Skip to main content
Advertisement
  • Loading metrics

Genome-wide physical activity interactions in adiposity ― A meta-analysis of 200,452 adults

  • Mariaelisa Graff ,

    Contributed equally to this work with: Mariaelisa Graff, Robert A. Scott, Anne E. Justice, Kristin L. Young

    migraff@email.unc.edu (MG); ruth.loos@mssm.edu (RJFL); tuomas.kilpelainen@sund.ku.dk (TOK)

    Affiliation Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America

  • Robert A. Scott ,

    Contributed equally to this work with: Mariaelisa Graff, Robert A. Scott, Anne E. Justice, Kristin L. Young

    Affiliation MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom

  • Anne E. Justice ,

    Contributed equally to this work with: Mariaelisa Graff, Robert A. Scott, Anne E. Justice, Kristin L. Young

    Affiliation Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America

  • Kristin L. Young ,

    Contributed equally to this work with: Mariaelisa Graff, Robert A. Scott, Anne E. Justice, Kristin L. Young

    Affiliations Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America, Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America

  • Mary F. Feitosa,

    Affiliation Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Llilda Barata,

    Affiliation Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Thomas W. Winkler,

    Affiliation Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany

  • Audrey Y. Chu,

    Affiliations National Heart, Lung, and Blood Institute, Framingham Heart Study, Framingham, Massachusetts, United States of America, Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America

  • Anubha Mahajan,

    Affiliation Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom

  • David Hadley,

    Affiliation Division of Population Health Sciences and Education, St. George's, University of London, London, United Kingdom

  • Luting Xue,

    Affiliations National Heart, Lung, and Blood Institute, Framingham Heart Study, Framingham, Massachusetts, United States of America, Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America

  • Tsegaselassie Workalemahu,

    Affiliation Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America

  • Nancy L. Heard-Costa,

    Affiliations National Heart, Lung, and Blood Institute, Framingham Heart Study, Framingham, Massachusetts, United States of America, Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America

  • Marcel den Hoed,

    Affiliations MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom, Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden

  • Tarunveer S. Ahluwalia,

    Affiliations 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

  • Qibin Qi,

    Affiliation Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, United States of America

  • Julius S. Ngwa,

    Affiliation Howard University, Department of Internal Medicine, Washington DC, United States of America

  • Frida Renström,

    Affiliations Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Malmö, Sweden, Department of Biobank Research, Umeå University, Umeå, Sweden

  • Lydia Quaye,

    Affiliation Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom

  • John D. Eicher,

    Affiliation Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, The Framingham Heart Study, Framingham, Massachusetts, United States of America

  • James E. Hayes,

    Affiliations Cell and Developmental Biology Graduate Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, New York, United States of America, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America

  • Marilyn Cornelis,

    Affiliations Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America, Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America

  • Zoltan Kutalik,

    Affiliations Institute of Social and Preventive Medicine, Lausanne University Hospital, Lausanne, Switzerland, Swiss Institute of Bioinformatics, Lausanne, Switzerland

  • Elise Lim,

    Affiliation Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America

  • Jian’an Luan,

    Affiliation MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom

  • Jennifer E. Huffman,

    Affiliations National Heart, Lung, and Blood Institute, Framingham Heart Study, Framingham, Massachusetts, United States of America, MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom

  • Weihua Zhang,

    Affiliations Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom, Department of Cardiology, Ealing Hospital HNS Trust, Middlesex, United Kingdom

  • Wei Zhao,

    Affiliation Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America

  • Paula J. Griffin,

    Affiliation Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America

  • Toomas Haller,

    Affiliation Estonian Genome Center, University of Tartu, Tartu, Estonia

  • Shafqat Ahmad,

    Affiliation Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Malmö, Sweden

  • Pedro M. Marques-Vidal,

    Affiliation Department of Internal Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland

  • Stephanie Bien,

    Affiliation Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America

  • Loic Yengo,

    Affiliation University of Lille, CNRS, Institut Pasteur de Lille, UMR 8199 - EGID, Lille, France

  • Alexander Teumer,

    Affiliations Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany, DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany

  • Albert Vernon Smith,

    Affiliations Icelandic Heart Association, Kopavogur, Iceland, Faculty of Medicine, University of Iceland, Reykjavik, Iceland

  • Meena Kumari,

    Affiliation ISER, University of Essex, Colchester, Essex, United Kingdom

  • Marie Neergaard Harder,

    Affiliation Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark

  • Johanne Marie Justesen,

    Affiliation Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark

  • Marcus E. Kleber,

    Affiliations Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany, Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany

  • Mette Hollensted,

    Affiliation Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark

  • Kurt Lohman,

    Affiliation Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America

  • Natalia V. Rivera,

    Affiliation Karolinska Institutet, Respiratory Unit, Department of Medicine Solna, Stockholm, Sweden

  • John B. Whitfield,

    Affiliation Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia

  • Jing Hua Zhao,

    Affiliation MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom

  • Heather M. Stringham,

    Affiliation Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America

  • Leo-Pekka Lyytikäinen,

    Affiliations Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland, Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere, Finland

  • Charlotte Huppertz,

    Affiliations Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands, EMGO+ Institute, Vrije Universiteit & VU University Medical Center, Amsterdam, The Netherlands, Department of Public and Occupational Health, VU University Medical Center, Amsterdam, The Netherlands

  • Gonneke Willemsen,

    Affiliations Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands, EMGO+ Institute, Vrije Universiteit & VU University Medical Center, Amsterdam, The Netherlands

  • Wouter J. Peyrot,

    Affiliation Department of Psychiatry, EMGO Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University Medical Center/GGZ InGeest, Amsterdam, The Netherlands

  • Ying Wu,

    Affiliation Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America

  • Kati Kristiansson,

    Affiliations National Institute for Health and Welfare, Department of Health, Helsinki, Finland, Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland

  • Ayse Demirkan,

    Affiliations Genetic Epidemiology Unit, Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands, Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands

  • Myriam Fornage,

    Affiliations Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America, Division of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, Texas, United States of America

  • Maija Hassinen,

    Affiliation Kuopio Research Institute of Exercise Medicine, Kuopio, Finland

  • Lawrence F. Bielak,

    Affiliation Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America

  • Gemma Cadby,

    Affiliation Centre for Genetic Origins of Health and Disease, University of Western Australia, Crawley, Western Australia, Australia

  • Toshiko Tanaka,

    Affiliation Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland, United States of America

  • Reedik Mägi,

    Affiliation Estonian Genome Center, University of Tartu, Tartu, Estonia

  • Peter J. van der Most,

    Affiliation Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands

  • Anne U. Jackson,

    Affiliation Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America

  • Jennifer L. Bragg-Gresham,

    Affiliation Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America

  • Veronique Vitart,

    Affiliation MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom

  • Jonathan Marten,

    Affiliation MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom

  • Pau Navarro,

    Affiliation MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom

  • Claire Bellis,

    Affiliations Human Genetics, Genome Institute of Singapore, Agency for Science, Technology and Research of Singapore, Singapore, Genomics Research Centre, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia

  • Dorota Pasko,

    Affiliation Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom

  • Åsa Johansson,

    Affiliation Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden

  • Søren Snitker,

    Affiliation Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, United States of America

  • Yu-Ching Cheng,

    Affiliations Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, United States of America, Veterans Affairs Maryland Health Care System, University of Maryland, Baltimore, Maryland, United States of America

  • Joel Eriksson,

    Affiliation Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden

  • Unhee Lim,

    Affiliation Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America

  • Mette Aadahl,

    Affiliations Research Centre for Prevention and Health, Glostrup University Hospital, Glostrup, Denmark, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark

  • Linda S. Adair,

    Affiliation Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America

  • Najaf Amin,

    Affiliation Genetic Epidemiology Unit, Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands

  • Beverley Balkau,

    Affiliation INSERM U-1018, CESP, Renal and Cardiovascular Epidemiology, UVSQ-UPS, Villejuif, France

  • Juha Auvinen,

    Affiliations Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland, Unit of Primary Care, Oulu University Hospital, Oulu, Finland

  • John Beilby,

    Affiliations Busselton Population Medical Research Institute, Nedlands, Western Australia, Australia, PathWest Laboratory Medicine of WA, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia, School of Pathology and Laboratory Medicine, The University of Western Australia, Crawley, Western Australia, Australia

  • Richard N. Bergman,

    Affiliation Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America

  • Sven Bergmann,

    Affiliations Swiss Institute of Bioinformatics, Lausanne, Switzerland, Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland

  • Alain G. Bertoni,

    Affiliations Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America

  • John Blangero,

    Affiliation Texas Biomedical Research Institute, San Antonio, Texas, United States of America

  • Amélie Bonnefond,

    Affiliation University of Lille, CNRS, Institut Pasteur de Lille, UMR 8199 - EGID, Lille, France

  • Lori L. Bonnycastle,

    Affiliation Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America

  • Judith B. Borja,

    Affiliations USC-Office of Population Studies Foundation, Inc., University of San Carlos, Cebu City, Philippines, Department of Nutrition and Dietetics, University of San Carlos, Cebu City, Philippines

  • Søren Brage,

    Affiliation MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom

  • Fabio Busonero,

    Affiliation Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale Delle Ricerche (CNR), Cittadella Universitaria di Monserrato, Monserrato, Italy

  • Steve Buyske,

    Affiliations Department of Genetics, Rutgers University, Piscataway, New Jersey, United States of America, Department of Statistics and Biostatistics, Rutgers University, Piscataway, New Jersey, United States of America

  • Harry Campbell,

    Affiliation Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, Edinburgh, Scotland

  • Peter S. Chines,

    Affiliation Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America

  • Francis S. Collins,

    Affiliation Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America

  • Tanguy Corre,

    Affiliations Swiss Institute of Bioinformatics, Lausanne, Switzerland, Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland

  • George Davey Smith,

    Affiliation MRC Integrative Epidemiology Unit & School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom

  • Graciela E. Delgado,

    Affiliation Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany

  • Nicole Dueker,

    Affiliation University of Maryland School of Medicine, Department of Epidemiology & Public Health, Baltimore, Maryland, United States of America

  • Marcus Dörr,

    Affiliations DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany, Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany

  • Tapani Ebeling,

    Affiliations Department of Medicine, Oulu University Hospital, Oulu, Finland, Institute of Clinical Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland

  • Gudny Eiriksdottir,

    Affiliation Icelandic Heart Association, Kopavogur, Iceland

  • Tõnu Esko,

    Affiliations Estonian Genome Center, University of Tartu, Tartu, Estonia, Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, United States of America, Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America, Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America

  • Jessica D. Faul,

    Affiliation Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, United States of America

  • Mao Fu,

    Affiliation Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, United States of America

  • Kristine Færch,

    Affiliation Steno Diabetes Center, Gentofte, Denmark

  • Christian Gieger,

    Affiliations Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany, Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany, Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany

  • Sven Gläser,

    Affiliation Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany

  • Jian Gong,

    Affiliation Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America

  • Penny Gordon-Larsen,

    Affiliations Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America, Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America

  • Harald Grallert,

    Affiliations Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany, Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany, German Center for Diabetes Research (DZD), München-Neuherberg, Germany

  • Tanja B. Grammer,

    Affiliation Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany

  • Niels Grarup,

    Affiliation Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark

  • Gerard van Grootheest,

    Affiliation Department of Psychiatry, EMGO Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University Medical Center/GGZ InGeest, Amsterdam, The Netherlands

  • Kennet Harald,

    Affiliation National Institute for Health and Welfare, Department of Health, Helsinki, Finland

