901
|
Zillikens MC, Demissie S, Hsu YH, Yerges-Armstrong LM, Chou WC, Stolk L, Livshits G, Broer L, Johnson T, Koller DL, Kutalik Z, Luan J, Malkin I, Ried JS, Smith AV, Thorleifsson G, Vandenput L, Hua Zhao J, Zhang W, Aghdassi A, Åkesson K, Amin N, Baier LJ, Barroso I, Bennett DA, Bertram L, Biffar R, Bochud M, Boehnke M, Borecki IB, Buchman AS, Byberg L, Campbell H, Campos Obanda N, Cauley JA, Cawthon PM, Cederberg H, Chen Z, Cho NH, Jin Choi H, Claussnitzer M, Collins F, Cummings SR, De Jager PL, Demuth I, Dhonukshe-Rutten RAM, Diatchenko L, Eiriksdottir G, Enneman AW, Erdos M, Eriksson JG, Eriksson J, Estrada K, Evans DS, Feitosa MF, Fu M, Garcia M, Gieger C, Girke T, Glazer NL, Grallert H, Grewal J, Han BG, Hanson RL, Hayward C, Hofman A, Hoffman EP, Homuth G, Hsueh WC, Hubal MJ, Hubbard A, Huffman KM, Husted LB, Illig T, Ingelsson E, Ittermann T, Jansson JO, Jordan JM, Jula A, Karlsson M, Khaw KT, Kilpeläinen TO, Klopp N, Kloth JSL, Koistinen HA, Kraus WE, Kritchevsky S, Kuulasmaa T, Kuusisto J, Laakso M, Lahti J, Lang T, Langdahl BL, Launer LJ, Lee JY, Lerch MM, Lewis JR, Lind L, Lindgren C, Liu Y, et alZillikens MC, Demissie S, Hsu YH, Yerges-Armstrong LM, Chou WC, Stolk L, Livshits G, Broer L, Johnson T, Koller DL, Kutalik Z, Luan J, Malkin I, Ried JS, Smith AV, Thorleifsson G, Vandenput L, Hua Zhao J, Zhang W, Aghdassi A, Åkesson K, Amin N, Baier LJ, Barroso I, Bennett DA, Bertram L, Biffar R, Bochud M, Boehnke M, Borecki IB, Buchman AS, Byberg L, Campbell H, Campos Obanda N, Cauley JA, Cawthon PM, Cederberg H, Chen Z, Cho NH, Jin Choi H, Claussnitzer M, Collins F, Cummings SR, De Jager PL, Demuth I, Dhonukshe-Rutten RAM, Diatchenko L, Eiriksdottir G, Enneman AW, Erdos M, Eriksson JG, Eriksson J, Estrada K, Evans DS, Feitosa MF, Fu M, Garcia M, Gieger C, Girke T, Glazer NL, Grallert H, Grewal J, Han BG, Hanson RL, Hayward C, Hofman A, Hoffman EP, Homuth G, Hsueh WC, Hubal MJ, Hubbard A, Huffman KM, Husted LB, Illig T, Ingelsson E, Ittermann T, Jansson JO, Jordan JM, Jula A, Karlsson M, Khaw KT, Kilpeläinen TO, Klopp N, Kloth JSL, Koistinen HA, Kraus WE, Kritchevsky S, Kuulasmaa T, Kuusisto J, Laakso M, Lahti J, Lang T, Langdahl BL, Launer LJ, Lee JY, Lerch MM, Lewis JR, Lind L, Lindgren C, Liu Y, Liu T, Liu Y, Ljunggren Ö, Lorentzon M, Luben RN, Maixner W, McGuigan FE, Medina-Gomez C, Meitinger T, Melhus H, Mellström D, Melov S, Michaëlsson K, Mitchell BD, Morris AP, Mosekilde L, Newman A, Nielson CM, O'Connell JR, Oostra BA, Orwoll ES, Palotie A, Parker SCJ, Peacock M, Perola M, Peters A, Polasek O, Prince RL, Räikkönen K, Ralston SH, Ripatti S, Robbins JA, Rotter JI, Rudan I, Salomaa V, Satterfield S, Schadt EE, Schipf S, Scott L, Sehmi J, Shen J, Soo Shin C, Sigurdsson G, Smith S, Soranzo N, Stančáková A, Steinhagen-Thiessen E, Streeten EA, Styrkarsdottir U, Swart KMA, Tan ST, Tarnopolsky MA, Thompson P, Thomson CA, Thorsteinsdottir U, Tikkanen E, Tranah GJ, Tuomilehto J, van Schoor NM, Verma A, Vollenweider P, Völzke H, Wactawski-Wende J, Walker M, Weedon MN, Welch R, Wichmann HE, Widen E, Williams FMK, Wilson JF, Wright NC, Xie W, Yu L, Zhou Y, Chambers JC, Döring A, van Duijn CM, Econs MJ, Gudnason V, Kooner JS, Psaty BM, Spector TD, Stefansson K, Rivadeneira F, Uitterlinden AG, Wareham NJ, Ossowski V, Waterworth D, Loos RJF, Karasik D, Harris TB, Ohlsson C, Kiel DP. Large meta-analysis of genome-wide association studies identifies five loci for lean body mass. Nat Commun 2017; 8:80. [PMID: 28724990 PMCID: PMC5517526 DOI: 10.1038/s41467-017-00031-7] [Show More Authors] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 05/02/2017] [Indexed: 12/25/2022] Open
Abstract
Lean body mass, consisting mostly of skeletal muscle, is important for healthy aging. We performed a genome-wide association study for whole body (20 cohorts of European ancestry with n = 38,292) and appendicular (arms and legs) lean body mass (n = 28,330) measured using dual energy X-ray absorptiometry or bioelectrical impedance analysis, adjusted for sex, age, height, and fat mass. Twenty-one single-nucleotide polymorphisms were significantly associated with lean body mass either genome wide (p < 5 × 10-8) or suggestively genome wide (p < 2.3 × 10-6). Replication in 63,475 (47,227 of European ancestry) individuals from 33 cohorts for whole body lean body mass and in 45,090 (42,360 of European ancestry) subjects from 25 cohorts for appendicular lean body mass was successful for five single-nucleotide polymorphisms in/near HSD17B11, VCAN, ADAMTSL3, IRS1, and FTO for total lean body mass and for three single-nucleotide polymorphisms in/near VCAN, ADAMTSL3, and IRS1 for appendicular lean body mass. Our findings provide new insight into the genetics of lean body mass.Lean body mass is a highly heritable trait and is associated with various health conditions. Here, Kiel and colleagues perform a meta-analysis of genome-wide association studies for whole body lean body mass and find five novel genetic loci to be significantly associated.
Collapse
Affiliation(s)
- M Carola Zillikens
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
| | - Serkalem Demissie
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Yi-Hsiang Hsu
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Laura M Yerges-Armstrong
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Wen-Chi Chou
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute, Cambridge, MA, 02142, USA
| | - Lisette Stolk
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
| | - Gregory Livshits
- Sackler Faculty of Medicine, Department of Anatomy and Anthropology, Tel Aviv University, Tel Aviv, 6997801, Israel
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Campus, London, WC2R 2LS, UK
| | - Linda Broer
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Toby Johnson
- Department of Medical Genetics, University of Lausanne, Lausanne, 1011, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
- Centre Hospitalier Universitaire (CHUV), University Institute for Social and Preventive Medicine, Lausanne, 1010, Switzerland
| | - Daniel L Koller
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Zoltán Kutalik
- Department of Medical Genetics, University of Lausanne, Lausanne, 1011, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
- Centre Hospitalier Universitaire (CHUV), University Institute for Social and Preventive Medicine, Lausanne, 1010, Switzerland
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
| | - Ida Malkin
- Sackler Faculty of Medicine, Department of Anatomy and Anthropology, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Janina S Ried
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | | | - Liesbeth Vandenput
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
| | - Weihua Zhang
- Department Epidemiology and Biostatistics, School of Public Health, Imperial College, London, SW7 2AZ, UK
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
| | - Ali Aghdassi
- Department of Medicine A, University of Greifswald, Greifswald, 17489, Germany
| | - Kristina Åkesson
- Department of Clinical Sciences, Lund University, Malmö, 22362, Sweden
- Department of Orthopedics, Skåne University Hospital, Malmö, S-205 02, Sweden
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Leslie J Baier
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, AZ, 85014, USA
| | - Inês Barroso
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
- NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK
- Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge Metabolic Research Laboratories, Cambridge, CB2 OQQ, UK
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Experimental & Integrative Genomics, University of Lübeck, Lübeck, 23562, Germany
- School of Public Health, Faculty of Medicine, Imperial College London, London, W6 8RP, UK
| | - Rainer Biffar
- Centre of Oral Health, Department of Prosthetic Dentistry, Gerodontology and Biomaterials, University of Greifswald, Greifswald, 17489, Germany
| | - Murielle Bochud
- Centre Hospitalier Universitaire (CHUV), University Institute for Social and Preventive Medicine, Lausanne, 1010, Switzerland
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ingrid B Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St Louis, MO, 63110, USA
- Division of Biostatistics, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Aron S Buchman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Liisa Byberg
- Department of Surgical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Harry Campbell
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, EH8 9AG, UK
| | | | - Jane A Cauley
- Department of Epidemiology Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Peggy M Cawthon
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| | - Henna Cederberg
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Zhao Chen
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, 85714, USA
| | - Nam H Cho
- Department of Preventive Medicine, Ajou University School of Medicine, Youngtong-Gu, Suwon, 16499, Korea
| | - Hyung Jin Choi
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju Si, Korea
| | - Melina Claussnitzer
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute, Cambridge, MA, 02142, USA
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, 02139, USA
- Institute of Human Genetics, MRI, Technische Universität München, Munich, 81675, Germany
- Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Francis Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, Bethesda, MD, 20892, USA
| | - Steven R Cummings
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| | - Philip L De Jager
- Harvard Medical School, Boston, MA, 02115, USA
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, 02142, USA
| | - Ilja Demuth
- Lipid Clinic at the Interdisciplinary Metabolism Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 13353, Germany
- Institute of Medical and Human Genetics, Charité - Universitätsmedizin Berlin, Berlin, 13353, Germany
| | | | - Luda Diatchenko
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, H3A 0G1, Canada
- Regional Center for Neurosensory Disorders, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | | | - Anke W Enneman
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Mike Erdos
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, Bethesda, MD, 20892, USA
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, 00014, Finland
- Unit of General Practice, Helsinki University Central Hospital, Helsinki, 00014, Finland
- Folkhalsan Research Centre, Helsinki, 00250, Finland
- Vasa Central Hospital, Vasa, 65130, Finland
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Joel Eriksson
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Karol Estrada
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Daniel S Evans
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Mao Fu
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Melissa Garcia
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute for Aging, Bethesda, MD, 20892, USA
| | - Christian Gieger
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Thomas Girke
- Institute for Integrative Genome Biology, University of California, Riverside, CA, 92521, USA
- Department of Botany and Plant Sciences, University of California, Riverside, CA, 92521, USA
| | - Nicole L Glazer
- Departments of Medicine and Epidemiology, Boston University School of Medicine and Public Health, Boston, MA, 02118, USA
| | - Harald Grallert
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Department of Botany and Plant Sciences, University of California, Riverside, CA, 92521, USA
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- CCG Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, 85764, Germany
- CCG Nutrigenomics and Type 2 Diabetes. Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Jagvir Grewal
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Bok-Ghee Han
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, 28159, Korea
| | - Robert L Hanson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, AZ, 85014, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, Scotland, EH4 2XU, UK
| | - Albert Hofman
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Eric P Hoffman
- Department of Pharmaceutical Sciences, SUNY Binghamton, Binghamton, NY, 13902, USA
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, 17487, Germany
| | - Wen-Chi Hsueh
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, AZ, 85014, USA
| | - Monica J Hubal
- Department of Exercise and Nutrition Sciences, George Washington University, Washington, DC, 20052, USA
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, 20052, USA
| | - Alan Hubbard
- Division of Biostatistics, School of Public Health, University of California, Berkeley, CA, 94720, USA
| | - Kim M Huffman
- Division of Rheumatology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Lise B Husted
- Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, DK 8000, Denmark
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Department of Human Genetics, Hannover Medical School, Hannover, 30625, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, 30625, Germany
| | - Erik Ingelsson
- Department of Medical Sciences, Uppsala University, Uppsala, 75185, Sweden
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Till Ittermann
- Institute for Community Medicine, University of Greifswald, Greifswald, 17489, Germany
| | - John-Olov Jansson
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE 405 30, Sweden
| | - Joanne M Jordan
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517, USA
| | - Antti Jula
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Magnus Karlsson
- Department of Clinical Sciences and Orthopaedics, Lund University, Skåne University Hospital SUS, Malmö, 22362, Sweden
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Tuomas O Kilpeläinen
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Copenhagen, 2100, Denmark
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Norman Klopp
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, 30625, Germany
| | | | - Heikki A Koistinen
- Department of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, 00029, Finland
- Endocrinology, Abdominal Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, 00029, Finland
- Department of Health, National Institute for Health and Welfare, Helsinki, 00271, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, 00290, Finland
| | - William E Kraus
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Stephen Kritchevsky
- Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Teemu Kuulasmaa
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Jari Lahti
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, FI00014, Finland
| | - Thomas Lang
- University of California San Francisco, San Francisco, CA, 94143, USA
| | - Bente L Langdahl
- Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, DK 8000, Denmark
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute for Aging, Bethesda, MD, 20892, USA
| | - Jong-Young Lee
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, 28159, Korea
| | - Markus M Lerch
- Department of Medicine A, University of Greifswald, Greifswald, 17489, Germany
| | - Joshua R Lewis
- School of Medicine and Pharmacology, University of Western Australia, Perth, 6009, Australia
- Centre for Kidney Research, School of Public Health, University of Sydney, Sydney, 2006, Australia
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Cecilia Lindgren
- Wellcome Trust Centre for Human Genetics, Oxford University, Oxford, OX3 7BN, UK
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, 27517, USA
| | - Tian Liu
- Max Planck Institute for Molecular Genetics, Berlin, 14195, Germany
- Max Planck Institute for Human Development, Berlin, 14195, Germany
| | - Youfang Liu
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517, USA
| | - Östen Ljunggren
- Department of Medical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Mattias Lorentzon
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Robert N Luben
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - William Maixner
- Regional Center for Neurosensory Disorders, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Fiona E McGuigan
- Department of Clinical Sciences, Lund University, Malmö, 22362, Sweden
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Thomas Meitinger
- Institute of Human Genetics, MRI, Technische Universität München, Munich, 81675, Germany
- Institute of Human Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Håkan Melhus
- Department of Medical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Dan Mellström
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Simon Melov
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, LA, CA, 90089, USA
| | - Karl Michaëlsson
- Department of Surgical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Braxton D Mitchell
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD, 21201, USA
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, Oxford University, Oxford, OX3 7BN, UK
- Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - Leif Mosekilde
- Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, DK 8000, Denmark
| | - Anne Newman
- Center for Aging and Population Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | | | - Jeffrey R O'Connell
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ben A Oostra
- Department of Clinical Genetics, Erasmus MC, Rotterdam, 300 CA, The Netherlands
- Centre for Medical Systems Biology and Netherlands Consortium on Healthy Aging, Leiden, RC2300, The Netherlands
| | - Eric S Orwoll
- Oregon Health & Science University, Portland, OR, 97239, USA
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, FI00014, Finland
| | - Stephen C J Parker
- Human Genetics and Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Munro Peacock
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Markus Perola
- National Institute for Health and Welfare, Helsinki, 00271, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
- Diabetes and Obesity Research Program, University of Helsinki, Helsinki, FI00014, Finland
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Ozren Polasek
- Faculty of Medicine, Department of Public Health, University of Split, Split, 21000, Croatia
| | - Richard L Prince
- School of Medicine and Pharmacology, University of Western Australia, Perth, 6009, Australia
- Department of Endocrinology and Diabetes, Sir Charles Gardiner Hospital, Perth, 6009, Australia
| | - Katri Räikkönen
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, FI00014, Finland
| | - Stuart H Ralston
- Molecular Medicine Centre, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, Scotland, EH4 2XU, UK
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
- Hjelt Institute, University of Helsinki, Helsinki, Finland
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - John A Robbins
- Department of Medicine, University of California at Davis, Sacramento, CA, 95817, USA
| | - Jerome I Rotter
- Institute for Translational Genomic and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor UCLA Medical Center, Torrance, CA, 90502, USA
| | - Igor Rudan
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, EH8 9AG, UK
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Suzanne Satterfield
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Eric E Schadt
- Department of Genetics and Genomic Science, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sabine Schipf
- Institute for Community Medicine, University of Greifswald, Greifswald, 17489, Germany
| | - Laura Scott
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joban Sehmi
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Jian Shen
- Oregon Health & Science University, Portland, OR, 97239, USA
| | - Chan Soo Shin
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Gunnar Sigurdsson
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Department of Endocrinology and Metabolism, Landspitali, The National University Hospital of Iceland, Reykjavik, 101, Iceland
| | - Shad Smith
- Center for Translational Pain Medicine, Department of Anesthiology, Duke University Medical Center, Durham, NC, 27110, USA
| | - Nicole Soranzo
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Alena Stančáková
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Elisabeth Steinhagen-Thiessen
- Lipid Clinic at the Interdisciplinary Metabolism Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 13353, Germany
| | - Elizabeth A Streeten
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Geriatric Research and Education Clinical Center (GRECC) - Veterans Administration Medical Center, Baltimore, MD, 21201, USA
| | | | - Karin M A Swart
- Department of Epidemiology and Biostatistics, and the EMGO Institute, VU University Medical Center, Amsterdam, BT1081, The Netherlands
| | - Sian-Tsung Tan
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Mark A Tarnopolsky
- Department of Medicine, McMaster University Medical Center, Hamilton, ON, Canada, L8N 3Z5
| | - Patricia Thompson
- Department of Pathology, Stony Brook School of Medicine, Stony Brook, NY, 11794, USA
| | - Cynthia A Thomson
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, 85714, USA
| | - Unnur Thorsteinsdottir
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- deCODE Genetics, Reykjavik, 101, Iceland
| | - Emmi Tikkanen
- National Institute for Health and Welfare, Helsinki, 00271, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
- Molecular Medicine Centre, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, Scotland, EH4 2XU, UK
| | - Gregory J Tranah
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| | - Jaakko Tuomilehto
- Vasa Central Hospital, Vasa, 65130, Finland
- Department of Neuroscience and Preventive Medicine, Danube-University Krems, Krems, 3500, Austria
- Diabetes Research Group, King Abdulaziz University, Jeddah, 12589, Saudi Arabia
- Dasman Diabetes Institute, Dasman, 15462, Kuwait
| | - Natasja M van Schoor
- Department of Epidemiology and Biostatistics, and the EMGO Institute, VU University Medical Center, Amsterdam, BT1081, The Netherlands
| | - Arjun Verma
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
| | - Peter Vollenweider
- Department of Medicine and Internal Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, CH-1011, Switzerland
| | - Henry Völzke
- Institute for Community Medicine, University of Greifswald, Greifswald, 17489, Germany
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
| | - Mark Walker
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Michael N Weedon
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, EX1 2LU, UK
| | - Ryan Welch
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - H-Erich Wichmann
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig-Maximilians-Universität, Munich, 81377, Germany
- Institute of Medical Statistics and Epidemiology, Technical University, Munich, 81675, Germany
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
| | - Frances M K Williams
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Campus, London, WC2R 2LS, UK
| | - James F Wilson
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, EH8 9AG, UK
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, Scotland, EH4 2XU, UK
| | - Nicole C Wright
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Weijia Xie
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, EX1 2LU, UK
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Yanhua Zhou
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - John C Chambers
- Department Epidemiology and Biostatistics, School of Public Health, Imperial College, London, SW7 2AZ, UK
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield NHS Foundation Trust and Imperial College, London, SW3 6NP, UK
- Imperial College Healthcare NHS Trust, London, W2 1NY, UK
| | - Angela Döring
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
- Centre for Medical Systems Biology and Netherlands Consortium on Healthy Aging, Leiden, RC2300, The Netherlands
| | - Michael J Econs
- Department of Medicine and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Jaspal S Kooner
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
- Imperial College Healthcare NHS Trust, London, W2 1NY, UK
| | - Bruce M Psaty
- Departments of Medicine, Epidemiology, and Health Services, Cardiovascular Health Research Unit, University of Washington, Seattle, WA, 98101, USA
- Kaiser Permanente Washington Health Research Institute, Washington, Seattle, WA, 98101, USA
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Campus, London, WC2R 2LS, UK
| | - Kari Stefansson
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- deCODE Genetics, Reykjavik, 101, Iceland
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
| | - Vicky Ossowski
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, AZ, 85014, USA
| | - Dawn Waterworth
- Medical Genetics, GlaxoSmithKline, Philadelphia, PA, 19112, USA
| | - Ruth J F Loos
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Institute of Child Health and Development, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Genetics of Obesity and Related Traits Program, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - David Karasik
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, 1311502, Israel
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute for Aging, Bethesda, MD, 20892, USA
| | - Claes Ohlsson
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Douglas P Kiel
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
- Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA.
