutagenesis and phenotyping programmes to specifically generate new recessive models of late onset or age-related disease. The emphasis will be on the exploration of phenotype space in aging mouse mutant populations providing us with the opportunity to: identify genes and pathways involved in age related disease, scrutinise these models for biomarkers of age related disease, and provide better platforms for pre-clinical assessment of new therapies for such diseases. Pedigrees will be aged up to 18 months und undergo phenotyping across a wide range of disease areas. Analysis will occur at several defined time points throughout the life of the mice and the phenotypes included in the screening include diabetes and metabolism, neurobehaviour, cognitive tests, bone analysis, renal function, cardiac disease, liver function, sensoneural (vision and hearing), and a comprehensive clinical chemistry screening. Other screens are also in development. Detailed histological analysis will be carried out as part of the terminal analysis and a biobank of samples taken at different ages will also be established for retrospective analysis. The age challenged mice will be an important resource for many research groups, identifying novel genes and pathways resulting in age-related phenotypes along with the potential to discover new biomarkers. 148 ABSTRACT 110 GELSOLIN PLAYS A ROLE IN THE ACTIN POLYMERISATION COMPLEX OF HAIR CELL STEREOCILIA Philomena Mburu1, MR Romero1, Helen Hilton1, Andrew Parker1, Stuart Townsend1, Yoshiaki Kikkawa2, and SDM Brown1 1 Medical Research Council Mammalian Genetics Unit, Harwell Science and Innovation Campus, Oxfordshire, UK 2 Department of Bioproduction, Tokyo University of Agriculture, Yasaka, Abashiri, Hokkaido, Japan A complex of proteins scaffolded by the PDZ protein, whirlin, reside at the stereocilia tip and are critical for stereocilia development and elongation. We have shown that in outer hair cells (OHCs) whirlin is part of a larger complex involving the MAGUK protein, p55, and protein 4.1R. Whirlin interacts with p55 which is expressed exclusively in outer hair cells (OHC) in both the long stereocilia that make up the stereocilia bundle proper as well as surrounding shorter microvilli that will eventually regress. In erythrocytes, p55 forms a tripartite complex with protein 4.1R and glycophorin C promoting the assembly of actin filaments and the interaction of whirlin with p55 indicates that it plays a similar role in OHC stereocilia. However, the components directly involved in actin filament regulation in stereocilia are unknown. We have investigated additional components of the whirlin interactome by identifying interacting partners to p55. We show that the actin capping and severing protein, gelsolin, is a part of the whirlin complex. Gelsolin is detected in OHC where it localizes to the the tips of the shorter rows but not to the longest row of stereocilia and the pattern of localisation at the apical hair cell surface is strikingly similar to p55. Like p55, gelsolin is ablated in the whirler and shaker2 mutants. Moreover, in a gelsolin mutant, stereocilia in the apex of the cochlea become long and straggly indicating defects in the regulation of stereocilia elongation. The identification of gelsolin provides for the first time a link between the whirlin scaffolding protein complex involved in stereocilia elongation and a known actin regulatory molecule. 149 ABSTRACT 111 NEW STERILE MODEL MOUSE FROM RIKEN ENU-BASED GENEDRIVEN MUTAGENESIS Takuya Murata, Shigeru Makino, Ryutaro Fukumura, and Yoichi Gondo RIKEN BRC, Ibaraki, Japan RIKEN ENU-based gene-driven mutagenesis, “a reverse genetics in the next generation”, is based on the two key resources; mutant genome DNA and frozen sperm of thousands of ENU-mutagenized G1 male. By high-throughput screening of mutant genomic DNA library, we can obtain allelic series of mutations on the target gene. Despite of randomly induced mutations, we can usually discover many missense mutations together with “null” mutation(s). Compared with the knockout method, it is efficient to find multiple mutations even in a cost effective way. The targeted mice are recovered by IVF technology from the frozen sperm. Since thousands of point mutations were introduced in all over the genome of each G1 mouse line, they are usually removed by continuous backcrosses at least six generations. Along the way of the backcrosses, the excess hetrozygotes at every generation may be utilized to the intercrosses in order to pre-examine the “phenotypes even still in progress.” This distinctive infrastructure has been widely opened to the research community since 2002 (http://www.brc.riken.jp/lab/mutants/genedriven.htm). WNT/β − catenin signaling is essential for various stages of development, including cell fate determination, cell proliferation (tumorigenesis), maintenance of the stem cell state, and so on. In the targeted analyses of the C429S mutant mouse line on the β−catenin gene, we found unique phenotypes observed in the homozygotes obtained from 12th backcrossed parents so far. The mutant homozygotes were born normally escaping from the embryonic lethality; however, most of them are sterile, despite oogenesis or spermatogenesis looked normal. In female, most homozygotes showed hydrometrocolpos (uterine edema) resulting from vaginal atresia (closure of vagina). In homozygous male, regardless of making vaginal plug, seminiferous seemed to be affected, because many sperm cells were found ectopically in seminal vesicle as well as its extra lobes. From the observation along the seminal vesicle formation, these abnormal lobes were originated from the caudal ends of the Wolffian duct, which normally develops to caudal vas deferens, namely that the cell fate change occurred in the caudal Wolffian duct of the homozygous male. The sterility, coinciding with persistent malformations of the internal genitalia, has been previously reported in various human cases, such as hydrometrocolpos (equivalent to our phenotype), PMDS (Persistent Mullerian Duct Syndrome), MKKS (McKusick-Kaufman Syndrome), and so on. However, the cases corresponding to our homozygous males have never reported in both mouse and human, suggesting a new sterile model in this field. 150 ABSTRACT 112 GENETIC DIVERSITY AMONG C57BL/6 SUBSTRAINS BASED ON SNPS Kazuyuki Mekada1, Ikuo Miura1, Takehide Murata1, Atsushi Toyoda2, Mayu Hirose1, Shigeharu Wakana1, Kuniya Abe1, and Atsushi Yoshiki1 1 RIKEN BioResource Center, Japan 2 National Institute of Genetics, Japan The C57BL/6 mouse strain is the best-known inbred strain and is commonly used to create congenic strains as the genetic background for spontaneous and induced mutations. Since the 1950s, C57BL/6 has diverged to several substrains including C57BL/6J and C57BL/6N substrains. Currently, over 20 substrains have been established worldwide, and their phenotypic and genetic differences have been reported. Large-scale mutagenesis programs to mutate all of the protein-encoding genes in the mouse using gene trapping and targeting in embryonic stem cells derived from C57BL/6N are ongoing. Therefore, it is required for the correct interpretation of the data from the functional analyses of these genes to clarify the genetic properties of the C57BL/6N substrains and the differences between C57BL/6N and C57BL/6J. We have already reported over 30 single nucleotide polymorphisms (SNPs) in several C57BL/6 substrains. Nevertheless, we have not yet recovered any SNP markers between C57BL/6J and C57BL/6N on chromosomes 1, 12 and Y. Besides, no SNP markers that can distinguish the C57BL/6N substrains have been identified. Recently, a more detailed genome sequence information of the C57BL/6N is available from the end sequencing program of C57BL/6N BAC clones by RIKEN BRC and the National Institute of Genetics with support of NBRP, MEXT Japan, and the resequencing project by the Wellcome Trust Sanger Institute. In this study, we report recovery of new SNPs between the C57BL/6J and C57BL/6N substrains using these information, and the genetic diversity among C57BL/6 substrains. 151 ABSTRACT 113 NOVEL ENU SCREENS FOR IMMUNE-RELATED PHENOTYPES Vera M Ripoll1, Marie Hutchison2, Paul Potter2, and Roger Cox1 1 Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, UK 2 Mary Lyon Centre, Medical Research Council, Harwell, Oxfordshire, UK The identification of novel alleles through ENU mutagenesis has provided clues to a number of genes responsible for the immune response, contributing substantially to our understanding of host defence, allergy and autoimmunity. We are undertaking two different ENU screens to expand our library of immune-related alleles and generate new models for inflammatory diseases. Our phenotype-driven screen aims to identify ENU induced mutations that result in abnormal blood leukocyte counts and cell ratios. To date we have screened 88 pedigrees (1700 animals) utilizing both haematology and Flow Cytometry analyzers. We have identified at least 17 mouse lines with abnormal monocyte, neutrophils and/or lymphocytes numbers. Five different pedigrees with distinctive blood phenotypes were selected for inheritance testing. We are currently undertaking further phenotypic analysis of these pedigrees as well as carrying out the mapping of the mutated chromosomal loci to identify potential gene candidates. We will describe up to date progress in these experiments. In a gene-driven approach, we have screened a DNA archive of 10000 samples from F1 ENU-mutagenized mice for novel mutations in macrophage-inflammatory genes. One of our genes of interest known as Mpeg1 (macrophage-expressed gene 1) is a macrophage-specific gene, involved in macrophage activation and responses to bacterial and viral infection. A mutant with a stop codon, which disrupts the most important protein domain, was re-derived. We have been carrying out primary immune-phenotyping experiments in Mpeg1 heterozygous and homozygous mutants. Preliminary data demonstrate that macrophages from Mpeg1 homozygous mice are unable to uptake bacteria and exhibit impaired phagocytosis rates. Mpeg1 homozygous macrophages secrete significantly higher levels of nitric oxide than Mpeg1 heterozygous and wild type macrophages. These results confirm the involvement of Mpeg1 in the endocytic pathways and suggest a role for Mpeg1 in the macrophage antibacterial responses. 152 ABSTRACT 114 GRB10 MODIFIES MPNST TUMORIGENESIS IN A MOUSE MODEL OF NF1 Karlyne M. Reilly, Jessica Van Schaick, Keiko Akagi, Christina DiFabio. Robert Tuskan, Sandra Burkett, and Jessica Walrath Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA As human GWAS studies make the identification of susceptibility genes for cancer more accessible, mouse models of cancer provide a unique opportunity to identify modifier genes for rare human cancers and modifiers that show complex epistatic interactions. Neurofibromatosis type 1 (NF1) is one of the most common genetics diseases affecting the nervous system, with patients developing cancers of both the central and peripheral nervous systems. Although these tumors are less common, they have very poor prognosis, and so a better understanding of the biology underlying nervous system tumors is needed to develop new therapeutic options. We are using a mouse model of NF1 in which the Nf1 and Trp53 gene are mutated on the same chromosome in cis (NPcis mice). These mice develop central and peripheral nervous system tumors characteristic of NF1 and can be used to identify modifier genes of these tumors. We have identified both strain-specific modifier effects and parent-of-originspecific modifier effects that show interesting patterns of interaction. We report here the identification of an imprinted modifier gene, Grb10, on mouse Chromosome 11. Grb10 is expressed at different levels in tumors from progeny of NPcis mutant mothers compared to tumors from progeny of NPcis mutant fathers and correlates with differences in tumor susceptibility. We show that mutation of Grb10 in the cis configuration with Nf1 and Trp53 alters susceptibility to peripheral nerve tumors in vivo. 153 ABSTRACT 115 HOMOLOGY MAPPING: DEVELOPMENT OF THE MOUSE HINDLIMB MUSCULOSKELETAL MODEL USING THE HUMAN MUSCULOSKELETAL MODEL AND SCLERAXIS (SCX)-GFP MOUSE Satoshi Oota1, Nobunori Kakusho2, Yosuke Ikegami3, Kazuyuki Mekada1, Koh Ayusawa3, Hirotaka Imagawa3, Yuichi