  • Nicholas D. Hastie,

    Affiliation MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom

  • Aki S. Havulinna,

    Affiliation National Institute for Health and Welfare, Department of Health, Helsinki, Finland

  • Dena Hernandez,

    Affiliation Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, United States of America

  • Lucia Hindorff,

    Affiliation Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America

  • Lynne J. Hocking,

    Affiliations Musculoskeletal Research Programme, Division of Applied Medicine, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom, Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom

  • Oddgeir L. Holmens,

    Affiliation St. Olav Hospital, Trondheim University Hospital, Trondheim, Norway

  • Christina Holzapfel,

    Affiliations Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany, Institute for Nutritional Medicine, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany

  • Jouke Jan Hottenga,

    Affiliations Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands, NCA Institute, VU University & VU Medical Center, Amsterdam, The Netherlands

  • Jie Huang,

    Affiliation Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom

  • Tao Huang,

    Affiliation Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America

  • Jennie Hui,

    Affiliations Busselton Population Medical Research Institute, Nedlands, Western Australia, Australia, PathWest Laboratory Medicine of WA, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia, School of Population Health, The University of Western Australia, Crawley, Western Australia, Australia

  • Cornelia Huth,

    Affiliations Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany, German Center for Diabetes Research (DZD), München-Neuherberg, Germany

  • Nina Hutri-Kähönen,

    Affiliations Department of Pediatrics, Tampere University Hospital, Tampere, Finland, Department of Pediatrics, University of Tampere School of Medicine, Tampere, Finland

  • Alan L. James,

    Affiliations Busselton Population Medical Research Institute, Nedlands, Western Australia, Australia, Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia, School of Medicine and Pharmacology, The University of Western Australia, Crawley, Western Australia, Australia

  • John-Olov Jansson,

    Affiliation Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden

  • Min A. Jhun,

    Affiliation Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America

  • Markus Juonala,

    Affiliations Department of Medicine, University of Turku, Turku, Finland, Division of Medicine, Turku University Hospital, Turku, Finland

  • Leena Kinnunen,

    Affiliation National Institute for Health and Welfare, Department of Health, Helsinki, Finland

  • Heikki A. Koistinen,

    Affiliations National Institute for Health and Welfare, Department of Health, Helsinki, Finland, Department of Medicine and Abdominal Center: Endocrinology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland, Minerva Foundation Institute for Medical Research, Helsinki, Finland

  • Ivana Kolcic,

    Affiliation Department of Public Health, Faculty of Medicine, University of Split, Split, Croatia

  • Pirjo Komulainen,

    Affiliation Kuopio Research Institute of Exercise Medicine, Kuopio, Finland

  • Johanna Kuusisto,

    Affiliation Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland

  • Kirsti Kvaløy,

    Affiliation HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway

  • Mika Kähönen,

    Affiliations Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland, Department of Clinical Physiology, University of Tampere School of Medicine, Tampere, Finland

  • Timo A. Lakka,

    Affiliations Kuopio Research Institute of Exercise Medicine, Kuopio, Finland, Institute of Biomedicine, Physiology, University of Eastern Finland, Kuopio Campus, Finland

  • Lenore J. Launer,

    Affiliation Neuroepidemiology Section, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America

  • Benjamin Lehne,

    Affiliation Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom

  • Cecilia M. Lindgren,

    Affiliations Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, United States of America, The Big Data Institute, University of Oxford, Oxford, United Kingdom

  • Mattias Lorentzon,

    Affiliations Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden, Geriatric Medicine, Sahlgrenska University Hospital, Mölndal, Sweden

  • Robert Luben,

    Affiliation Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom

  • Michel Marre,

    Affiliations INSERM U-1138, Équipe 2: Pathophysiology and Therapeutics of Vascular and Renal diseases Related to Diabetes, Centre de Recherche des Cordeliers, Paris, France, Department of Endocrinology, Diabetology, Nutrition, and Metabolic Diseases, Bichat Claude Bernard Hospital, Paris, France

  • Yuri Milaneschi,

    Affiliation Department of Psychiatry, EMGO Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University Medical Center/GGZ InGeest, Amsterdam, The Netherlands

  • Keri L. Monda,

    Affiliations Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America, Center for Observational Research, Amgen Inc., Thousand Oaks, California, United States of America

  • Grant W. Montgomery,

    Affiliation Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia

  • Marleen H. M. De Moor,

    Affiliations EMGO+ Institute, Vrije Universiteit & VU University Medical Center, Amsterdam, The Netherlands, Section of Clinical Child and Family Studies, Department of Educational and Family Studies, Vrije Universiteit, Amsterdam, The Netherlands

  • Antonella Mulas,

    Affiliations Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale Delle Ricerche (CNR), Cittadella Universitaria di Monserrato, Monserrato, Italy, Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy

  • Martina Müller-Nurasyid,

    Affiliations Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany, Department of Medicine I, Ludwig-Maximilians-Universität, Munich, Germany, DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany

  • A. W. Musk,

    Affiliations Busselton Population Medical Research Institute, Nedlands, Western Australia, Australia, School of Population Health, The University of Western Australia, Crawley, Western Australia, Australia, Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia

  • Reija Männikkö,

    Affiliation Kuopio Research Institute of Exercise Medicine, Kuopio, Finland

  • Satu Männistö,

    Affiliation National Institute for Health and Welfare, Department of Health, Helsinki, Finland

  • Narisu Narisu,

    Affiliation Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America

  • Matthias Nauck,

    Affiliations DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany, Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany

  • Jennifer A. Nettleton,

    Affiliation Division of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, Texas, United States of America

  • Ilja M. Nolte,

    Affiliation Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands

  • Albertine J. Oldehinkel,

    Affiliation Interdisciplinary Center Psychopathology and Emotion Regulation (ICPE), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands

  • Matthias Olden,

    Affiliation Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany

  • Ken K. Ong,

    Affiliation MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom

  • Sandosh Padmanabhan,

    Affiliations Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom, Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom

  • Lavinia Paternoster,

    Affiliation MRC Integrative Epidemiology Unit & School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom

  • Jeremiah Perez,

    Affiliation Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America

  • Markus Perola,

    Affiliations National Institute for Health and Welfare, Department of Health, Helsinki, Finland, Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland, University of Tartu, Estonian Genome Centre, Tartu, Estonia

  • Annette Peters,

    Affiliations Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany, German Center for Diabetes Research (DZD), München-Neuherberg, Germany, DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany

  • Ulrike Peters,

    Affiliation Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America

  • Patricia A. Peyser,

    Affiliation Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America

  • Inga Prokopenko,

    Affiliation Genomics of Common Disease, Imperial College London, London, United Kingdom

  • Hannu Puolijoki,

    Affiliation South Ostrobothnia Central Hospital, Seinäjoki, Finland

  • Olli T. Raitakari,

    Affiliations Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland

  • Tuomo Rankinen,

    Affiliation Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States of America

  • Laura J. Rasmussen-Torvik,

    Affiliation Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America

  • Rajesh Rawal,

    Affiliations Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany, Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany, Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany

  • Paul M. Ridker,

    Affiliations Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America, Harvard Medical School, Boston, Massachusetts, United States of America

  • Lynda M. Rose,

    Affiliation Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America

  • Igor Rudan,

    Affiliation Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, Edinburgh, Scotland

  • Cinzia Sarti,

    Affiliation Social Services and Health Care Department, City of Helsinki, Helsinki, Finland

  • Mark A. Sarzynski,

    Affiliation Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States of America

  • Kai Savonen,

    Affiliation Kuopio Research Institute of Exercise Medicine, Kuopio, Finland

  • William R. Scott,

    Affiliation Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom

  • Serena Sanna,

    Affiliation Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale Delle Ricerche (CNR), Cittadella Universitaria di Monserrato, Monserrato, Italy

  • Alan R. Shuldiner,

    Affiliations Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, United States of America, Veterans Affairs Maryland Health Care System, University of Maryland, Baltimore, Maryland, United States of America

  • Steve Sidney,

    Affiliation Division of Research, Kaiser Permanente Northern California, Oakland, California, United States of America

  • Günther Silbernagel,

    Affiliation Division of Angiology, Department of Internal Medicine, Medical University Graz, Austria

  • Blair H. Smith,

    Affiliations Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom, School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, Scotland

  • Jennifer A. Smith,

    Affiliation Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America

  • Harold Snieder,

    Affiliation Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands

  • Alena Stančáková,

    Affiliation Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland

  • Barbara Sternfeld,

    Affiliation Division of Research, Kaiser Permanente Northern California, Oakland, California, United States of America

  • Amy J. Swift,

    Affiliation Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America

  • Tuija Tammelin,

    Affiliation LIKES Research Center for Sport and Health Sciences, Jyväskylä, Finland

  • Sian-Tsung Tan,

    Affiliation National Heart and Lung Institute, Imperial College London, United Kingdom

  • Barbara Thorand,

    Affiliations Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany, German Center for Diabetes Research (DZD), München-Neuherberg, Germany

  • Dorothée Thuillier,

    Affiliation University of Lille, CNRS, Institut Pasteur de Lille, UMR 8199 - EGID, Lille, France

  • Liesbeth Vandenput,

    Affiliation Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden

  • Henrik Vestergaard,

    Affiliations 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

  • Jana V. van Vliet-Ostaptchouk,

    Affiliation Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands

  • Marie-Claude Vohl,

    Affiliations Institute of Nutrition and Functional Foods, Quebec, Canada, School of Nutrition, Laval University, Quebec, Canada

  • Uwe Völker,

    Affiliations DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Germany

  • Gérard Waeber,

    Affiliation Department of Internal Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland

  • Mark Walker,

    Affiliation Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom

  • Sarah Wild,

    Affiliation Centre for Population Health Sciences, Usher Institute for Population Health Sciences and Informatics, Teviot Place, Edinburgh, Scotland

  • Andrew Wong,

    Affiliation MRC Unit for Lifelong Health and Ageing at UCL, London, United Kingdom

  • Alan F. Wright,

    Affiliation MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom

  • M. Carola Zillikens,

    Affiliation Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands

  • Niha Zubair,

    Affiliation Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America

  • Christopher A. Haiman,

    Affiliation Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America

  • Loic Lemarchand,

    Affiliation Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America

  • Ulf Gyllensten,

    Affiliation Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden

  • Claes Ohlsson,

    Affiliation Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden

  • Albert Hofman,

    Affiliations Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands, Netherlands Consortium for Healthy Aging, Leiden University Medical Center, Leiden, The Netherlands

  • Fernando Rivadeneira,

    Affiliations Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands, Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands, Netherlands Consortium for Healthy Aging, Leiden University Medical Center, Leiden, The Netherlands

  • André G. Uitterlinden,

    Affiliations Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands, Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands

  • Louis Pérusse,

    Affiliations Institute of Nutrition and Functional Foods, Quebec, Canada, Department of Kinesiology, Laval University, Quebec, Canada

  • James F. Wilson,

    Affiliations MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom, Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, Edinburgh, Scotland

  • Caroline Hayward,

    Affiliation MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom

  • Ozren Polasek,

    Affiliations Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, Edinburgh, Scotland, Department of Public Health, Faculty of Medicine, University of Split, Split, Croatia

  • Francesco Cucca,

    Affiliations Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale Delle Ricerche (CNR), Cittadella Universitaria di Monserrato, Monserrato, Italy, Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy

  • Kristian Hveem,

    Affiliation HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway

  • Catharina A. Hartman,

    Affiliation Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands

  • Anke Tönjes,

    Affiliation University of Leipzig, Medical Department, Leipzig, Germany

  • Stefania Bandinelli,

    Affiliation Geriatric Unit, Azienda Sanitaria Firenze, Florence, Italy

  • Lyle J. Palmer,

    Affiliation School of Public Health, University of Adelaide, Adelaide, South Australia, Australia

  • Sharon L. R. Kardia,

    Affiliation Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America

  • Rainer Rauramaa,

    Affiliations Kuopio Research Institute of Exercise Medicine, Kuopio, Finland, Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland

  • Thorkild I. A. Sørensen,

    Affiliations 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 Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark, MRC Integrative Epidemiology Unit & School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom, Department of Clinical Epidemiology, Bispebjerg and Frederiksberg Hospitals, The Capital Region, Copenhagen, Denmark

  • Jaakko Tuomilehto,

    Affiliations National Institute for Health and Welfare, Department of Health, Helsinki, Finland, Centre for Vascular Prevention, Danube-University Krems, Krems, Austria, Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia

  • Veikko Salomaa,

    Affiliation National Institute for Health and Welfare, Department of Health, Helsinki, Finland

  • Brenda W. J. H. Penninx,

    Affiliation Department of Psychiatry, EMGO Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University Medical Center/GGZ InGeest, Amsterdam, The Netherlands

  • Eco J. C. de Geus,

    Affiliations Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands, EMGO+ Institute, Vrije Universiteit & VU University Medical Center, Amsterdam, The Netherlands

  • Dorret I. Boomsma,

    Affiliations Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands, NCA Institute, VU University & VU Medical Center, Amsterdam, The Netherlands

  • Terho Lehtimäki,

    Affiliations Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland, Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere, Finland

  • Massimo Mangino,

    Affiliations Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom, National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas' Foundation Trust, London, United Kingdom

  • Markku Laakso,

    Affiliation Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland

  • Claude Bouchard,

    Affiliation Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States of America

  • Nicholas G. Martin,

    Affiliation Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia

  • Diana Kuh,

    Affiliation MRC Unit for Lifelong Health and Ageing at UCL, London, United Kingdom

  • Yongmei Liu,

    Affiliation Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America

  • Allan Linneberg,

    Affiliations Research Centre for Prevention and Health, Glostrup University Hospital, Glostrup, Denmark, Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark, Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark

  • Winfried März,

    Affiliations Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany, Synlab Academy, Synlab Services LLC, Mannheim, Germany, Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria

  • Konstantin Strauch,

    Affiliations Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany, Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany

  • Mika Kivimäki,

    Affiliation Department of Epidemiology and Public Health, University College London, London, United Kingdom

  • Tamara B. Harris,

    Affiliation Laboratory of Epidemiology and Population Science, National Institute on Aging, Bethesda, Maryland, United States of America

  • Vilmundur Gudnason,

    Affiliations Icelandic Heart Association, Kopavogur, Iceland, Faculty of Medicine, University of Iceland, Reykjavik, Iceland

  • Henry Völzke,

    Affiliations Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany, DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany

  • Lu Qi,

    Affiliation Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America

  • Marjo-Riitta Järvelin,

    Affiliations Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom, Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland, Unit of Primary Care, Oulu University Hospital, Oulu, Finland, Biocenter Oulu, University of Oulu, Oulu, Finland, MRC-PHE Centre for Environment and Health, Imperial College London, London, United Kingdom

  • John C. Chambers,

    Affiliations Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom, Department of Cardiology, Ealing Hospital HNS Trust, Middlesex, United Kingdom, Imperial College Healthcare NHS Trust, London, United Kingdom

  • Jaspal S. Kooner,

    Affiliations Department of Cardiology, Ealing Hospital HNS Trust, Middlesex, United Kingdom, National Heart and Lung Institute, Imperial College London, United Kingdom, Imperial College Healthcare NHS Trust, London, United Kingdom

  • Philippe Froguel,

    Affiliations University of Lille, CNRS, Institut Pasteur de Lille, UMR 8199 - EGID, Lille, France, Hammersmith Hospital, London, United Kingdom

  • Charles Kooperberg,

    Affiliation Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America

  • Peter Vollenweider,

    Affiliation Department of Internal Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland

  • Göran Hallmans,

    Affiliation Department of Biobank Research, Umeå University, Umeå, Sweden

  • Torben Hansen,

    Affiliation Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark

  • Oluf Pedersen,

    Affiliation Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark

  • Andres Metspalu,

    Affiliation Estonian Genome Center, University of Tartu, Tartu, Estonia

  • Nicholas J. Wareham,

    Affiliation MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom

  • Claudia Langenberg,

    Affiliation MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom

  • David R. Weir,

    Affiliation Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, United States of America

  • David J. Porteous,

    Affiliations Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom

  • Eric Boerwinkle,

    Affiliation Division of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, Texas, United States of America

  • Daniel I. Chasman,

    Affiliations Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America, Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America, Harvard Medical School, Boston, Massachusetts, United States of America

  • CHARGE Consortium,
  • EPIC-InterAct Consortium,
  • PAGE Consortium ,

    Membership is listed in the Supporting Information.

  • Gonçalo R. Abecasis,

    Affiliation Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America

  • Inês Barroso,

    Affiliations Wellcome Trust Sanger Institute, Hinxton, United Kingdom, NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom, The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, United Kingdom

  • Mark I. McCarthy,

    Affiliations Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom, Oxford NIHR Biomedical Research Centre, Oxford, United Kingdom

  • Timothy M. Frayling,

    Affiliation Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom

  • Jeffrey R. O’Connell,

    Affiliation Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, United States of America

  • Cornelia M. van Duijn,

    Affiliations Genetic Epidemiology Unit, Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands, Netherlands Consortium for Healthy Aging, Leiden University Medical Center, Leiden, The Netherlands, Center of Medical Systems Biology, Leiden, The Netherlands

  • Michael Boehnke,

    Affiliation Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America

  • Iris M. Heid,

    Affiliation Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany

  • Karen L. Mohlke,

    Affiliation Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America

  • David P. Strachan,

    Affiliation Population Health Research Institute, St. George's University of London, London, United Kingdom

  • Caroline S. Fox,

    Affiliation Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, The Framingham Heart Study, Framingham, Massachusetts, United States of America

  • Ching-Ti Liu,

    Affiliation Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America

  • Joel N. Hirschhorn,

    Affiliations Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America, Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America, Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts, United States of America

  • Robert J. Klein,

    Affiliation Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America

  • Andrew D. Johnson,

    Affiliations National Heart, Lung, and Blood Institute, Framingham Heart Study, Framingham, Massachusetts, United States of America, Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, The Framingham Heart Study, Framingham, Massachusetts, United States of America

  • Ingrid B. Borecki,

    Affiliation Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Paul W. Franks,

    Affiliations Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America, Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Malmö, Sweden, Department of Public Health & Clinical Medicine, Umeå University, Umeå, Sweden

  • Kari E. North,

    Affiliation Carolina Center for Genome Sciences, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America

  • L. Adrienne Cupples,

    Affiliations National Heart, Lung, and Blood Institute, Framingham Heart Study, Framingham, Massachusetts, United States of America, Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America

  • Ruth J. F. Loos ,

    migraff@email.unc.edu (MG); ruth.loos@mssm.edu (RJFL); tuomas.kilpelainen@sund.ku.dk (TOK)

    ‡ These authors jointly supervised this work.

    Affiliations MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom, Genetics of Obesity and Related Metabolic Traits Program, Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America, The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America

  •  [ ... ],
  • Tuomas O. Kilpeläinen

    migraff@email.unc.edu (MG); ruth.loos@mssm.edu (RJFL); tuomas.kilpelainen@sund.ku.dk (TOK)

    ‡ These authors jointly supervised this work.

    Affiliations MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark, The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America

  • [ view all ]
  • [ view less ]

Correction

23 Aug 2017: Graff M, Scott RA, Justice AE, Young KL, Feitosa MF, et al. (2017) Correction: Genome-wide physical activity interactions in adiposity ― A meta-analysis of 200,452 adults. PLOS Genetics 13(8): e1006972. https://doi.org/10.1371/journal.pgen.1006972 View correction

Abstract

Physical activity (PA) may modify the genetic effects that give rise to increased risk of obesity. To identify adiposity loci whose effects are modified by PA, we performed genome-wide interaction meta-analyses of BMI and BMI-adjusted waist circumference and waist-hip ratio from up to 200,452 adults of European (n = 180,423) or other ancestry (n = 20,029). We standardized PA by categorizing it into a dichotomous variable where, on average, 23% of participants were categorized as inactive and 77% as physically active. While we replicate the interaction with PA for the strongest known obesity-risk locus in the FTO gene, of which the effect is attenuated by ~30% in physically active individuals compared to inactive individuals, we do not identify additional loci that are sensitive to PA. In additional genome-wide meta-analyses adjusting for PA and interaction with PA, we identify 11 novel adiposity loci, suggesting that accounting for PA or other environmental factors that contribute to variation in adiposity may facilitate gene discovery.

Author summary

Decline in daily physical activity is thought to be a key contributor to the global obesity epidemic. However, the impact of sedentariness on adiposity may be in part determined by a person’s genetic constitution. The specific genetic variants that are sensitive to physical activity and regulate adiposity remain largely unknown. Here, we aimed to identify genetic variants whose effects on adiposity are modified by physical activity by examining ~2.5 million genetic variants in up to 200,452 individuals. We also tested whether adjusting for physical activity as a covariate could lead to the identification of novel adiposity variants. We find robust evidence of interaction with physical activity for the strongest known obesity risk-locus in the FTO gene, of which the body mass index-increasing effect is attenuated by ~30% in physically active individuals compared to inactive individuals. Our analyses indicate that other similar gene-physical activity interactions may exist, but better measurement of physical activity, larger sample sizes, and/or improved analytical methods will be required to identify them. Adjusting for physical activity, we identify 11 novel adiposity variants, suggesting that accounting for physical activity or other environmental factors that contribute to variation in adiposity may facilitate gene discovery.

Introduction

In recent decades, we have witnessed a global obesity epidemic that may be driven by changes in lifestyle such as easier access to energy-dense foods and decreased physical activity (PA) [1]. However, not everyone becomes obese in obesogenic environments. Twin studies suggest that changes in body weight in response to lifestyle interventions are in part determined by a person’s genetic constitution [24]. Nevertheless, the genes that are sensitive to environmental influences remain largely unknown.

Previous studies suggest that genetic susceptibility to obesity, assessed by a genetic risk score for BMI, may be attenuated by PA [5, 6]. A large-scale meta-analysis of the FTO obesity locus in 218,166 adults showed that being physically active attenuates the BMI-increasing effect of this locus by ~30% [7]. While these findings suggest that FTO, and potentially other previously established BMI loci, may interact with PA, it has been hypothesized that loci showing the strongest main effect associations in genome-wide association studies (GWAS) may be the least sensitive to environmental and lifestyle influences, and may therefore not make the best candidates for interactions [8]. Yet no genome-wide search for novel loci exhibiting SNP×PA interaction has been performed. A genome-wide meta-analysis of genotype-dependent phenotypic variance of BMI, a marker of sensitivity to environmental exposures, in ~170,000 participants identified FTO, but did not show robust evidence of environmental sensitivity for other loci [9]. Recent genome-wide meta-analyses of adiposity traits in >320,000 individuals uncovered loci interacting with age and sex, but also suggested that very large sample sizes are required for interaction studies to be successful [10].