| |
Collapse
|
902
|
Tseng F, Filev D, Chinnam RB. A mutual information based online evolving clustering approach and its applications. EVOLVING SYSTEMS 2017. [DOI: 10.1007/s12530-017-9191-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
903
|
Robinson MR, English G, Moser G, Lloyd-Jones LR, Triplett MA, Zhu Z, Nolte IM, van Vliet-Ostaptchouk JV, Snieder H, Esko T, Milani L, Mägi R, Metspalu A, Magnusson PKE, Pedersen NL, Ingelsson E, Johannesson M, Yang J, Cesarini D, Visscher PM. Genotype-covariate interaction effects and the heritability of adult body mass index. Nat Genet 2017; 49:1174-1181. [PMID: 28692066 DOI: 10.1038/ng.3912] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 06/12/2017] [Indexed: 12/18/2022]
Abstract
Obesity is a worldwide epidemic, with major health and economic costs. Here we estimate heritability for body mass index (BMI) in 172,000 sibling pairs and 150,832 unrelated individuals and explore the contribution of genotype-covariate interaction effects at common SNP loci. We find evidence for genotype-age interaction (likelihood ratio test (LRT) = 73.58, degrees of freedom (df) = 1, P = 4.83 × 10-18), which contributed 8.1% (1.4% s.e.) to BMI variation. Across eight self-reported lifestyle factors, including diet and exercise, we find genotype-environment interaction only for smoking behavior (LRT = 19.70, P = 5.03 × 10-5 and LRT = 30.80, P = 1.42 × 10-8), which contributed 4.0% (0.8% s.e.) to BMI variation. Bayesian association analysis suggests that BMI is highly polygenic, with 75% of the SNP heritability attributable to loci that each explain <0.01% of the phenotypic variance. Our findings imply that substantially larger sample sizes across ages and lifestyles are required to understand the full genetic architecture of BMI.
Collapse
Affiliation(s)
- Matthew R Robinson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.,Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Geoffrey English
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Gerhard Moser
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Luke R Lloyd-Jones
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Marcus A Triplett
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Zhihong Zhu
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jana V van Vliet-Ostaptchouk
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Tonu Esko
- Estonian Genome Center, University of Tartu, Tartu, Estonia.,Division of Endocrinology, Boston Children's Hospital, Cambridge, Massachusetts, USA.,Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Lili Milani
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu, Estonia.,Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | | | - Jian Yang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - David Cesarini
- Center for Experimental Social Science, Department of Economics, New York University, New York, New York, USA
| | - Peter M Visscher
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| |
Collapse
|
904
|
Prakash J, Gabdulina G, Trofimov S, Livshits G. Quantitative genetics of circulating Hyaluronic Acid (HA) and its correlation with hand osteoarthritis and obesity-related phenotypes in a community-based sample. Ann Hum Biol 2017; 44:522-530. [PMID: 28535729 DOI: 10.1080/03014460.2017.1334822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND One of the potential molecular biomarkers of osteoarthritis (OA) is hyaluronic acid (HA). HA levels may be related to the severity and progression of OA. However, little is known about the contribution of major risk factors for osteoarthritis, e.g. obesity-related phenotypes and genetics to HA variation. AIM To clarify the quantitative effect of these factors on HA. SUBJECTS AND METHODS An ethnically homogeneous sample of 911 apparently healthy European-derived individuals, assessed for radiographic hand osteoarthritis (RHOA), HA, leptin, adiponectin, and several anthropometrical measures of obesity-related phenotypes was studied. Model-based quantitative genetic analysis was used to reveal genetic and shared environmental factors affecting the variation of the study's phenotypes. RESULTS The HA levels significantly correlated with the age, RHOA, adiponectin, obesity-related phenotypes, and the waist-to-hip ratio. The putative genetic effects contributed significantly to the variation of HA (66.2 ± 9.3%) and they were also significant factors in the variations of all the other studied phenotypes, with the heritability estimate ranging between 0.122 ± 4.4% (WHR) and 45.7 ± 2.2% (joint space narrowing). CONCLUSIONS This is the first study to report heritability estimates of HA variation and its correlation with obesity-related phenotypes, ADP and RHOA. However, the nature of genetic effects on HA and its correlation with other study phenotypes require further clarification.
Collapse
Affiliation(s)
- Jai Prakash
- a Human Population Biology Research Unit, Department of Anatomy and Anthropology , Tel Aviv University , Tel Aviv , Israel
| | - Gulzhan Gabdulina
- b Department of Internal Medicine , Asfendiyarov Kazakh National Medical University , Almigty , Kazakhstan
| | - Svetlana Trofimov
- a Human Population Biology Research Unit, Department of Anatomy and Anthropology , Tel Aviv University , Tel Aviv , Israel
| | - Gregory Livshits
- a Human Population Biology Research Unit, Department of Anatomy and Anthropology , Tel Aviv University , Tel Aviv , Israel.,c Lilian and Marcel Pollak Chair of Biological Anthropology, Sackler Faculty of Medicine , Tel Aviv University , Tel Aviv , Israel
| |
Collapse
|
905
|
Visscher PM, Wray NR, Zhang Q, Sklar P, McCarthy MI, Brown MA, Yang J. 10 Years of GWAS Discovery: Biology, Function, and Translation. Am J Hum Genet 2017; 101:5-22. [PMID: 28686856 DOI: 10.1016/j.ajhg.2017.06.005] [Citation(s) in RCA: 2115] [Impact Index Per Article: 264.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Application of the experimental design of genome-wide association studies (GWASs) is now 10 years old (young), and here we review the remarkable range of discoveries it has facilitated in population and complex-trait genetics, the biology of diseases, and translation toward new therapeutics. We predict the likely discoveries in the next 10 years, when GWASs will be based on millions of samples with array data imputed to a large fully sequenced reference panel and on hundreds of thousands of samples with whole-genome sequencing data.
Collapse
|
906
|
Wang M, Hancock TP, MacLeod IM, Pryce JE, Cocks BG, Hayes BJ. Putative enhancer sites in the bovine genome are enriched with variants affecting complex traits. Genet Sel Evol 2017; 49:56. [PMID: 28683716 PMCID: PMC5499214 DOI: 10.1186/s12711-017-0331-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/26/2017] [Indexed: 12/31/2022] Open
Abstract
Background Enhancers are non-coding DNA sequences, which when they are bound by specific proteins increase the level of gene transcription. Enhancers activate unique gene expression patterns within cells of different types or under different conditions. Enhancers are key contributors to gene regulation, and causative variants that affect quantitative traits in humans and mice have been located in enhancer regions. However, in the bovine genome, enhancers as well as other regulatory elements are not yet well defined. In this paper, we sought to improve the annotation of bovine enhancer regions by using publicly available mammalian enhancer information. To test if the identified putative bovine enhancer regions are enriched with functional variants that affect milk production traits, we performed genome-wide association studies using imputed whole-genome sequence data followed by meta-analysis and enrichment analysis. Results We produced a library of candidate bovine enhancer regions by using publicly available bovine ChIP-Seq enhancer data in combination with enhancer data that were identified based on sequence homology with human and mouse enhancer databases. We found that imputed whole-genome sequence variants associated with milk production traits in 16,581 dairy cattle were enriched with enhancer regions that were marked by bovine-liver H3K4me3 and H3K27ac histone modifications from both permutation tests and gene set enrichment analysis. Enhancer regions that were identified based on sequence homology with human and mouse enhancer regions were not as strongly enriched with trait-associated sequence variants as the bovine ChIP-Seq candidate enhancer regions. The bovine ChIP-Seq enriched enhancer regions were located near genes and quantitative trait loci that are associated with pregnancy, growth, disease resistance, meat quality and quantity, and milk quality and quantity traits in dairy and beef cattle. Conclusions Our results suggest that sequence variants within enhancer regions that are located in bovine non-coding genomic regions contribute to the variation in complex traits. The level of enrichment was higher in bovine-specific enhancer regions that were identified by detecting histone modifications H3K4me3 and H3K27ac in bovine liver tissues than in enhancer regions identified by sequence homology with human and mouse data. These results highlight the need to use bovine-specific experimental data for the identification of enhancer regions. Electronic supplementary material The online version of this article (doi:10.1186/s12711-017-0331-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Min Wang
- School of Applied Systems Biology, La Trobe University, Bundoora, VIC, 3083, Australia.
| | - Timothy P Hancock
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
| | - Iona M MacLeod
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
| | - Jennie E Pryce
- School of Applied Systems Biology, La Trobe University, Bundoora, VIC, 3083, Australia.,Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
| | - Benjamin G Cocks
- School of Applied Systems Biology, La Trobe University, Bundoora, VIC, 3083, Australia.,Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
| | - Benjamin J Hayes
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
| |
Collapse
|
907
|
Selzam S, Dale PS, Wagner RK, DeFries JC, Cederlöf M, O’Reilly PF, Krapohl E, Plomin R. Genome-Wide Polygenic Scores Predict Reading Performance Throughout the School Years. SCIENTIFIC STUDIES OF READING : THE OFFICIAL JOURNAL OF THE SOCIETY FOR THE SCIENTIFIC STUDY OF READING 2017; 21:334-349. [PMID: 28706435 PMCID: PMC5490720 DOI: 10.1080/10888438.2017.1299152] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
It is now possible to create individual-specific genetic scores, called genome-wide polygenic scores (GPS). We used a GPS for years of education (EduYears) to predict reading performance assessed at UK National Curriculum Key Stages 1 (age 7), 2 (age 12) and 3 (age 14) and on reading tests administered at ages 7 and 12 in a UK sample of 5,825 unrelated individuals. EduYears GPS accounts for up to 5% of the variance in reading performance at age 14. GPS predictions remained significant after accounting for general cognitive ability and family socioeconomic status. Reading performance of children in the lowest and highest 12.5% of the EduYears GPS distribution differed by a mean growth in reading ability of approximately two school years. It seems certain that polygenic scores will be used to predict strengths and weaknesses in education.
Collapse
|
908
|
Gonen S, Battagin M, Johnston SE, Gorjanc G, Hickey JM. The potential of shifting recombination hotspots to increase genetic gain in livestock breeding. Genet Sel Evol 2017; 49:55. [PMID: 28676070 PMCID: PMC5496647 DOI: 10.1186/s12711-017-0330-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/26/2017] [Indexed: 01/01/2023] Open
Abstract
Background This study uses simulation to explore and quantify the potential effect of shifting recombination hotspots on genetic gain in livestock breeding programs. Methods We simulated three scenarios that differed in the locations of quantitative trait nucleotides (QTN) and recombination hotspots in the genome. In scenario 1, QTN were randomly distributed along the chromosomes and recombination was restricted to occur within specific genomic regions (i.e. recombination hotspots). In the other two scenarios, both QTN and recombination hotspots were located in specific regions, but differed in whether the QTN occurred outside of (scenario 2) or inside (scenario 3) recombination hotspots. We split each chromosome into 250, 500 or 1000 regions per chromosome of which 10% were recombination hotspots and/or contained QTN. The breeding program was run for 21 generations of selection, after which recombination hotspot regions were kept the same or were shifted to adjacent regions for a further 80 generations of selection. We evaluated the effect of shifting recombination hotspots on genetic gain, genetic variance and genic variance. Results Our results show that shifting recombination hotspots reduced the decline of genetic and genic variance by releasing standing allelic variation in the form of new allele combinations. This in turn resulted in larger increases in genetic gain. However, the benefit of shifting recombination hotspots for increased genetic gain was only observed when QTN were initially outside recombination hotspots. If QTN were initially inside recombination hotspots then shifting them decreased genetic gain. Discussion Shifting recombination hotspots to regions of the genome where recombination had not occurred for 21 generations of selection (i.e. recombination deserts) released more of the standing allelic variation available in each generation and thus increased genetic gain. However, whether and how much increase in genetic gain was achieved by shifting recombination hotspots depended on the distribution of QTN in the genome, the number of recombination hotspots and whether QTN were initially inside or outside recombination hotspots. Conclusions Our findings show future scope for targeted modification of recombination hotspots e.g. through changes in zinc-finger motifs of the PRDM9 protein to increase genetic gain in production species. Electronic supplementary material The online version of this article (doi:10.1186/s12711-017-0330-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Serap Gonen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, Scotland, UK
| | - Mara Battagin
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, Scotland, UK
| | - Susan E Johnston
- Institute of Evolutionary Biology, The University of Edinburgh, Charlotte Auerbach Road, Edinburgh, EH9 3FL, UK
| | - Gregor Gorjanc
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, Scotland, UK
| | - John M Hickey
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, Scotland, UK.
| |
Collapse
|
909
|
Capellini TD, Chen H, Cao J, Doxey AC, Kiapour AM, Schoor M, Kingsley DM. Ancient selection for derived alleles at a GDF5 enhancer influencing human growth and osteoarthritis risk. Nat Genet 2017; 49:1202-1210. [PMID: 28671685 PMCID: PMC6556117 DOI: 10.1038/ng.3911] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 06/12/2017] [Indexed: 12/19/2022]
Abstract
Variants in GDF5 are associated with human arthritis and decreased height, but the causal mutations are still unknown. We surveyed the Gdf5 locus for regulatory regions in transgenic mice and fine-mapped separate enhancers controlling expression in joints versus growing ends of long bones. A large downstream regulatory region contains a novel growth enhancer (GROW1), which is required for normal Gdf5 expression at ends of developing bones and for normal bone lengths in vivo. Human GROW1 contains a common base-pair change that decreases enhancer activity and colocalizes with peaks of positive selection in humans. The derived allele is rare in Africa but common in Eurasia and is found in Neandertals and Denisovans. Our results suggest that an ancient regulatory variant in GROW1 has been repeatedly selected in northern environments and that past selection on growth phenotypes explains the high frequency of a GDF5 haplotype that also increases arthritis susceptibility in many human populations.
Collapse
Affiliation(s)
- Terence D Capellini
- Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA.,Department of Developmental Biology, Stanford University, Stanford, California, USA
| | - Hao Chen
- Department of Developmental Biology, Stanford University, Stanford, California, USA
| | - Jiaxue Cao
- Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Andrew C Doxey
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Ata M Kiapour
- Department of Orthopedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Schoor
- Department of Developmental Biology, Stanford University, Stanford, California, USA
| | - David M Kingsley
- Department of Developmental Biology, Stanford University, Stanford, California, USA.,Howard Hughes Medical Institute, Stanford University, Stanford, California, USA
| |
Collapse
|
910
|
Gibert JM, Blanco J, Dolezal M, Nolte V, Peronnet F, Schlötterer C. Strong epistatic and additive effects of linked candidate SNPs for Drosophila pigmentation have implications for analysis of genome-wide association studies results. Genome Biol 2017; 18:126. [PMID: 28673357 PMCID: PMC5496195 DOI: 10.1186/s13059-017-1262-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/19/2017] [Indexed: 01/01/2023] Open
Abstract
Background The mapping resolution of genome-wide association studies (GWAS) is limited by historic recombination events and effects are often assigned to haplotype blocks rather than individual SNPs. It is not clear how many of the SNPs in the block, and which ones, are causative. Drosophila pigmentation is a powerful model to dissect the genetic basis of intra-specific and inter-specific phenotypic variation. Three tightly linked SNPs in the t-MSE enhancer have been identified in three D. melanogaster populations as major contributors to female abdominal pigmentation. This enhancer controls the expression of the pigmentation gene tan (t) in the abdominal epidermis. Two of the three SNPs were confirmed in an independent study using the D. melanogaster Genetic Reference Panel established from a North American population. Results We determined the functional impact of SNP1, SNP2, and SNP3 using transgenic lines to test all possible haplotypes in vivo. We show that all three candidate SNPs contribute to female Drosophila abdominal pigmentation. Interestingly, only two SNPs agree with the effect predicted by GWAS; the third one goes in the opposite direction because of linkage disequilibrium between multiple functional SNPs. Our experimental design uncovered strong additive effects for the three SNPs, but we also found significant epistatic effects explaining up to 11% of the total variation. Conclusions Our results suggest that linked causal variants are important for the interpretation of GWAS and functional validation is needed to understand the genetic architecture of traits. Electronic supplementary material The online version of this article (doi:10.1186/s13059-017-1262-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jean-Michel Gibert
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, Biologie du Développement Paris Seine-Institut de Biologie Paris Seine (LBD-IBPS), case 24, 9 quai St-Bernard, 75005, Paris, France
| | - Jorge Blanco
- Institut für Populationsgenetik, Vetmeduni Vienna, Veterinärplatz 1, 1210, Wien, Austria
| | - Marlies Dolezal
- Institut für Populationsgenetik, Vetmeduni Vienna, Veterinärplatz 1, 1210, Wien, Austria
| | - Viola Nolte
- Institut für Populationsgenetik, Vetmeduni Vienna, Veterinärplatz 1, 1210, Wien, Austria
| | - Frédérique Peronnet
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, Biologie du Développement Paris Seine-Institut de Biologie Paris Seine (LBD-IBPS), case 24, 9 quai St-Bernard, 75005, Paris, France
| | - Christian Schlötterer
- Institut für Populationsgenetik, Vetmeduni Vienna, Veterinärplatz 1, 1210, Wien, Austria.
| |
Collapse
|
911
|
Roetker NS, Armasu SM, Pankow JS, Lutsey PL, Tang W, Rosenberg MA, Palmer TM, MacLehose RF, Heckbert SR, Cushman M, de Andrade M, Folsom AR. Taller height as a risk factor for venous thromboembolism: a Mendelian randomization meta-analysis. J Thromb Haemost 2017; 15:1334-1343. [PMID: 28445597 PMCID: PMC5504700 DOI: 10.1111/jth.13719] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Indexed: 12/22/2022]
Abstract
Essentials Observational data suggest taller people have a higher risk of venous thromboembolism (VTE). We used Mendelian randomization techniques to further explore this association in three studies. Risk of VTE increased by 30-40% for each 10 cm increment in height. Height was more strongly associated with deep vein thrombosis than with pulmonary embolism. SUMMARY Background Taller height is associated with a greater risk of venous thromboembolism (VTE). Objectives To use instrumental variable (IV) techniques (Mendelian randomization) to further explore this relationship. Methods Participants of European ancestry were included from two cohort studies (Atherosclerosis Risk in Communities [ARIC] study and Cardiovascular Health Study [CHS]) and one case-control study (Mayo Clinic VTE Study [Mayo]). We created two weighted genetic risk scores (GRSs) for height; the full GRS included 668 single-nucleotide polymorphisms (SNPs) from a previously published meta-analysis, and the restricted GRS included a subset of 362 SNPs not associated with weight independently of height. Standard logistic regression and IV models were used to estimate odds ratios (ORs) for VTE per 10-cm increment in height. ORs were pooled across the three studies by the use of inverse variance-weighted random effects meta-analysis. Results Among 9143 ARIC and 3180 CHS participants free of VTE at baseline, there were 367 and 109 incident VTE events. There were 1143 VTE cases and 1292 controls included from Mayo. The pooled ORs from non-IV models and models using the full and restricted GRSs as IVs were 1.27 (95% confidence interval [CI] 1.11-1.46), 1.34 (95% CI 1.04-1.73) and 1.45 (95% CI 1.04-2.01) per 10-cm greater height, respectively. Conclusions Taller height is associated with an increased risk of VTE in adults of European ancestry. Possible explanations for this association, including taller people having a greater venous surface area, a higher number of venous valves, or greater hydrostatic pressure, need to be explored further.