Obata1, Ryutaro Himeno4, Hideo Yokota2, Yoshihiko Nakamura3, and Atsushi Yoshiki1 1 BioResource center, RIKEN, Tsukuba, Japan 2 Advanced Science Institute, RIKEN, Wako, Japan 3 Department of Mechano-Infomatics, The University of Tokyo, Tokyo, Japan 4 Advanced Center for Computing and Communication, RIKEN, Wako, Japan Recently, the hypothesis-driven strategy has become useful for analyzing movement disorders of laboratory mice. This new strategy possesses a complementary role for the traditional comprehensive battery strategy. In order to acquire detailed data on motor function required by this new strategy, biomechanics is an ideal framework. However, difficulties in the direct application of biomechanics to laboratory mice remain. One burdensome difficulty is the development of a musculoskeletal model that can be used to perform biomechanical analyses on subjects. Because of the heavy requirements in the medical and athletic areas, detailed human musculoskeletal models have been elaborated. On the other hand, no fine-grained musculoskeletal model of the laboratory mouse has been developed to date. Unlike humans, knowledge of the functional anatomy of the mouse is limited. Although MRI scan is a conventional methodology for obtaining data on functional anatomy, its spatial resolution is insufficient for mice due to their considerably small sizes. To overcome this problem, we developed two alternative methods: (1)Homology mapping. The human and mouse skeletal systems are evolutionarily conserved. Theoretically, it is possible to map in silico part of the human muscular system onto an existing mouse skeletal model according to evolutionary relationships. We developed an algorithm to homologically map the human muscular system on the mouse skeleton. (2)Scanning transgenic mice. Using transgenic mice (the limb tendon-specific Scleraxis (Scx)-GFP reporter line), we obtained 3D geometries of limb tendons using a 3-dimensional internal structure microscope. We estimated musculotendon geometries with regard to skeletal elements by integrating them with X-ray CT scan data. These methods are not only useful for developing a musculoskeletal model but also provide insights on evolutionary difference between humans and mice. 154 ABSTRACT 116 MUTATION IDENTIFICATION IN MICE BY EXOME SEQUENCING Laura Reinholdt1, David Bergstrom1, Steve Murray1, Michelle Curtain1, Carol Bult1, Joel Richardson1, Lucy Rowe1, Mary Barter1, Daniel J. Gerhardt2, Mark D'Ascenzo2, Todd Richmond2, Tom J. Albert2, Jeffrey A. Jeddeloh2, Benjamin Kile4, Ivo Gut5, Jay Shendure3, and Leah Rae Donahue1 1 The Jackson Laboratory, Bar Harbor, ME, USA 2 Roche NimbleGen, Inc., Madison, WI, USA 3 University of Washington, Department of Genome Sciences, Seattle, WA USA 4 Walter and Eliza Hall Institute of Medical Research, Parkville Victoria, Australia 5 Centre National de Génotypage, Evry, France Traditionally, mutations in the mouse are identified via genetic mapping, a process that can take several years and requires ample vivarium space. However, with the advent of high throughput sequencing technology, mutations can be discovered with minimal mapping data. Potential high throughput sequencing approaches to mutation discovery include whole genome sequencing or targeted re-sequencing via capture technologies. Currently, the sequencing and analysis of whole genomes remains burdensome and expensive for many laboratories. Targeted re-sequencing is less expensive and the data are compensatingly more manageable, but this technique requires substantial genetic mapping and the design and purchase of custom capture tools (i.e. arrays or probe pools). Targeted re-sequencing of the coding portion of the genome, i.e. the ‘exome’, provides an opportunity to resequence mouse mutants with minimal mapping data and alleviates the need for a custom array / probe pool for each mutant. This is critical for large sets of mice, such as those in The Jackson Laboratory’s Mouse Mutant Resource (MMR); a program, which has been funded to provide spontaneous and induced mutant mouse models for 50 years. To significantly reduce the time, effort and cost of mutation detection, we have collaborated to develop a sequence capture probe pool representing the mouse exome. This exome is a collection of 203,225 exonic regions comprising just over 54.3 Mb of target sequence, and is based on a unified gene set. The unique design consists of non-redundant gene predictions from NCBI, Ensembl and Vega. Our preliminary data show that capture sensitivity is high, >96.7% of the targeted bases are covered with just one lane of 75 bp paired end on the Illumina GAIIx (per 4 Gbp of mappable mate-pairs). The capture specificity is also high, >75% reads from the lane come from the targeted bases. The consequence of this high efficacy is that the platform provides sufficient coverage to detect heterozygosity. We have found the genetic background to only modestly impact the performance of a capture probe pool based upon the C57BL6(mm9) reference. Lastly, and most importantly, we have successfully used this approach to identify multiple spontaneous and induced mutations in the mouse. 155 ABSTRACT 117 STATUS OF THE US LINES OF THE COLLABORATIVE CROSS: COMPLETED LINES Darla R Miller1, Ryan J Buus1, Jennifer N Shockley1, Stephanie D Hansen1, Ginger D Shaw1, Terry J Gooch1, Jason S Spence1, Timothy A Bell1, Catherine E Welsh2, Leonard McMillan2, Wei Wang2, Jeremy Wang2, Eric Yi Liu2, Kenneth F Manly3, Gary A Churchill4, David W Threadgill5, and Fernando Pardo-Manuel de Villena1 1 Department of Genetics, Lineberger Comprehensive Cancer Center, Carolina Center for Genome Science 2 Department of Computer Science; University of North Carolina Chapel Hill, NC, USA 3 Department of Biostatistics, University at Buffalo, Buffalo NY, USA 4 The Jackson Laboratory, Bar Harbor, ME, USA 5 Department of Genetics, North Carolina State University, Raleigh, NC, USA The Collaborative Cross (CC) is a large panel of recently established multiparental recombinant inbred (RI) mouse lines specifically designed to overcome the limitations of existing mouse genetic resources for analysis of phenotypes caused by combinatorial allele effects. The CC is the only existing mammalian resource that has high and uniform genome-wide variation effectively randomized across a large, heterogeneous, and infinitely reproducible population in order to support data integration across environmental and biological perturbations, across space (different labs) and time (temporal studies). Since the last status report published in 2008 there have been significant changes in the production of the US arm of the CC, including the relocation of the CC to UNC, the constitution of an External Advisory Board, the naming scheme for completed CC lines and the establishment of an MTA to facilitate access, distribution and preservation of CC lines from the different arms. There have been changes in the breeding scheme in later generations of inbreeding, the development of a medium density SNP panel optimized for the CC and the use of this panel to accelerate inbreeding through selective mating based on marker genotypes. We will report on the completion of the first CC line, the status of the lines undergoing marker assisted inbreeding and access to CC mice, highdensity genotypes and haplotype reconstructions of CC related resources (http://csbio.unc.edu/CCstatus/). 156 ABSTRACT 118 WHOLE GENOME RESEQUENCING OF ESTONIAN DAIRY COW (ESTONIAN HOLSTEIN) Rutt Lilleoja1,2, Ü Jaakma1, and S Kõks1,2 1 Estonian University of Life Sciences, Tartu, Estonia 2 University of Tartu, Tartu, Estonia The bovine reference genome currently available from public databases is one of a Hereford female (Btau_4.0). We present here whole genome resequencing of Estonian dairy cow using next generation sequencing. Whole genome sequence of a female Estonian Holstein was generated using massively parallel sequencing technology. 700 million short sequencing reads were obtained from 50-bp mate-paired library with a mean insert size of 1500 bp. All data was aligned against Btau_4.0 Hereford reference sequence using Bioscope v1.2. We were able to map over 75% of the reads to the reference, that is 42.3 gigabases of sequence resulting in an avarage of 16-fold coverage. Further analysis of our data is ongoing and the results will be presented during the conference meeting. 157 ABSTRACT 119 HYPOTHALAMIC MITOCHONDRIAL DYSFUNCTION, A POSSIBLE CAUSE OF ANOREXIA Charlotte E Lindfors1#, Ida A Nilsson1,2#*, Pablo M Garcia-Roves3, Aamir R Zuberi4,5, Mohsen Karimi6, Lea R Donahue4, Derry C Roopenian4, Jan Mulder7, Mathias Uhlén8, Tomas J Ekström6, Muriel T Davisson4, Tomas Hökfelt2, Martin Schalling1, and Jeanette E Johansen1 1 Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden 2 Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden 3 Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden 4 The Jackson laboratory, Bar Harbor, Maine, USA 5 Present address: Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA 6 Department of Clinical Neuroscience, Karolinska Institutet Stockholm, Sweden 7 European Neuroscience Institute at Aberdeen, University of Aberdeen, Aberdeen, UK 8 Department of Biotechnology, AlbaNova University Center, Stockholm, Sweden #equal contribution Human dysfunction of the oxidative phosphorylation system (OXPHOS) and complex I (CI) can be associated with poor feeding and failure to thrive (FTT). These phenotypic traits are also seen in the anx/anx mouse which exhibits disturbed feeding behavior and aberrances, including signs of neurodegeneration, in peptidergic neurons and neurotransmitter systems important for the regulation of food intake in the hypothalamic arcuate nucleus (Arc). In rodents, the Arc neuronal networks develop primarily during the first three postnatal weeks, a very energy demanding period. We therefore hypothesized that the starvation and degenerative phenotypes in the anx/anx mouse could be related to defects in the OXPHOS and possibly CI deficiency. Via an Affymetrix expression analysis on Arc and mapping of the anx locus, we identified one of the CI assembly factors, the Ndufaf1 gene, as an excellent anx candidate gene. By Taqman realtime-PCR and western blots we showed that the Ndufaf1 gene and protein is approximately 50% downregulated in anx/anx mice and we also showed that it is specific to the anx-allele. Furthermore, native Western blots demonstrate decreased hypothalamic levels of fully assembled CI, and increased levels of partly assembled CI in anx/anx mice. Finally, we show evidence for oxidative stress and reduced efficiency of CI in anx/anx hypothalamus. These and previous results provide evidence that the anorexia and FTT of the anx/anx mouse are caused by hypothalamic neurodegeneration following dysfunction of mitochondrial CI respiration. 158 ABSTRACT 120 HIGH-THROUGHPUT GENOTYPING AND QUALITY CONTROL OF GENE-TARGETED MUTATIONS IN THE MOUSE Edward Ryder, Diane Gleeson, Debarati Bhattacharjee, Ross Cook and the Mouse Genetics Programme Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK The Sanger Institute is an active member of the international KOMP and EUCOMM consortiums, whose goal is to produce conditional knockouts in ES cell lines and mice as a resource for the research community. As part of these projects and the EUMODIC European phenotyping collaboration, the Mouse Genetics Programme (MGP) is involved in the generation of mouse mutants from the ES cell stage to primary phenotyped mice. To establish a mutant colony progeny from chimeras must be initially genotyped to determine whether they carry the mutation in the germ line, and in subsequent generations to see if they are wild type, heterozygous or homozygous. Tissue samples are genotyped by a combination of Neo count qPCR and short range PCR, which allows us to process up to 10,000 samples per week. Once a sufficient number of mice have been generated they are sent as cohorts to the phenotyping pipelines for further study and analysis, returning to genotyping for verification at necropsy. QC methods are discussed for confirming the targeting and mutant allele structure prior to injection, and in mouse colonies prior to release to the phenotyping pipelines, external laboratories and the EMMA and KOMP repositories. This is in conjunction with the newly established EUCOMM minimum genotyping standard for mice generated by the collaborating laboratories. Experiments to determine FRT-recombination rates in the mutant alleles, and gene expression studies are also discussed. 