Here, we report results from a large-scale genome-wide meta-analysis of SNP×PA interactions in adiposity in up to 200,452 adults. As part of these interaction analyses, we also examine whether adjusting for PA or jointly testing for SNP’s main effect and interaction with PA may identify novel adiposity loci.

Results

Identification of loci interacting with PA

We performed meta-analyses of results from 60 studies, including up to 180,423 adults of European descent and 20,029 adults of other ancestries to assess interactions between ~2.5 million genotyped or HapMap-imputed SNPs and PA on BMI and BMI-adjusted waist circumference (WCadjBMI) and waist-hip ratio (WHRadjBMI) (S1S5 Tables). Similar to a previous meta-analysis of the interaction between FTO and PA [7], we standardized PA by categorizing it into a dichotomous variable where on average ~23% of participants were categorized as inactive and ~77% as physically active (see Methods and S6 Table). On average, inactive individuals had 0.99 kg/m2 higher BMI, 3.46 cm higher WC, and 0.018 higher WHR than active individuals (S4 and S5 Tables).

Each study first performed genome-wide association analyses for each SNP’s effect on BMI in the inactive and active groups separately. Corresponding summary statistics from each cohort were subsequently meta-analyzed, and the SNP×PA interaction effect was estimated by calculating the difference in the SNP’s effect between the inactive and active groups. To identify sex-specific SNP×PA interactions, we performed the meta-analyses separately in men and women, as well as in the combined sample. In addition, we carried out meta-analyses in European-ancestry studies only and in European and other-ancestry studies combined.

We used two approaches to identify loci whose effects are modified by PA. In the first approach, we searched for genome-wide significant SNP×PA interaction effects (PINT<5x10-8). As shown in Fig 1, this approach yielded the highest power to identify cross-over interaction effects where the SNP’s effect is directionally opposite between the inactive and active groups. However, this approach has low power to identify interaction effects where the SNP’s effect is directionally concordant between the inactive and active groups (Fig 1). We identified a genome-wide significant interaction between rs986732 in cadherin 12 (CDH12) and PA on BMI in European-ancestry studies (betaINT = -0.076 SD/allele, PINT = 3.1x10-8, n = 134,767) (S7 Table). The interaction effect was directionally consistent but did not replicate in an independent sample of 31,097 individuals (betaINT = -0.019 SD/allele, PINT = 0.52), and the pooled association P value for the discovery and replication stages combined did not reach genome-wide significance (NTOTAL = 165,864; PINT-TOTAL = 3x10-7) (S1 Fig). No loci showed genome-wide significant interactions with PA on WCadjBMI or WHRadjBMI. CDH12 encodes an integral membrane protein mediating calcium-dependent cell-cell adhesion in the brain, where it may play a role in neurogenesis [11]. While CDH12 rs4701252 and rs268972 SNPs have shown suggestive associations with waist circumference (P = 2x10-6) and BMI (P = 5x10-5) in previous GWAS [12, 13], the SNPs are not in LD with rs986732 (r2<0.1).

thumbnail
Fig 1. Power to identify PA-adjusted main, joint or GxPA interaction effects in 200,000 individuals (45,000 inactive, 155,000 active).

The plots compare power to identify genome-wide significant main effects (PadjPA<5x10-8, dashed black), joint effects (PJOINT<5x10-8, dotted green) or GxPA interaction effects (PINT<5x10-8, solid magenta) as well as the power to identify Bonferroni-corrected interaction effects (PINT<0.05/number of loci, solid orange) for the SNPs that reached a genome-wide significant PA-adjusted main effect association (PadjPA<5x10-8). The power computations were based on analytical power formulae provided elsewhere [50] and were conducted a-priori based on various types of known realistic BMI effect sizes [51]. Panels A, C, E: Assuming an effect in inactive individuals similar to a small (, comparable to the known BMI effect of the NUDT3 locus), medium (, comparable to the known BMI effect of the BDNF locus) and large (, comparable to the known BMI effect of the FTO locus) realistic effect on BMI and for various effects in physically active individuals (varied on the x axis); Panels B,D,F: Assuming an effect in physically active individuals similar to the small, medium and large realistic effects of the NUDT3, BDNF and FTO loci on BMI and for various effects in inactive individuals (varied on x axis).

https://doi.org/10.1371/journal.pgen.1006528.g001

In our second approach, we tested interaction for loci showing a genome-wide significant main effect on BMI, WCadjBMI or WHRadjBMI (S7S12 Tables). We adjusted the significance threshold for SNP×PA interaction by Bonferroni correction (P = 0.05/number of SNPs tested). As shown in Fig 1, this approach enhanced our power to identify interaction effects where there is a difference in the magnitude of the SNP’s effect between inactive and active groups when the SNP’s effect is directionally concordant between the groups. We identified a significant SNP×PA interaction of the FTO rs9941349 SNP on BMI in the meta-analysis of European-ancestry individuals; the BMI-increasing effect was 33% smaller in active individuals (betaACTIVE = 0.072 SD/allele) than in inactive individuals (betaINACTIVE = 0.106 SD/allele, PINT = 4x10-5). The rs9941349 SNP is in strong LD (r2 = 0.87) with FTO rs9939609 for which interaction with PA has been previously established in a meta-analysis of 218,166 adults [7]. We identified no loci interacting with PA for WCadjBMI or WHRadjBMI.

In a previously published meta-analysis [7], the FTO locus showed a geographic difference for the interaction effect where the interaction was more pronounced in studies from North America than in those from Europe. To test for geographic differences in the present study, we performed additional meta-analyses for the FTO rs9941349 SNP, stratified by geographic origin (North America vs. Europe). While the interaction effect was more pronounced in studies from North America (betaINT = 0.052 SD/allele, P = 5x10-4, N = 63,896) than in those from Europe (betaINT = 0.028 SD/allele, P = 0.006, N = 109,806), we did not find a statistically significant difference between the regions (P = 0.14).

Explained phenotypic variance in inactive and active individuals.

We tested whether the variance explained by ~1.1 million common variants (MAF≥1%) differed between the inactive and active groups for BMI, WCadjBMI, and WHRadjBMI [14]. In the physically active individuals, the variants explained ~20% less of variance in BMI than in inactive individuals (12.4% vs. 15.7%, respectively; Pdifference = 0.046), suggesting that PA may reduce the impact of genetic predisposition to adiposity overall. There was no significant difference in the variance explained between active and inactive groups for WCadjBMI (8.6% for active, 9.3% for inactive; Pdifference = 0.70) or WHRadjBMI (6.9% for active, 8.0% for inactive; Pdifference = 0.59).

To further investigate differences in explained variance between the inactive and active groups, we calculated variance explained by subsets of SNPs selected based on significance thresholds (ranging from P = 5x10-8 to P = 0.05) of PA-adjusted SNP association with BMI, WCadjBMI or WHRadjBMI [15] (S13 Table). We found 17–26% smaller explained variance for BMI in the active group than in the inactive group at all P value thresholds (S13 Table).

Identification of novel loci when adjusting for PA or when jointly testing for SNP main effect and interaction with PA

Physical activity contributes to variation in BMI, WCadjBMI, and WHRadjBMI, hence, adjusting for PA as a covariate may enhance power to identify novel adiposity loci. To that extent, each study performed genome-wide analyses for association with BMI, WCadjBMI, and WHRadjBMI while adjusting for PA. Subsequently, we performed meta-analyses of the study-specific results. We discovered 10 genome-wide significant loci (2 for BMI, 1 for WCadjBMI, 7 for WHRadjBMI) that have not been reported in previous GWAS of adiposity traits (Table 1, S2S4 Figs).

thumbnail
Table 1. Novel loci achieving genome-wide significance (P<5x10-8) in meta-analyses for PA-adjusted SNP main effect (PadjPA) or the joint test of SNP main effect and SNP-PA interaction (Pjoint).

https://doi.org/10.1371/journal.pgen.1006528.t001

To establish whether additionally accounting for SNP×PA interactions would identify novel loci, we calculated the joint significance of PA-adjusted SNP main effect and SNP×PA interaction using the method of Aschard et al [16]. As illustrated in Fig 1, the joint test enhanced our power to identify loci where the SNP shows simultaneously a main effect and an interaction effect. We identified a novel BMI locus near ELAVL2 in men (PJOINT = 4x10-8), which also showed suggestive evidence of interaction with PA (PINT = 9x10-4); the effect of the BMI-increasing allele was attenuated by 71% in active as compared to inactive individuals (betaINACTIVE = 0.087 SD/allele, betaACTIVE = 0.025 SD/allele) (Table 1, S2S4 Figs).

To evaluate the effect of PA adjustment on the results for the 11 novel loci, we performed a look-up in published GIANT consortium meta-analyses for BMI, WCadjBMI, and WHRadjBMI that did not adjust for PA [17, 18] (S22 Table). All 11 loci showed a consistent direction of effect between the present PA-adjusted and the previously published PA-unadjusted results, but the PA-unadjusted associations were less pronounced despite up to 40% greater sample size, suggesting that adjustment for PA may have increased our power to identify these loci.

The biological relevance of putative candidate genes in the novel loci, based on our thorough searches of the literature, GWAS catalog look-ups, and analyses of eQTL enrichment and overlap with functional regulatory elements, are described in Tables 2 and 3. As the novel loci were identified in a PA-adjusted model, where adjusting for PA may have contributed to their identification, we examined whether the lead SNPs in these loci are associated with the level of PA. More specifically, we performed look-ups in GWAS analyses for the levels of moderate-to-vigorous intensity leisure-time PA (n = 80,035), TV-viewing time (n = 28,752), and sedentary behavior at work (n = 59,381) or during transportation (n = 15,152) [personal communication with Marcel den Hoed, Marilyn Cornelis, and Ruth Loos]. However, we did not find significant associations when correcting for the number of loci that were examined (P>0.005) (S16 Table).

thumbnail
Table 2. Genes of biological interest within 500 kb of lead SNPs associated with BMI.

https://doi.org/10.1371/journal.pgen.1006528.t002

thumbnail
Table 3. Genes of biological interest within 500 kb of lead SNPs associated with WCadjBMI or WHRadjBMI.

https://doi.org/10.1371/journal.pgen.1006528.t003

Identification of secondary signals

In addition to uncovering 11 novel adiposity loci, our PA-adjusted GWAS and the joint test of SNP main effect and SNP×PA interaction confirmed 148 genome-wide significant loci (50 for BMI, 58 for WCadjBMI, 40 for WHRadjBMI) that have been established in previous main effect GWAS for adiposity traits (S7S12 Tables, S4 Fig). The lead SNPs in eight of the previously established loci (5 for BMI, 3 for WCadjBMI), however, showed no LD or only weak LD (r2<0.3) with the published lead SNP, suggesting they could represent novel secondary signals in known loci (S17 Table). To test whether these eight signals are independent of the previously published signals, we performed conditional analyses [19]. Three of the eight SNPs we examined, in/near NDUFS4, MEF2C-AS1 and CPA1, were associated with WCadjBMI with P<5x10-8 in our PA-adjusted GWAS even after conditioning on the published lead SNP, hence representing novel secondary signals in these loci (S17 Table).