Collapse
Affiliation(s)
- N S Roetker
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - S M Armasu
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - J S Pankow
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - P L Lutsey
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - W Tang
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - M A Rosenberg
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Aurora, CO, USA
- Division of Cardiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - T M Palmer
- Department of Mathematics and Statistics, Lancaster University, Lancaster, UK
| | - R F MacLehose
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - S R Heckbert
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - M Cushman
- Department of Medicine, University of Vermont, Burlington, VT, USA
- Department of Pathology, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | - M de Andrade
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - A R Folsom
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
912
|
Abstract
Pharmacogenomics (PGx), a substantial component of "personalized medicine", seeks to understand each individual's genetic composition to optimize drug therapy -- maximizing beneficial drug response, while minimizing adverse drug reactions (ADRs). Drug responses are highly variable because innumerable factors contribute to ultimate phenotypic outcomes. Recent genome-wide PGx studies have provided some insight into genetic basis of variability in drug response. These can be grouped into three categories. [a] Monogenic (Mendelian) traits include early examples mostly of inherited disorders, and some severe (idiosyncratic) ADRs typically influenced by single rare coding variants. [b] Predominantly oligogenic traits represent variation largely influenced by a small number of major pharmacokinetic or pharmacodynamic genes. [c] Complex PGx traits resemble most multifactorial quantitative traits -- influenced by numerous small-effect variants, together with epigenetic effects and environmental factors. Prediction of monogenic drug responses is relatively simple, involving detection of underlying mutations; due to rarity of these events and incomplete penetrance, however, prospective tests based on genotype will have high false-positive rates, plus pharmacoeconomics will require justification. Prediction of predominantly oligogenic traits is slowly improving. Although a substantial fraction of variation can be explained by limited numbers of large-effect genetic variants, uncertainty in successful predictions and overall cost-benefit ratios will make such tests elusive for everyday clinical use. Prediction of complex PGx traits is almost impossible in the foreseeable future. Genome-wide association studies of large cohorts will continue to discover relevant genetic variants; however, these small-effect variants, combined, explain only a small fraction of phenotypic variance -- thus having limited predictive power and clinical utility.
Collapse
Affiliation(s)
- Ge Zhang
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, United States.
| | - Daniel W Nebert
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, United States; Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati School of Medicine, Cincinnati, OH 45267-0056, United States.
| |
Collapse
|
913
|
Lu Q, Powles RL, Abdallah S, Ou D, Wang Q, Hu Y, Lu Y, Liu W, Li B, Mukherjee S, Crane PK, Zhao H. Systematic tissue-specific functional annotation of the human genome highlights immune-related DNA elements for late-onset Alzheimer's disease. PLoS Genet 2017; 13:e1006933. [PMID: 28742084 PMCID: PMC5546707 DOI: 10.1371/journal.pgen.1006933] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 08/07/2017] [Accepted: 07/18/2017] [Indexed: 12/31/2022] Open
Abstract
Continuing efforts from large international consortia have made genome-wide epigenomic and transcriptomic annotation data publicly available for a variety of cell and tissue types. However, synthesis of these datasets into effective summary metrics to characterize the functional non-coding genome remains a challenge. Here, we present GenoSkyline-Plus, an extension of our previous work through integration of an expanded set of epigenomic and transcriptomic annotations to produce high-resolution, single tissue annotations. After validating our annotations with a catalog of tissue-specific non-coding elements previously identified in the literature, we apply our method using data from 127 different cell and tissue types to present an atlas of heritability enrichment across 45 different GWAS traits. We show that broader organ system categories (e.g. immune system) increase statistical power in identifying biologically relevant tissue types for complex diseases while annotations of individual cell types (e.g. monocytes or B-cells) provide deeper insights into disease etiology. Additionally, we use our GenoSkyline-Plus annotations in an in-depth case study of late-onset Alzheimer's disease (LOAD). Our analyses suggest a strong connection between LOAD heritability and genetic variants contained in regions of the genome functional in monocytes. Furthermore, we show that LOAD shares a similar localization of SNPs to monocyte-functional regions with Parkinson's disease. Overall, we demonstrate that integrated genome annotations at the single tissue level provide a valuable tool for understanding the etiology of complex human diseases. Our GenoSkyline-Plus annotations are freely available at http://genocanyon.med.yale.edu/GenoSkyline.
Collapse
Affiliation(s)
- Qiongshi Lu
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Ryan L. Powles
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, United States of America
| | - Sarah Abdallah
- Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Derek Ou
- Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Qian Wang
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, United States of America
| | - Yiming Hu
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Yisi Lu
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Wei Liu
- School of Life Sciences, Peking University, Beijing, China
| | - Boyang Li
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Shubhabrata Mukherjee
- Division of General Internal Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Paul K. Crane
- Division of General Internal Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, United States of America
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- VA Cooperative Studies Program Coordinating Center, West Haven, Connecticut, United States of America
| |
Collapse
|
914
|
Carrying the baton: Evolution science and a contextual behavioral analysis of language and cognition. JOURNAL OF CONTEXTUAL BEHAVIORAL SCIENCE 2017. [DOI: 10.1016/j.jcbs.2017.01.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
915
|
Affiliation(s)
- Laurie R Braun
- Pediatric Endocrine Unit and.,Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | | |
Collapse
|
916
|
Hayden LP, Cho MH, McDonald MLN, Crapo JD, Beaty TH, Silverman EK, Hersh CP. Susceptibility to Childhood Pneumonia: A Genome-Wide Analysis. Am J Respir Cell Mol Biol 2017; 56:20-28. [PMID: 27508494 DOI: 10.1165/rcmb.2016-0101oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Previous studies have indicated that in adult smokers, a history of childhood pneumonia is associated with reduced lung function and chronic obstructive pulmonary disease. There have been few previous investigations using genome-wide association studies to investigate genetic predisposition to pneumonia. This study aims to identify the genetic variants associated with the development of pneumonia during childhood and over the course of the lifetime. Study subjects included current and former smokers with and without chronic obstructive pulmonary disease participating in the COPDGene Study. Pneumonia was defined by subject self-report, with childhood pneumonia categorized as having the first episode at <16 years. Genome-wide association studies for childhood pneumonia (843 cases, 9,091 control subjects) and lifetime pneumonia (3,766 cases, 5,659 control subjects) were performed separately in non-Hispanic whites and African Americans. Non-Hispanic white and African American populations were combined in the meta-analysis. Top genetic variants from childhood pneumonia were assessed in network analysis. No single-nucleotide polymorphisms reached genome-wide significance, although we identified potential regions of interest. In the childhood pneumonia analysis, this included variants in NGR1 (P = 6.3 × 10-8), PAK6 (P = 3.3 × 10-7), and near MATN1 (P = 2.8 × 10-7). In the lifetime pneumonia analysis, this included variants in LOC339862 (P = 8.7 × 10-7), RAPGEF2 (P = 8.4 × 10-7), PHACTR1 (P = 6.1 × 10-7), near PRR27 (P = 4.3 × 10-7), and near MCPH1 (P = 2.7 × 10-7). Network analysis of the genes associated with childhood pneumonia included top networks related to development, blood vessel morphogenesis, muscle contraction, WNT signaling, DNA damage, apoptosis, inflammation, and immune response (P ≤ 0.05). We have identified genes potentially associated with the risk of pneumonia. Further research will be required to confirm these associations and to determine biological mechanisms. CLINICAL TRIAL REGISTRATION NCT00608764.
Collapse
Affiliation(s)
- Lystra P Hayden
- 1 Division of Respiratory Diseases, Boston Children's Hospital, Boston, Massachusetts.,2 Channing Division of Network Medicine and
| | - Michael H Cho
- 2 Channing Division of Network Medicine and.,3 Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | - Terri H Beaty
- 5 Johns Hopkins University, Bloomberg School of Public Health, Baltimore, Maryland
| | - Edwin K Silverman
- 2 Channing Division of Network Medicine and.,3 Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Craig P Hersh
- 2 Channing Division of Network Medicine and.,3 Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
917
|
Hisado-Oliva A, Ruzafa-Martin A, Sentchordi L, Funari MFA, Bezanilla-López C, Alonso-Bernáldez M, Barraza-García J, Rodriguez-Zabala M, Lerario AM, Benito-Sanz S, Aza-Carmona M, Campos-Barros A, Jorge AAL, Heath KE. Mutations in C-natriuretic peptide (NPPC): a novel cause of autosomal dominant short stature. Genet Med 2017; 20:91-97. [PMID: 28661490 DOI: 10.1038/gim.2017.66] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 04/05/2017] [Indexed: 12/21/2022] Open
Abstract
PurposeC-type natriuretic peptide (CNP) and its principal receptor, natriuretic peptide receptor B (NPR-B), have been shown to be important in skeletal development. CNP and NPR-B are encoded by natriuretic peptide precursor-C (NPPC) and natriuretic peptide receptor 2 (NPR2) genes, respectively. While NPR2 mutations have been described in patients with skeletal dysplasias and idiopathic short stature (ISS), and several Npr2 and Nppc skeletal dysplasia mouse models exist, no mutations in NPPC have been described in patients to date.MethodsNPPC was screened in 668 patients (357 with disproportionate short stature and 311 with autosomal dominant ISS) and 29 additional ISS families in an ongoing whole-exome sequencing study.ResultsTwo heterozygous NPPC mutations, located in the highly conserved CNP ring, were identified. Both showed significant reductions in cyclic guanosine monophosphate synthesis, confirming their pathogenicity. Interestingly, one has been previously linked to skeletal abnormalities in the spontaneous Nppc mouse long-bone abnormality (lbab) mutant.ConclusionsOur results demonstrate, for the first time, that NPPC mutations cause autosomal dominant short stature in humans. The NPPC mutations cosegregated with a short stature and small hands phenotype. A CNP analog, which is currently in clinical trials for the treatment of achondroplasia, seems a promising therapeutic approach, since it directly replaces the defective protein.
Collapse
Affiliation(s)
- Alfonso Hisado-Oliva
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, U753), Instituto Carlos III, Madrid, Spain.,Multidisciplinary Skeletal Dysplasia Unit (UMDE), Hospital Universitario La Paz, Madrid, Spain
| | - Alba Ruzafa-Martin
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain
| | - Lucia Sentchordi
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain.,Multidisciplinary Skeletal Dysplasia Unit (UMDE), Hospital Universitario La Paz, Madrid, Spain.,Department of Pediatrics, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Mariana F A Funari
- Laboratorio de Hormonios e Genetica Molecular (LIM42), Hospital das Clinicas da Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, Brazil
| | | | - Marta Alonso-Bernáldez
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain
| | - Jimena Barraza-García
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, U753), Instituto Carlos III, Madrid, Spain.,Multidisciplinary Skeletal Dysplasia Unit (UMDE), Hospital Universitario La Paz, Madrid, Spain
| | - Maria Rodriguez-Zabala
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain
| | - Antonio M Lerario
- Unidade de Endocrinologia Genetica (LIM25), Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil.,Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Sara Benito-Sanz
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, U753), Instituto Carlos III, Madrid, Spain.,Multidisciplinary Skeletal Dysplasia Unit (UMDE), Hospital Universitario La Paz, Madrid, Spain
| | - Miriam Aza-Carmona
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, U753), Instituto Carlos III, Madrid, Spain.,Multidisciplinary Skeletal Dysplasia Unit (UMDE), Hospital Universitario La Paz, Madrid, Spain
| | - Angel Campos-Barros
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, U753), Instituto Carlos III, Madrid, Spain
| | - Alexander A L Jorge
- Laboratorio de Hormonios e Genetica Molecular (LIM42), Hospital das Clinicas da Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, Brazil.,Unidade de Endocrinologia Genetica (LIM25), Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Karen E Heath
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, U753), Instituto Carlos III, Madrid, Spain.,Multidisciplinary Skeletal Dysplasia Unit (UMDE), Hospital Universitario La Paz, Madrid, Spain
| |
Collapse
|
918
|
Latorre P, Varona L, Burgos C, Carrodeguas JA, López-Buesa P. O-GlcNAcylation mediates the control of cytosolic phosphoenolpyruvate carboxykinase activity via Pgc1α. PLoS One 2017; 12:e0179988. [PMID: 28644880 PMCID: PMC5482481 DOI: 10.1371/journal.pone.0179988] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 06/07/2017] [Indexed: 11/19/2022] Open
Abstract
PGC1α is a coactivator of many transcription factors and cytosolic phosphoenolpyruvate carboxykinase (PCK1) is a key enzyme for gluconeogenesis. PGC1α interacts with the transcription factor PPARγ to stimulate PCK1 expression and thus de novo glucose synthesis. These proteins are not only important for central energy metabolism but also for supplying intermediates for other metabolic pathways, including lipidogenesis and protein synthesis and might therefore be important factors in the ethiopathogenesis of metabolic disorders like diabetes but also in other pathologies like cancer. Since polymorphisms in these proteins have been related to some phenotypic traits in animals like pigs and PGC1α G482S polymorphism increases fat deposition in humans, we have investigated the molecular basis of such effects focusing on a commonly studied polymorphism in pig Pgc1α, which changes a cysteine at position 430 (WT) of the protein to a serine (C430S). Biochemical analyses show that Pgc1α WT stimulates higher expression of human PCK1 in HEK293T and HepG2 cells. Paradoxically, Pgc1α WT is less stable than Pgc1α p.C430S in HEK293T cells. However, the study of different post-translational modifications shows a higher O-GlcNAcylation level of Pgc1α p.C430S. This higher O-GlcNAcylation level significantly decreases the interaction between Pgc1α and PPARγ demonstrating the importance of post-translational glycosylation of PGC1α in the regulation of PCK1 activity. This, furthermore, could explain at least in part the observed epistatic effects between PGC1α and PCK1 in pigs.
Collapse
Affiliation(s)
- Pedro Latorre
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), BIFIIQFR (CSIC) Joint Unit, Universidad de Zaragoza, Zaragoza, Spain
| | - Luis Varona
- Departamento de Anatomía, Embriología y Genética, Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón (IA2), Zaragoza, Spain
| | - Carmen Burgos
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), BIFIIQFR (CSIC) Joint Unit, Universidad de Zaragoza, Zaragoza, Spain
| | - José A. Carrodeguas
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), BIFIIQFR (CSIC) Joint Unit, Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- IIS Aragón, Zaragoza, Spain
| | - Pascual López-Buesa
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), BIFIIQFR (CSIC) Joint Unit, Universidad de Zaragoza, Zaragoza, Spain
| |
Collapse
|
919
|
|
920
|
Sun S, Hood M, Scott L, Peng Q, Mukherjee S, Tung J, Zhou X. Differential expression analysis for RNAseq using Poisson mixed models. Nucleic Acids Res 2017; 45:e106. [PMID: 28369632 PMCID: PMC5499851 DOI: 10.1093/nar/gkx204] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 03/02/2017] [Accepted: 03/17/2017] [Indexed: 12/13/2022] Open
Abstract
Identifying differentially expressed (DE) genes from RNA sequencing (RNAseq) studies is among the most common analyses in genomics. However, RNAseq DE analysis presents several statistical and computational challenges, including over-dispersed read counts and, in some settings, sample non-independence. Previous count-based methods rely on simple hierarchical Poisson models (e.g. negative binomial) to model independent over-dispersion, but do not account for sample non-independence due to relatedness, population structure and/or hidden confounders. Here, we present a Poisson mixed model with two random effects terms that account for both independent over-dispersion and sample non-independence. We also develop a scalable sampling-based inference algorithm using a latent variable representation of the Poisson distribution. With simulations, we show that our method properly controls for type I error and is generally more powerful than other widely used approaches, except in small samples (n <15) with other unfavorable properties (e.g. small effect sizes). We also apply our method to three real datasets that contain related individuals, population stratification or hidden confounders. Our results show that our method increases power in all three data compared to other approaches, though the power gain is smallest in the smallest sample (n = 6). Our method is implemented in MACAU, freely available at www.xzlab.org/software.html.
Collapse
Affiliation(s)
- Shiquan Sun
- Systems Engineering Institute, Xi’an Jiaotong University, Xi’an, Shaanxi 710049, P.R. China
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michelle Hood
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Laura Scott
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Qinke Peng
- Systems Engineering Institute, Xi’an Jiaotong University, Xi’an, Shaanxi 710049, P.R. China
| | - Sayan Mukherjee
- Departments of Statistical Science, Mathematics, and Computer Science, Duke University, Durham, NC 27708, USA
| | - Jenny Tung
- Departments of Evolutionary Anthropology and Biology, Duke University, Durham, NC 27708, USA
- Duke University Population Research Institute, Duke University, Durham, NC 27708, USA
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
921
|
Abstract
Many factors affect eukaryotic gene expression. Transcription factors, histone codes, DNA folding, and noncoding RNA modulate expression. Those factors interact in large, broadly connected regulatory control networks. An engineer following classical principles of control theory would design a simpler regulatory network. Why are genomes overwired? Neutrality or enhanced robustness may lead to the accumulation of additional factors that complicate network architecture. Dynamics progresses like a ratchet. New factors get added. Genomes adapt to the additional complexity. The newly added factors can no longer be removed without significant loss of fitness. Alternatively, highly wired genomes may be more malleable. In large networks, most genomic variants tend to have a relatively small effect on gene expression and trait values. Many small effects lead to a smooth gradient, in which traits may change steadily with respect to underlying regulatory changes. A smooth gradient may provide a continuous path from a starting point up to the highest peak of performance. A potential path of increasing performance promotes adaptability and learning. Genomes gain by the inductive process of natural selection, a trial and error learning algorithm that discovers general solutions for adapting to environmental challenge. Similarly, deeply and densely connected computational networks gain by various inductive trial and error learning procedures, in which the networks learn to reduce the errors in sequential trials. Overwiring alters the geometry of induction by smoothing the gradient along the inductive pathways of improving performance. Those overwiring benefits for induction apply to both natural biological networks and artificial deep learning networks.
Collapse
Affiliation(s)
- Steven A Frank
- Department of Ecology and Evolutionary Biology, University of California, Irvine, CA, 92697-2525, USA
| |
Collapse
|
922
|
Abstract
A central goal of genetics is to understand the links between genetic variation and disease. Intuitively, one might expect disease-causing variants to cluster into key pathways that drive disease etiology. But for complex traits, association signals tend to be spread across most of the genome-including near many genes without an obvious connection to disease. We propose that gene regulatory networks are sufficiently interconnected such that all genes expressed in disease-relevant cells are liable to affect the functions of core disease-related genes and that most heritability can be explained by effects on genes outside core pathways. We refer to this hypothesis as an "omnigenic" model.
Collapse
Affiliation(s)
- Evan A Boyle
- Department of Genetics, Stanford University, Stanford, CA 94305, USA.
| | - Yang I Li
- Department of Genetics, Stanford University, Stanford, CA 94305, USA.
| | - Jonathan K Pritchard
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
923
|
Boyle EA, Li YI, Pritchard JK. An Expanded View of Complex Traits: From Polygenic to Omnigenic. Cell 2017; 169:1177-1186. [PMID: 28622505 PMCID: PMC5536862 DOI: 10.1016/j.cell.2017.05.038] [Citation(s) in RCA: 1773] [Impact Index Per Article: 221.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/16/2017] [Accepted: 05/24/2017] [Indexed: 12/13/2022]
Abstract
A central goal of genetics is to understand the links between genetic variation and disease. Intuitively, one might expect disease-causing variants to cluster into key pathways that drive disease etiology. But for complex traits, association signals tend to be spread across most of the genome-including near many genes without an obvious connection to disease. We propose that gene regulatory networks are sufficiently interconnected such that all genes expressed in disease-relevant cells are liable to affect the functions of core disease-related genes and that most heritability can be explained by effects on genes outside core pathways. We refer to this hypothesis as an "omnigenic" model.