159 ABSTRACT 121 THE SANGER MOUSE GENETICS THROUGHPUT CHARACTERISATION PHENOTYPES IN KNOCKOUT MICE Mark Lucas, C Podrini, E Cambridge, S Clare, L of the Mouse Genetics Programme The Sanger M.G.P., Wellcome Trust Sanger Hinxton, Cambridgeshire, United Kingdom PROGRAMME: HIGH OF IMMUNOLOGICAL Mottram, JK White on behalf Institute, Genome Campus, The Sanger Institute Mouse Genetics Programme is committed to making a significant contribution to the functional annotation of the mammalian genome. We are using the growing knockout first conditional ready targeted ES cell resource currently being created at Sanger, to generate, characterise and archive 200 lines of knockout mice per year. In addition to studying the role of each gene in normal development and function, the breadth and depth of our phenotyping platform ensures that phenotypic data on a spectrum of disease conditions are obtained for each mouse line without the need for any prior assumptions about function. One area of focus is the immune system. Whole blood is collected from mice that have undergone a standardised battery of phenotyping tests. Multi-staining FACS analysis is used to profile the major immune cell populations from these immunologically naïve bloods, whilst haematological abnormalities are assessed through complete blood counts. Furthermore, the immunoglobulin profile of plasma from these mice is assessed. A panel of 42 tissues and organs including the spleen, thymus and mesenteric lymph nodes are collected at necropsy and processed through to paraffin blocks for storage in our Biobank as a stable, long term resource. An additional cohort of mice from each mutant strain is also subjected to pathogenic challenge to assess susceptibility to two common pathogens, Citrobacter rodentium and Salmonella enterica serovar Typhimurium. Complementing this analysis, we identify the expression profile of each gene using the lacZ reporter gene, expressed under the control of the endogenous promoter of the target gene. To date the PBL FACS analysis has completed 197 mutant lines, with 14 demonstrating a phenotype (7.7% hit rate) and the Plasma Immunoglobulin analysis has completed 44 mutant lines with 5 lines showing a phenotype (12.8% hit rate). Here we present a summary of the phenotyping data available to date including examples of novel findings from a subset of immunologically altered mutant lines. 160 ABSTRACT 122 THE MOUSE REFERENCE GENOME ASSEMBLY: UPDATES AND RESOURCES Valerie A Schneider and DM Church on behalf of the Genome Reference Consortium and NCBI Genome Annotation Team NCBI, Bethesda, MD; The Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK The Genome Center At Washington University, St. Louis, MO; EBI, Hinxton, Cambridge, UK The current version of the mouse reference genome, MGSCv37, is among the highest quality genomes in existence. Released in 2007, this assembly representing the C57BL/6J strain is comprised of >96% finished, clone-based sequence with <1% of the genome consisting of gaps. This valuable resource plays critical roles in murine research and enables comparative analyses with the genomes of humans and other organisms. The reference genome also serves as the framework for the assembly of other mouse genomes sequenced by various short-read technologies. The many research advances facilitated by the reference genome have led to the recognition that some regions of the mouse genome are not optimally represented in the current assembly. Regions recalcitrant to cloning in BAC vectors remain as gaps, while highly complex repetitive structures have caused mis-assembly at others. Due to inter-strain variation, some loci are insufficiently represented by the single strain of the reference genome. Correcting genome problems and providing sequence representation for these strain-variable regions is a major focus of the Genome Reference Consortium (GRC), the group responsible for the mouse reference genome. The GRC strives to make its work transparent and welcomes community input and feedback. We will present examples of regions under review by the GRC and discuss plans for future releases of the mouse reference genome. NCBI provides bioinformatics support for the GRC and also performs annotation of the mouse reference assembly through its genome annotation pipeline. Annotated features include genes, RefSeq transcripts, repeats, genomic sequences not used in the assembly and genomic, gene trap and MICER clones. NCBI also provides annotation for several partial assemblies from other mouse strains in regions with significant inter-strain variation. The NCBI MapViewer can be used to view this annotation and permits the integrated display of maps and assemblies that use different coordinate systems, while the CloneFinder tool can be used to identify clones that map to a defined genomic region as specified by coordinate or annotated feature. We will present information on these tools and on recent NCBI efforts to re-annotate MGSCv37. 161 ABSTRACT 123 DBVAR AND DBSNP: NCBI DATABASES OF SIMPLE AND STRUCTURAL VARIATIONS Valerie Schneider, Tim Hefferon, John Garner, Azat Mardanov, Ming Ward, Aleksey Vinokurov, Melvin Quintos, Mike Kholodov, David Shao, John Lopez, Steve Sherry, and Deanna Church National Center for Biotechnology Information, National Library of Medicine, Bethesda, Maryland, USA The National Center for Biotechnology Information (NCBI) creates and maintains a set of databases that archive, process, display and report information related to germline and somatic variants. These databases, the Database of Genomic Structural Variations (dbVar) and the Database of Single Nucleotide Polymorphisms (dbSNP), are integrated with many resources at NCBI including Gene, GeneTests, OMIM, PubMed, and Nucleotide. This presentation focuses on dbVar and dbSNP, summarizing current function and highlighting recent improvements. Key to both databases are the archival and processing functions. Each submission is assigned a database identifier (nssv# and ss#) based either on flanking invariant sequence or locations asserted on reference sequences. Data in dbSNP are then processed to aggregate information from multiple submitters (assign rs#) and to calculate locations on each version of a genome and on NCBI Reference Sequences (RefSeqs). Because these stable, public accessions are citable in publications, they facilitate aggregation of information across studies. Researchers and genetic testers are encouraged to submit their variation data and to cite their submissions in manuscripts and on the web. Once data are accessioned, they are made available in diverse ways: Entrez searches, studyspecific reports, annotation on the genome, human gene-specific displays such as Variation Viewer, and ftp transfer. Advanced search functions to allow users to find variants with specific qualities are available using our limits and advanced search functions. Acknowledgments Work at NCBI is supported by the NIH Intramural Research Program and the National Library of Medicine. 162 ABSTRACT 124 CURRENT PROGRSSS OF PHENOTYPIC ANALYSES IN JAPAN MOUSE CLINIC Tomohiro Suzuki, Hideki Kaneda, Kimio Kobayashi, Ikuo Miura, Tamio Furuse, Ikuko Yamada, Osamu Minowa, Hideaki Toki, Hiromi Motegi, Maki Inoue, Tetsuo Noda, Hiroshi Maduya, and Sigeharu Wakana RIKEN BioResource Center, Japan Japan Mouse clinic (JMC) has been established for comprehensive mouse phenotyping with national mouse resources since 2008. As far as we set up phenotyping pipeline, standard operating procedures (SOPs), and home-page to receive a request as a first step, and high-through put mouse reproduction system based on in vitro fertilization technique and high-through put genome scanning system for checking and integration of mouse genetic background as a second step, and data capture and publication system of mouse phenotypic information as a last step. Until now we have received requests of comprehensive phenotypic analysis about 112 strains and have performed about 47 strains. The phenotypic information about a part of the 47 strains, including 13 standard strains, has been published on a phenotypic viewing site named Pheno-Pub on JMC homepage (http://phenopub.brc.riken.jp/). In this meeting we will show the results obtained from enormous phenotypic data in JMC about variances among trials on all test items in JMC using phenotypic data obtained from C57BL/6J, phenotypic differences between C57BL/6 sub-strains and among breeding colonies, and frequencies of phenotypic differences among genotypes (homozygote, heterozygote, and wild-type). In addition, we will introduce the future plan about the construction of phenotyping platform to analyze gene-environmental interactions between gene-function and environmental factor such as nutrition, facility condition, and various stress for understanding genetic predispositions for environmental influences. 163 ABSTRACT 125 HYPERACTIVE MICE SHOW A CLUSTER OF ASSOCIATED METABOLIC FEATURES Jacqueline K White, Anna-Karin Gerdin, Christine Podrini on behalf of the Mouse Genetics Programme, and Karen P Steel Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK Mutant mice with balance problems due to vestibular system defects show a characteristic behavioural response of head-bobbing, circling and apparent hyperactivity. They have long been known to have smaller body weight, presumably due to their increased rate of exercise. We have taken a selection of these mutant lines through our primary phenotypic screen to ask what other features might be affected. The lines used included mutations in Myo6, Myo7a, Cdh23, Grxcr1 and Mir96, among others. The mutant lines with hyperactivity showed a characteristic cluster of other features with high frequency. Most showed hearing impairment, low body weight, reduced fat mass, reduced plasma cholesterol and lipoprotein levels, improved tolerance of glucose in the intraperitoneal glucose tolerance test, increased latency to respond to heat in the hot plate test, and increased activity, VO2 and VCO2 in the indirect calorimetry test. A separate group of mutants selected for deafness alone without a balance problem showed none of these features. A further set of mutant lines selected for small body weight showed only reduced fat mass and increased VO2 and VCO2, and none of the other features shown by the hyperactive group. The genes involved are expressed in a variety of tissues with the only common site being the inner ear. These data support a causal relationship between hyperactivity and the other metabolic features observed. 164 ABSTRACT 126 MULTIPLICITY OF PHENOTYPES IN EARLY GENERATIONS OF THE MOUSE DIVERSITY OUTCROSS POPULATION Karen L Svenson and GA Churchill The Jackson Laboratory, Bar Harbor, Maine USA The Diversity Outcross is currently being developed at The Jackson Laboratory as a new mouse resource population. The DO is a heterogeneous mouse stock derived from partially inbred ORNL Collaborative Cross strains. The population utilizes a randomized outcrossing strategy with avoidance of sibling matings. The DO is designed to compliment the CC strain panel by providing a high resolution mapping resource. Groups of DO will approximate the genetic diversity found in human populations. Each individual in the DO will be genetically unique, representing a novel combination of alleles present in the eight CC founder strains. Although individual DO animals cannot be replicated, mapping results can be validated and permanent models constructed using the CC strains or their F1 progeny. The randomized outcrossing scheme used to create and maintain the DO population confers significantly greater genetic variation than that found in common outbred strains of mice. The JAX DO is currently producing the fifth outcross generation and is being maintained as 160 breeding pairs. We have obtained animals from the third outcross population and have collected over 50 phenotypic measurements under both standard chow and high fat feeding conditions. Many of these measurements describe features of the metabolic syndrome, namely plasma glucose, insulin, triglycerides and HDL cholesterol, and body composition (% fat and weight). As expected, DO animals display a wide spectrum of variation in these and other parameters, including whole blood analysis and behavioral traits such as home cage nesting and aggressiveness. The phenotypic variation seen in the DO is comparable to that observed in a large-scale inbred mouse strain survey and mimics that of human populations. With rapidly emerging strategies for high throughput sequencing of both DNA and RNA, the assignment of coincident and relevant disease phenotypes to causative functional variants will also be accelerated, thereby enhancing the development of well defined disease models. 