Enrichment of the identified loci with functional regulatory elements

Epigenetic variation may underlie gene-environment interactions observed in epidemiological studies [20] and PA has been shown to induce marked epigenetic changes in the genome [21]. We examined whether the BMI or WHRadjBMI loci reaching P<1x10-5 for interaction with PA (13 loci for BMI, 5 for WHRadjBMI) show overall enrichment with chromatin states in adipose, brain and muscle tissues available from the Roadmap Epigenomics Consortium [22]. However, we did not find significant enrichment (S18 and S19 Tables), which may be due to the limited number of identified loci. The lack of significant findings may also be due to the assessment of chromatin states in the basal state, which may not reflect the dynamic changes that occur when cells are perturbed by PA [23].

We also tested whether the loci reaching P<5x10-8 in our PA-adjusted GWAS of BMI or WHRadjBMI show enrichment with chromatin states and found significant enrichment of the BMI loci with enhancer, weak transcription, and polycomb-repressive elements in several brain cell lines, and with enhancer elements in three muscle cell lines (S20 and S21 Tables). We also found significant enrichment of the WHRadjBMI loci with enhancer elements in three adipose and six muscle cell lines, with active transcription start sites in two adipose cell lines, and with polycomb-repressive elements in seven brain cell lines. The enrichment of our PA-adjusted main effect results with chromatin annotations in skeletal muscle in particular, the tissue most affected by PA, could highlight regulatory mechanisms that may be influenced by PA.

Discussion

In this genome-wide meta-analysis of more than 200,000 adults, we do not find evidence of interaction with PA for loci other than the established FTO locus. However, when adjusting for PA or jointly testing for SNP main effect and interaction with PA, we identify 11 novel adiposity loci, suggesting that accounting for PA or other environmental factors that contribute to variation in adiposity may increase power for gene discovery.

Our results suggest that if SNP×PA interaction effects for common variants exist, they are unlikely to be of greater magnitude than observed for FTO, the BMI-increasing effect of which is attenuated by ~30% in physically active individuals. The fact that common SNPs explain less of the BMI variance among physically active compared to inactive individuals indicates that further interactions may exist, but larger meta-analyses, more accurate and precise measurement of PA, and/or improved analytical methods will be required to identify them. We found no difference between inactive and active individuals in variance explained by common SNPs in aggregate for WCadjBMI or WHRadjBMI, and no loci interacted with PA on WCadjBMI or WHRadjBMI. Therefore, PA may not modify genetic influences as strongly for body fat distribution as for overall adiposity. Furthermore, while differences in variance explained by common variants may be due to genetic effects being modified by PA, it is important to note that heritability can change in the absence of changes in genetic effects, if environmental variation differs between the inactive and active groups. Therefore, the lower BMI variance explained in the active group could be partly due to a potentially greater environmental variation in this group.

While we replicated the previously observed interaction between FTO and PA [7], it remains unclear what biological mechanisms underlie the attenuation in FTO’s effect in physically active individuals, and whether the interaction is due to PA or due to confounding by other environmental exposures. While some studies suggest that FTO may interact with diet [2426], a recent meta-analysis of 177,330 individuals did not find interaction between FTO and dietary intakes of total energy, protein, carbohydrate or fat [27]. The obesity-associated FTO variants are located in a super-enhancer region [28] and have been associated with DNA methylation levels [2931], suggesting that this region may be sensitive to epigenetic effects that could mediate the interaction between FTO and PA.

In genome-wide analyses for SNP main effects adjusting for PA, or when testing for the joint significance of SNP main effect and SNPxPA interaction, we identify 11 novel adiposity loci, even though our sample size was up to 40% smaller than in the largest published main effect meta-analyses [17, 18]. Our findings suggest that accounting for PA may facilitate the discovery of novel adiposity loci. Similarly, accounting for other environmental factors that contribute to variation in adiposity could lead to the discovery of additional loci.

In the present meta-analyses, statistical power to identify SNPxPA interactions may have been limited due to challenges relating to the measurement and statistical modeling of PA [5]. Of the 60 participating studies, 56 assessed PA by self-report while 4 used wearable PA monitors. Measurement error and bias inherent in self-report estimates of PA [32] can attenuate effect sizes for SNP×PA interaction effects towards the null [33]. Measurement using PA monitors provides more consistent results, but the monitors are not able to cover all types of activities and the measurement covers a limited time span compared to questionnaires [34]. As sample size requirements increase nonlinearly when effect sizes decrease, any factor that leads to a deflation in the observed interaction effect estimates may make their detection very difficult, even when very large population samples are available for analysis. Finally, because of the wide differences in PA assessment tools used among the participating studies, we treated PA as a dichotomous variable, harmonizing PA into inactive and active individuals. Considerable loss of power is anticipated when a continuous PA variable is dichotomized [35]. Our power could be enhanced by using a continuous PA variable if a few larger studies with equivalent, quantitative PA measurements were available.

In summary, while our results suggest that adjusting for PA or other environmental factors that contribute to variation in adiposity may increase power for gene discovery, we do not find evidence of SNP×PA interaction effects stronger than that observed for FTO. While other SNP×PA interaction effects on adiposity are likely to exist, combining many small studies with varying characteristics and PA assessment tools may be inefficient for identifying such effects [5]. Access to large cohorts with quantitative, equivalent PA variables, measured with relatively high accuracy and precision, may be necessary to uncover novel SNP×PA interactions.

Methods

Main analyses

Ethics statement.

All studies were conducted according to the Declaration of Helsinki. The studies were approved by the local ethical review boards and all study participants provided written informed consent for the collection of samples and subsequent analyses.

Outcome traits—BMI, WCadjBMI and WHRadjBMI.

We examined three anthropometric traits related to overall adiposity (BMI) or body fat distribution (WCadjBMI and WHRadjBMI) [36] that were available from a large number of studies. Before the association analyses, we calculated sex-specific residuals by adjusting for age, age2, BMI (for WCadjBMI and WHRadjBMI traits only), and other necessary study-specific covariates, such as genotype-derived principal components. Subsequently, we normalized the distributions of sex-specific trait residuals using inverse normal transformation.

Physical activity.

Physical activity was assessed and quantified in various ways in the participating studies of the meta-analysis (S1 and S6 Tables). Aiming to amass as large a sample size as possible, we harmonized PA by categorizing it into a simple dichotomous variable—physically inactive vs. active—that could be derived in a relatively consistent way in all participating studies, and that would be consistent with previous findings on gene-physical activity interactions and the relationship between activity levels and health outcomes. In studies with categorical PA data, individuals were defined inactive if they reported having a sedentary occupation and being sedentary during transport and leisure-time (<1 h of moderate intensity leisure-time or commuting PA per week). All other individuals were defined physically active. Previous studies in large-scale individual cohorts have demonstrated that the interaction between FTO, or a BMI-increasing genetic risk score, with physical activity, is most pronounced approximately at this activity level [6, 37, 38]. In studies with continuous PA data, PA variables were standardized by defining individuals belonging to the lowest sex- and age-adjusted quintile of PA levels as inactive, and all other individuals as active. The study-specific coding of the dichotomous PA variable in each study is described in S6 Table.

Study-specific association analyses.

We included 42 studies with genome-wide data, 10 studies with Metabochip data, and eight studies with both genome-wide and Metabochip data. If both genome-wide and Metabochip data were available for the same individual, we only included the genome-wide data (S1 Table). Studies with genome-wide genotyped data used either Affymetrix or Illumina arrays (S2 Table). Following study-specific quality control measures, the genotype data were imputed using the HapMap phase II reference panel (S2 Table). Studies with Metabochip data used the custom Illumina HumanCardio-Metabo BeadChip containing ~195K SNPs designed to support large-scale follow-up of known associations with metabolic and cardiovascular traits [39]. Each study ran autosomal SNP association analyses with BMI, WCadjBMI and WHRadjBMI across their array of genetic data using the following linear regression models in men and women separately: 1) active individuals only; 2) inactive individuals only; and 3) active and inactive individuals combined, adjusting for the PA stratum. In studies that included families or closely related individuals, regression coefficients were estimated using a variance component model that modeled relatedness in men and women combined, with sex as a covariate, in addition to the sex-specific analyses. The additive genetic effect for each SNP and phenotype association was estimated using linear regression. For studies with a case-control design (S1 Table), cases and controls were analyzed separately.

Quality control of study-specific association results.

All study-specific files for the three regression models listed above were processed through a standardized quality control protocol using the EasyQC software [40]. The study-specific quality control measures included checks on file completeness, range of test statistics, allele frequencies, trait transformation, population stratification, and filtering out of low quality data. Checks on file completeness included screening for missing alleles, effect estimates, allele frequencies, and other missing data. Checks on range of test statistics included screening for invalid statistics such as P-values >1 or <0, negative standard errors, or SNPs with low minor allele count (MAC, calculated as MAF*N, where MAF is the minor allele frequency and N is the sample size) and where SNPs with MAC<5 in the inactive or the active group were removed. The correctness of trait transformation to inverse normal was examined by plotting 2/median of the standard error with the square root of the sample size. Population stratification was examined by calculating the study specific genomic control inflation factor (λGC) [41]. If a study had λGC>1.1, the study analyst was contacted and asked to revise the analyses by adjusting for principal components. The allele frequencies in each study were examined for strand issues and miscoded alleles by plotting effect allele frequencies against the corresponding allele frequencies from the HapMap2 reference panel. Finally, low quality data were filtered out by removing monomorphic SNPs, imputed SNPs with poor imputation quality (r2_hat <0.3 in MACH [42], observed/expected dosage variance <0.3 in BIMBAM [43], proper_info <0.4 in IMPUTE [44]), and genotyped SNPs with a low call-rate (<95%) or that were out of Hardy-Weinberg equilibrium (P<10−6).

Meta-analyses.

Beta-coefficients and standard errors were combined by an inverse-variance weighted fixed effect method, implemented using the METAL software [45]. We performed meta-analyses for each of the three models (active, inactive, active + inactive adjusted for PA) in men only, in women only, and in men and women combined. Study-specific GWAS results were corrected for genomic control using all SNPs. Study-specific Metabochip results as well as the meta-analysis results for GWAS and Metabochip combined were corrected for genomic control using 4,425 SNPs included on the Metabochip for replication of associations with QT-interval, a phenotype not correlated with BMI, WCadjBMI or WHRadjBMI, after pruning of SNPs within 500 kb of an anthropometry replication SNP. We excluded SNPs that 1) were not available in at least half of the maximum sample size in each stratum; 2) had a heterogeneity I2 >75%, or 3) were missing chromosomal and base position annotation in dbSNP.

Calculation of the significance of SNP×PA interaction and of the joint significance of SNP main effect and SNP×PA interaction.

To identify SNP×PA interactions, we used the EasyStrata R package [46] to test for the difference in meta-analyzed beta-coefficients between the active and inactive groups for the association of each SNP with BMI, WCadjBMI and WHRadjBMI. Easystrata tests for differences in effect estimates between the active and inactive strata by subtracting one beta from the other (βactive−βinactive,) and dividing by the overall standard error of the difference as follows: where r is the Spearman rank correlation coefficient between βactive and βinactive for all genome-wide SNPs. The joint significance of the SNP main and SNP×PA interaction effects was estimated using the method by Aschard et al. [16] which is a joint test for genetic main effects and gene-environment interaction effects where gene-environment interaction is calculated as the difference in effect estimates between two exposure strata, accounting for 2 degrees of freedom.