Collapse
Affiliation(s)
- Evan A Boyle
- Department of Genetics, Stanford University, Stanford, CA 94305, USA.
| | - Yang I Li
- Department of Genetics, Stanford University, Stanford, CA 94305, USA.
| | - Jonathan K Pritchard
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
924
|
Hannon E, Weedon M, Bray N, O’Donovan M, Mill J. Pleiotropic Effects of Trait-Associated Genetic Variation on DNA Methylation: Utility for Refining GWAS Loci. Am J Hum Genet 2017; 100:954-959. [PMID: 28528868 PMCID: PMC5473725 DOI: 10.1016/j.ajhg.2017.04.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 04/19/2017] [Indexed: 11/23/2022] Open
Abstract
Most genetic variants identified in genome-wide association studies (GWASs) of complex traits are thought to act by affecting gene regulation rather than directly altering the protein product. As a consequence, the actual genes involved in disease are not necessarily the most proximal to the associated variants. By integrating data from GWAS analyses with those from genetic studies of regulatory variation, it is possible to identify variants pleiotropically associated with both a complex trait and measures of gene regulation. In this study, we used summary-data-based Mendelian randomization (SMR), a method developed to identify variants pleiotropically associated with both complex traits and gene expression, to identify variants associated with complex traits and DNA methylation. We used large DNA methylation quantitative trait locus (mQTL) datasets generated from two different tissues (blood and fetal brain) to prioritize genes for >40 complex traits with robust GWAS data and found considerable overlap with the results of SMR analyses performed with expression QTL (eQTL) data. We identified multiple examples of variable DNA methylation associated with GWAS variants for a range of complex traits, demonstrating the utility of this approach for refining genetic association signals.
Collapse
|
925
|
Abstract
Short stature is a common and heterogeneous condition that is often genetic in etiology. For most children with genetic short stature, the specific molecular causes remain unknown; but with advances in exome/genome sequencing and bioinformatics approaches, new genetic causes of growth disorders have been identified, contributing to the understanding of the underlying molecular mechanisms of longitudinal bone growth and growth failure. Identifying new genetic causes of growth disorders has the potential to improve diagnosis, prognostic accuracy, and individualized management, and help avoid unnecessary testing for endocrine and other disorders.
Collapse
Affiliation(s)
- Youn Hee Jee
- Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive MSC 1103, Bethesda, MD 20892-1103, USA.
| | - Anenisia C Andrade
- Division of Pediatric Endocrinology, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Solnavägen 1, Solna 171 77, Sweden
| | - Jeffrey Baron
- Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive MSC 1103, Bethesda, MD 20892-1103, USA
| | - Ola Nilsson
- Division of Pediatric Endocrinology, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Solnavägen 1, Solna 171 77, Sweden; University Hospital, Örebro University, Södra Grev Rosengatan, Örebro 701 85, Sweden
| |
Collapse
|
926
|
Greenbaum J, Wu K, Zhang L, Shen H, Zhang J, Deng HW. Increased detection of genetic loci associated with risk predictors of osteoporotic fracture using a pleiotropic cFDR method. Bone 2017; 99:62-68. [PMID: 28373146 PMCID: PMC5488332 DOI: 10.1016/j.bone.2017.03.052] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/26/2017] [Accepted: 03/30/2017] [Indexed: 11/28/2022]
Abstract
Although GWAS have been successful in identifying some osteoporosis associated loci, the findings explain only a small fraction of the total genetic variance. In this study we use a recently developed novel pleiotropic conditional false discovery rate (cFDR) method to identify novel genetic loci associated with two risk traits for osteoporotic fracture (the clinical outcome and end result of osteoporosis), Height (HT) and Femoral Neck (FNK) BMD. The cFDR method allows us to improve the detection of associated variants by incorporating any potentially shared genetic mechanisms between the two associated traits. We analyzed the summary statistics from two GWAS meta-analyses for single nucleotide polymorphisms (SNPs) that are associated with HT and FNK BMD. Using the cFDR method, we show enrichment in the identification of SNPs associated with each trait conditioned on their strength of association with the second trait. The findings revealed 18 SNPs that are associated with both HT and FNK BMD, 4 of which had not previously been reported to play a role in bone health. The novel SNPs located at KIF1B and the intergenic region between FERD3L and TWISTNB are noteworthy as these genes may be associated with processes that are functionally important in bone metabolism. By leveraging GWAS results from related phenotypes we identified several novel loci that may contribute to the proportion of variability explained for each trait, although we cannot speculate about these potential contributions to heritability based on this analysis alone.
Collapse
Affiliation(s)
- Jonathan Greenbaum
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | - Kehao Wu
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | - Lan Zhang
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | - Hui Shen
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | - Jigang Zhang
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | - Hong-Wen Deng
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
927
|
Tachmazidou I, Süveges D, Min JL, Ritchie GRS, Steinberg J, Walter K, Iotchkova V, Schwartzentruber J, Huang J, Memari Y, McCarthy S, Crawford AA, Bombieri C, Cocca M, Farmaki AE, Gaunt TR, Jousilahti P, Kooijman MN, Lehne B, Malerba G, Männistö S, Matchan A, Medina-Gomez C, Metrustry SJ, Nag A, Ntalla I, Paternoster L, Rayner NW, Sala C, Scott WR, Shihab HA, Southam L, St Pourcain B, Traglia M, Trajanoska K, Zaza G, Zhang W, Artigas MS, Bansal N, Benn M, Chen Z, Danecek P, Lin WY, Locke A, Luan J, Manning AK, Mulas A, Sidore C, Tybjaerg-Hansen A, Varbo A, Zoledziewska M, Finan C, Hatzikotoulas K, Hendricks AE, Kemp JP, Moayyeri A, Panoutsopoulou K, Szpak M, Wilson SG, Boehnke M, Cucca F, Di Angelantonio E, Langenberg C, Lindgren C, McCarthy MI, Morris AP, Nordestgaard BG, Scott RA, Tobin MD, Wareham NJ, Burton P, Chambers JC, Smith GD, Dedoussis G, Felix JF, Franco OH, Gambaro G, Gasparini P, Hammond CJ, Hofman A, Jaddoe VWV, Kleber M, Kooner JS, Perola M, Relton C, Ring SM, Rivadeneira F, Salomaa V, Spector TD, Stegle O, Toniolo D, Uitterlinden AG, Barroso I, Greenwood CMT, Perry JRB, et alTachmazidou I, Süveges D, Min JL, Ritchie GRS, Steinberg J, Walter K, Iotchkova V, Schwartzentruber J, Huang J, Memari Y, McCarthy S, Crawford AA, Bombieri C, Cocca M, Farmaki AE, Gaunt TR, Jousilahti P, Kooijman MN, Lehne B, Malerba G, Männistö S, Matchan A, Medina-Gomez C, Metrustry SJ, Nag A, Ntalla I, Paternoster L, Rayner NW, Sala C, Scott WR, Shihab HA, Southam L, St Pourcain B, Traglia M, Trajanoska K, Zaza G, Zhang W, Artigas MS, Bansal N, Benn M, Chen Z, Danecek P, Lin WY, Locke A, Luan J, Manning AK, Mulas A, Sidore C, Tybjaerg-Hansen A, Varbo A, Zoledziewska M, Finan C, Hatzikotoulas K, Hendricks AE, Kemp JP, Moayyeri A, Panoutsopoulou K, Szpak M, Wilson SG, Boehnke M, Cucca F, Di Angelantonio E, Langenberg C, Lindgren C, McCarthy MI, Morris AP, Nordestgaard BG, Scott RA, Tobin MD, Wareham NJ, Burton P, Chambers JC, Smith GD, Dedoussis G, Felix JF, Franco OH, Gambaro G, Gasparini P, Hammond CJ, Hofman A, Jaddoe VWV, Kleber M, Kooner JS, Perola M, Relton C, Ring SM, Rivadeneira F, Salomaa V, Spector TD, Stegle O, Toniolo D, Uitterlinden AG, Barroso I, Greenwood CMT, Perry JRB, Walker BR, Butterworth AS, Xue Y, Durbin R, Small KS, Soranzo N, Timpson NJ, Zeggini E. Whole-Genome Sequencing Coupled to Imputation Discovers Genetic Signals for Anthropometric Traits. Am J Hum Genet 2017; 100:865-884. [PMID: 28552196 PMCID: PMC5473732 DOI: 10.1016/j.ajhg.2017.04.014] [Show More Authors] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 04/21/2017] [Indexed: 01/05/2023] Open
Abstract
Deep sequence-based imputation can enhance the discovery power of genome-wide association studies by assessing previously unexplored variation across the common- and low-frequency spectra. We applied a hybrid whole-genome sequencing (WGS) and deep imputation approach to examine the broader allelic architecture of 12 anthropometric traits associated with height, body mass, and fat distribution in up to 267,616 individuals. We report 106 genome-wide significant signals that have not been previously identified, including 9 low-frequency variants pointing to functional candidates. Of the 106 signals, 6 are in genomic regions that have not been implicated with related traits before, 28 are independent signals at previously reported regions, and 72 represent previously reported signals for a different anthropometric trait. 71% of signals reside within genes and fine mapping resolves 23 signals to one or two likely causal variants. We confirm genetic overlap between human monogenic and polygenic anthropometric traits and find signal enrichment in cis expression QTLs in relevant tissues. Our results highlight the potential of WGS strategies to enhance biologically relevant discoveries across the frequency spectrum.
Collapse
Affiliation(s)
- Ioanna Tachmazidou
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Dániel Süveges
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Josine L Min
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Graham R S Ritchie
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK; Usher Institute of Population Health Sciences & Informatics, University of Edinburgh, Edinburgh EH16 4UX, UK; MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH16 4UX, UK
| | - Julia Steinberg
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Klaudia Walter
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Valentina Iotchkova
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK; European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SD, UK
| | | | - Jie Huang
- Boston VA Research Institute, Boston, MA 02130, USA
| | - Yasin Memari
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Shane McCarthy
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Andrew A Crawford
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK; BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Cristina Bombieri
- Department of Neurological, Biomedical and Movement Sciences, University of Verona, Verona 37134, Italy
| | - Massimiliano Cocca
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste 34100, Italy
| | - Aliki-Eleni Farmaki
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens 17671, Greece
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Pekka Jousilahti
- Department of Health, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Marjolein N Kooijman
- The Generation R Study Group, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Pediatrics, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Benjamin Lehne
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK
| | - Giovanni Malerba
- Department of Neurological, Biomedical and Movement Sciences, University of Verona, Verona 37134, Italy
| | - Satu Männistö
- Department of Health, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Angela Matchan
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Carolina Medina-Gomez
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Internal Medicine, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Sarah J Metrustry
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Abhishek Nag
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Ioanna Ntalla
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Nigel W Rayner
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK; Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Cinzia Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan 20132, Italy
| | - William R Scott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK; Department of Cardiology, Ealing Hospital NHS Trust, Middlesex UB1 3EU, UK
| | - Hashem A Shihab
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Lorraine Southam
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK; Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Beate St Pourcain
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK; Max Planck Institute for Psycholinguistics, Nijmegen 6500, the Netherlands
| | - Michela Traglia
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Katerina Trajanoska
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Internal Medicine, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Gialuigi Zaza
- Renal Unit, Department of Medicine, Verona University Hospital, Verona 37126, Italy
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK; Department of Cardiology, Ealing Hospital NHS Trust, Middlesex UB1 3EU, UK
| | - María S Artigas
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Narinder Bansal
- Cardiovascular Epidemiology Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Marianne Benn
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | - Zhongsheng Chen
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Petr Danecek
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | - Wei-Yu Lin
- Cardiovascular Epidemiology Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Adam Locke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO 63108, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Alisa K Manning
- Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA; Department of Medicine, Harvard University Medical School, Boston, MA 02115, USA
| | - Antonella Mulas
- Istituto di Ricerca Genetica e Biomedica (IRGB-CNR), Cagliari 09100, Italy; Università degli Studi di Sassari, Sassari 07100, Italy
| | - Carlo Sidore
- Istituto di Ricerca Genetica e Biomedica (IRGB-CNR), Cagliari 09100, Italy
| | - Anne Tybjaerg-Hansen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | - Anette Varbo
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | | | - Chris Finan
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, UK
| | | | - Audrey E Hendricks
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK; Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO 80204, USA
| | - John P Kemp
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK; University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD 4072, Australia
| | - Alireza Moayyeri
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK; Institute of Health Informatics, University College London, London NW1 2DA, UK
| | | | - Michal Szpak
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Scott G Wilson
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK; School of Medicine and Pharmacology, The University of Western Australia, Crawley, WA 6009, Australia; Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica (IRGB-CNR), Cagliari 09100, Italy; Università degli Studi di Sassari, Sassari 07100, Italy
| | - Emanuele Di Angelantonio
- Cardiovascular Epidemiology Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge CB1 8RN, UK; The National Institute for Health Research Blood and Transplant Unit (NIHR BTRU) in Donor Health and Genomics at the University of Cambridge, Cambridge CB1 8RN, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Cecilia Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Li Ka Shing Centre for Health Information and Discovery, The Big Data Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK; Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Department of Biostatistics, University of Liverpool, Liverpool L69 3GL, UK; Estonian Genome Center, University of Tartu, Tartu, Tartumaa 51010, Estonia
| | - Børge G Nordestgaard
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | - Robert A Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Martin D Tobin
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK; National Institute for Health Research (NIHR) Leicester Respiratory Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | | | | | - Paul Burton
- D2K Research Group, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - John C Chambers
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK; Department of Cardiology, Ealing Hospital NHS Trust, Middlesex UB1 3EU, UK; Imperial College Healthcare NHS Trust, London W2 1NY, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens 17671, Greece
| | - Janine F Felix
- The Generation R Study Group, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Pediatrics, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Oscar H Franco
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Giovanni Gambaro
- Division of Nephrology and Dialysis, Columbus-Gemelli University Hospital, Catholic University, Rome 00168, Italy
| | - Paolo Gasparini
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste 34100, Italy; Medical Genetics, Institute for Maternal and Child Health IRCCS "Burlo Garofolo", Trieste 34100, Italy
| | - Christopher J Hammond
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Pediatrics, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Marcus Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital NHS Trust, Middlesex UB1 3EU, UK; Imperial College Healthcare NHS Trust, London W2 1NY, UK; National Heart and Lung Institute, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Markus Perola
- Department of Health, National Institute for Health and Welfare, Helsinki 00271, Finland; Estonian Genome Center, University of Tartu, Tartu, Tartumaa 51010, Estonia; Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki 00290, Finland
| | - Caroline Relton
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Susan M Ring
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Internal Medicine, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Veikko Salomaa
- Department of Health, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Oliver Stegle
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SD, UK
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan 20132, Italy
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands; Department of Internal Medicine, Erasmus Medical Center, University Medical Center, Rotterdam 3000 CA, the Netherlands
| | | | | | | | - Inês Barroso
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK; University of Cambridge Metabolic Research Laboratories, and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Celia M T Greenwood
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC H3T 1E2, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, QC H3A 1A2, Canada; Department of Oncology, McGill University, Montréal, QC H2W 1S6, Canada
| | - John R B Perry
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK; MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Brian R Walker
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Adam S Butterworth
- Cardiovascular Epidemiology Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge CB1 8RN, UK; The National Institute for Health Research Blood and Transplant Unit (NIHR BTRU) in Donor Health and Genomics at the University of Cambridge, Cambridge CB1 8RN, UK
| | - Yali Xue
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Richard Durbin
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Nicole Soranzo
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK; The National Institute for Health Research Blood and Transplant Unit (NIHR BTRU) in Donor Health and Genomics at the University of Cambridge, Cambridge CB1 8RN, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0AH, UK
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Eleftheria Zeggini
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK.
| |
Collapse
|
928
|
Albuquerque EVA, Scalco RC, Jorge AAL. MANAGEMENT OF ENDOCRINE DISEASE: Diagnostic and therapeutic approach of tall stature. Eur J Endocrinol 2017; 176:R339-R353. [PMID: 28274950 DOI: 10.1530/eje-16-1054] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/23/2017] [Accepted: 03/08/2017] [Indexed: 12/17/2022]
Abstract
Tall stature is defined as a height of more than 2 standard deviations (s.d.) above average for same sex and age. Tall individuals are usually referred to endocrinologists so that hormonal disorders leading to abnormal growth are excluded. However, the majority of these patients have familial tall stature or constitutional advance of growth (generally associated with obesity), both of which are diagnoses of exclusion. It is necessary to have familiarity with a large number of rarer overgrowth syndromes, especially because some of them may have severe complications such as aortic aneurysm, thromboembolism and tumor predisposition and demand-specific follow-up approaches. Additionally, endocrine disorders associated with tall stature have specific treatments and for this reason their recognition is mandatory. With this review, we intend to provide an up-to-date summary of the genetic conditions associated with overgrowth to emphasize a practical diagnostic approach of patients with tall stature and to discuss the limitations of current growth interruption treatment options.
Collapse
Affiliation(s)
- Edoarda V A Albuquerque
- Unidade de Endocrinologia GenéticaLaboratório de Endocrinologia Celular e Molecular (LIM/25), Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Renata C Scalco
- Unidade de Endocrinologia do DesenvolvimentoLaboratório de Hormônios e Genética Molecular (LIM/42) do Hospital das Clinicas, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Disciplina de Endocrinologia da Faculdade de Ciências Médicas da Santa Casa de São PauloSão Paulo, Brazil
| | - Alexander A L Jorge
- Unidade de Endocrinologia GenéticaLaboratório de Endocrinologia Celular e Molecular (LIM/25), Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
929
|
Hagenaars SP, Gale CR, Deary IJ, Harris SE. Cognitive ability and physical health: a Mendelian randomization study. Sci Rep 2017; 7:2651. [PMID: 28572633 PMCID: PMC5453939 DOI: 10.1038/s41598-017-02837-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/19/2017] [Indexed: 12/25/2022] Open
Abstract
Causes of the association between cognitive ability and health remain unknown, but may reflect a shared genetic aetiology. This study examines the causal genetic associations between cognitive ability and physical health. We carried out two-sample Mendelian randomization analyses using the inverse-variance weighted method to test for causality between later life cognitive ability, educational attainment (as a proxy for cognitive ability in youth), BMI, height, systolic blood pressure, coronary artery disease, and type 2 diabetes using data from six independent GWAS consortia and the UK Biobank sample (N = 112 151). BMI, systolic blood pressure, coronary artery disease and type 2 diabetes showed negative associations with cognitive ability; height was positively associated with cognitive ability. The analyses provided no evidence for casual associations from health to cognitive ability. In the other direction, higher educational attainment predicted lower BMI, systolic blood pressure, coronary artery disease, type 2 diabetes, and taller stature. The analyses indicated no causal association from educational attainment to physical health. The lack of evidence for causal associations between cognitive ability, educational attainment, and physical health could be explained by weak instrumental variables, poorly measured outcomes, or the small number of disease cases.
Collapse
Affiliation(s)
- Saskia P Hagenaars
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
- Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
- Division of Psychiatry, University of Edinburgh, Edinburgh, EH10 5HF, UK
| | - Catharine R Gale
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
- Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
- Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK.