165 ABSTRACT 127 SDOP-DB: A DATABASE FOR INTERNATIONAL SHARING AND STANDARDIZATION OF MOUSE PHENOTYPING PROTOCOLS Nobuhiko Tanaka1, Kazunori Waki1, Hideki Kaneda2, Tomohiro Suzuki2, Ikuko Yamada2, Tamio Furuse2, Kimio Kobayashi2, Hiromi Motegi3, Hideaki Toki3, Maki Inoue3, Osamu Minowa3, Tetsuo Noda3, Shigeharu Wakana2, and Hiroshi Masuya1 1 Technology and Development Unit for Knowledge Base of Mouse Phenotype, BRC, RIKEN, Japan, 2Technology and Development Team for Mouse Phenotype Analysis, JMC, BRC, RIKEN, Japan, 3Team for Advanced Development and Evaluation of Human Disease Models, BRC, RIKEN, Japan Because differences in data between different protocols have often serious effects on their derived results, there are crucial needs for sharing detailed contents of protocols to integrate experimental data in experimental research communities. For that purpose, we developed a new data format termed Standardized Description of Operating Procedures (SDOPs), which provides an assay-specific descriptive framework and enables direct and detailed comparison of procedural parameters. Using this data format, we further developed a comparative standardized-protocol database, SDOP-DB (http://www.brc. riken.jp/lab/bpmp/SDOP/), as a practical application to share, compare and evaluate the contents of protocols to enable data comparisons and analyses in the field of mouse phenotypic analyses (Bioinformatics, 26(8), 1133-1134, 2010). The SDOP-DB has the following features: (1). The SDOP-DB is helpful for identifying procedural parameter(s) that can result in differences in data between different protocols. Users can easily access phenotype data through hyperlinks to mouse phenotype databases such as ‘Pheno-Pub’ (http://www.brc.riken.jp/ lab/jmc/mouse_clinic/en/m-strain_en.html) in Japan Mouse Clinic and ‘EuroPhenome’ (http://www.europhenome.org/databrowser/baselineViewer.jsp ) in European Mouse Disease Clinic, allowing association of the data with each user's own in-house data.(2). For all analyses to be covered in SDOP-DB, users can submit in-house standardized protocols by filling in MS-Excel form downloaded from the website. Therefore, SDOP-DB can act as a web-based repository of mouse phenotypic protocols. (3). Users can freely download SDOP system from the website, enabling users to use as the application tool for protocol version management both within and between laboratories. The many uses of the system in the research community can accelerate standardization of mouse phenotyping protocols.(4). To provide an interoperable framework for sharing and reuse of mouse phenotyping protocols among laboratories, we provide the SDOP-DB data in the Phenotyping Procedures Markup Language (PPML) format, a standardized data format for describing experimental protocols developed by Mouse Phenotype Database Integration Consortium (InterPhenome). The above-described features in the SDOP-DB are helpful for the international sharing and standardization of mouse phenotyping protocols. 166 ABSTRACT 128 MODELING HUMAN CHROMOSOME 21-ASSOCIATED MENTAL RETARDATION DISORDERS IN MICE Tao Yu1, Zhongyou Li1, Steven J Clapcote2, Zhengping Jia2, Chunhong Liu1, Annie Pao1, Sei-ichi Matsui1, Norma J Nowak1, John C Roder2, Chu Chen3, Allan Bradley4, and Eugene Y Yu1 1 Roswell Park Cancer Institute, Buffalo, NY, USA 2 University of Toronto, Toronto, Canada 3 Louisiana State University, New Orleans, LA, USA 4 Wellcome Trust Sanger Institute, Cambridge, UK Copy-number variation in the human genome can be disease-causing or phenotypically neutral. This type of genetic rearrangement associated with human chromosome 21 (Hsa21) includes partial Monosomy 21 and Trisomy 21, which are genomic causes for intellectual disability. The regions on Hsa21 are syntenically conserved with approximately 2.3, 22.9 and 1.1-Mb regions located on mouse chromosome 10 (Mmu10), Mmu16 and Mmu17, respectively. In this project, we generated chromosomal deletions and the reciprocal duplications spanning the entire Hsa21 syntenic regions on Mmu10, Mmu16 and Mmu17 in mouse ES cells using Cre/loxP-mediated chromosome engineering, which led to viable mutant mice carrying the corresponding deletions on Mmu10 or Mmu17 as well as duplications on Mmu10, Mmu16, or Mmu17. The phenotypic analysis of the mutant mice carrying individual rearrangement or compound rearrangements shows intellectual disability-related phenotypes. Mutant mice carrying either deletion exhibit impairment in learning and memory. The mutant mice trisomic for all Hsa21 syntenic regions exhibit Down syndrome-related neurological defects, including impaired cognitive behaviors, reduced hippocampal long-term potentiation and hydrocephalus. These results suggest that when all the mouse orthologs of the Hsa21 genes are triplicated, an abnormal cognitively relevant phenotype is the final outcome of the elevated expressions of these orthologs as well as all the possible functional interactions among themselves and/or with other mouse genes. Because of the desirable genotypes and phenotypes, these deletion and duplication mouse models may serve as powerful tools for further understanding intellectual disability associated with partial Monosomy 21 or Trisomy 21 and may also be used for developing novel therapeutic interventions for the clinical manifestation of the disorders. 167 ABSTRACT 129 TAIWAN MOUSE CLINIC—AN EXPERIMENT FOR PHENOTYPING SERVICE Jeffrey Jong-Young Yen, Yen-Hui Chen, and Ya-Wen Hsaio Taiwan Mouse Clinic, NRPGM, and IBMS, AS, Taipei, Taiwan MOUSE The mouse has emerged as one of the most popular human disease models recently since the availability of enormous amount of mutant mouse lines generated by spontaneous mutation, induced mutagenesis, transgenics and insertional mutagenesis, and gene targeting strategy. However, it is also well recognized that capitalizing of these mutant resources critically dependents upon our capability of comprehensive phenotyping for general health and in-depth characterization for a special physiological function. Since May 2008, we have established a comprehensive mouse phenotyping core facility in Taipei, called Taiwan Mouse Clinic, to promote local biomedical researches using the mouse as model by the funding through the National Research Program on Genomic Medicine (NRPGM), NSC, Taiwan. There are three major goals of our project: one, to construct a centralized, modular and user-friendly mouse phenotyping platform; two, to provide services, education and consultation for both entrylevel general physical checkup and advance phenotyping techniques; three, to provide a unique local, national and international resource for studying mouse models of human diseases. The key component of this project is the establishment of the Mouse Hotel which honors the health certificates from all mouse vivaria and only requests a minimum essential specific pathogen list and greatly reduces the turn-around time of animal trafficking within mouse clinic. All animals in hotel are all-in and all-out and are handled as if in P2 vivarium, and both animals and equipments in phenotyping laboratories are monitored for pathogen contamination routinely. In the past one and half year there are more than 50 laboratories all over Taiwan requested our services and the regular services have grown up to 5000 items per month and have reached more than 78% of our maximum service capacity. We have also obtained the base line comprehensive phenotyping data for the C57BL/6JNarl strain and have revised the standard operational procedures for home cage activity analysis with Clever System and for metabolic rate analysis with LabMaster Calorimetry Module. Finally, we have engaged enthusiastically in regional and international collaboration in exchanging phenotyping platforms, databases, and resources through the networks of Asia Mouse Phenotyping Consortium (AMPC) and International Mouse Phenotyping Consortium (IMPC). 168 ABSTRACT 130 INITIAL ASSEMBLY OF 17 MOUSE STRAINS FROM THE MOUSE GENOMES PROJECT Guy Slater1, Thomas M Keane1, Jared Simpson1, Aylwyn Scally1, Xiangchao Gan2, Richard Mott2, Jonathan Flint2, and David J Adams2 1 Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK 2 Wellcome Trust Centre for Human Genetics, Oxford University, Oxford, UK The Mouse Genomes Project has sequenced the genomes of 17 inbred mouse strains to between 20-35x coverage using paired-end Illumina sequencing. The reads vary in length from 36 to 108bp, and the read pairs contain insert sizes ranging from 150bp to 3kb. We present initial sequence assemblies of these 17 mouse strains. Assembly of such datasets remains challenging and a pure denovo approach is impractical. Therefore, we have developed a low memory assembly pipeline using a reference guided approach, whereby read pairs are mapped to the reference and then split into groups corresponding to the region to which they have been mapped. These groups of reads were de novo assembled using velvet, and the resulting contigs are refined by filling gaps between contigs by using alignment-guided local reassembly. For each strain, there is a varying sized pool of reads that do not map to the C57BL/6J reference and these are assembled de novo separately. The initial results from the assembly pipeline produced contigs with an N50 of approx.5kb. We are then using a mixed scaffolding approach that uses a combination of the reference and longer insert reads (3kb-40kb) to produce chromosome sized scaffolds per strain. The quality of the assemblies are evaluated by comparison of the C57BL/6NJ assembly to the C57BL/6N reference, and also by comparison of the NOD assembly to 10Mbp of manually finished BACs. We estimate that between 2-18Mbp of sequence is novel and not contained in the reference sequence, with some novel contigs being up to 10kbp in length and containing novel protein coding sequences. 169 ABSTRACT 131 CLUSTERED PROTOCADHERIN GENES: GENETIC CODES FOR GENERATING THE COMPLEX BRAIN Takeshi Yagi, Shunsuke Toyoda, Yasushi Itoga, Keizo Hirano, Atsushi Okayama, Shinichi Yokota, Shota Katori, Teruyoshi Hirayama, and Takahiro Hirabayashi Osaka University, Graduate School for Frontier Biosciences, Lab of KOKOROBiology, Japan The brain is a complex system composed of enormous numbers of differentiated neurons. To examine the molecular mechanisms underlying complex organization of the brain, it is important to identify the molecules involved in generating neural diversity and organization. The clustered protocadherin (Pcdh) genes encode diverse cadherin-related proteins that express in the brain. In mammals, the clustered Pcdh family consists of three gene clusters: Pcdh-a, Pcdh-b, and Pcdh-g. At the single-cell level, several dozen Pcdh-a, -b and -g mRNAs are regulated monoallelically, resulting in the combinatorial expression of distinct variable exons. The PCDH-Aand PCDH-G proteins also form oligomers, further increasing the molecular diversity at the cell surface. Loss-of function mice in the clustered Pcdh genes had neurological abnormalities for generating neural networks. Also we found epigenetic regulations of clustered Pcdh genes in the individual neurons. Here I would like to discuss genetic codes for generating the complex brain system. 