Testing for secondary signals.

Approximate conditional analyses were conducted using GCTA version 1.24 [19]. In the analyses for SNPs identified in our meta-analyses of European-ancestry individuals only, LD correlations between SNPs were estimated using a reference sample comprised of European-ancestry participants of the Atherosclerosis Risk in Communities (ARIC) study. In the analyses for SNPs identified in our meta-analyses of all ancestries combined, the reference sample comprised 93% of European-ancestry individuals and 6% of African ancestry participants from ARIC, as well as 1% of CHB and JPT samples from the HapMap2 panel, to approximate the ancestry mixture in our all ancestry meta-analyses. To test if our identified SNPs were independent secondary signals that fell within 1 Mbp of a previously established signal, we used the GCTA—cojo-cond command to condition our lead SNPs on each previously established SNP in the same locus.

Replication analysis for the CDH12 locus.

The replication analysis for the CDH12 locus included participants from the EPIC-Norfolk (NINACTIVE = 4,755, NACTIVE = 11,526) and Fenland studies (NINACTIVE = 1,213, NACTIVE = 4,817), and from the random subcohort of the EPIC-InterAct Consortium (NINACTIVE = 2,154, NACTIVE = 6,632). PA stratum-specific estimates of the association of CDH12 with BMI were assessed and meta-analyzed by fixed effects meta-analyses, and the differences between the PA-strata were determined as described above.

Examining the influence of BMI, WCadjBMI and WHRadjBMI-associated loci on other complex traits and their potential functional roles

NHGRI-EBI GWAS catalog lookups.

To identify associations of the novel BMI, WCadjBMI or WHRadjBMI loci with other complex traits in published GWAS, we extracted previously reported GWAS associations within 500 kb and r2>0.6 with any of the lead SNPs, from the GWAS Catalog of the National Human Genome Research Institute and European Bioinformatics Institute [47] (S14 Table).

eQTLs.

We examined the cis-associations of the novel BMI, WCadjBMI or WHRadjBMI loci with the expression of nearby genes from various tissues by performing a look-up in a library of >100 published expression datasets, as described previously by Zhang et al [48]. In addition, we examined cis-associations using gene expression data derived from fasting peripheral whole blood in the Framingham Heart Study [49] (n = 5,206), adjusting for PA, age, age2, sex and cohort. For each novel locus, we evaluated the association of all transcripts ±1 Mb from the lead SNP. To minimize the potential for false positives, we only considered associations where our lead SNP or its proxy (r2>0.8) was either the peak SNP associated with the expression of a gene transcript in the region, or in strong LD (r2>0.8) with the peak SNP.

Overlap with functional regulatory elements.

We used the Uncovering Enrichment Through Simulation method to combine the genetic association data with the Roadmap Epigenomics Project segmentation data [22]. First, 10,000 sets of random SNPs were selected among HapMap2 SNPs with a MAF >0.05 that matched the original input SNPs based on proximity to a transcription start site and the number of LD partners (r2>0.8 in individuals of European ancestry in the 1000 Genomes Project). The LD partners were combined with their original lead SNPs to create 10,000 sets of matched random SNPs and their respective LD partners. These sets were intersected with the 15-state ChromHMM data from the Roadmap Epigenomics Project and resultant co-localizations were collapsed from total SNPs down to loci, which were then used to calculate an empirical P value when comparing the original SNPs to the random sets. We examined the enrichment for all loci reaching P<10−5 for SNP×PA interaction combined, and for all loci reaching P<5x10-8 in the PA-adjusted SNP main effect model combined. In addition, we examined the variant-specific overlap with regulatory elements for each of the index SNPs of the novel BMI, WCadjBMI and WHRadjBMI loci and variants in strong LD (r2>0.8).

Estimation of variance explained in inactive and active groups.

We compared variance explained for BMI, WCadjBMI and WHRadjBMI between the active and inactive groups using two approaches. First, we used a method previously reported by Kutalik et al [15], and selected subsets of SNPs based on varying P value thresholds (ranging from 5x10-8 to 0.05) from the SNP main effect model adjusted for PA. Each subset of SNPs was clumped into independent regions using a physical distance criterion of <500kb, and the most significant lead SNP within the respective region was selected. For each lead SNP, the explained variance was calculated as: in the active and inactive groups separately, where N is the sample size and P is the P value for SNP main effect in active or inactive strata. Finally, the variance explained by each subset of SNPs in the active and inactive strata was estimated by summing up the variance explained by the SNPs.

Second, we applied the LD Score regression tool developed by Bulik-Sullivan et al [14] to quantify the proportion of inflation due to polygenicity (heritability) rather than confounding (cryptic relatedness or population stratification) using meta-analysis summary results. LD Score regression leverages LD between causal and index variants to distinguish true signals by regressing meta-analysis summary results on an ‘LD Score’, i.e. the cumulative genetic variation that an index SNP tags. To obtain heritability estimates by PA strata, we regressed our summary results from the genome-wide meta-analyses of BMI, WCadjBMI and WHRadjBMI, stratified by PA status (active and inactive), on pre-calculated LD Scores available in HapMap3 reference samples of up to 1,061,094 variants with MAF≥1% and N>10th percentile of the total sample size.

Supporting information

S1 Fig. Interaction between the CDH12 locus and physical activity on BMI in the discovery genome-wide meta-analysis (n = 134,767), in the independent replication sample (n = 31,097), and in the discovery and replication samples combined.

https://doi.org/10.1371/journal.pgen.1006528.s002

(DOCX)

S2 Fig. Quantile-Quantile and Manhattan plots for the genome-wide meta-analysis results of the SNP main effect adjusting for physical activity (SNPadjPA), interaction between SNP and physical activity, and the joint effect of SNP main effect and SNP×PA interaction (Joint2df) in men and women of European-ancestry combined.

https://doi.org/10.1371/journal.pgen.1006528.s003

(DOCX)

S3 Fig. Regional association plots for novel BMI, WCadjBMI or WHRadjBMI loci showing either a genome-wide significant SNP main effect when adjusting for physical activity as a covariate, or a genome-wide significant joint effect of physical activity-adjusted SNP main effect and SNP × physical activity interaction.

https://doi.org/10.1371/journal.pgen.1006528.s004

(DOCX)

S4 Fig. Heatmap of P values for the physical activity-adjusted SNP main effect model (PadjPA), the joint model (Pjoint), and the SNPxPA interaction model (Pint).

https://doi.org/10.1371/journal.pgen.1006528.s005

(DOCX)

S1 Table. Basic study information and description of outcome assessment (BMI, WC, WHR) and Physical activity assessment.

https://doi.org/10.1371/journal.pgen.1006528.s006

(XLSX)

S2 Table. Genotyping and imputation platforms of the participating studies.

https://doi.org/10.1371/journal.pgen.1006528.s007

(XLSX)

S3 Table. Population characteristics for inactive and active individuals combined in the participating studies.

https://doi.org/10.1371/journal.pgen.1006528.s008

(XLSX)

S4 Table. Population characteristics for inactive individuals in the participating studies.

https://doi.org/10.1371/journal.pgen.1006528.s009

(XLSX)

S5 Table. Population characteristics for active individuals in the participating studies.

https://doi.org/10.1371/journal.pgen.1006528.s010

(XLSX)

S6 Table. Methods used for measuring physical activity and definitions of inactive for studies participating in the meta-analyses.

https://doi.org/10.1371/journal.pgen.1006528.s011

(XLSX)

S7 Table. All SNPs that met significance for BMI in the European only analyses for at least one of the approaches tested: interaction, adjusted for physical activity, or jointly accounting for the main and interaction effects.

https://doi.org/10.1371/journal.pgen.1006528.s012

(XLSX)

S8 Table. All SNPs that met significance for BMI in the all ancestry analyses for at least one of the approaches tested: interaction, adjusted for physical activity, or jointly accounting for the main and interaction effects.

https://doi.org/10.1371/journal.pgen.1006528.s013

(XLSX)

S9 Table. All SNPs that met significance for waist circumference adjusted for BMI in the European only analyses for at least one of the approaches tested: interaction, adjusted for physical activity, or jointly accounting for the main and interaction effects.

https://doi.org/10.1371/journal.pgen.1006528.s014

(XLSX)

S10 Table. All SNPs that met significance for waist circumference adjusted for BMI in the all ancestry analyses for at least one of the approaches tested: interaction, adjusted for physical activity, or jointly accounting for the main and interaction effects.

https://doi.org/10.1371/journal.pgen.1006528.s015

(XLSX)

S11 Table. All SNPs that met significance for waist-to-hip ratio adjusted for BMI in the European only analyses for at least one of the approaches tested: interaction, adjusted for physical activity, or jointly accounting for the main and interaction effects.

https://doi.org/10.1371/journal.pgen.1006528.s016

(XLSX)

S12 Table. All SNPs that met significance for waist-to-hip ratio adjusted for BMI in the all ancestry analyses for at least one of the approaches tested: interaction, adjusted for physical activity, or jointly accounting for the main and interaction effects.

https://doi.org/10.1371/journal.pgen.1006528.s017

(XLSX)

S13 Table. Variance explained using P value thresholds.

https://doi.org/10.1371/journal.pgen.1006528.s018

(XLSX)

S14 Table. GWAS catalog lookups for novel loci and new secondary signal in known loci.

https://doi.org/10.1371/journal.pgen.1006528.s019

(XLSX)

S15 Table. Association of the novel loci with cis gene expression (cis-eQTL).

https://doi.org/10.1371/journal.pgen.1006528.s020

(XLSX)

S16 Table. Association of loci identified for interaction with physical activity, for physical activity-adjusted SNP main effect, or for joint association of SNP main effect and physical activity interaction, with physical activity and sedentary behaviour.

https://doi.org/10.1371/journal.pgen.1006528.s021

(XLSX)

S17 Table. Results for approximate conditional analyses to identify secondary signals in the novel BMI, WCadjBMI or WHRadjBMI-associated locia.

https://doi.org/10.1371/journal.pgen.1006528.s022

(XLSX)

S18 Table. Enrichment of loci interacting with PA (Pint<10−5) on the level of BMI with functional genomic elements in adipose, brain, and muscle tissue cell lines from the Roadmap Epigenomics Project.

https://doi.org/10.1371/journal.pgen.1006528.s023

(XLSX)

S19 Table. Enrichment of loci interacting with PA (Pint<10−5) on the level of WHRadjBMI with functional genomic elements in adipose, brain, and muscle tissue cell lines from the Roadmap Epigenomics Project.

https://doi.org/10.1371/journal.pgen.1006528.s024

(XLSX)

S20 Table. Enrichment of loci showing association with BMI (PadjPA<5x10-8) with functional genomic elements in adipose, brain, and muscle tissue cell lines from the Roadmap Epigenomics Project.

https://doi.org/10.1371/journal.pgen.1006528.s025

(XLSX)

S21 Table. Enrichment of loci showing association with WHRadjBMI (PadjPA<5x10-8) with functional genomic elements in adipose, brain, and muscle tissue cell lines from the Roadmap Epigenomics Project.

https://doi.org/10.1371/journal.pgen.1006528.s026

(XLSX)

S22 Table. Association of novel loci identified for interaction with physical activity, for physical activity-adjusted SNP main effect, or for joint association of the SNP main effect and physical activity interaction, in GIANT results not accounting for physical activity.