- Medical Genetics Section, University of Edinburgh Centre for Genomic and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK.
| |
Collapse
|
930
|
Dias C, Giordano M, Frechette R, Bellone S, Polychronakos C, Legault L, Deal CL, Goodyer CG. Genetic variations at the human growth hormone receptor (GHR) gene locus are associated with idiopathic short stature. J Cell Mol Med 2017; 21:2985-2999. [PMID: 28557176 PMCID: PMC5661101 DOI: 10.1111/jcmm.13210] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/17/2017] [Indexed: 12/15/2022] Open
Abstract
GH plays an essential role in the growing child by binding to the growth hormone receptor (GHR) on target cells and regulating multiple growth promoting and metabolic effects. Mutations in the GHR gene coding regions result in GH insensitivity (dwarfism) due to a dysfunctional receptor protein. However, children with idiopathic short stature (ISS) show growth impairment without GH or GHR defects. We hypothesized that decreased expression of the GHR gene may be involved. To test this, we investigated whether common genetic variants (microsatellites, SNPs) in regulatory regions of the GHR gene region were associated with the ISS phenotype. Genotyping of a GT‐repeat microsatellite in the GHR 5′UTR in a Montreal ISS cohort (n = 37 ISS, n = 105 controls) revealed that the incidence of the long/short (L/S) genotype was 3.3× higher in ISS children than controls (P = 0.04, OR = 3.85). In an Italian replication cohort (n = 143 ISS, n = 282 controls), the medium/short (M/S) genotype was 1.9× more frequent in the male ISS than controls (P = 0.017, OR = 2.26). In both ISS cohorts, logistic regression analysis of 27 SNPs showed an association of ISS with rs4292454, while haplotype analysis revealed specific risk haplotypes in the 3′ haploblocks. In contrast, there were no differences in GT genotype frequencies in a cohort of short stature (SS) adults versus controls (CARTaGENE: n = 168 SS, n = 207 controls) and the risk haplotype in the SS cohort was located in the most 5′ haploblock. These data suggest that the variants identified are potentially genetic markers specifically associated with the ISS phenotype.
Collapse
Affiliation(s)
- Christel Dias
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Mara Giordano
- Laboratory of Human Genetics, Department of Health Science, University of Eastern Piedmont, Novara, Italy
| | | | - Simonetta Bellone
- Division of Pediatrics, Department of Health Science, University of Eastern Piedmont, Novara, Italy
| | - Constantin Polychronakos
- Departments of Experimental Medicine, Human Genetics and Pediatrics, McGill University, Montreal, QC, Canada
| | - Laurent Legault
- Department of Pediatrics, McGill University, Montreal, QC, Canada
| | - Cheri L Deal
- CHU Ste-Justine Research Centre and Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
| | - Cynthia Gates Goodyer
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada.,Departments of Experimental Medicine and Pediatrics, McGill University, Montreal, QC, Canada
| |
Collapse
|
931
|
Southam L, Gilly A, Süveges D, Farmaki AE, Schwartzentruber J, Tachmazidou I, Matchan A, Rayner NW, Tsafantakis E, Karaleftheri M, Xue Y, Dedoussis G, Zeggini E. Whole genome sequencing and imputation in isolated populations identify genetic associations with medically-relevant complex traits. Nat Commun 2017; 8:15606. [PMID: 28548082 PMCID: PMC5458552 DOI: 10.1038/ncomms15606] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 03/28/2017] [Indexed: 01/26/2023] Open
Abstract
Next-generation association studies can be empowered by sequence-based imputation and by studying founder populations. Here we report ∼9.5 million variants from whole-genome sequencing (WGS) of a Cretan-isolated population, and show enrichment of rare and low-frequency variants with predicted functional consequences. We use a WGS-based imputation approach utilizing 10,422 reference haplotypes to perform genome-wide association analyses and observe 17 genome-wide significant, independent signals, including replicating evidence for association at eight novel low-frequency variant signals. Two novel cardiometabolic associations are at lead variants unique to the founder population sequences: chr16:70790626 (high-density lipoprotein levels beta −1.71 (SE 0.25), P=1.57 × 10−11, effect allele frequency (EAF) 0.006); and rs145556679 (triglycerides levels beta −1.13 (SE 0.17), P=2.53 × 10−11, EAF 0.013). Our findings add empirical support to the contribution of low-frequency variants in complex traits, demonstrate the advantage of including population-specific sequences in imputation panels and exemplify the power gains afforded by population isolates. Isolated populations can provide useful information on low-frequency variants for dissecting genetic architecture of complex traits. Here, Zeggini and colleagues show enrichment of rare and low-frequency variants and 8 novel low-frequency variant signals for cardiometabolic traits in two Greek isolated populations
Collapse
Affiliation(s)
- Lorraine Southam
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton CB10 1SA, UK.,Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Arthur Gilly
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton CB10 1SA, UK
| | - Dániel Süveges
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton CB10 1SA, UK
| | - Aliki-Eleni Farmaki
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens 17671, Greece
| | | | | | - Angela Matchan
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton CB10 1SA, UK
| | - Nigel W Rayner
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton CB10 1SA, UK.,Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | | | | | - Yali Xue
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton CB10 1SA, UK
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens 17671, Greece
| | | |
Collapse
|
932
|
Hojo H, Chung UI, Ohba S. Identification of the gene-regulatory landscape in skeletal development and potential links to skeletal regeneration. Regen Ther 2017; 6:100-107. [PMID: 30271844 PMCID: PMC6134913 DOI: 10.1016/j.reth.2017.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 12/21/2022] Open
Abstract
A class of gene-regulatory elements called enhancers are the main mediators controlling quantitative, temporal and spatial gene expressions. In the course of evolution, the enhancer landscape of higher organisms such as mammals has become quite complex, exerting biological functions precisely and coordinately. In mammalian skeletal development, the master transcription factors Sox9, Runx2 and Sp7/Osterix function primarily through enhancers on the genome to achieve specification and differentiation of skeletal cells. Recently developed genome-scale analyses have shed light on multiple layers of gene regulations, uncovering not only the primary mode of actions of these transcription factors on skeletal enhancers, but also the relation of the epigenetic landscape to three-dimensional chromatin architecture. Here, we review findings on the emerging framework of gene-regulatory networks involved in skeletal development. We further discuss the power of genome-scale analyses to provide new insights into genetic diseases and regenerative medicine in skeletal tissues. Skeletal development is coordinated by master transcription factors. ChIP-seq analyses for the skeletal regulators identified their modes of actions. Analyses of epigenetic landscape features distinct cell types in skeletal tissues. Integrated analyses of the gene regulatory networks link to skeletal regeneration.
Collapse
Affiliation(s)
- Hironori Hojo
- Department of Bioengineering, The University of Tokyo Graduate School of Engineering, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Ung-Il Chung
- Department of Bioengineering, The University of Tokyo Graduate School of Engineering, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.,Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Shinsuke Ohba
- Department of Bioengineering, The University of Tokyo Graduate School of Engineering, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.,Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
933
|
Wu Y, Zheng Z, Visscher PM, Yang J. Quantifying the mapping precision of genome-wide association studies using whole-genome sequencing data. Genome Biol 2017; 18:86. [PMID: 28506277 PMCID: PMC5432979 DOI: 10.1186/s13059-017-1216-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/20/2017] [Indexed: 01/21/2023] Open
Abstract
Background Understanding the mapping precision of genome-wide association studies (GWAS), that is the physical distances between the top associated single-nucleotide polymorphisms (SNPs) and the causal variants, is essential to design fine-mapping experiments for complex traits and diseases. Results Using simulations based on whole-genome sequencing (WGS) data from 3642 unrelated individuals of European descent, we show that the association signals at rare causal variants (minor allele frequency ≤ 0.01) are very unlikely to be mapped to common variants in GWAS using either WGS data or imputed data and vice versa. We predict that at least 80% of the common variants identified from published GWAS using imputed data are within 33.5 Kbp of the causal variants, a resolution that is comparable with that using WGS data. Mapping precision at these loci will improve with increasing sample sizes of GWAS in the future. For rare variants, the mapping precision of GWAS using WGS data is extremely high, suggesting WGS is an efficient strategy to detect and fine-map rare variants simultaneously. We further assess the mapping precision by linkage disequilibrium between GWAS hits and causal variants and develop an online tool (gwasMP) to query our results with different thresholds of physical distance and/or linkage disequilibrium (http://cnsgenomics.com/shiny/gwasMP). Conclusions Our findings provide a benchmark to inform future design and development of fine-mapping experiments and technologies to pinpoint the causal variants at GWAS loci. Electronic supplementary material The online version of this article (doi:10.1186/s13059-017-1216-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yang Wu
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Zhili Zheng
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.,The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Peter M Visscher
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Jian Yang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia. .,Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
934
|
Yaghootkar H, Bancks MP, Jones SE, McDaid A, Beaumont R, Donnelly L, Wood AR, Campbell A, Tyrrell J, Hocking LJ, Tuke MA, Ruth KS, Pearson ER, Murray A, Freathy RM, Munroe PB, Hayward C, Palmer C, Weedon MN, Pankow JS, Frayling TM, Kutalik Z. Quantifying the extent to which index event biases influence large genetic association studies. Hum Mol Genet 2017; 26:1018-1030. [PMID: 28040731 DOI: 10.1093/hmg/ddw433] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/19/2016] [Indexed: 11/12/2022] Open
Abstract
As genetic association studies increase in size to 100 000s of individuals, subtle biases may influence conclusions. One possible bias is 'index event bias' (IEB) that appears due to the stratification by, or enrichment for, disease status when testing associations between genetic variants and a disease-associated trait. We aimed to test the extent to which IEB influences some known trait associations in a range of study designs and provide a statistical framework for assessing future associations. Analyzing data from 113 203 non-diabetic UK Biobank participants, we observed three (near TCF7L2, CDKN2AB and CDKAL1) overestimated (body mass index (BMI) decreasing) and one (near MTNR1B) underestimated (BMI increasing) associations among 11 type 2 diabetes risk alleles (at P < 0.05). IEB became even stronger when we tested a type 2 diabetes genetic risk score composed of these 11 variants (-0.010 standard deviations BMI per allele, P = 5 × 10- 4), which was confirmed in four additional independent studies. Similar results emerged when examining the effect of blood pressure increasing alleles on BMI in normotensive UK Biobank samples. Furthermore, we demonstrated that, under realistic scenarios, common disease alleles would become associated at P < 5 × 10- 8 with disease-related traits through IEB alone, if disease prevalence in the sample differs appreciably from the background population prevalence. For example, some hypertension and type 2 diabetes alleles will be associated with BMI in sample sizes of >500 000 if the prevalence of those diseases differs by >10% from the background population. In conclusion, IEB may result in false positive or negative genetic associations in very large studies stratified or strongly enriched for/against disease cases.
Collapse
Affiliation(s)
- Hanieh Yaghootkar
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Michael P Bancks
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Sam E Jones
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Aaron McDaid
- Institute of Social and Preventive Medicine, Lausanne University Hospital, Lausanne 1010, Switzerland
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Robin Beaumont
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Louise Donnelly
- Division of Cardiovascular & Diabetes Medicine, Medical Research Institute, University of Dundee, Dundee, Scotland, UK
| | - Andrew R Wood
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Archie Campbell
- Generation Scotland, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, UK
| | - Jessica Tyrrell
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Lynne J Hocking
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Marcus A Tuke
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Katherine S Ruth
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Ewan R Pearson
- Division of Cardiovascular & Diabetes Medicine, Medical Research Institute, University of Dundee, Dundee, Scotland, UK
| | - Anna Murray
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Rachel M Freathy
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Patricia B Munroe
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Biomedical Research Unit, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Caroline Hayward
- Generation Scotland, MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, UK
| | - Colin Palmer
- Division of Cardiovascular & Diabetes Medicine, Medical Research Institute, University of Dundee, Dundee, Scotland, UK
| | - Michael N Weedon
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - James S Pankow
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine, Lausanne University Hospital, Lausanne 1010, Switzerland
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| |
Collapse
|
935
|
Abstract
There is great potential for genome sequencing to enhance patient care through improved diagnostic sensitivity and more precise therapeutic targeting. To maximize this potential, genomics strategies that have been developed for genetic discovery - including DNA-sequencing technologies and analysis algorithms - need to be adapted to fit clinical needs. This will require the optimization of alignment algorithms, attention to quality-coverage metrics, tailored solutions for paralogous or low-complexity areas of the genome, and the adoption of consensus standards for variant calling and interpretation. Global sharing of this more accurate genotypic and phenotypic data will accelerate the determination of causality for novel genes or variants. Thus, a deeper understanding of disease will be realized that will allow its targeting with much greater therapeutic precision.
Collapse
Affiliation(s)
- Euan A Ashley
- Center for Inherited Cardiovascular Disease, Falk Cardiovascular Research Building, Stanford Medicine, 870 Quarry Road, Stanford, California 94305, USA
| |
Collapse
|
936
|
Zeng P, Zhou X, Huang S. Prediction of gene expression with cis-SNPs using mixed models and regularization methods. BMC Genomics 2017; 18:368. [PMID: 28490319 PMCID: PMC5425981 DOI: 10.1186/s12864-017-3759-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 05/03/2017] [Indexed: 12/25/2022] Open
Abstract
Background It has been shown that gene expression in human tissues is heritable, thus predicting gene expression using only SNPs becomes possible. The prediction of gene expression can offer important implications on the genetic architecture of individual functional associated SNPs and further interpretations of the molecular basis underlying human diseases. Methods We compared three types of methods for predicting gene expression using only cis-SNPs, including the polygenic model, i.e. linear mixed model (LMM), two sparse models, i.e. Lasso and elastic net (ENET), and the hybrid of LMM and sparse model, i.e. Bayesian sparse linear mixed model (BSLMM). The three kinds of prediction methods have very different assumptions of underlying genetic architectures. These methods were evaluated using simulations under various scenarios, and were applied to the Geuvadis gene expression data. Results The simulations showed that these four prediction methods (i.e. Lasso, ENET, LMM and BSLMM) behaved best when their respective modeling assumptions were satisfied, but BSLMM had a robust performance across a range of scenarios. According to R2 of these models in the Geuvadis data, the four methods performed quite similarly. We did not observe any clustering or enrichment of predictive genes (defined as genes with R2 ≥ 0.05) across the chromosomes, and also did not see there was any clear relationship between the proportion of the predictive genes and the proportion of genes in each chromosome. However, an interesting finding in the Geuvadis data was that highly predictive genes (e.g. R2 ≥ 0.30) may have sparse genetic architectures since Lasso, ENET and BSLMM outperformed LMM for these genes; and this observation was validated in another gene expression data. We further showed that the predictive genes were enriched in approximately independent LD blocks. Conclusions Gene expression can be predicted with only cis-SNPs using well-developed prediction models and these predictive genes were enriched in some approximately independent LD blocks. The prediction of gene expression can shed some light on the functional interpretation for identified SNPs in GWASs.
Collapse
Affiliation(s)
- Ping Zeng
- Department of Epidemiology and Biostatistics, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, Jiangsu, 221004, China. .,Department of Biostatistics, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48104, USA.
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48104, USA
| | - Shuiping Huang
- Department of Epidemiology and Biostatistics, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
937
|
Percival CJ, Marangoni P, Tapaltsyan V, Klein O, Hallgrímsson B. The Interaction of Genetic Background and Mutational Effects in Regulation of Mouse Craniofacial Shape. G3 (BETHESDA, MD.) 2017; 7:1439-1450. [PMID: 28280213 PMCID: PMC5427488 DOI: 10.1534/g3.117.040659] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 03/03/2017] [Indexed: 11/18/2022]
Abstract
Inbred genetic background significantly influences the expression of phenotypes associated with known genetic perturbations and can underlie variation in disease severity between individuals with the same mutation. However, the effect of epistatic interactions on the development of complex traits, such as craniofacial morphology, is poorly understood. Here, we investigated the effect of three inbred backgrounds (129X1/SvJ, C57BL/6J, and FVB/NJ) on the expression of craniofacial dysmorphology in mice (Mus musculus) with loss of function in three members of the Sprouty family of growth factor negative regulators (Spry1, Spry2, or Spry4) in order to explore the impact of epistatic interactions on skull morphology. We found that the interaction of inbred background and the Sprouty genotype explains as much craniofacial shape variation as the Sprouty genotype alone. The most severely affected genotypes display a relatively short and wide skull, a rounded cranial vault, and a more highly angled inferior profile. Our results suggest that the FVB background is more resilient to Sprouty loss of function than either C57 or 129, and that Spry4 loss is generally less severe than loss of Spry1 or Spry2 While the specific modifier genes responsible for these significant background effects remain unknown, our results highlight the value of intercrossing mice of multiple inbred backgrounds to identify the genes and developmental interactions that modulate the severity of craniofacial dysmorphology. Our quantitative results represent an important first step toward elucidating genetic interactions underlying variation in robustness to known genetic perturbations in mice.
Collapse
Affiliation(s)
- Christopher J Percival
- Alberta Children's Hospital Institute for Child and Maternal Health, University of Calgary, Alberta T2N 4N1, Canada
- The McCaig Bone and Joint Institute, University of Calgary, Alberta T2N 4Z6, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Alberta T2N 4N1, Canada
| | - Pauline Marangoni
- Department of Orofacial Sciences, University of California, San Francisco, California 94143
- Program in Craniofacial Biology, University of California, San Francisco, California 94143
| | - Vagan Tapaltsyan
- Department of Orofacial Sciences, University of California, San Francisco, California 94143
- Program in Craniofacial Biology, University of California, San Francisco, California 94143
- Department of Preventive and Restorative Dental Sciences, University of California, San Francisco, California 94143
| | - Ophir Klein
- Department of Orofacial Sciences, University of California, San Francisco, California 94143
- Program in Craniofacial Biology, University of California, San Francisco, California 94143
- Department of Pediatrics, University of California, San Francisco, California 94143
- Institute for Human Genetics, University of California, San Francisco, California 94143
| | - Benedikt Hallgrímsson
- Alberta Children's Hospital Institute for Child and Maternal Health, University of Calgary, Alberta T2N 4N1, Canada
- The McCaig Bone and Joint Institute, University of Calgary, Alberta T2N 4Z6, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
938
|
Tatton-Brown K, Loveday C, Yost S, Clarke M, Ramsay E, Zachariou A, Elliott A, Wylie H, Ardissone A, Rittinger O, Stewart F, Temple IK, Cole T, Mahamdallie S, Seal S, Ruark E, Rahman N. Mutations in Epigenetic Regulation Genes Are a Major Cause of Overgrowth with Intellectual Disability. Am J Hum Genet 2017; 100:725-736. [PMID: 28475857 PMCID: PMC5420355 DOI: 10.1016/j.ajhg.2017.03.010] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/24/2017] [Indexed: 12/04/2022] Open
Abstract
To explore the genetic architecture of human overgrowth syndromes and human growth control, we performed experimental and bioinformatic analyses of 710 individuals with overgrowth (height and/or head circumference ≥+2 SD) and intellectual disability (OGID). We identified a causal mutation in 1 of 14 genes in 50% (353/710). This includes HIST1H1E, encoding histone H1.4, which has not been associated with a developmental disorder previously. The pathogenic HIST1H1E mutations are predicted to result in a product that is less effective in neutralizing negatively charged linker DNA because it has a reduced net charge, and in DNA binding and protein-protein interactions because key residues are truncated. Functional network analyses demonstrated that epigenetic regulation is a prominent biological process dysregulated in individuals with OGID. Mutations in six epigenetic regulation genes—NSD1, EZH2, DNMT3A, CHD8, HIST1H1E, and EED—accounted for 44% of individuals (311/710). There was significant overlap between the 14 genes involved in OGID and 611 genes in regions identified in GWASs to be associated with height (p = 6.84 × 10−8), suggesting that a common variation impacting function of genes involved in OGID influences height at a population level. Increased cellular growth is a hallmark of cancer and there was striking overlap between the genes involved in OGID and 260 somatically mutated cancer driver genes (p = 1.75 × 10−14). However, the mutation spectra of genes involved in OGID and cancer differ, suggesting complex genotype-phenotype relationships. These data reveal insights into the genetic control of human growth and demonstrate that exome sequencing in OGID has a high diagnostic yield.