170 ABSTRACT 132 COMPREHENSIVE PHENOTYPING OF MOUSE MODELS Tania Sorg, Marie-France Champy, Elodie Bedu, Roy Combe, Hamid Meziane, and Yann Herault Institut Clinique de la Souris, Illkirch, France The Institut Clinique de la Souris (ICS) / Mouse Clinical Institute (MCI) is a technology platform that provides a comprehensive set of highly specialized mouse services to scientists from academia and industry. The ICS combines the capacity of generating mutant mice on a large scale with a high-throughput and comprehensive phenotypic analysis of mice. The ICS phenotyping platforms are adapted for the study of genetically engineered mouse models (GEMMs), but can also be used for pharmacological and toxicological studies in the mouse. The ICS services will ultimately help the scientific community to use the mouse to develop a complete functional annotation of the human genome and to employ this to better understand human diseases and their underlying physiological and pathological basis. Achieving this goal will underpin and speed the biopharmaceutical and biotech industries that increasingly depend upon advanced, mouse-based analysis in drug development. The ICS has successfully assembled a comprehensive phenotyping platform, which at present is composed of the 5 following core units: - Clinical Chemistry laboratory. The activities of this laboratory cover clinical chemistry, hematology, coagulation, immunology, endocrinology and other metabolites. - Metabolic exploration. This service is set up to phenotype the metabolic function, such as in diabetes and obesity models. Currently tests are in place to analyze the body composition, glucose homeostasis, and energy expenditure, as well as the skeleto-muscular and the uro-genital systems, and the gastro-intestinal tract. - Cardiovascular and respiratory exploration. This service is set up to phenotype the function of the cardio-respiratory system. Currently tests are in place to analyze the cardiac function and anatomy, as well as the respiratory system such as in asthma models. - Behavior and nervous system. This service explores the central and peripheral nervous system, sensory systems, as well as behavior in mice. This core has developed a comprehensive tests battery to evaluate: general CNS function, affective behaviors (anxiety, depression), cognitive function, sensory thresholds and analgesia. Assays have also been set up to analyze the sensory systems (visual and auditory functions) - Histology and pathology. This service provides a comprehensive histological and histopathological analysis of mutant and control mice, as well as embryology studies. The assays performed by each core unit will be presented, as well as validated flow schemes for applications in therapeutic areas. 171 ABSTRACT 133 TARGETED AND WHOLE EXOME RESEQUENCING OF MOUSE MONOGENIC MUTANTS, QUANTITATIVE TRAIT LOCI AND CANCER MODELS David R. Beier1, Jennifer L. Moran2, Evan Mauceli2, Snaevar Sigurdsson2, Tim Fennell2, Lauren Ambrogio2, Miriam H. Meisler3, John C. Schimenti4, Karen Cichowski1, Laura Reinholdt5, Jane Wilkinson2, Stacey B. Gabriel2, Federica di Palma2, and Kerstin Lindblad-Toh3 1 Genetics Division, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA 2 The Broad Institute, Cambridge, MA, USA 3 Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA 4 Center for Vertebrate Genomics, Cornell University, Ithaca, NY, USA 5 The Jackson Laboratory, Bar Harbor, ME, USA We have established a mouse mutant resequencing initiative in order to apply next generation sequencing technology to the identification of ENU-induced and spontaneous mutations, quantitative trait loci (QTL) and cancer mutations. Funding of the initiative at the Broad Institute through The Large-Scale Genome Sequencing Program [National Human Genome Research Institute (NHGRI); NIH] has enabled pilot studies to be performed and future sequencing of approved models to be free to the mouse community. In two pilot studies, targeted resequencing of 13 monogenic mutants mapped to <5 Mb intervals was performed by solution-based hybrid selection of genomic DNA libraries and Agilent oligonucleotide RNA baits followed by Illumina sequencing. For spontaneous mutants and ENU-induced mutants in which a mutation in a noncoding region was suspected, the entire non-recombinant intervals were sequenced in mutant and control strains. For most ENU-induced mutants, all RefSeq exons, UTRs and promoters in non-recombinant intervals were sequenced. Over 5 Mb of genomic DNA covering 565 genes was sequenced in pilot work. Of 9 ENU-induced mutants analyzed, causal mutations were identified in 5 mutants. We have now designed two mouse exome baits covering all coding regions of a set of ~21,500 curated genes on the mm9 genome assembly. The more optimal exome design will be used in production for this initiative at the Broad and will also be available to the community through purchase from Agilent. An application process will be implemented for 1) sequencing of cancer models and mapped or unmapped monogenic mutants by whole exome sequencing and 2) Nimblegen-based targeted genomic regionbased sequencing of mutants or mapped quantitative trait loci. 172 ABSTRACT 134 DYNAMIC TRANSCRIPTOMES DURING NEURAL DIFFERENTIATION OF HUMAN EMBRYONIC STEM CELLS REVEALED BY SHORT, LONG, AND PAIRED-END SEQUENCING Jia Qian Wu 1, Lukas Habegger2, Parinya Noisa3, Anna Szekely4, Caihong Qiu5, Stephen Hutchison6, Debasish Raha7, Michael Egholm6, Haifan Lin5, Sherman Weissman4, Wei Cui3, Mark Gerstein2,8,9, and Michael Snyder1 1 Department of Genetics, Stanford University School of Medicine, Stanford, CA 2 Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 3 Institute of Reproductive and Developmental Biology, Imperial College London, London, UK 4 Genetics Department, Yale University, New Haven, CT 5 Yale Stem Cell Center, Yale University, New Haven, CT 6 454 Life Sciences Sequencing Centre, Branford, CT 7 Molecular, Cellular and Developmental Biology Department, Yale University, New Haven, CT 8 olecular Biophysics and Biochemistry Department, Yale University, New Haven, CT 9 Computer Science Department, Yale University, New Haven, CT In order to examine the fundamental mechanisms governing neural differentiation, we analyzed the transcriptome changes that occur during the differentiation of human embryonic stem cells (hESCs) into the neural lineage. Undifferentiated hESCs as well as cells at three stages of early neural differentiation, N1 (early initiation), N2 (neural progenitor), and N3 (early gliallike) were analyzed using a combination of single read, paired-end read, and long read RNA sequencing. The results revealed enormous complexity in gene transcription and splicing dynamics during neural cell differentiation. We found previously unannotated transcripts and spliced isoforms specific for each stage of differentiation. Interestingly, splicing isoform diversity is highest in undifferentiated hESCs and decreases upon differentiation, a phenomenon we call “isoform specialization.” During neural differentiation, we observed differential expression of many types of genes including those involved in key signaling pathways, and a large number of extracellular receptors exhibit stagespecific regulation. These results provide a valuable resource for studying neural differentiation and reveal new insights into the mechanisms underlying in vitro neural differentiation of hESCs, such as neural fate specification, NPC identity maintenance and the transition from a predominantly neuronal state into one with increased gliogenic potential. 173 ABSTRACT 135 IMPROVING MOUSE GENOME ANNOTATION THROUGH NEW TECHNOLOGIES Laurens Wilming, Gary Saunders, Markus Brosch, Gavin Laird, Adam Frankish, and Jennifer Harrow Wellcome Trust Sanger Institute, Cambridgeshire, UK New technologies - RNA-seq, protein mass spectrometry and multiple genome sequencing - provide researchers with a treasure trove of new data. Examining gene families such as MUP (Major Urinary Protein) or regions of interest such as the MHC (Major Histocompatibility Complex), we compare how the Mouse Genomes project (sequencing 17 mouse inbred strains) performs in these complex regions. New RNA-seq data is helping us identify new transcribed regions and offers the ability of correlating splice variants with tissues, cell types or developmental stages. Mouse proteome data produced by WTSI, combined with Peptide Atlas data, revealed over 10 new protein-coding genes not present in any database, and also evidence for exapted pseudogene loci that produce peptides. New proteome and transcriptome data allows us to complete and refine annotation by indicating new variants and extensions to incomplete genes and by showing previously separate gene fragments that can be joined into single loci. Apart from aiding annotation in general, this also helps us resolve CCDS (Consensus Coding Sequence project) conflicts. 174 ABSTRACT 136 GENOMIC ALTERATIONS IN TUMORS OF THE MCM4CHAOS3 BREAST CANCER MOUSE MODEL Marsha D. Wallace1, Ethan Cerami2, Stefan Stefanov1, Lishuang Shen1, and John C. Schimenti1 1 Cornell University, Ithaca, NY, USA 2 Memorial Sloan-Kettering Cancer Center, New York, NY, USA Our Chaos3 model in the defined C3H background contains the only endogenous gene mutation in mice known to lead exclusively to mammary tumors. Chaos3 is a point mutation of a highly conserved residue in the minichromosome maintenance 4 (Mcm4) gene, detected in a screen for Genomic Instability (GIN). MCM4 is a subunit of the Mcm2-7 complex that acts as the helicase to unwind DNA during DNA replication. It is also required for DNA replication licensing, which restricts replication to only once per cell cycle. When homozygous Chaos3 female C3H mice are aged, >80% develop exclusively breast cancer at ~12 months. To determine the underlying biological basis by which a point mutation in a DNA replication gene ultimately leads to breast cancer, we have employed Nimblegen Sequence Capture technology and CGH arrays to analyze Chaos3 tumors. We designed a Seq Cap chip containing ~1200 candidate cancer related genes to find mutations contributing to carcinogenesis. Furthermore, we analyzed Copy Number Variations (CNVs) in 11 Chaos3 tumors using Nimblegen 720K CGH arrays. While few mutations were uncovered, interestingly we find CNVs shared among many Chaos3 tumors. Virtually all Chaos3 tumors shared amplified regions on Chromosome 12 and 16. Also, specific regions on Chromosome 4, 5, and 11 are deleted in the majority of Chaos3 mammary tumors specifically, suggesting deletion of genes in these regions may be responsible for driving mammary transformation in the model. These genes are excellent candidates for future studies to validate susceptibility genes underlying spontaneous or heritable forms of breast cancer, demonstrating Chaos3 mice are a unique and powerful model to investigate mammary tumor carcinogenesis. 175 ABSTRACT 137 A MISSENSE MUTATION IN THE NOVEL WD-REPEATCONTAINING PROTEIN, WDR81, CAUSES PURKINJE CELL AND PHOTORECEPTOR CELL DEATH IN THE ENU-INDUCED NEUROLOGICAL MOUSE MUTANT NUR5 Maria Traka1, Kathleen J. Millen2, and Brian Popko1 1 Department of Neurology, The University of Chicago Center for Peripheral Neuropathy, The University of Chicago, Chicago, USA 2 Department of Human Genetics, The University of Chicago, Chicago, IL, USA To identify genes that affect neuronal survival in the central nervous system (CNS), we characterized the N-ethyl-N-nitrosourea-(ENU) induced autosomal recessive mutant mouse line nur5, which develops an adult-onset motor coordination disorder that significantly worsens with age. Histological analysis of these mice showed progressive postnatal Purkinje cell and photoreceptor cell death. The nur5 mutation was initially localized to a 24 cM region of Chromosome 11 between the Trp53 and Wnt3 genes. To fine-map the mutation, we performed genetic linkage analysis of polymorphic markers at the Trp53 Wnt3 interval that identified an L302P mutation in the WD-repeat-containing protein, WDR81. We confirmed that the nur5 phenotype is caused by the mutation in the Wdr81 gene by transgenic BAC rescue. Therefore, Wdr81nur5 was designated as a mutant allele of the Wdr81 gene. We show that WDR81 is expressed in Purkinje cells and photoreceptor cells, as well as other CNS neurons, and its expression pattern, based on immunohistochemical analysis, is not affected by the Wdr81nur5mutation. EM analysis demonstrated a prominent accumulation of large membranous particles in the mutant Purkinje cell dendrites that precedes neuronal loss, suggesting that dendrite defects may result in Purkinje cell death in Wdr81nur5 mutants. To discover potential binding partners of WDR81, we performed a yeast-two hybrid screen and coimmunoprecipitation analysis that revealed an association of this protein with the spectrin-repeat containing protein SYNE1. SYNE1 is expressed in Purkinje cells and is known to be important for Purkinje cell physiology, as the premature termination of SYNE1 was recently identified to be the cause of human adultonset autosomal recessive ataxia type 1 (ARCA-1) with cerebellar atrophy. The association of WDR81 with SYNE1 suggests that these proteins are part of a multimolecular complex that is critical for neuronal survival. 