https://doi.org/10.1371/journal.pgen.1006528.s027

(XLSX)

Author Contributions

  1. Conceptualization: TOK RJFL.
  2. Data curation: TOK LAC RJFL KLMon.
  3. Formal analysis: RAS KLY AEJ TWW AMa DHa NLHC JSN TSA LQ TW FRe MdH TOK QQ MG TSA LX AYC LAC MC RJFL MFF KEN JDE ADJ JEHa RJK.
  4. Investigation: AVS TBH GE LJL VG KEN MG AJ KLMon KLY EB PGL JBW NGM GWM DPS GC LJP JHua AWM ALJ JB MFo CBe HMS TR SSn BS YW JBB LSA ZK PMMV TC SBi GWi PV KKr ASH KHa SM VS MP JM IR CHup VV IK OPo LY DT BB MMar AB PF RRaw TAL PK MHo KSa RMag THal TEb AMa JL RAS SBr NJW JHZ RL SBu AD NA CMvD IBB MFF LB RJFL LX NLHC EL JP PJG CSF CTL LAC MB FSC KLMol RNB JT LK TEs HP CS HAK LFB SLRK MAJ PAP SA FRe IB GH PWF JE CO MLo AEJ JOJ TSA LPa GDS TIAS JEHu DJP SP LJH BHS SSa YCC MFu JRO ARS MHa MA AL HVe LQu THu QQ JAS JDF WZhan DRW KHv OLH AUJ LLB AJS KKv TTan DHe SBa MNH JMJ KF NG OPe TWW IMH MO CG HG AP KSt CHo CHut RRau BT MMN WZhao BL WRS STT JCC JSK MEK GED TBG GS JHui WM UL CHa LL LH KL AGB LJRT JAN YL MLa JK AS PSC NN WJP GvG YM BWJHP AMu CML MIM TTam JA MRJ LQi THan DK KKO AW ÅJ UG EJCdG MHMdM JJH DIB GWa PN AFW NDH SW HC JFW CBo MCV LPe DP MW TMF NVR MCZ FRi AH AGU JLBG SSi FB AMe GRA FC ATe HVo UV MD SG MN RMan IP ATo PJvdM JVvVO IMN HS AJO CAH MMan DIC AYC LMR PMR SBi NZ JG UP CK MKu MKi CL LPL NHK MJ MKä OTR TL.
  5. Supervision: GRA IB MB IBB CSF TMF IMH RJFL MIM KLMon KEN JRO DPS CMvD JNH.
  6. Writing – original draft: MG RAS AEJ KLY MFF LB LQu PWF RJFL TOK.
  7. Writing – review & editing: MG RAS AEJ KLY MFF TWW LB AYC AMa DHa LX TW NLHC TSA MdH QQ JSN FRe LQu JDE JEHa MC ZK EL JL JEHu WZhan WZhao PJG THal SA PMMV SBi LY ATe AVS MKu MNH JMJ MEK MHo NVR JBW JHZ HS LL CHup GWi WJP YW KKr AD KL YL MFo MHa LFB GC TTan RMag PJvdM AUJ JLBG VV JM PN CBe DP ÅJ SSn YCC JE UL MA LSA NA BB JA JB RNB AGB JB AB LLB JBB SBr FB SBu HC PSC FSC TC GDS GED ND MD TEb GE TEs JDF MFu KF CG SG JG PGL HG TBG NG GvG THu KHa NDH ASH DHe LH LJH OLH CHo JJH JHua THan JHui CHut NHK ALJ JOJ MAH MJ LK HAK IK PK JK KKv MKa TAL LJL BL CML MLo RL MMar YM KLMon GWM MHMdM AMu AMe RMan SM NN MN JAN IMN AJO MO KKO SP LPa JP MP AP UP PAP IP HP OTR TR LJRT RRaw PMR LMR IR CS MAS KSa WRS SSa ARS SSi GS BHS JAS HS AS BS AJS TTam STT BT DT LV HVe JVvVO MCV UV GWa MW SW AW AFW MCZ NZ CAHai LL UG CO AH FRi AGU LPe JFW CHa OPo FC KHv CAHar ATo SBa LJP SLRK RRau TIAS JT VS BWJHP EJCdG DIB TL MMan MLa CBo NGM DK AL WM KSt MKi TBH VG HVo LQi MRJ JCC JSK PF CK PV GH OPe NJW CL DRW DJP EB DIC GRA IB MIM TMF JRO CMvD MB IMH KLMol DPS CSF CTL JNH RJK ADJ IBB PWF KEN LAC RJFL TOK.