Collapse
Affiliation(s)
- Katrina Tatton-Brown
- Division of Genetics and Epidemiology, Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, UK; South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London SW17 0QT, UK
| | - Chey Loveday
- Division of Genetics and Epidemiology, Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, UK
| | - Shawn Yost
- Division of Genetics and Epidemiology, Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, UK
| | - Matthew Clarke
- Division of Genetics and Epidemiology, Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, UK
| | - Emma Ramsay
- Division of Genetics and Epidemiology, Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, UK
| | - Anna Zachariou
- Division of Genetics and Epidemiology, Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, UK
| | - Anna Elliott
- Division of Genetics and Epidemiology, Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, UK
| | - Harriet Wylie
- Division of Genetics and Epidemiology, Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, UK
| | - Anna Ardissone
- Child Neurology Unit, Foundation IRCCS C Besta Neurological Institute, Milan 20133, Italy
| | - Olaf Rittinger
- Landeskrankenanstalten Salzburg, Kinderklinik Department of Pediatrics, Klinische Genetik, Salzburg 5020, Austria
| | - Fiona Stewart
- Northern Ireland Regional Genetics Service, Belfast City Hospital, Belfast BT9 7AB, Northern Ireland
| | - I Karen Temple
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK; Wessex Clinical Genetics Service, University Hospital Southampton NHS Trust, Southampton SO16 6YD, UK
| | - Trevor Cole
- West Midlands Regional Genetics Service, Birmingham Women's Hospital NHS Foundation Trust and University of Birmingham, Birmingham Health Partners, Birmingham B15 2TG, UK
| | - Shazia Mahamdallie
- Division of Genetics and Epidemiology, Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, UK
| | - Sheila Seal
- Division of Genetics and Epidemiology, Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, UK
| | - Elise Ruark
- Division of Genetics and Epidemiology, Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, UK
| | - Nazneen Rahman
- Division of Genetics and Epidemiology, Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, UK; Cancer Genetics Unit, Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK.
| |
Collapse
|
939
|
Alacevich C, Tarozzi A. Child height and intergenerational transmission of health: Evidence from ethnic Indians in England. ECONOMICS AND HUMAN BIOLOGY 2017; 25:65-84. [PMID: 27836569 DOI: 10.1016/j.ehb.2016.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 10/17/2016] [Accepted: 10/17/2016] [Indexed: 06/06/2023]
Abstract
A large literature documents a widespread prevalence of small stature among Indian children as well as adults. We show that a height gap relative to a richer population such as whites in England also exists, although substantially reduced, among adult immigrants of Indian ethnicity in England. This is despite positive height selection into migration, demonstrated by ethnic Indian adults in England being on average 6-7cm taller than in India. However, the difference between natives and ethnic Indians in England disappears among their younger sons and daughters, although it re-appears among adolescents. We estimate that, conditional on age, gender and parental height, ethnic Indian children of age 2-4 in England are 6-8% taller than in India. Such degree of catch up in one generation is remarkable, also because in England children of ethnic Indians have much smaller birthweight than whites, by about 0.4kg on average.
Collapse
Affiliation(s)
| | - Alessandro Tarozzi
- Universitat Pompeu Fabra, Spain; Barcelona GSE; Centro de Investigación en Economía y Salud (CRES-UPF), Spain.
| |
Collapse
|
940
|
Abstract
Primary sclerosing cholangitis (PSC) is a chronic disease leading to fibrotic scarring of the intrahepatic and extrahepatic bile ducts, causing considerable morbidity and mortality via the development of cholestatic liver cirrhosis, concurrent IBD and a high risk of bile duct cancer. Expectations have been high that genetic studies would determine key factors in PSC pathogenesis to support the development of effective medical therapies. Through the application of genome-wide association studies, a large number of disease susceptibility genes have been identified. The overall genetic architecture of PSC shares features with both autoimmune diseases and IBD. Strong human leukocyte antigen gene associations, along with several susceptibility genes that are critically involved in T-cell function, support the involvement of adaptive immune responses in disease pathogenesis, and position PSC as an autoimmune disease. In this Review, we survey the developments that have led to these gene discoveries. We also elaborate relevant interpretations of individual gene findings in the context of established disease models in PSC, and propose relevant translational research efforts to pursue novel insights.
Collapse
|
941
|
Kominakis A, Hager-Theodorides AL, Zoidis E, Saridaki A, Antonakos G, Tsiamis G. Combined GWAS and 'guilt by association'-based prioritization analysis identifies functional candidate genes for body size in sheep. Genet Sel Evol 2017; 49:41. [PMID: 28454565 PMCID: PMC5408376 DOI: 10.1186/s12711-017-0316-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 04/19/2017] [Indexed: 12/30/2022] Open
Abstract
Background Body size in sheep is an important indicator of productivity, growth and health as well as of environmental adaptation. It is a composite quantitative trait that has been studied with high-throughput genomic methods, i.e. genome-wide association studies (GWAS) in various mammalian species. Several genomic markers have been associated with body size traits and genes have been identified as causative candidates in humans, dog and cattle. A limited number of related GWAS have been performed in various sheep breeds and have identified genomic regions and candidate genes that partly account for body size variability. Here, we conducted a GWAS in Frizarta dairy sheep with phenotypic data from 10 body size measurements and genotypic data (from Illumina ovineSNP50 BeadChip) for 459 ewes. Results The 10 body size measurements were subjected to principal component analysis and three independent principal components (PC) were constructed, interpretable as width, height and length dimensions, respectively. The GWAS performed for each PC identified 11 significant SNPs, at the chromosome level, one on each of the chromosomes 3, 8, 9, 10, 11, 12, 19, 20, 23 and two on chromosome 25. Nine out of the 11 SNPs were located on previously identified quantitative trait loci for sheep meat, production or reproduction. One hundred and ninety-seven positional candidate genes within a 1-Mb distance from each significant SNP were found. A guilt-by-association-based (GBA) prioritization analysis (PA) was performed to identify the most plausible functional candidate genes. GBA-based PA identified 39 genes that were significantly associated with gene networks relevant to body size traits. Prioritized genes were identified in the vicinity of all significant SNPs except for those on chromosomes 10 and 12. The top five ranking genes were TP53, BMPR1A, PIK3R5, RPL26 and PRKDC. Conclusions The results of this GWAS provide evidence for 39 causative candidate genes across nine chromosomal regions for body size traits, some of which are novel and some are previously identified candidates from other studies (e.g. TP53, NTN1 and ZNF521). GBA-based PA has proved to be a useful tool to identify genes with increased biological relevance but it is subjected to certain limitations. Electronic supplementary material The online version of this article (doi:10.1186/s12711-017-0316-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Antonios Kominakis
- Department of Animal Science and Aquaculture, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece
| | - Ariadne L Hager-Theodorides
- Department of Animal Science and Aquaculture, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece.
| | - Evangelos Zoidis
- Department of Animal Science and Aquaculture, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece
| | - Aggeliki Saridaki
- Department of Environmental and Natural Resources Management, University of Patras, Seferi 2, 30100, Agrinio, Greece
| | - George Antonakos
- Agricultural and Livestock Union of Western Greece, 13rd Km N.R. Agrinio-Ioannina, 30100, Lepenou, Greece
| | - George Tsiamis
- Department of Environmental and Natural Resources Management, University of Patras, Seferi 2, 30100, Agrinio, Greece
| |
Collapse
|
942
|
Abstract
Despite thousands of genetic loci identified to date, a large proportion of genetic variation predisposing to complex disease and traits remains unaccounted for. Advances in sequencing technology enable focused explorations on the contribution of low-frequency and rare variants to human traits. Here we review experimental approaches and current knowledge on the contribution of these genetic variants in complex disease and discuss challenges and opportunities for personalised medicine.
Collapse
Affiliation(s)
- Lorenzo Bomba
- Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, CB10 1HH, UK
| | - Klaudia Walter
- Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, CB10 1HH, UK
| | - Nicole Soranzo
- Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, CB10 1HH, UK. .,Department of Haematology, University of Cambridge, Hills Rd, Cambridge, CB2 0AH, UK. .,The National Institute for Health Research Blood and Transplant Unit (NIHR BTRU) in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Strangeways Research Laboratory, Wort's Causeway, Cambridge, CB1 8RN, UK.
| |
Collapse
|
943
|
Justice AE, Winkler TW, Feitosa MF, Graff M, Fisher VA, Young K, Barata L, Deng X, Czajkowski J, Hadley D, Ngwa JS, Ahluwalia TS, Chu AY, Heard-Costa NL, Lim E, Perez J, Eicher JD, Kutalik Z, Xue L, Mahajan A, Renström F, Wu J, Qi Q, Ahmad S, Alfred T, Amin N, Bielak LF, Bonnefond A, Bragg J, Cadby G, Chittani M, Coggeshall S, Corre T, Direk N, Eriksson J, Fischer K, Gorski M, Neergaard Harder M, Horikoshi M, Huang T, Huffman JE, Jackson AU, Justesen JM, Kanoni S, Kinnunen L, Kleber ME, Komulainen P, Kumari M, Lim U, Luan J, Lyytikäinen LP, Mangino M, Manichaikul A, Marten J, Middelberg RPS, Müller-Nurasyid M, Navarro P, Pérusse L, Pervjakova N, Sarti C, Smith AV, Smith JA, Stančáková A, Strawbridge RJ, Stringham HM, Sung YJ, Tanaka T, Teumer A, Trompet S, van der Laan SW, van der Most PJ, Van Vliet-Ostaptchouk JV, Vedantam SL, Verweij N, Vink JM, Vitart V, Wu Y, Yengo L, Zhang W, Hua Zhao J, Zimmermann ME, Zubair N, Abecasis GR, Adair LS, Afaq S, Afzal U, Bakker SJL, Bartz TM, Beilby J, Bergman RN, Bergmann S, Biffar R, Blangero J, Boerwinkle E, Bonnycastle LL, Bottinger E, Braga D, Buckley BM, Buyske S, Campbell H, et alJustice AE, Winkler TW, Feitosa MF, Graff M, Fisher VA, Young K, Barata L, Deng X, Czajkowski J, Hadley D, Ngwa JS, Ahluwalia TS, Chu AY, Heard-Costa NL, Lim E, Perez J, Eicher JD, Kutalik Z, Xue L, Mahajan A, Renström F, Wu J, Qi Q, Ahmad S, Alfred T, Amin N, Bielak LF, Bonnefond A, Bragg J, Cadby G, Chittani M, Coggeshall S, Corre T, Direk N, Eriksson J, Fischer K, Gorski M, Neergaard Harder M, Horikoshi M, Huang T, Huffman JE, Jackson AU, Justesen JM, Kanoni S, Kinnunen L, Kleber ME, Komulainen P, Kumari M, Lim U, Luan J, Lyytikäinen LP, Mangino M, Manichaikul A, Marten J, Middelberg RPS, Müller-Nurasyid M, Navarro P, Pérusse L, Pervjakova N, Sarti C, Smith AV, Smith JA, Stančáková A, Strawbridge RJ, Stringham HM, Sung YJ, Tanaka T, Teumer A, Trompet S, van der Laan SW, van der Most PJ, Van Vliet-Ostaptchouk JV, Vedantam SL, Verweij N, Vink JM, Vitart V, Wu Y, Yengo L, Zhang W, Hua Zhao J, Zimmermann ME, Zubair N, Abecasis GR, Adair LS, Afaq S, Afzal U, Bakker SJL, Bartz TM, Beilby J, Bergman RN, Bergmann S, Biffar R, Blangero J, Boerwinkle E, Bonnycastle LL, Bottinger E, Braga D, Buckley BM, Buyske S, Campbell H, Chambers JC, Collins FS, Curran JE, de Borst GJ, de Craen AJM, de Geus EJC, Dedoussis G, Delgado GE, den Ruijter HM, Eiriksdottir G, Eriksson AL, Esko T, Faul JD, Ford I, Forrester T, Gertow K, Gigante B, Glorioso N, Gong J, Grallert H, Grammer TB, Grarup N, Haitjema S, Hallmans G, Hamsten A, Hansen T, Harris TB, Hartman CA, Hassinen M, Hastie ND, Heath AC, Hernandez D, Hindorff L, Hocking LJ, Hollensted M, Holmen OL, Homuth G, Jan Hottenga J, Huang J, Hung J, Hutri-Kähönen N, Ingelsson E, James AL, Jansson JO, Jarvelin MR, Jhun MA, Jørgensen ME, Juonala M, Kähönen M, Karlsson M, Koistinen HA, Kolcic I, Kolovou G, Kooperberg C, Krämer BK, Kuusisto J, Kvaløy K, Lakka TA, Langenberg C, Launer LJ, Leander K, Lee NR, Lind L, Lindgren CM, Linneberg A, Lobbens S, Loh M, Lorentzon M, Luben R, Lubke G, Ludolph-Donislawski A, Lupoli S, Madden PAF, Männikkö R, Marques-Vidal P, Martin NG, McKenzie CA, McKnight B, Mellström D, Menni C, Montgomery GW, Musk AW(B, Narisu N, Nauck M, Nolte IM, Oldehinkel AJ, Olden M, Ong KK, Padmanabhan S, Peyser PA, Pisinger C, Porteous DJ, Raitakari OT, Rankinen T, Rao DC, Rasmussen-Torvik LJ, Rawal R, Rice T, Ridker PM, Rose LM, Bien SA, Rudan I, Sanna S, Sarzynski MA, Sattar N, Savonen K, Schlessinger D, Scholtens S, Schurmann C, Scott RA, Sennblad B, Siemelink MA, Silbernagel G, Slagboom PE, Snieder H, Staessen JA, Stott DJ, Swertz MA, Swift AJ, Taylor KD, Tayo BO, Thorand B, Thuillier D, Tuomilehto J, Uitterlinden AG, Vandenput L, Vohl MC, Völzke H, Vonk JM, Waeber G, Waldenberger M, Westendorp RGJ, Wild S, Willemsen G, Wolffenbuttel BHR, Wong A, Wright AF, Zhao W, Zillikens MC, Baldassarre D, Balkau B, Bandinelli S, Böger CA, Boomsma DI, Bouchard C, Bruinenberg M, Chasman DI, Chen YD, Chines PS, Cooper RS, Cucca F, Cusi D, Faire UD, Ferrucci L, Franks PW, Froguel P, Gordon-Larsen P, Grabe HJ, Gudnason V, Haiman CA, Hayward C, Hveem K, Johnson AD, Wouter Jukema J, Kardia SLR, Kivimaki M, Kooner JS, Kuh D, Laakso M, Lehtimäki T, Marchand LL, März W, McCarthy MI, Metspalu A, Morris AP, Ohlsson C, Palmer LJ, Pasterkamp G, Pedersen O, Peters A, Peters U, Polasek O, Psaty BM, Qi L, Rauramaa R, Smith BH, Sørensen TIA, Strauch K, Tiemeier H, Tremoli E, van der Harst P, Vestergaard H, Vollenweider P, Wareham NJ, Weir DR, Whitfield JB, Wilson JF, Tyrrell J, Frayling TM, Barroso I, Boehnke M, Deloukas P, Fox CS, Hirschhorn JN, Hunter DJ, Spector TD, Strachan DP, van Duijn CM, Heid IM, Mohlke KL, Marchini J, Loos RJF, Kilpeläinen TO, Liu CT, Borecki IB, North KE, Cupples LA. Genome-wide meta-analysis of 241,258 adults accounting for smoking behaviour identifies novel loci for obesity traits. Nat Commun 2017; 8:14977. [PMID: 28443625 PMCID: PMC5414044 DOI: 10.1038/ncomms14977] [Show More Authors] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 02/15/2017] [Indexed: 02/07/2023] Open
Abstract
Few genome-wide association studies (GWAS) account for environmental exposures, like smoking, potentially impacting the overall trait variance when investigating the genetic contribution to obesity-related traits. Here, we use GWAS data from 51,080 current smokers and 190,178 nonsmokers (87% European descent) to identify loci influencing BMI and central adiposity, measured as waist circumference and waist-to-hip ratio both adjusted for BMI. We identify 23 novel genetic loci, and 9 loci with convincing evidence of gene-smoking interaction (GxSMK) on obesity-related traits. We show consistent direction of effect for all identified loci and significance for 18 novel and for 5 interaction loci in an independent study sample. These loci highlight novel biological functions, including response to oxidative stress, addictive behaviour, and regulatory functions emphasizing the importance of accounting for environment in genetic analyses. Our results suggest that tobacco smoking may alter the genetic susceptibility to overall adiposity and body fat distribution.