176 ABSTRACT 138 USING WHOLE GENOME NEXT-GENERATION SEQUENCING AS A TOOL TO FIND ENU-INDUCED MUTATIONS Michelle Simon, S Greenaway, P Denny, A-M Mallon, and JM Hancock Medical Research Council, Mammalian Genetics Unit, Harwell, Oxfordshire, U.K. Phenotype-driven screens after chemical mutagenesis of males with N-ethyl-Nnitrosourea (ENU) at MRC Harwell have been incredibly productive in identifying mouse mutants of biological interest. Nevertheless, identification of the causative mutations by conventional linkage analysis and Sanger sequencing of genes in the minimal genetic interval remains a bottleneck. We have been using next generation sequencing, using the Illumina Genome Analyser 2x platform, to accelerate this process of mutation detection. A sequence analysis pipeline, based on existing packages (i.e. Bowtie and Samtools) has been used to align reads to the C57BL/6J reference sequence, automatically identify unique variants, populate a custom sequence database and identify low and high confidence SNPs. Further developments will include assessment of the impact of putative mutations on splicing efficiency, predicted protein structure and integration with Mousebook – the MRC Harwell web portal. We will present the design and implementation of the re-sequencing pipeline, the methodology used to categorise high confidence SNPs and the identification of novel natural variants. 177 ABSTRACT 139 THE SANGER MOUSE GENETICS PROGRAMME; HIGH THROUGHPUT MORPHOLOGICAL ANALYSIS OF THE EYE Valerie E Vancollie1, J Estabel1, D Sunter1, VB Mahajan2,3, SH Tsang4, MC Naumann4, M Mahajan2,3, JK White1, on behalf of the Mouse Genetics Programme 1 The Sanger Mouse Genetics Programme, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, United Kingdom 2 Department of Ophthalmology and Visual Sciences, University of Iowa, IA USA 3 Omics Laboratory, University of Iowa, IA USA 4 Bernard and Shirlee Brown Glaucoma Laboratory, Department of Pathology and Cell Biology AND Department of Ophthalmology, College of Physicians and Surgeons, Columbia University, New York, NY USA The Sanger Mouse Genetics Programme aims to make a significant impact on our understanding of the function of genes and their role in disease by generating, characterising and archiving in the order of 200 lines of knockout mice per year. Knockout mice are phenotyped using a battery of tests relevant to key disease areas including diabetes, obesity, hearing and vision disorders, immune disorders, pain and motor function. In addition to studying the role of each gene in normal development and function, the breadth and depth of our phenotyping platform ensures that phenotypic data on a spectrum of disease conditions are obtained for each mouse line without the need for any prior assumptions about gene function. Here we will report on the utility of our high throughput approach for the morphological examination of eyes. Eyes are first screened using a slit lamp following a standardised list of parameters in order to view the lids, cornea, pupil and iris. Eyes are then dilated to view the lens and images of abnormalities are taken with a slit lamp mounted camera. The screen continues with eyes in the dilated state using an ophthalmoscope to view the retina. Any abnormalities observed are then imaged using Topical Endoscopic Fundus Imaging. Complementing this analysis, we identify the expression profile of each gene in adults and at E14.5, using the lacZ reporter gene, expressed under the control of the endogenous promoter of the target gene. Through collaboration, detailed histological analysis of all knockout lines that enter our pipeline is performed with follow up analysis on lines where lacZ expression has been noted in eye wholemounts. As a result of the combination of our core high-throughput eye morphology screen and collaboration on histological analyses we have observed phenotypic abnormalities in ~7% of all mutant lines examined to date. 178 ABSTRACT 140 A CELLULAR GENETICS PLATFORM TO IDENTIFY PHARMACOGENETIC TOXICITY PATHWAYS O Suzuki1, N Butz1, M Pletcher2, A Su3, B Steffy1, D Scoville1, A Frick1, J Trask4, R Thomas4, and Tim Wiltshire1 1 University of North Carolina at Chapel Hill, Division of Pharmacotherapy and Experimental Therapeutics 2 Pfizer Incorporated Groton, Connecticut 3 Genomics Institute of Novartis Research Foundation at La Jolla, California 4 The Hamner Institute for Health Sciences, Research Triangle Park, North Carolina, USA Pharmacogenetic studies have successfully identified genetic variants that contribute to variation in susceptibility to drug responses, but it is still a complex and challenging task to evaluate broadly across the human genome to identify the genetic components of response to drugs. Despite limitations there is a pressing need to identify genetic components that contribute to the efficacy and toxicity of drugs, across a wide spectrum of agents. We have proposed that using an alternative model population approach will provide many underlying mechanisms and pathways that are implicated in drug activity and responses. We have developed a platform of both in-vitro and in-vivo assays from genetically well defined mouse strains which will enable us to assess effects of toxicity and efficacy of current and novel agents in drug therapies. Here, we present results obtained using a new strategy of cellular genetics to identify genes and gene pathways that underlie susceptibility to cellular-level adverse drug reaction and toxicities. We have the ability to pinpoint genetic components that contribute to, or underlie, the toxicity phenotypes. We have screened 75 toxicants/drugs in a high-content imaging screen that determines changes in specific cell-health status phenotypes (nuclear changes, membrane permeability, mitochondrial membrane potential and apoptosis). Primary cell lines from 32 inbred mouse strains were phenotyped and the drug responses were used in genome-wide association analyses to identify loci that underlie the toxicity traits. We identify a number of potential candidate genes for specific drug toxicity phenotypes and provide validation of targets using over-expression and siRNA knockdown of specific gene targets. Genes were selected and prioritized for validation based on network analysis and expression in mouse embryonic fibroblasts. In addition, we identify broader cross-drug toxicity pathways. Gene-set enrichment analysis of the candidate loci revealed an overrepresentation of genes involved in cell-cycle regulation. 179 ABSTRACT 141 LESSONS FROM PHENOTYPE OF KO MOUSE, THE ROLE OF SNUR4 ON ADIPOGENESIS AND INSULIN SENSITIVITY Jae Hoon Shin1, Il Yong Kim1, Yo Na Kim1,2, Ji Won Choi1, Kyung Jin Roh1, Mi Ra Sohn4, Cheol soo Choi2,3, Yun Soo Bae4, and Je Kyung Seong1 1 Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Seoul National University, Seoul, Korea 2 Lee Gil Ya Cancer and Diabetes Institute, 3Division of Endocrinology Gil Medical Center, Gachon University of Medicine and Science, Incheon 4 Laboratory of Molecular Biochemistry Division of Molecular Life Sciences Ewha Woman’s University, Seoul, Korea Recently we developed SNUR4 KO mice. The body weight of SNUR4 KO mice were decreased with growth retardation due to decreased abdominal body fat. To validate the anti-obesity role of SNUR4, HFD (High fat diet) and LFD (Low fat diet) were fed for 12 weeks to SNUR4 knock-out (KO) mice (-/-), hetero KO (+/-), and wild-type mice. The body weight of HFD-fed SNUR4 KO mice showed significantly reduced with the rate of weight gain compared to HFD-fed wild-type mice. The ratio of epididymal fat weight to body weight and adipocyte size significantly decreased in HFD-fed SNUR4 KO compared with the HFD-fed wild-type mice. However, the fatty liver change was observed both two groups. Also the expression level of transcriptional factors and enzymes leading to lipogenesis including Acrp30, aP2, PPARr, FAS, and SREBP was decreased in adipose tissue and liver from HFD-fed SNUR4 KO mice. Next, we test metabolic phenotyping using mouse metabolic phenotype system to identify the cause of resistance to diet-induced obesity in SNUR4 KO mice. To determine metabolic rate in SNUR4 KO, we measured oxygen consumption (VO2), respiratory exchange ratio (RER) and energy expenditure. There are no differences between SNUR4 KO and wild-type mice in RER and energy expenditure. Locomotor activity and food intake were also similar between two groups. We performed hyperinsulinemic-euglycemic clamps to determine insulin dependent glucose uptake in SNUR4 KO mice. The glucose infusion rate was increased in SNUR4 KO mice than wild-type mice. Hepatic glucose output was decreased in SNUR4 KO mice than wild-type mice in clamped condition but no difference was detected in basal condition. SNUR4 KO mice were found to take up significantly more glucose and this glucose into glycolysis and glycogen synthesis. Our finding suggests that SNUR4 KO mice are resistance to diet-induced obesity in adipose tissue. Also, insulin-stimulated glucose uptake and metabolism were more sensitive in SNUR4 KO mice than wild-type mice. 180 ABSTRACT 142 EPIBLAST-ABLATION OF SOX2 EXPRESSION LEADS TO NEUROCRISTOPATHIES IN MOUSE EMBRYOS Nikolaos Mandalos1,*, Marannia Saridaki1*, Jessica Lea Harper1, Anastasia Kotsoni1, Aris N. Economides2, and Eumorphia Remboutsika1 1 Stem Cell Biology Laboratory, Institute of Molecular Biology and Genetics, Biomedical Sciences Research Centre “Alexander Fleming”, Vari, Greece 2 Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA *equal contribution Sox2 is a single exon gene essential for the maintenance of pluripotency, the development of the epiblast and trachea, lung and nervous system development and homeostasis. Precise genetic manipulations in the Sox2 locus to alter the expression of Sox2 are essential due (a) to the fact that the proximal promoter and the coding region of the Sox2 gene are embedded in a CpG island and (b) to the presence of a miRNA (mmu-mir1897) and another non-coding RNA transcript (Sox2ot) in the region. Here, we describe the ablation of Sox2 in the epiblast using a novel Conditional by Inversion Sox2 allele (Sox2COIN). Sox2COIN/COIN mice are normal and fertile and epiblast-specific inversion of the Sox2COIN allele using Sox2-Cre generates normal heterozygote animals (Sox2INV/+:TgSox2CRE). Sox2INV/+ intercrosses generate a true Sox2 null allele as homozygote mice (Sox2INV/INV) are not viable and embryos die around implantation. Sox2INV/mosaic embryos die around E11 and exhibit a range craniofacial and heart defects as well as abnormal growth structures in the central nervous system. These defects are described in human conditions known as neurocristopathies and have been attributed to defects in neural crest development. Our results demonstrate that COIN is a method of choice for precise manipulations of single exon genes and reveals a novel role for Sox2 in neurocristopathies. 