Collapse
Affiliation(s)
- Anne E. Justice
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Thomas W. Winkler
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine; St. Louis, Missouri, 63108 USA
| | - Misa Graff
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Virginia A. Fisher
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Kristin Young
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Llilda Barata
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine; St. Louis, Missouri, 63108 USA
| | - Xuan Deng
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Jacek Czajkowski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine; St. Louis, Missouri, 63108 USA
| | - David Hadley
- Population Health Research Institute, St. George's, University of London, London, SW17 0RE, UK
- TransMed Systems, Inc., Cupertino, California 95014, USA
| | - Julius S. Ngwa
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore Maryland, USA
| | - Tarunveer S. Ahluwalia
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center, Gentofte, Denmark
| | - Audrey Y. Chu
- NHLBI Framingham Heart Study, Framingham, Massachusetts, 01702 USA
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
| | - Nancy L. Heard-Costa
- NHLBI Framingham Heart Study, Framingham, Massachusetts, 01702 USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Elise Lim
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Jeremiah Perez
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - John D. Eicher
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, The Framingham Heart Study, Framingham, Massachusetts, USA
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine (IUMSP), Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss instititute of Bioinformatics
| | - Luting Xue
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Frida Renström
- Department of Biobank Research, Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, SE-205 02 Malmö, Sweden
| | - Joseph Wu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Shafqat Ahmad
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, SE-205 02 Malmö, Sweden
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Tamuno Alfred
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- The Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Najaf Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3015GE, The Netherlands
| | - Lawrence F. Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Amelie Bonnefond
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
| | - Jennifer Bragg
- Internal Medicine - Nephrology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Gemma Cadby
- Centre for Genetic Origins of Health and Disease, University of Western Australia, Crawley, Australia
| | - Martina Chittani
- Department of Health Sciences, University of Milan,Via A. Di Rudiní, 8 20142, Milano, Italy
| | - Scott Coggeshall
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
| | - Tanguy Corre
- Institute of Social and Preventive Medicine (IUMSP), Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss instititute of Bioinformatics
| | - Nese Direk
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Psychiatry, Dokuz Eylul University, Izmir, Turkey
| | - Joel Eriksson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Krista Fischer
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Mathias Gorski
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Marie Neergaard Harder
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Momoko Horikoshi
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Tao Huang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Epidemiology Domain, Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
| | - Jennifer E. Huffman
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, The Framingham Heart Study, Framingham, Massachusetts, USA
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Anne U. Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Johanne Marie Justesen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Leena Kinnunen
- Department of Health, National Institute for Health and Welfare, Helsinki, FI-00271 Finland
| | - Marcus E. Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Meena Kumari
- ISER, University of Essex, Colchester CO43SQ, UK
- Department of Epidemiology and Public Health, UCL, London, WC1E 6BT, UK
| | - Unhee Lim
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii 96813, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33520, Finland
- Department of Clinical Chemistry, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33014, Finland
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- NIHR Biomedical Research Centre at Guy's and St. Thomas' Foundation Trust, London, UK
| | - Ani Manichaikul
- Center for Public Health Genomics and Biostatistics Section, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Jonathan Marten
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Rita P. S. Middelberg
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Pau Navarro
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Louis Pérusse
- Department of Kinesiology, Faculty of Medicine, Université Laval, Québec, Canada
- Institute of Nutrition and Functional Foods, Université Laval, Québec, Canada
| | - Natalia Pervjakova
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia
| | - Cinzia Sarti
- Department of Social and Health Care, City of Helsinki, Helsinki, Finland
| | - Albert Vernon Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Alena Stančáková
- Department of Medicine, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - Rona J. Strawbridge
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Heather M. Stringham
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St Louis, Missouri, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, Baltimore Maryland, USA
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Germany
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, The Netherlands
- Department of Gerontology and Geriatrics, Leiden University Medical Center, The Netherlands
| | - Sander W. van der Laan
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
| | - Peter J. van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | | | - Sailaja L. Vedantam
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
| | - Niek Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Jacqueline M. Vink
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
- Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Ying Wu
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Loic Yengo
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
| | - Weihua Zhang
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Martina E. Zimmermann
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
| | - Niha Zubair
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Gonçalo R. Abecasis
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Linda S. Adair
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Saima Afaq
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
| | - Uzma Afzal
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
| | - Stephan J. L. Bakker
- Department of Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Traci M. Bartz
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington 98101, USA
| | - John Beilby
- Busselton Population Medical Research Institute, Nedlands, Western Australia 6009, Australia
- PathWest Laboratory Medicine of WA, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
- School of Pathology and Laboraty Medicine, The University of Western Australia, 35 Stirling Hwy, Crawley, Western Australia 6009, Australia
| | - Richard N. Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sven Bergmann
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss instititute of Bioinformatics
| | - Reiner Biffar
- Clinic for Prosthetic Dentistry, Gerostomatology and Material Science, University Medicine Greifswald, Germany
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Eric Boerwinkle
- Human Genetics Center, The University of Texas Health Science Center, PO Box 20186, Houston, Texas 77225, USA
| | - Lori L. Bonnycastle
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Erwin Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Daniele Braga
- Department of Health Sciences, University of Milan,Via A. Di Rudiní, 8 20142, Milano, Italy
| | - Brendan M. Buckley
- Department of Pharmacology and Therapeutics, University College Cork, Ireland
| | - Steve Buyske
- Department of Genetics, Rutgers University, Piscataway, New Jersey 08854, USA
- Department of Statistics and Biostatistics, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Harry Campbell
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - John C. Chambers
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Francis S. Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Joanne E. Curran
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Gert J. de Borst
- Department of Vascular Surgery, Division of Surgical Specialties, UMC Utrecht, The Netherlands
| | - Anton J. M. de Craen
- Department of Gerontology and Geriatrics, Leiden University Medical Center, The Netherlands
| | - Eco J. C. de Geus
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
- EMGO+ Institute Vrije Universiteit & Vrije Universiteit Medical Center
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Graciela E. Delgado
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hester M. den Ruijter
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
| | | | - Anna L. Eriksson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
| | - Jessica D. Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, UK
| | - Terrence Forrester
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona, JMAAW15 Jamaica
| | - Karl Gertow
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Bruna Gigante
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nicola Glorioso
- Hypertension and Related Disease Centre, AOU-University of Sassari
| | - Jian Gong
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- German Center for Diabetes Research, D-85764 Neuherberg, Germany
| | - Tanja B. Grammer
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Saskia Haitjema
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
| | - Göran Hallmans
- Department of Public Health and Clinical Medicine, Section for Nutritional Research, Umeå University, Umeå, Sweden
| | - Anders Hamsten
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tamara B. Harris
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Catharina A. Hartman
- Interdisciplinary Center Psychopathology and Emotion Regulation (ICPE), University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Maija Hassinen
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Nicholas D. Hastie
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Andrew C. Heath
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Lucia Hindorff
- Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Lynne J. Hocking
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Mette Hollensted
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Germany
| | - Jouke Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Jie Huang
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Joseph Hung
- School of Medicine and Pharmacology, The University of Western Australia, 25 Stirling Hwy, Crawley, Western Australia 6009, Australia
- Department of Cardiovascular Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
| | - Nina Hutri-Kähönen
- Department of Pediatrics, Tampere University Hospital, Tampere 33521, Finland
- Department of Pediatrics, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33014, Finland
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, 751 85, Sweden
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
- Science for Life Laboratory, Uppsala University, Uppsala, 750 85, Sweden
| | - Alan L. James
- Busselton Population Medical Research Institute, Nedlands, Western Australia 6009, Australia
- School of Medicine and Pharmacology, The University of Western Australia, 25 Stirling Hwy, Crawley, Western Australia 6009, Australia
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
| | - John-Olov Jansson
- Department of Physiology, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, MRC–PHE Centre for Environment & Health, School of Public Health, Imperial College London, UK
- Center for Life Course Epidemiology, Faculty of Medicine, University of OuluP.O.Box 5000, FI-90014, Finland
- Biocenter Oulu, University of Oulu, Finland
- Unit of Primary Care, Oulu University Hospital, Kajaanintie 50, P.O.Box 20, FI-90220, 90029 Oulu, Finland
| | - Min A. Jhun
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Markus Juonala
- Department of Medicine, University of Turku, Turku 20520 Finland
- Division of Medicine, Turku University Hospital, Turku 20521, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere 33521, Finland
- Department of Clinical Physiology, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33014, Finland
| | - Magnus Karlsson
- Clinical and Molecular Osteoporosis Research Unit, Department of Orthopedics and Clinical Sciences, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Heikki A. Koistinen
- Department of Health, National Institute for Health and Welfare, Helsinki, FI-00271 Finland
- Department of Medicine and Abdominal Center: Endocrinology, University of Helsinki and Helsinki University Central Hospital, Helsinki, FI-00029 Finland
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, FI-00290 Finland
| | - Ivana Kolcic
- Department of Public Health, Faculty of Medicine, University of Split, Croatia
| | - Genovefa Kolovou
- Department of Cardiology, Onassis Cardiac Surgery Center, Athens, Greece
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Bernhard K. Krämer
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Kirsti Kvaløy
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, 7600 Levanger, Norway
| | - Timo A. Lakka
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Institute of Biomedicine/Physiology, University of Eastern Finland, Kuopio Campus, Finland
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Lenore J. Launer
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Karin Leander
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nanette R. Lee
- USC-Office of Population Studies Foundation, Inc., University of San Carlos, Cebu City 6000, Philippines
- Department of Anthropology, Sociology and History, University of San Carlos, Cebu City 6000, Philippines
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala 751 85, Sweden
| | - Cecilia M. Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Li Ka Shing Centre for Health Information and Discovery, The Big Data Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Allan Linneberg
- Research Centre for Prevention and Health, the Capital Region of Denmark, Copenhagen, Denmark
- Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stephane Lobbens
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
| | - Marie Loh
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore 138648, Singapore
| | - Mattias Lorentzon
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Robert Luben
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Gitta Lubke
- Department of Psychology, University of Notre Dame, Notre Dame, USA
| | - Anja Ludolph-Donislawski
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Sara Lupoli
- Department of Health Sciences, University of Milan,Via A. Di Rudiní, 8 20142, Milano, Italy
| | - Pamela A. F. Madden
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Reija Männikkö
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne university hospital (CHUV), Lausanne, Switzerland
| | - Nicholas G. Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Colin A. McKenzie
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona, JMAAW15 Jamaica
| | - Barbara McKnight
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington 98101, USA
- Program in Biostatistics and Biomathematics, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Dan Mellström
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Grant W. Montgomery
- Molecular Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - AW (Bill) Musk
- Busselton Population Medical Research Institute, Nedlands, Western Australia 6009, Australia
- School of Population Health, The University of Western Australia, 35 Stirling Hwy, Crawley, Western Australia 6009, Australia
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
| | - Narisu Narisu
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Germany
| | - Ilja M. Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Albertine J. Oldehinkel
- Interdisciplinary Center Psychopathology and Emotion Regulation (ICPE), University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Matthias Olden
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
| | - Ken K. Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Sandosh Padmanabhan
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland
| | - Patricia A. Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Charlotta Pisinger
- Research Center for Prevention and Health, Glostrup Hospital, Glostrup Denmark
- Department of Public Health, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - David J. Porteous
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh
| | - Olli T. Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20521, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20520, Finland
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - D. C. Rao
- Division of Biostatistics, Washington University School of Medicine, St Louis, Missouri, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Laura J. Rasmussen-Torvik
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rajesh Rawal
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Treva Rice
- Division of Biostatistics, Washington University School of Medicine, St Louis, Missouri, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
- Division of Cardiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Lynda M. Rose
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
| | - Stephanie A. Bien
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Igor Rudan
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - Serena Sanna
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale Delle Ricerche (CNR), Cittadella Universitaria di Monserrato, 09042, Monserrato, Italy
| | - Mark A. Sarzynski
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, Faculty of Medicine, Glasgow, UK
| | - Kai Savonen
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - David Schlessinger
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Salome Scholtens
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- The Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Bengt Sennblad
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
- Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Marten A. Siemelink
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
| | - Günther Silbernagel
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Austria
| | - P Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Jan A. Staessen
- Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Science , University of Leuven, Campus Sint Rafael, Kapucijnenvoer 35, Leuven; Belgium
- R&D VitaK Group, Maastricht University, Brains Unlimited Building, Oxfordlaan 55, Maastricht, The Netherlands
| | - David J. Stott
- Institute of Cardiovascular and Medical Sciences, Faculty of Medicine, University of Glasgow, UK
| | - Morris A. Swertz
- Department of Genetics, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Amy J. Swift
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Kent D. Taylor
- Center for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor/UCLA Medical Center, Torrance, California, USA
- Department of Pediatrics, University of California Los Angeles, Los Angeles, California, USA
| | - Bamidele O. Tayo
- Department of Public Health Sciences, Stritch School of Medicine, Loyola University of Chicago, Maywood, Illinois 61053, USA
| | - Barbara Thorand
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- German Center for Diabetes Research, D-85764 Neuherberg, Germany
| | - Dorothee Thuillier
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
| | - Jaakko Tuomilehto
- Research Division, Dasman Diabetes Institute, Dasman, Kuwait
- Department of Neurosciences and Preventive Medicine, Danube-University Krems, 3500 Krems, Austria
- Saudi Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Andre G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Liesbeth Vandenput
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods, Université Laval, Québec, Canada
- School of Nutrition, Université Laval, Québec, Canada
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Germany
| | - Judith M. Vonk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Gérard Waeber
- Department of Medicine, Internal Medicine, Lausanne university hospital (CHUV), Lausanne, Switzerland
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - R. G. J. Westendorp
- Department of Public Health, and Center for Healthy Ageing, University of Copenhagen, Denmark
| | - Sarah Wild
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Bruce H. R. Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCL, 33 Bedford Place, London, WC1B 5JU, UK
| | - Alan F. Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Damiano Baldassarre
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Milan, Italy
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | | | - Carsten A. Böger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Dorret I. Boomsma
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Marcel Bruinenberg
- Lifelines Cohort Study, PO Box 30001, 9700 RB Groningen, The Netherlands
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
- Division of Genetics, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Yii-DerIda Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute and Department of Pediatrics, Harbor-UCLA, Torrance, California 90502, USA
| | - Peter S. Chines
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Richard S. Cooper
- Department of Public Health Sciences, Stritch School of Medicine, Loyola University of Chicago, Maywood, Illinois 61053, USA
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale Delle Ricerche (CNR), Cittadella Universitaria di Monserrato, 09042, Monserrato, Italy
- Dipartimento di Scienze Biomediche, Universita' degli Studi di Sassari, Sassari, Italy
| | - Daniele Cusi
- Sanipedia srl, Bresso (Milano), Italy and Institute of Biomedical Technologies National Centre of Research Segrate, Milano, Italy
| | - Ulf de Faire
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore Maryland, USA
| | - Paul W. Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, SE-205 02 Malmö, Sweden
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Department of Public Health & Clinical Medicine, Umeå University, Umeå, Sweden
| | - Philippe Froguel
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
- Department of Genomics of Common Disease, Imperial College London, London, UK
| | - Penny Gordon-Larsen
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill North Carolina, 27516, USA
| | - Hans- Jörgen Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Christopher A. Haiman
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, 90089, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Kristian Hveem
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, 7600 Levanger, Norway
| | - Andrew D. Johnson
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, The Framingham Heart Study, Framingham, Massachusetts, USA
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, The Netherlands
- Durrer Center for Cardiogenetic Research, Amsterdam, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Sharon L. R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, UCL, London, WC1E 6BT, UK
| | - Jaspal S. Kooner
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
- Imperial College Healthcare NHS Trust, London, UK
- Faculty of Med, National Heart & Lung Institute, Cardiovascular Science, Hammersmith Campus, Hammersmith Hospital, Hammersmith Campus, Imperial College London, UK
| | - Diana Kuh
- MRC Unit for Lifelong Health and Ageing at UCL, 33 Bedford Place, London, WC1B 5JU, UK
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33520, Finland
- Department of Clinical Chemistry, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33014, Finland
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii 96813, USA
| | - Winfried März
- Synlab Academy, Synlab Services GmbH, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
- Oxford National Institute for Health Research (NIHR) Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Department of Biostatistics, University of Liverpool, Liverpool L69 3GL, UK
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Lyle J. Palmer
- School of Public Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Gerard Pasterkamp
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
- Laboratory of Clinical Chemistry and Hematology, Division Laboratories & Pharmacy, UMC Utrecht, The Netherlands
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- German Center for Diabetes Research, D-85764 Neuherberg, Germany
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Ozren Polasek
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
- Department of Public Health, Faculty of Medicine, University of Split, Croatia
| | - Bruce M. Psaty
- Department of Medicine, University of Washington, Seattle, Washington 98195, USA
- Department of Epidemiology, University of Washington, Seattle, Washington 98101, USA
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington 98101, USA
| | - Lu Qi
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Rainer Rauramaa
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Blair H. Smith
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
- Division of Population Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4RB, Scotland
| | - Thorkild I. A. Sørensen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Epidemiology (formerly Institute of Preventive Medicine), Bispebjerg and Frederiksberg Hospital (2000 Frederiksberg), The Capital Region, Copenhagen, Denmark
- MRC Integrative Epidemiology Unit, Bristol University, Bristol, UK
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Henning Tiemeier
- Department of Psychiatry Erasmus Medical Center, Rotterdam, The Netherlands
| | - Elena Tremoli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Milan, Italy
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, The Netherlands
- Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, The Netherlands
| | - Henrik Vestergaard
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center, Gentofte, Denmark
| | - Peter Vollenweider
- Department of Medicine, Internal Medicine, Lausanne university hospital (CHUV), Lausanne, Switzerland
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - David R. Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - John B. Whitfield
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - James F. Wilson
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - Jessica Tyrrell
- Genetics of Complex Traits, University of Exeter Medical School, RILD Building University of Exeter, Exeter, EX2 5DW, UK
- European Centre for Environment and Human Health, University of Exeter Medical School, The Knowledge Spa, Truro TR1 3HD, UK
| | - Timothy M. Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - Inês Barroso
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Level 4, Institute of Metabolic Science Box 289 Addenbrooke's Hospital Cambridge CB2 OQQ, UK
- University of Cambridge Metabolic Research Laboratories, Level 4, Institute of Metabolic Science Box 289 Addenbrooke's Hospital Cambridge CB2 OQQ, UK
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Panagiotis Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Caroline S. Fox
- NHLBI Framingham Heart Study, Framingham, Massachusetts, 01702 USA
| | - Joel N. Hirschhorn
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
- Department of Genetics, Harvard Medical School, Boston Massachusetts 02115, USA
| | - David J. Hunter
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - David P. Strachan
- Population Health Research Institute, St. George's, University of London, London, SW17 0RE, UK
- Division of Population Health Sciences and Education, St George's, University of London, London SW17 0RE, UK
| | - Cornelia M. van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3015GE, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA). Leiden, The Netherlands
- Center for Medical Systems Biology, Leiden, The Netherlands
| | - Iris M. Heid
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Karen L. Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- The Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, USA
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
- Mount Sinai School of Medicine, New York 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Tuomas O. Kilpeläinen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
- Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Ingrid B. Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine; St. Louis, Missouri, 63108 USA
| | - Kari E. North
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
- NHLBI Framingham Heart Study, Framingham, Massachusetts, 01702 USA
| |
Collapse
|
944
|
Ng MCY, Graff M, Lu Y, Justice AE, Mudgal P, Liu CT, Young K, Yanek LR, Feitosa MF, Wojczynski MK, Rand K, Brody JA, Cade BE, Dimitrov L, Duan Q, Guo X, Lange LA, Nalls MA, Okut H, Tajuddin SM, Tayo BO, Vedantam S, Bradfield JP, Chen G, Chen WM, Chesi A, Irvin MR, Padhukasahasram B, Smith JA, Zheng W, Allison MA, Ambrosone CB, Bandera EV, Bartz TM, Berndt SI, Bernstein L, Blot WJ, Bottinger EP, Carpten J, Chanock SJ, Chen YDI, Conti DV, Cooper RS, Fornage M, Freedman BI, Garcia M, Goodman PJ, Hsu YHH, Hu J, Huff CD, Ingles SA, John EM, Kittles R, Klein E, Li J, McKnight B, Nayak U, Nemesure B, Ogunniyi A, Olshan A, Press MF, Rohde R, Rybicki BA, Salako B, Sanderson M, Shao Y, Siscovick DS, Stanford JL, Stevens VL, Stram A, Strom SS, Vaidya D, Witte JS, Yao J, Zhu X, Ziegler RG, Zonderman AB, Adeyemo A, Ambs S, Cushman M, Faul JD, Hakonarson H, Levin AM, Nathanson KL, Ware EB, Weir DR, Zhao W, Zhi D, The Bone Mineral Density in Childhood Study (BMDCS) Group, Arnett DK, Grant SFA, Kardia SLR, Oloapde OI, Rao DC, Rotimi CN, Sale MM, Williams LK, Zemel BS, Becker DM, Borecki IB, et alNg MCY, Graff M, Lu Y, Justice AE, Mudgal P, Liu CT, Young K, Yanek LR, Feitosa MF, Wojczynski MK, Rand K, Brody JA, Cade BE, Dimitrov L, Duan Q, Guo X, Lange LA, Nalls MA, Okut H, Tajuddin SM, Tayo BO, Vedantam S, Bradfield JP, Chen G, Chen WM, Chesi A, Irvin MR, Padhukasahasram B, Smith JA, Zheng W, Allison MA, Ambrosone CB, Bandera EV, Bartz TM, Berndt SI, Bernstein L, Blot WJ, Bottinger EP, Carpten J, Chanock SJ, Chen YDI, Conti DV, Cooper RS, Fornage M, Freedman BI, Garcia M, Goodman PJ, Hsu YHH, Hu J, Huff CD, Ingles SA, John EM, Kittles R, Klein E, Li J, McKnight B, Nayak U, Nemesure B, Ogunniyi A, Olshan A, Press MF, Rohde R, Rybicki BA, Salako B, Sanderson M, Shao Y, Siscovick DS, Stanford JL, Stevens VL, Stram A, Strom SS, Vaidya D, Witte JS, Yao J, Zhu X, Ziegler RG, Zonderman AB, Adeyemo A, Ambs S, Cushman M, Faul JD, Hakonarson H, Levin AM, Nathanson KL, Ware EB, Weir DR, Zhao W, Zhi D, The Bone Mineral Density in Childhood Study (BMDCS) Group, Arnett DK, Grant SFA, Kardia SLR, Oloapde OI, Rao DC, Rotimi CN, Sale MM, Williams LK, Zemel BS, Becker DM, Borecki IB, Evans MK, Harris TB, Hirschhorn JN, Li Y, Patel SR, Psaty BM, Rotter JI, Wilson JG, Bowden DW, Cupples LA, Haiman CA, Loos RJF, North KE. Discovery and fine-mapping of adiposity loci using high density imputation of genome-wide association studies in individuals of African ancestry: African Ancestry Anthropometry Genetics Consortium. PLoS Genet 2017; 13:e1006719. [PMID: 28430825 PMCID: PMC5419579 DOI: 10.1371/journal.pgen.1006719] [Show More Authors] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 05/05/2017] [Accepted: 03/29/2017] [Indexed: 11/20/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified >300 loci associated with measures of adiposity including body mass index (BMI) and waist-to-hip ratio (adjusted for BMI, WHRadjBMI), but few have been identified through screening of the African ancestry genomes. We performed large scale meta-analyses and replications in up to 52,895 individuals for BMI and up to 23,095 individuals for WHRadjBMI from the African Ancestry Anthropometry Genetics Consortium (AAAGC) using 1000 Genomes phase 1 imputed GWAS to improve coverage of both common and low frequency variants in the low linkage disequilibrium African ancestry genomes. In the sex-combined analyses, we identified one novel locus (TCF7L2/HABP2) for WHRadjBMI and eight previously established loci at P < 5×10−8: seven for BMI, and one for WHRadjBMI in African ancestry individuals. An additional novel locus (SPRYD7/DLEU2) was identified for WHRadjBMI when combined with European GWAS. In the sex-stratified analyses, we identified three novel loci for BMI (INTS10/LPL and MLC1 in men, IRX4/IRX2 in women) and four for WHRadjBMI (SSX2IP, CASC8, PDE3B and ZDHHC1/HSD11B2 in women) in individuals of African ancestry or both African and European ancestry. For four of the novel variants, the minor allele frequency was low (<5%). In the trans-ethnic fine mapping of 47 BMI loci and 27 WHRadjBMI loci that were locus-wide significant (P < 0.05 adjusted for effective number of variants per locus) from the African ancestry sex-combined and sex-stratified analyses, 26 BMI loci and 17 WHRadjBMI loci contained ≤ 20 variants in the credible sets that jointly account for 99% posterior probability of driving the associations. The lead variants in 13 of these loci had a high probability of being causal. As compared to our previous HapMap imputed GWAS for BMI and WHRadjBMI including up to 71,412 and 27,350 African ancestry individuals, respectively, our results suggest that 1000 Genomes imputation showed modest improvement in identifying GWAS loci including low frequency variants. Trans-ethnic meta-analyses further improved fine mapping of putative causal variants in loci shared between the African and European ancestry populations. Genome-wide association studies (GWAS) have identified >300 genetic regions that influence body size and shape as measured by body mass index (BMI) and waist-to-hip ratio (WHR), respectively, but few have been identified in populations of African ancestry. We conducted large scale high coverage GWAS and replication of these traits in 52,895 and 23,095 individuals of African ancestry, respectively, followed by additional replication in European populations. We identified 10 genome-wide significant loci in all individuals, and an additional seven loci by analyzing men and women separately. We combined African and European ancestry GWAS and were able to narrow down 43 out of 74 African ancestry associated genetic regions to contain small number of putative causal variants. Our results highlight the improvement of applying high density genome coverage and combining multiple ancestries in the identification and refinement of location of genetic regions associated with adiposity traits.
Collapse
Affiliation(s)
- Maggie C. Y. Ng
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Mariaelisa Graff
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, Icachn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Anne E. Justice
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Poorva Mudgal
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States of America
| | - Kristin Young
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Lisa R. Yanek
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis MO, United States of America
| | - Mary K. Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis MO, United States of America
| | - Kristin Rand
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Brian E. Cade
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Latchezar Dimitrov
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Qing Duan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Leslie A. Lange
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Michael A. Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States of America
- Data Tecnica International, Glen Echo, MD, United States of America
| | - Hayrettin Okut
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Salman M. Tajuddin
- National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - Bamidele O. Tayo
- Department of Public Health Sciences, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States of America
| | - Sailaja Vedantam
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, MA, United States of America
- Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
| | - Jonathan P. Bradfield
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Wei-Min Chen
- Department of Public Health Sciences and Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Alessandra Chesi
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Marguerite R. Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Badri Padhukasahasram
- Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, MI, United States of America
| | - Jennifer A. Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Matthew A. Allison
- Division of Preventive Medicine, Department of Family Medicine and Public Health, University of California San Diego, La Jolla, CA, United States of America
| | - Christine B. Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, United States of America
| | - Elisa V. Bandera
- Department of Population Science, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States of America
| | - Traci M. Bartz
- Cardiovascular Health Research Unit, Departments of Medicine and Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States of America
| | - Leslie Bernstein
- Beckman Research Institute of the City of Hope, Duarte, CA, United States of America
| | - William J. Blot
- International Epidemiology Institute, Rockville, MD, United States of America
| | - Erwin P. Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icachn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - John Carpten
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, United States of America
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - David V. Conti
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Richard S. Cooper
- Department of Public Health Sciences, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States of America
| | - Myriam Fornage
- Center for Human Genetics, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Barry I. Freedman
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Melissa Garcia
- National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - Phyllis J. Goodman
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Yu-Han H. Hsu
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, MA, United States of America
- Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
- Program in Bioinformatics and Integrative Genomics, Harvard Medical School, Boston, MA, United States of America
| | - Jennifer Hu
- Sylvester Comprehensive Cancer Center, University of Miami Leonard Miller School of Medicine, Miami, FL, United States of America
- Department of Public Health Sciences, University of Miami Leonard Miller School of Medicine, Miami, FL, United States of America
| | - Chad D. Huff
- Department of Epidemiology, University of Texas M.D. Anderson Cancer Center, Houston, TX, United States of America
| | - Sue A. Ingles
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States of America
| | - Esther M. John
- Cancer Prevention Institute of California, Fremont, CA, United States of America
- Department of Health Research and Policy (Epidemiology) and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Rick Kittles
- Division of Urology, Department of Surgery, The University of Arizona, Tucson, AZ, United States of America
| | - Eric Klein
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, United States of America
| | - Jin Li
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, United States of America
| | - Barbara McKnight
- Cardiovascular Health Research Unit, Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Uma Nayak
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Barbara Nemesure
- Department of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States of America
| | | | - Andrew Olshan
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, Chapel Hill, NC, United States of America
| | - Michael F. Press
- Department of Pathology and Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, United States of America
| | - Rebecca Rohde
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Benjamin A. Rybicki
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, United States of America
| | | | - Maureen Sanderson
- Department of Family and Community Medicine, Meharry Medical College, Nashville, TN, United States of America
| | - Yaming Shao
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - David S. Siscovick
- The New York Academy of Medicine, New York, NY, United States of America
| | - Janet L. Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States of America
| | - Victoria L. Stevens
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, United States of America
| | - Alex Stram
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Sara S. Strom
- Department of Epidemiology, University of Texas M.D. Anderson Cancer Center, Houston, TX, United States of America
| | - Dhananjay Vaidya
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- Department of Epidemiology, Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - John S. Witte
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, United States of America
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, United States of America
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Xiaofeng Zhu
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, United States of America
| | - Regina G. Ziegler
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Alan B. Zonderman
- National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - Adebowale Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, National Cancer Institute, Bethesda, MD, United States of America
| | - Mary Cushman
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, United States of America
| | - Jessica D. Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, United States of America
| | - Hakon Hakonarson
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Albert M. Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, United States of America
| | - Katherine L. Nathanson
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Erin B. Ware
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, United States of America
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, United States of America
| | - David R. Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, United States of America
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Degui Zhi
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | | | - Donna K. Arnett
- School of Public Health, University of Kentucky, Lexington, KY, United States of America
| | - Struan F. A. Grant
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Division of Endocrinology, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Sharon L. R. Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Olufunmilayo I. Oloapde
- Center for Clinical Cancer Genetics, Department of Medicine and Human Genetics, University of Chicago, Chicago, IL, United States of America
| | - D. C. Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Charles N. Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Michele M. Sale
- Department of Public Health Sciences and Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - L. Keoki Williams
- Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, MI, United States of America
- Department of Internal Medicine, Henry Ford Health System, Detroit, MI, United States of America
| | - Babette S. Zemel
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Diane M. Becker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Ingrid B. Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis MO, United States of America
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Inc, United States of America
| | - Michele K. Evans
- National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - Tamara B. Harris
- National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - Joel N. Hirschhorn
- Division of Endocrinology and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, MA, United States of America
- Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
- Departments of Genetics and Pediatrics, Harvard Medical School, Boston, MA, United States of America
| | - Yun Li
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Sanjay R. Patel
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, WA, United States of America
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, United States of America
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States of America
- Division of Genomic Outcomes, Departments of Pediatrics and Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Los Angeles, CA, United States of America
| | - James G. Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Donald W. Bowden
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - L. Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States of America
- NHLBI Framingham Heart Study, Framingham, MA, United States of America
| | - Christopher A. Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States of America
- * E-mail: (CAH); (RJFL); (KEN)
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icachn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Mindich Child Health and Development Institute, Ichan School of Medicine at Mount Sinai, New York, NY, United States of America
- * E-mail: (CAH); (RJFL); (KEN)
| | - Kari E. North
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, United States of America
- * E-mail: (CAH); (RJFL); (KEN)
| |
Collapse
|
945
|
Zimmermann MT, Kennedy RB, Grill DE, Oberg AL, Goergen KM, Ovsyannikova IG, Haralambieva IH, Poland GA. Integration of Immune Cell Populations, mRNA-Seq, and CpG Methylation to Better Predict Humoral Immunity to Influenza Vaccination: Dependence of mRNA-Seq/CpG Methylation on Immune Cell Populations. Front Immunol 2017; 8:445. [PMID: 28484452 PMCID: PMC5399034 DOI: 10.3389/fimmu.2017.00445] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/31/2017] [Indexed: 12/21/2022] Open
Abstract
The development of a humoral immune response to influenza vaccines occurs on a multisystems level. Due to the orchestration required for robust immune responses when multiple genes and their regulatory components across multiple cell types are involved, we examined an influenza vaccination cohort using multiple high-throughput technologies. In this study, we sought a more thorough understanding of how immune cell composition and gene expression relate to each other and contribute to interindividual variation in response to influenza vaccination. We first hypothesized that many of the differentially expressed (DE) genes observed after influenza vaccination result from changes in the composition of participants' peripheral blood mononuclear cells (PBMCs), which were assessed using flow cytometry. We demonstrated that DE genes in our study are correlated with changes in PBMC composition. We gathered DE genes from 128 other publically available PBMC-based vaccine studies and identified that an average of 57% correlated with specific cell subset levels in our study (permutation used to control false discovery), suggesting that the associations we have identified are likely general features of PBMC-based transcriptomics. Second, we hypothesized that more robust models of vaccine response could be generated by accounting for the interplay between PBMC composition, gene expression, and gene regulation. We employed machine learning to generate predictive models of B-cell ELISPOT response outcomes and hemagglutination inhibition (HAI) antibody titers. The top HAI and B-cell ELISPOT model achieved an area under the receiver operating curve (AUC) of 0.64 and 0.79, respectively, with linear model coefficients of determination of 0.08 and 0.28. For the B-cell ELISPOT outcomes, CpG methylation had the greatest predictive ability, highlighting potentially novel regulatory features important for immune response. B-cell ELISOT models using only PBMC composition had lower performance (AUC = 0.67), but highlighted well-known mechanisms. Our analysis demonstrated that each of the three data sets (cell composition, mRNA-Seq, and DNA methylation) may provide distinct information for the prediction of humoral immune response outcomes. We believe that these findings are important for the interpretation of current omics-based studies and set the stage for a more thorough understanding of interindividual immune responses to influenza vaccination.
Collapse
Affiliation(s)
- Michael T Zimmermann
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA.,Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| | | | - Diane E Grill
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA.,Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| | - Ann L Oberg
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA.,Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| | - Krista M Goergen
- Department of Health Science Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | | | | | - Gregory A Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
946
|
Jasper H. Past, Present, and Future in the Relationship between Growth Retardation and the IGF System: Excerpts from the Cesar Bergada Lecture Given during the SLEP 2015 Annual Meeting. Horm Res Paediatr 2017; 86:291-299. [PMID: 27820935 DOI: 10.1159/000449287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/17/2016] [Indexed: 11/19/2022] Open
Abstract
This mini review presents a personal view about the past, the present and the future of the relationship between growth retardation and the IGF system. Looking back, it is pertinent to include a brief look at the evolution of the somatomedin hypothesis, the use of IGF-I determinations in the clinic, and a review of the literature beginning in the late 1980s with the description of mutations in the Growth Hormone Receptor (GHR) gene. The present possibly started in the mid-1990s with the description of mutations in the IGF-I gene, followed in 2003 by reports of mutations in the genes coding for the IGF-I receptor and in the signal transducer and activator of transcription 5b (STAT5b). Finally, in 2004, mutations in the IGFALS gene were described. A diffuse limit between the present and the future might have been reached (the author's arbitrary decision) with the clinical applications of whole exome sequencing, which rapidly showed mutations in genes coding for STAT3, PAPP-A2 (pregnancy-associated plasma protein A2), and IGF-II.
Collapse
Affiliation(s)
- Héctor Jasper
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), Buenos Aires, Argentina
| |
Collapse
|
947
|
de With SAJ, Pulit SL, Staal WG, Kahn RS, Ophoff RA. More than 25 years of genetic studies of clozapine-induced agranulocytosis. THE PHARMACOGENOMICS JOURNAL 2017; 17:304-311. [PMID: 28418011 DOI: 10.1038/tpj.2017.6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 12/23/2016] [Accepted: 01/18/2017] [Indexed: 12/18/2022]
Abstract
Clozapine is one of the most effective atypical antipsychotic drugs prescribed to patients with treatment-resistant schizophrenia. Approximately 1% of patients experience potential life-threatening adverse effects in the form of agranulocytosis, greatly hindering its applicability in clinical practice. The etiology of clozapine-induced agranulocytosis (CIA) remains unclear, but is thought to be a heritable trait. We reviewed the genetic studies of CIA published thus far. One recurrent finding from early candidate gene study to more recent genome-wide analysis is that of the involvement of human leukocyte antigen locus. We conclude that CIA is most likely a complex, polygenic trait, which may hamper efforts to the development of a genetic predictor test with clinical relevance. To decipher the genetic architecture of CIA, it is necessary to apply more rigorous standards of phenotyping and study much larger sample sizes.
Collapse
Affiliation(s)
- S A J de With
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - S L Pulit
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - W G Staal
- Department of Cognitive Neuroscience, Radboud University Nijmegen Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands.,Department of Psychiatry, Radboud University Nijmegen Medical Center and Karakter, Center for Child and Adolescent Psychiatry, Nijmegen, The Netherlands
| | - R S Kahn
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - R A Ophoff
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.,UCLA Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| |
Collapse
|
948
|
Nolte IM, van der Most PJ, Alizadeh BZ, de Bakker PI, Boezen HM, Bruinenberg M, Franke L, van der Harst P, Navis G, Postma DS, Rots MG, Stolk RP, Swertz MA, Wolffenbuttel BH, Wijmenga C, Snieder H. Missing heritability: is the gap closing? An analysis of 32 complex traits in the Lifelines Cohort Study. Eur J Hum Genet 2017; 25:877-885. [PMID: 28401901 DOI: 10.1038/ejhg.2017.50] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 02/03/2017] [Accepted: 02/14/2017] [Indexed: 01/08/2023] Open
Abstract
Despite the recent explosive rise in number of genetic markers for complex disease traits identified in genome-wide association studies, there is still a large gap between the known heritability of these traits and the part explained by these markers. To gauge whether this 'heritability gap' is closing, we first identified genome-wide significant SNPs from the literature and performed replication analyses for 32 highly relevant traits from five broad disease areas in 13 436 subjects of the Lifelines Cohort. Next, we calculated the variance explained by multi-SNP genetic risk scores (GRSs) for each trait, and compared it to their broad- and narrow-sense heritabilities captured by all common SNPs. The majority of all previously-associated SNPs (median=75%) were significantly associated with their respective traits. All GRSs were significant, with unweighted GRSs generally explaining less phenotypic variance than weighted GRSs, for which the explained variance was highest for height (15.5%) and varied between 0.02 and 6.7% for the other traits. Broad-sense common-SNP heritability estimates were significant for all traits, with the additive effect of common SNPs explaining 48.9% of the variance for height and between 5.6 and 39.2% for the other traits. Dominance effects were uniformly small (0-1.5%) and not significant. On average, the variance explained by the weighted GRSs accounted for only 10.7% of the common-SNP heritability of the 32 traits. These results indicate that GRSs may not yet be ready for accurate personalized prediction of complex disease traits limiting widespread adoption in clinical practice.
Collapse
Affiliation(s)
- Ilja M Nolte
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter J van der Most
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Behrooz Z Alizadeh
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Paul Iw de Bakker
- Department of Medical Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - H Marike Boezen
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerjan Navis
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dirkje S Postma
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marianne G Rots
- Department of Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ronald P Stolk
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Morris A Swertz
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bruce Hr Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Harold Snieder
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
949
|
Martin AR, Gignoux CR, Walters RK, Wojcik GL, Neale BM, Gravel S, Daly MJ, Bustamante CD, Kenny EE. Human Demographic History Impacts Genetic Risk Prediction across Diverse Populations. Am J Hum Genet 2017. [PMID: 28366442 DOI: 10.1016/j.ajhg] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023] Open
Abstract
The vast majority of genome-wide association studies (GWASs) are performed in Europeans, and their transferability to other populations is dependent on many factors (e.g., linkage disequilibrium, allele frequencies, genetic architecture). As medical genomics studies become increasingly large and diverse, gaining insights into population history and consequently the transferability of disease risk measurement is critical. Here, we disentangle recent population history in the widely used 1000 Genomes Project reference panel, with an emphasis on populations underrepresented in medical studies. To examine the transferability of single-ancestry GWASs, we used published summary statistics to calculate polygenic risk scores for eight well-studied phenotypes. We identify directional inconsistencies in all scores; for example, height is predicted to decrease with genetic distance from Europeans, despite robust anthropological evidence that West Africans are as tall as Europeans on average. To gain deeper quantitative insights into GWAS transferability, we developed a complex trait coalescent-based simulation framework considering effects of polygenicity, causal allele frequency divergence, and heritability. As expected, correlations between true and inferred risk are typically highest in the population from which summary statistics were derived. We demonstrate that scores inferred from European GWASs are biased by genetic drift in other populations even when choosing the same causal variants and that biases in any direction are possible and unpredictable. This work cautions that summarizing findings from large-scale GWASs may have limited portability to other populations using standard approaches and highlights the need for generalized risk prediction methods and the inclusion of more diverse individuals in medical genomics.
Collapse
Affiliation(s)
- Alicia R Martin
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | | | - Raymond K Walters
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | | | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Simon Gravel
- Department of Human Genetics, McGill University, Montreal, QC H3A 0G1, Canada; McGill University and Genome Quebec Innovation Centre, Montreal, QC H3A 0G1, Canada
| | - Mark J Daly
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | | | - Eimear E Kenny
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center of Statistical Genetics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
950
|
Human Demographic History Impacts Genetic Risk Prediction across Diverse Populations. Am J Hum Genet 2017; 100:635-649. [PMID: 28366442 DOI: 10.1016/j.ajhg.2017.03.004] [Citation(s) in RCA: 892] [Impact Index Per Article: 111.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 03/10/2017] [Indexed: 01/10/2023] Open
Abstract
The vast majority of genome-wide association studies (GWASs) are performed in Europeans, and their transferability to other populations is dependent on many factors (e.g., linkage disequilibrium, allele frequencies, genetic architecture). As medical genomics studies become increasingly large and diverse, gaining insights into population history and consequently the transferability of disease risk measurement is critical. Here, we disentangle recent population history in the widely used 1000 Genomes Project reference panel, with an emphasis on populations underrepresented in medical studies. To examine the transferability of single-ancestry GWASs, we used published summary statistics to calculate polygenic risk scores for eight well-studied phenotypes. We identify directional inconsistencies in all scores; for example, height is predicted to decrease with genetic distance from Europeans, despite robust anthropological evidence that West Africans are as tall as Europeans on average. To gain deeper quantitative insights into GWAS transferability, we developed a complex trait coalescent-based simulation framework considering effects of polygenicity, causal allele frequency divergence, and heritability. As expected, correlations between true and inferred risk are typically highest in the population from which summary statistics were derived. We demonstrate that scores inferred from European GWASs are biased by genetic drift in other populations even when choosing the same causal variants and that biases in any direction are possible and unpredictable. This work cautions that summarizing findings from large-scale GWASs may have limited portability to other populations using standard approaches and highlights the need for generalized risk prediction methods and the inclusion of more diverse individuals in medical genomics.
Collapse
|