181 ABSTRACT 143 A COMPREHENSIVE CATALOGUE OF STRUCTURAL VARIATION FROM 17 INBRED MOUSE STRAINS Kim Wong1,$ $B Yalcin2,$ $T Keane1,$ $M Goodson2,$ $A Agam2,$ $C Nellaker2,$ $J Stalker1,$ $J Flint2,$ $and D Adams1 1 The Wellcome Trust Sanger Institute,$ $Hinxton,$ $Cambridgeshire,$ $UK 1 Wellcome Trust Centre for Human Genetics,$ $Oxford,$ $UK The genomes of 17 key inbred mouse strains have been sequenced by the Mouse Genomes Project (http://www.sanger.ac.uk/mousegenomes/ ) using next-generation sequencing technology. The strains, which include 4 wildderived strains (CAST/EiJ, Spretus/EiJ, PWK/PhJ and WSB/EiJ ) and 13 laboratory strains (129P2/O1a, 129S1/SvImJ, 129S5, A/J, AKR/J, BALB/cJ, C3H/HeJ, C57BL/6NJ, CBA/J, DBA/2J, LP/J, OD/ShiLtJ, NZO/HiLtJ ), were sequenced to 20-35x depth. This has enabled us to compare all 17 strains to the reference genome to identify a highly confident set of large (>100bp) structural variants (SVs ) such as insertions, deletions, inversions, and copy number gains. Currently, several methods are available to identify SVs from mapped read data, however, independently, none are able to identify all types and sizes of SVs. We have developed a pipeline, SVMerge, which enabled us to utilize a suite of software tools to generate a more comprehensive set of SVs. We have computationally validated each SV by performing local assembly, which also enabled better localization of the breakpoints. We have performed experimental validation on each class of automated calls to evaluate the false discovery rates, and performed manual SV annotation to determine false negative rates. In total, we have identified a non-redundant catalogue of over 400,000 SVs among the 17 strains. The majority of SVs in the laboratory strains are shared with at least one other strain. The wild-derived strains, however, harbor a greater number of unique SVs and more genes are disrupted by SVs. Deletions, for example, disrupt approximately 200 protein-coding genes in each laboratory strain, and up to 700 genes in the wild-derived strains. Finally, by observing the strain distribution patterns of these SV events and correlating them with inter-strain gene expression differences, we have been able to determine the potential phenotypic consequences of individual SVs. This is the most comprehensive collection of large, fine-scale mouse SVs to date, and together with the complete assemblies, SNPs, and short indels, will be a valuable resource for the mouse genetics community. 182 ABSTRACT 144 COLLECTION AND DISTRIBUTION OF CRE/FLP-DRIVERS AT THE RIKEN BIORESOURCE CENTER Atsushi Yoshiki, Kazuyuki Mekada, Hatsumi Nakata, Ayumi Murakami, Masayo Kadota, Fumio Ike, Noriko Hiraiwa, Kaoru Fukami-Kobayashi, Yuichi Obata RIKEN BioResource Center, Tsukuba, Japan The RIKEN Bioresource Center (RBRC) is the core facility for mouse resources in Japan under the National BioResource Project (NBRP, http://www.nbrp.jp) of the Japanese Ministry of Education, Culture, Sports, Science, and Technology, and is also a member of the International Coordination Group of the CREATE consortium (http://www.creline.org). RBRC collects, develops, and distributes useful Cre/Flp-drivers for functional genomics studies in collaboration with the international scientific community. Currently, the characterization of the tissue specific expression of these drivers and the associated phenotype data is a challenging issue that will be used to enrich the value of the Cre/Flp-drivers. RBRC has so far collected 73 Cre-drivers and 4 Flp-drivers, and has distributed these lines worldwide via our website (http://www.brc.riken.jp/lab/animal/en), the International Mouse Strain Resource (IMSR, http://www.findmice.org), and Biomart (http://www.creline.org/search_cre_mice). The 73 Cre-drivers consist of 59 transgenic and 14 targeted knock-in mice. The most popular lines include deleter mice that are used in crosses with conditional knockout mice such as B6Tg(CAG-Cre)CZ-MO2Osb and B6-Tg(CAG-FLPe), and other tissue specific Cre-drivers such as Emx1-Cre, Mesp1-Cre, Nestin-Cre, and Neurog3-Cre. In addition, we are developing new Cre-drivers with different promoters, such as Ins1, Krt14, Tagln, Tek/Tie2, Vil1, and Wap, as well as fluorescent reporters using transgenesis and knock-in methods on the C57BL/6NCrlj background. In this study, we report the updated list of these Cre/Flp-drivers with data for their tissue-specificity and the status of our development program for Cre-drivers and reporters at RBRC. 183 ABSTRACT 145 ANALYSIS OF THE EXPRESSION PROFILE OF CRH-POMC SYSTEM GENES IN VITILIGO SKIN BIOPSIES Ene Reimann1,2,, K Kingo2, M Karelson2, T Salum3, E Aunin1, P Reemann1,2, K Abram2, E Vasar1, 4, H Silm2, and S Kõks1,4,5 1 University of Tartu, Estonia, Department of Physiology 2 University of Tartu, Estonia, Clinic of Dermatology 3 Tartu University Hospital, United Laboratories, Estonia 4 University of Tartu, Estonia, Centre of Translational Medicine 5 Estonian University of Life Sciences, Estonia, Institute of Veterinary Medicine and Animal Sciences Background/Aims: The corticotrophin-releasing hormone-proopiomelanocortin (CRH-POMC) system coordinates stress response and melanogenesis in skin. Potential role of this system in the pathogenesis of vitiligo is not clear. This study analyzes the expression of genes related to the CRH-POMC system, including melanin-concentrating hormone (PMCH), endogenous opioids and their receptors. Methods: With quantitative real-time-PCR the mRNA expression of twelve genes were measured in samples extracted from skin biopsies of vitiligo patients and healthy controls. Results: The mRNA expression level of prepronociceptin (PNOC) and its receptor opioid receptor like 1 (OPRL1) was higher in vitiligo involved skin compared to healthy controls. The expression level of (PMCH) has increased in uninvolved and involved vitiligo skin; melanin concentrating hormone receptor 1 (MCHR1) expression was higher in uninvolved and lower in involved vitiligo skin compared to healthy control skin. Conclusions: It was found that in addition to CRH-POMC system genes, the expression of some genes from related systems (MCH and PNOC system) is also altered in the skin of vitiligo patients and both MCH and endogenous opioids derived from PNOC gene may participate in the pathogenesis of vitiligo. Our study supports the focal role of CRH-POMC-MCH system in the skin in the maintenance of skin homeostasis. 184 ABSTRACT 146 EFFECTS OF A HIGH SPONTANEOUS MUTATION RATE IN MAMMALIAN GERMLINE BY USING MUTATOR MICE MODIFIED REPLICATIVE DNA POLYMERASE DELTA Arikuni Uchimura1, Yuko Hidaka1, Ikuo Miura2, Shigeharu Wakana2, and Takeshi Yagi1 1 Graduate School of Frontier Biosciences, Osaka University, 2Technology and Development Team for Mouse Phenotype Analysis, Japan Mouse Clinic, RIKEN BioResource Center Germline mutation rate is a very important parameter in mammalian genetics and evolutionary biology. Arising frequent mutations cause an increase in patients suffered from various congenital diseases and a decrease in mean fitness among population. Although a spontaneous deleterious mutation rate is known to be tremendously high in a mammalian germline, we know little about what effect is caused by such a frequent mutation through several generations. To investigate this, we established a new experimental model. In this model, we generated a mutator mouse strain on a C57BL/6 background, in which the spontaneous mutation rate was increased by modifying a replicative DNA polymerase ƒÂ with a gene targeting method. The mutator mice were then bred by mating between siblings for several generations, producing several mutation accumulation mouse lines (MA lines). In these MA lines, after 4 years breeding we found several phenotypic changes, a decrease in reproductive ability, an increase in the number of mice suffered from severe hydrocephalous, and sudden death without visible defects. In addition, we also found some phenotypic variations and several heritable abnormal phenotypes in each MA line. The heritable abnormal phenotypes included a short leg, reduced toe number and a chirping behavior (of which mutants emitted clearly audible sounds in the human hearing frequency range like a songbird). Our novel experimental model could provide a clue to understand relationships between the germline mutation rate and mammalian evolution and a risk of high mutation rate for mammalian prosperity. 185 ABSTRACT 147 TOOLING UP MGI FOR A DELUGE IN MOUSE PHENOTYPE AND DISEASE MODEL DATA Anna Anagnostopoulos, Janan Eppig, and Mouse Genome Informatics Group The Jackson Laboratory, Bar Harbor, ME, USA Global efforts to functionally annotate the mouse genome through large-scale mutagenesis and phenotyping consortia have catapulted the size and diversity of new mouse resources aimed at building predictive models of human disease. The Mouse Genome Informatics resource (MGI, www.informatics.jax.org) for the genetics, genomics and biology of the laboratory mouse grants free access to current integrated biological knowledge spanning from sequence to phenotype and disease model information to unified data on worldwide mouse resource holdings via IMSR. MGI curates aberrant mouse phenotypes in the context of mutations, strain variations, QTLs and complex traits shaped by intricate epigenome-environment networks. To support the data deluge fueled by ongoing translational research, MGI has devised automated processes to gradually incorporate all multipurpose alleles generated by various IKMC partners, and built a Recombinase Data Portal (www.creportal.org) to provide expression and specificity data for each cre-containing transgene and knock-in allele, aiming to integrate phenotype characterization of newly generated mutant mouse lines as available. MGI users can access mouse phenotypes, alleles and disease models of preclinical value using a suite of tools, including an enhanced Quick Search Tool, Mouse GBrowse, web-based vocabulary browsers and data-specific query forms. Robust search inputs include standardized terms from the Mammalian Phenotype Ontology, a widely adopted ontological model that enables phenotype annotations to background-specific allelic genotypes at varying degrees of granularity. Use of human disease (OMIM) terms serves to harness associations between phenodeviant mouse features and orthologous human gene mutations or disease syndromes for which defined mouse genotypes model the human condition. We will review MGI phenotype viewing options, customized retrieval of complex phenogenomic datasets and disease model mining from various perspectives. Supported by NIH grant HG000330 186 Appendix 1 BIOINFORMATICS WORKSHOPS Co-Organizers: Carol Bult, Ph.D. (The Jackson Laboratory, Bar Harbor, ME, USA) Deanna Church, Ph.D. (National Center for Biotechnology Information, Bethesda, MD, USA) Special Guest Organizer for 2010: Valerie Schneider, Ph.D. (National Center for Biotechnology Information, Bethesda, MD, USA) Tutorial 1 Room “Polymnia” Getting the Most from the Mouse Reference Genome Assembly Instructor: Valerie Schneider, Ph.D. (National Center for Biotechnology Information, Bethesda, Maryland, USA) The mouse reference assembly is among the highest quality genomes ever sequenced, and plays diverse roles in research. In 2007, the Genome Reference Consortium (GRC) assumed responsibility for the mouse reference genome and is engaged in ongoing efforts to update and improve this assembly. This workshop is designed to provide attendees with the knowledge and tools needed to make the most of this valuable and dynamic resource. Topics covered in this workshop will include: • • • • • • • Creating the Mouse Genome Assembly Sequencing Technologies and Assembly Methods Assessing Assemblies Assembly Management Updates to the Reference Assembly Reporting Problems with the Assembly Tools for Accessing the Assembly 187 Tutorial 2 Room “Erato” Mouse Phenome Database Instructor: Molly Bogue, Ph.D. (The Jackson Laboratory, Bar Harbor, Maine, USA) The Mouse Phenome Database (MPD; phenome.jax.org), maintained at The Jackson Laboratory, is the product of an international community effort to collect quantitative phenotypic and genotypic data on laboratory mouse strains. Since last year, MPD has added several comprehensive datasets, including autism-relevant behaviors, toxicogenetic analysis of drug-induced liver injury, susceptibility to alcohol intoxication, aging-related phenotypes, reproductive parameters, cardiovascular responses to beta-adrenergic blockade or activation, adiposity, and vestibular and eye parameters. MPD is also in the process of integrating gene expression datasets, which are eligible for analysis with MPD tools. MPD is indispensable for helping researchers select optimal strains for many research applications. MPD contains: • Data for over 600 strains of mice (inbred, RI, consomic, etc.) • Hundreds of baseline measurements of biomedically-relevant phenotypes • A growing collection of data from treated mice, e.g., drugs; carcinogenic or toxic compounds; high-fat diet • Detailed protocols and environmental conditions of the test animals • New SNP datasets (more strains and more genomic locations) • Gene expression microarray data • Pathology survey of aged inbred strains of mice Topics covered in this workshop will include: • Introduce the Mouse Phenome Project and MPD • Find data sets related to specific areas of biology • Find all data available in MPD for a particular strain • Find details about a project submitted to MPD • Perform basic statistical analysis on MPD data sets • Combine phenotype measurement data with large-scale SNP data • Compare SNPs across multiple strains • Query SNPs based on MGI objects (genes, MIT markers, microRNAs, and QTLs) • Search for phenotype correlations across the entire MPD • Create a “shopping cart” of user-defined data for custom analysis • Download data from MPD • Making suggestions to MPD 188 Tutorial 3 Room “Kalia” A Tour of Mammalian Genome Annotation Instructors: Laurens Wilming, Ph.D. & Mark Thomas, Ph.D. (Havana group, Wellcome Trust Sanger Institute, Hinxton, UK) A reference gene set of accurately, detailed and comprehensively mapped loci with associated features is crucial for genomic and genetic research. The Wellcome Trust Sanger Institute is involved in producing reference gene sets in human and mouse that includes VEGA, Ensembl and CCDS. The human genome is annotated and analysed as part of the ENCODE (encyclopedia of DNA elements) consortium. As part of the International Knock-out Mouse Consortium (IKMC) we provide the annotation for the mouse knock-out projects, together with collaborators at Washington University and the University of Manitoba. We also lead the Mouse Genomes Project, sequencing and analysing the genomes of 17 laboratory mouse strains that differ with varying degrees from the reference C57BL/6J strain. In this workshop we will present the following: • • • • Explanation of annotation, biotypes and Ensembl-VEGA gene merges, specifically in the context of the IKMC, ENCODE, CCDS and community annotation projects Accessing genome annotation using genome browsers (VEGA/Ensembl and UCSC) and other web resources Identification of knock-out mouse designs and associated resources using genome browsers and the IKMC web portal Walk-in clinic, help desk or Q&A: interactive sessions helping delegates improve genome annotation and answer their annotation questions. Delegates are encouraged to bring us their annotation queries for discussion during the “walk-in clinic” session or anytime during the conference or afterwards. 189 PARTICIPANTS 59 45 101 60 42,147 22 61 62 37,63,133 64 65,66,67 107 86 19 28 68 30 6 58 32 69 70 71 56 36 99 98 7 72 73 90 74 Acosta, Deanna - Albert Einstein College of Medicine, USA Adams, Niels - Mary Lyon Center, UK Agami, Reuven - The Netherlands Cancer Institute, Netherlands Aidinis, Vassilis - BSRC Alexander Fleming, Greece Al-Mahdawi, Sahar - Brunel University, UK Amato, Roberto - University of Naples Federico II, Italy Anagnostopoulos, Anna - The Jackson Laboratory, USA Angel, Joe - The University of Texas, USA Arkell, Ruth - Early Mammalian Development, Australia Ball, Simon - Medical Research Council, UK Balling , Rudi - Luxembourg Center for Systems Biomedicine, Luxembourg Bauer, Beth - University of Missouri, USA Beier, David - Harvard Medical School, USA Benavides, Fernando - M.D. Anderson Cancer Center, USA Bergstrom, David - The Jackson Laboratory, USA Birling, Marie-Christine - ICS, France Boivin, Gregory - McGill University, Canada Brodnicki, Thomas - St. Vincents Institute, Australia Brown, Steve - Medical Research Council (MRC), UK Bryda, Elizabeth - University of Missouri, USA Bucan, Maja - Univ. of Pennsylvania, USA Cabin, Deborah - McLaughlin Research Institute, USA Calaway, John - University of North Carolina, USA Carlson, George - McLaughlin Research Institute, USA Charizopoulou, Nikoletta - NIDCD/NIH, USA Chen, Yen-Hui - Taiwan Mouse Clinic, Taiwan Clare, Simon - Sanger Institute, UK Connolly, Diana - Albert Einstein College of Med. of Yeshiva University, USA Copeland, Neal - Inst. Of Molecular and Cell Biology, Singapore Croniger, Colleen - Case Western Reserve University, USA Cross, Sally - MRC Human Genetics Unit, UK Danecek, Petr - Wellcome Trust Sanger Institute, UK Das, Sabyasachi - Emory University, USA de la Casa Esperon, Elena - University of Castilla-La Mancha, ES Didion, John - Univ. of North Carolina, USA Doerner, Stephanie - Case Western Reserve University, USA Donahue, Leah Rae - The Jackson Laboratory, USA Douni, Eleni - BSRC Alexander Fleming, Greece Eaton, Sally - Medical Research Council, UK 190 42 76 47 75 53 77 52 78 26 82 79 48 23 57 100 39 55 13 9 87 89 92 97 16 96 15 3 91 93 Economides, Aris - Regeneron Pharmaceuticals Inc., USA Ekker, Marc - University of Ottawa, Canada Elliott, Rosemary - Roswell Park Cancer Inst, USA Engel, Nora - Temple University School of Medicine, USA Estabel, Jeanne - Wellcome Trust Sanger Institute, UK Foote, Simon - Menzies Research Inst., Australia Forejt, Jiri - Instututre of Molecular Genetics AS CR, CZ Fukumura, Ryutaro - RIKEN BRC, Japan Galas, David - Inst. For Systems Bio, USA Gerdin, Anna-Karin - Wellcome Trust Sanger Institute, UK Goggolidou, Paraskevi - Medical Research Council, UK Gondo, Yoichi - RIKEN, Japan Gunn, Teresa - McLaughlin Research Institute, USA Hagn, Michael - Helmholtz Zentrum München, Germany Hassan, Ahmad - Medical Research Council (MRC), UK Heaney, Jason - Case Wester Reserve University, USA Hentges, Kathryn - The University of Manchester, UK Herman, Gail - The Ohio State University, USA Horvat, Simon - University of Ljubljana, Animal Science, Slovenia Houri-Haddad, Yael - Boston University, USA Hrabe de Angelis, Martin - Helmholtz Zentrum München, Germany Humphrey, Neil - The Univ. of Manchester, UK Hunter, Kent - National Cancer Institute, USA Ioakeimidis, Fotios - BSRC "Alexander Fleming", Greece Iraqi, Fuad - Tel Aviv University, Israel Ishida, Saeko - Kyoto University, Japan Ivanov, Andrey - GEOKHI RAS, Russia Jackson, Ian - MRC Human Genetics Unit, UK Jaubert, Jean - Institut Pasteur, France Jenkins, Nancy - Indt. Of Molecular and Cell Biology, Singapore Johansen, Jeanette - Karolinska Institutet, Sweden Kaerst, Stefan - Humboldt-Universität zu Berlin, Germany Kane, Leanne - Wellcome Trust Sanger Inst., UK Karalis, Katia - BRFAA Greece and Childrens Hospital, Greece Keane, Thomas - Wellcome Trust Sanger Institute, UK Keegan, Catherine - Pediatrics and Human Genetics, USA Kember, Rachel - SGDP, UK Khrameeva, Ekaterina - Moscow State University, Russia Koide, Tsuyoshi - Mouse Genomics Resource Laboratory, Japan Kong, Philip - Kennedy Institute, UK Korthaus, Dirk - Takeda SingaporePte Ltd., Singapore 191 33 83 43 95 102 118 119 14 121 18 85 54 110 112 117 17 4 111 35105 106 11 115 104 51 24 46 94 108,109 38 114 145 116 Kuramoto, Takashi - Kyoto University, Japan Lad, Heena - MRC Harwell, UK Leake, John - Rady Children's Hospital San Diego, USA LeBoeuf, Renee - University of Washington, USA Lewis, Morag - Wellcome Trust Sanger Institute, UK Libert, Claude - Ghent University, Belgium Lilleoja, Rutt - University of Tartu, EE Lindfors, Charlotte - Molecular Medicine and Surgery, Sweden Lo , Chiao-Ling - Purdue University, USA Logan, Darren - Wellcome Trust Sanger Institute, UK Lossie, Amy - Purdue University, USA Lucas, Mark - Sanger Institute, UK Maddatu, Terry - The Jackson Laboratory, USA Marton, Jennifer - McGill University, Canada Mashimo, Tomoji - Kyoto University, Japan Masuya, Hiroshi - RIKEN, Japan Matin, Angabin - MD Anderson Cancer Center, USA Mburu, Philomena - MRC Harwell, UK Mekada, Kazuyuki - RIKEN BioResource Center, Japan Miller, Darla - UNC at Chapel Hill, USA Mock, Beverly - NCI, NIH, USA Mohammadi, Amir - The Australian National University, Australia Montagutelli, Xavier - Institut Pasteur, France Murata, Takuya - RIKEN BRC, Japan Murray, Stephen - The Jackson Laboratory, USA Nadeau, Joseph - Inst. For Systems Bio, USA Neuhold, Lisa - National Eye Inst./NIH/HHS, USA Ning , Li - BGI Shenzhen, China Niti, Alexandra - BSRC Alexander Fleming, Greece Noben-Trauth, Konrad - National Institutes of Health, USA Oota, Satoshi - RIKEN, Japan Osawa, Gail - University of Michigan, USA Pardo Manuel De Villena, Fernando - Univ. of North Carolina, USA Parsons, Michael - MRC Harwell, UK Peters, Jo - MRC Harwell, UK Pook, Mark - Brunel University, UK Potter, Paul - Medical Research Council, UK Ramirez-Solis, Ramiro - Wellcome Trust Sanger Insti, UK Reilly, Karlyne - NCI-Frederick, USA Reimann, Ene - Tartu, Estonia, Estonia Reinholdt, Laura - The Jackson Laboratory, USA 192 113 120 122,123 1 138 40 130 5 132 21 125 8 12 124 126 31 49 127 84 34 27 146 10 139 25 44 136 20 135 2 Ripoll, Vera - Medical Research Council, UK Rowe, Lucy - The Jackson Laboratory, USA Ryder, Edward - Sanger Institute, UK Schneider, Valerie - DHHS/NIH/NLM/NCBI, USA Schughart, Klaus - Helmholtz Centre for Infection Research, Germany shusterman, ariel - Department of Prosthodontics, IL Simon, Michelle - Medical Research Council, UK Skarnes, William - The Wellcome Trust Sanger, UK Slater, Guy - Wellcome Trust Sanger Institute, UK Smith, Rebecca - Kings College London, Institute of Psychiatry, UK Snaith, Mike - Medimmune Ltd, UK Snoek, Margriet - Netherlands Cancer Institute, NL Sorg, Tania - ICS, France Spiezio, Sabrina - Case Western Reserve Univ., USA Stamova, Boryana - University of California, USA Steel, Karen - Wellcome Trust Sanger Institute, UK Stephen, Louise - University of Manchester, UK Su, Zhaoliang - China Sunter, David - Wellcome Trust Sanger Institute, UK Suzuki, Tomohiro - RIKEN, Japan Svenson, Karen - The Jackson Laboratory, USA Szalai, Gabor - University of South Carolina, USA Takada, Toyoyuki - National Institute of Genetics, Japan Tanaka, Nobuhiko - RIKEN BRC, Japan Tateossian, Hilda - MRC Harwell, UK Thomas, Mark - Wellcome Trust Sanger Institute, UK Threadgill, David - North Carolina State Univ., USA Tinsley, Louise - Medical Research Council (MRC), UK Traka, Maria - The University of Chicago, USA Uchimura, Arikuni - Osaka University, Japan Van Schaick, Jessica - The George Washington University, USA Vancollie, Valerie - Wellcome Trust Sanger Inst., UK Vlangos, Christopher - University of Michigan, USA WAKANA, Shigeharu - RIKEN BRC, Japan Wakeland, Edward - UT Southwestern Medical Center, USA Wallace, Marsha - Cornell University, USA Weiss, Ervin - The Hebrew University, Israel Westrick, Randal - University of Michigan, USA White, Jacqul - Wellcome Trust Sanger Inst., UK Wilming, Laurens - Wellcome Trust Sanger Institute, UK Wiltshire, Sean - McGill University, Canada 193 29,140 143 134 131 80,81 129 128 50 Wiltshire, Tim - University of North Carolina, USA Wong, Kim - Wellcome Trust Sanger Institute, UK Wu, Jiaqian - Stanford University, USA Xuengang, Wang - BGI Shenzhen, China Yagi, Takeshi - Osaka University, Japan Yang-Yen, Hsin-Fang - Academia Sinica, Taiwan Yann, Herault - ICS/IGBMC-TAAM, France Yen, Jeffrey - Institute of Biomedical Sciences, Taiwan Yu, Eugene - Roswell Park Cancer Institute, USA Yuan, Rong - The Jackson Laboratory, USA 194 SPONSORS Aegean Conferences and the IMGS wish to gratefully acknowledge the following sponsors for their generous support of this meeting. Funding for student scholarships was made possible by: 2R13HG002394 from the following ICs at NIH: NHGRI, NIMH, NICHD, NIAID, NIEHS, NCRR, AND NINDS BGI SHENZHEN Cambridge Journals Genomics Genesis Genetics Society of America Genome Research Mammalian Genome The Ellison Medical Foundation 195 UPCOMING EVENTS 4th International Conference on Tissue Engineering Minoa Palace Conference Center, May 27-June 1, 2011 6th Leukocyte Signal Transduction Workshop Minoa Palace Conference Center, June 1-5, 2011 9th International Conference Pathways, Networks, and Systems Medicine Minoa Palace Conference Center, June 5-10, 2011 8th International Conference Innate Immunity Minoa Palace Conference Center, June 15-20, 2011 5th International Workshop on Complement Associated Diseases, Animal Models, and Therapeutics Sheraton Conference Center, Rhodes, Greece June 22-27, 2011 3rd International Summit on Hurricanes and Climate Change Sheraton Conference Center, Ixia, Rhodes, Greece June 27-July 2, 2011 4th International Conference on Crossroads between Innate and Adaptive Immunity Royal Myconian Hotel. Chania, Crete, Greece, Sept. 11-16, 2011 1st International Conference on Immunometabolism: Molecular and Cellular Immunology of Metabolism Aldemar Knossos Royal Village, Crete, Greece, Sept 18-23, 2011 5th International Conference on Autoimmunity: Mechanisms and Novel Treatments Aldemar Knossos Royal Village, Crete, Greece, Sept 18-23, 2011 196 CONFERENCE CENTER MAP 197 ALDEMAR HOTEL MAP 198 Aegean Conference Proceedings 199 To purchase using the Aegean Conferences discount download the form: http://www.aegeanconferences.org/Publications/AegeanConferencesFlyer.pdf 200