References

  1. 1. WHO. Obesity: preventing and managing the global epidemic. Report of a WHO consultation. World Health Organization technical report series. 2000;894:i–xii, 1–253. Epub 2001/03/10. pmid:11234459
  2. 2. Bouchard C, Tremblay A. Genetic influences on the response of body fat and fat distribution to positive and negative energy balances in human identical twins. The Journal of nutrition. 1997;127(5 Suppl):943S–7S. Epub 1997/05/01. pmid:9164270
  3. 3. Bouchard C, Tremblay A, Despres JP, Nadeau A, Lupien PJ, Theriault G, et al. The response to long-term overfeeding in identical twins. The New England journal of medicine. 1990;322(21):1477–82. Epub 1990/05/24. pmid:2336074
  4. 4. Hainer V, Stunkard AJ, Kunesova M, Parizkova J, Stich V, Allison DB. Intrapair resemblance in very low calorie diet-induced weight loss in female obese identical twins. International journal of obesity and related metabolic disorders: journal of the International Association for the Study of Obesity. 2000;24(8):1051–7. Epub 2000/08/22.
  5. 5. Ahmad S, Rukh G, Varga TV, Ali A, Kurbasic A, Shungin D, et al. Gene x physical activity interactions in obesity: combined analysis of 111,421 individuals of European ancestry. PLoS genetics. 2013;9(7):e1003607. Epub 2013/08/13. pmid:23935507
  6. 6. Li S, Zhao JH, Luan J, Ekelund U, Luben RN, Khaw KT, et al. Physical activity attenuates the genetic predisposition to obesity in 20,000 men and women from EPIC-Norfolk prospective population study. PLoS medicine. 2010;7(8). Epub 2010/09/09.
  7. 7. Kilpelainen TO, Qi L, Brage S, Sharp SJ, Sonestedt E, Demerath E, et al. Physical activity attenuates the influence of FTO variants on obesity risk: a meta-analysis of 218,166 adults and 19,268 children. PLoS medicine. 2011;8(11):e1001116. Epub 2011/11/10. pmid:22069379
  8. 8. Scott RA, Chu AY, Grarup N, Manning AK, Hivert MF, Shungin D, et al. No interactions between previously associated 2-hour glucose gene variants and physical activity or BMI on 2-hour glucose levels. Diabetes. 2012;61(5):1291–6. Epub 2012/03/15. pmid:22415877
  9. 9. Yang J, Loos RJ, Powell JE, Medland SE, Speliotes EK, Chasman DI, et al. FTO genotype is associated with phenotypic variability of body mass index. Nature. 2012;490(7419):267–72. Epub 2012/09/18. pmid:22982992
  10. 10. Winkler TW, Justice AE, Graff M, Barata L, Feitosa MF, Chu S, et al. The Influence of Age and Sex on Genetic Associations with Adult Body Size and Shape: A Large-Scale Genome-Wide Interaction Study. PLoS genetics. 2015;11(10):e1005378. Epub 2015/10/02. pmid:26426971
  11. 11. Selig S, Lidov HG, Bruno SA, Segal MM, Kunkel LM. Molecular characterization of Br-cadherin, a developmentally regulated, brain-specific cadherin. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(6):2398–403. Epub 1997/03/18. pmid:9122206
  12. 12. Heard-Costa NL, Zillikens MC, Monda KL, Johansson A, Harris TB, Fu M, et al. NRXN3 is a novel locus for waist circumference: a genome-wide association study from the CHARGE Consortium. PLoS genetics. 2009;5(6):e1000539. Epub 2009/06/27. pmid:19557197
  13. 13. Ng MC, Hester JM, Wing MR, Li J, Xu J, Hicks PJ, et al. Genome-wide association of BMI in African Americans. Obesity (Silver Spring, Md). 2012;20(3):622–7. Epub 2011/06/28.
  14. 14. Bulik-Sullivan BK, Loh PR, Finucane HK, Ripke S, Yang J, Patterson N, et al. LD Score regression distinguishes confounding from polygenicity in genome-wide association studies. Nature genetics. 2015;47(3):291–5. Epub 2015/02/03. pmid:25642630
  15. 15. Kutalik Z, Whittaker J, Waterworth D, Beckmann JS, Bergmann S. Novel method to estimate the phenotypic variation explained by genome-wide association studies reveals large fraction of the missing heritability. Genetic epidemiology. 2011;35(5):341–9. Epub 2011/04/06. pmid:21465548
  16. 16. Aschard H, Hancock DB, London SJ, Kraft P. Genome-wide meta-analysis of joint tests for genetic and gene-environment interaction effects. Human heredity. 2010;70(4):292–300. Epub 2011/02/05. pmid:21293137
  17. 17. Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, Day FR, et al. Genetic studies of body mass index yield new insights for obesity biology. Nature. 2015;518(7538):197–206. Epub 2015/02/13. pmid:25673413
  18. 18. Shungin D, Winkler TW, Croteau-Chonka DC, Ferreira T, Locke AE, Magi R, et al. New genetic loci link adipose and insulin biology to body fat distribution. Nature. 2015;518(7538):187–96. Epub 2015/02/13. pmid:25673412
  19. 19. Yang J, Ferreira T, Morris AP, Medland SE, Madden PA, Heath AC, et al. Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nature genetics. 2012;44(4):369–75, s1–3. Epub 2012/03/20. pmid:22426310
  20. 20. Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nature genetics. 2003;33 Suppl:245–54. Epub 2003/03/01.
  21. 21. Ling C, Ronn T. Epigenetic adaptation to regular exercise in humans. Drug discovery today. 2014;19(7):1015–8. Epub 2014/03/19. pmid:24632002
  22. 22. Kundaje A, Meuleman W, Ernst J, Bilenky M, Yen A, Heravi-Moussavi A, et al. Integrative analysis of 111 reference human epigenomes. Nature. 2015;518(7539):317–30. Epub 2015/02/20. pmid:25693563
  23. 23. Barres R, Yan J, Egan B, Treebak JT, Rasmussen M, Fritz T, et al. Acute exercise remodels promoter methylation in human skeletal muscle. Cell metabolism. 2012;15(3):405–11. Epub 2012/03/13. pmid:22405075
  24. 24. Ahmad T, Lee IM, Pare G, Chasman DI, Rose L, Ridker PM, et al. Lifestyle interaction with fat mass and obesity-associated (FTO) genotype and risk of obesity in apparently healthy U.S. women. Diabetes care. 2011;34(3):675–80. Epub 2011/01/27. pmid:21266646
  25. 25. Corella D, Arnett DK, Tucker KL, Kabagambe EK, Tsai M, Parnell LD, et al. A high intake of saturated fatty acids strengthens the association between the fat mass and obesity-associated gene and BMI. The Journal of nutrition. 2011;141(12):2219–25. Epub 2011/11/04. pmid:22049296
  26. 26. Sonestedt E, Roos C, Gullberg B, Ericson U, Wirfalt E, Orho-Melander M. Fat and carbohydrate intake modify the association between genetic variation in the FTO genotype and obesity. The American journal of clinical nutrition. 2009;90(5):1418–25. Epub 2009/09/04. pmid:19726594
  27. 27. Qi Q, Kilpelainen TO, Downer MK, Tanaka T, Smith CE, Sluijs I, et al. FTO genetic variants, dietary intake and body mass index: insights from 177,330 individuals. Human molecular genetics. 2014;23(25):6961–72. Epub 2014/08/12. pmid:25104851
  28. 28. Claussnitzer M, Dankel SN, Kim KH, Quon G, Meuleman W, Haugen C, et al. FTO Obesity Variant Circuitry and Adipocyte Browning in Humans. The New England journal of medicine. 2015;373(10):895–907. Epub 2015/08/20. pmid:26287746
  29. 29. Almen MS, Jacobsson JA, Moschonis G, Benedict C, Chrousos GP, Fredriksson R, et al. Genome wide analysis reveals association of a FTO gene variant with epigenetic changes. Genomics. 2012;99(3):132–7. Epub 2012/01/12. pmid:22234326
  30. 30. Bell CG, Finer S, Lindgren CM, Wilson GA, Rakyan VK, Teschendorff AE, et al. Integrated genetic and epigenetic analysis identifies haplotype-specific methylation in the FTO type 2 diabetes and obesity susceptibility locus. PloS one. 2010;5(11):e14040. Epub 2010/12/03. pmid:21124985
  31. 31. Toperoff G, Aran D, Kark JD, Rosenberg M, Dubnikov T, Nissan B, et al. Genome-wide survey reveals predisposing diabetes type 2-related DNA methylation variations in human peripheral blood. Human molecular genetics. 2012;21(2):371–83. Epub 2011/10/14. pmid:21994764
  32. 32. Helmerhorst HJ, Brage S, Warren J, Besson H, Ekelund U. A systematic review of reliability and objective criterion-related validity of physical activity questionnaires. The international journal of behavioral nutrition and physical activity. 2012;9:103. Epub 2012/09/04. pmid:22938557
  33. 33. Lundberg M, Hallqvist J, Diderichsen F. Exposure-dependent misclassification of exposure in interaction analyses. Epidemiology (Cambridge, Mass). 1999;10(5):545–9. Epub 1999/09/01.
  34. 34. Skender S, Ose J, Chang-Claude J, Paskow M, Bruhmann B, Siegel EM, et al. Accelerometry and physical activity questionnaires—a systematic review. BMC public health. 2016;16:515. Epub 2016/06/17. pmid:27306667
  35. 35. Ragland DR. Dichotomizing continuous outcome variables: dependence of the magnitude of association and statistical power on the cutpoint. Epidemiology (Cambridge, Mass). 1992;3(5):434–40. Epub 1992/09/01.
  36. 36. Heid IM, Jackson AU, Randall JC, Winkler TW, Qi L, Steinthorsdottir V, et al. Meta-analysis identifies 13 new loci associated with waist-hip ratio and reveals sexual dimorphism in the genetic basis of fat distribution. Nature genetics. 2010;42(11):949–60. Epub 2010/10/12. pmid:20935629
  37. 37. Andreasen CH, Stender-Petersen KL, Mogensen MS, Torekov SS, Wegner L, Andersen G, et al. Low physical activity accentuates the effect of the FTO rs9939609 polymorphism on body fat accumulation. Diabetes. 2008;57(1):95–101. Epub 2007/10/19. pmid:17942823
  38. 38. Vimaleswaran KS, Li S, Zhao JH, Luan J, Bingham SA, Khaw KT, et al. Physical activity attenuates the body mass index-increasing influence of genetic variation in the FTO gene. The American journal of clinical nutrition. 2009;90(2):425–8. Epub 2009/06/26. pmid:19553294
  39. 39. Voight BF, Kang HM, Ding J, Palmer CD, Sidore C, Chines PS, et al. The metabochip, a custom genotyping array for genetic studies of metabolic, cardiovascular, and anthropometric traits. PLoS genetics. 2012;8(8):e1002793. Epub 2012/08/10. pmid:22876189
  40. 40. Winkler TW, Day FR, Croteau-Chonka DC, Wood AR, Locke AE, Magi R, et al. Quality control and conduct of genome-wide association meta-analyses. Nature protocols. 2014;9(5):1192–212. Epub 2014/04/26. pmid:24762786
  41. 41. Devlin B, Roeder K. Genomic control for association studies. Biometrics. 1999;55(4):997–1004. Epub 2001/04/21. pmid:11315092
  42. 42. Li Y, Willer CJ, Ding J, Scheet P, Abecasis GR. MaCH: using sequence and genotype data to estimate haplotypes and unobserved genotypes. Genetic epidemiology. 2010;34(8):816–34. Epub 2010/11/09. pmid:21058334
  43. 43. Guan Y, Stephens M. Practical issues in imputation-based association mapping. PLoS genetics. 2008;4(12):e1000279. Epub 2008/12/06. pmid:19057666
  44. 44. Marchini J, Howie B, Myers S, McVean G, Donnelly P. A new multipoint method for genome-wide association studies by imputation of genotypes. Nature genetics. 2007;39(7):906–13. Epub 2007/06/19. pmid:17572673
  45. 45. Willer CJ, Li Y, Abecasis GR. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics (Oxford, England). 2010;26(17):2190–1. Epub 2010/07/10.
  46. 46. Winkler TW, Kutalik Z, Gorski M, Lottaz C, Kronenberg F, Heid IM. EasyStrata: evaluation and visualization of stratified genome-wide association meta-analysis data. Bioinformatics (Oxford, England). 2015;31(2):259–61. Epub 2014/09/28.
  47. 47. Welter D, MacArthur J, Morales J, Burdett T, Hall P, Junkins H, et al. The NHGRI GWAS Catalog, a curated resource of SNP-trait associations. Nucleic acids research. 2014;42(Database issue):D1001–6. Epub 2013/12/10. pmid:24316577
  48. 48. Zhang X, Gierman HJ, Levy D, Plump A, Dobrin R, Goring HH, et al. Synthesis of 53 tissue and cell line expression QTL datasets reveals master eQTLs. BMC genomics. 2014;15:532. Epub 2014/06/30. pmid:24973796
  49. 49. Joehanes R, Ying S, Huan T, Johnson AD, Raghavachari N, Wang R, et al. Gene expression signatures of coronary heart disease. Arteriosclerosis, thrombosis, and vascular biology. 2013;33(6):1418–26. Epub 2013/03/30. pmid:23539218
  50. 50. Behrens G, Winkler TW, Gorski M, Leitzmann MF, Heid IM. To stratify or not to stratify: power considerations for population-based genome-wide association studies of quantitative traits. Genetic epidemiology. 2011;35(8):867–79. Epub 2011/11/30. pmid:22125224
  51. 51. Speliotes EK, Willer CJ, Berndt SI, Monda KL, Thorleifsson G, Jackson AU, et al. Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nature genetics. 2010;42(11):937–48. Epub 2010/10/12. pmid:20935630
  52. 52. Gibbs J, Young RC, Smith GP. Cholecystokinin decreases food intake in rats. Journal of comparative and physiological psychology. 1973;84(3):488–95. Epub 1973/09/01. pmid:4745816
  53. 53. Muurahainen N, Kissileff HR, Derogatis AJ, Pi-Sunyer FX. Effects of cholecystokinin-octapeptide (CCK-8) on food intake and gastric emptying in man. Physiology & behavior. 1988;44(4–5):645–9. Epub 1988/01/01.
  54. 54. de Krom M, van der Schouw YT, Hendriks J, Ophoff RA, van Gils CH, Stolk RP, et al. Common genetic variations in CCK, leptin, and leptin receptor genes are associated with specific human eating patterns. Diabetes. 2007;56(1):276–80. Epub 2006/12/29. pmid:17192493
  55. 55. Okada Y, Kubo M, Ohmiya H, Takahashi A, Kumasaka N, Hosono N, et al. Common variants at CDKAL1 and KLF9 are associated with body mass index in east Asian populations. Nature genetics. 2012;44(3):302–6. Epub 2012/02/22. pmid:22344221
  56. 56. Gao FB, Keene JD. Hel-N1/Hel-N2 proteins are bound to poly(A)+ mRNA in granular RNP structures and are implicated in neuronal differentiation. Journal of cell science. 1996;109 (Pt 3):579–89. Epub 1996/03/01.
  57. 57. Nikpay M, Goel A, Won HH, Hall LM, Willenborg C, Kanoni S, et al. A comprehensive 1,000 Genomes-based genome-wide association meta-analysis of coronary artery disease. Nature genetics. 2015;47(10):1121–30. Epub 2015/09/08. pmid:26343387
  58. 58. Cha JY, Kim HJ, Yu JH, Xu J, Kim D, Paul BD, et al. Dexras1 mediates glucocorticoid-associated adipogenesis and diet-induced obesity. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(51):20575–80. Epub 2013/12/04. pmid:24297897
  59. 59. Murgia M, Serrano AL, Calabria E, Pallafacchina G, Lomo T, Schiaffino S. Ras is involved in nerve-activity-dependent regulation of muscle genes. Nature cell biology. 2000;2(3):142–7. Epub 2000/03/09. pmid:10707084
  60. 60. Zhang W, Thompson BJ, Hietakangas V, Cohen SM. MAPK/ERK signaling regulates insulin sensitivity to control glucose metabolism in Drosophila. PLoS genetics. 2011;7(12):e1002429. Epub 2012/01/14. pmid:22242005
  61. 61. Freyer J, Behrensen M, Zouhair A, Schunkert H, Erdmann J. Abstract 15307: Mras-knockout leads to obesity and a lack of B-cell function. Circulation. 2012;126:A15307.
  62. 62. Shi X, Sun X, Liu M, Li D, Aneja R, Zhou J. CEP70 protein interacts with gamma-tubulin to localize at the centrosome and is critical for mitotic spindle assembly. The Journal of biological chemistry. 2011;286(38):33401–8. Epub 2011/07/29. pmid:21795687
  63. 63. Dupuy D, Duperat VG, Arveiler B. SCAN domain-containing 2 gene (SCAND2) is a novel nuclear protein derived from the zinc finger family by exon shuffling. Gene. 2002;289(1–2):1–6. Epub 2002/05/31. pmid:12036577
  64. 64. Christians JK, Bath AK, Amiri N. Pappa2 deletion alters IGFBPs but has little effect on glucose disposal or adiposity. Growth hormone & IGF research: official journal of the Growth Hormone Research Society and the International IGF Research Society. 2015;25(5):232–9. Epub 2015/07/15.
  65. 65. Christians JK, de Zwaan DR, Fung SH. Pregnancy associated plasma protein A2 (PAPP-A2) affects bone size and shape and contributes to natural variation in postnatal growth in mice. PloS one. 2013;8(2):e56260. Epub 2013/03/05. pmid:23457539
  66. 66. Conover CA, Boldt HB, Bale LK, Clifton KB, Grell JA, Mader JR, et al. Pregnancy-associated plasma protein-A2 (PAPP-A2): tissue expression and biological consequences of gene knockout in mice. Endocrinology. 2011;152(7):2837–44. Epub 2011/05/19. pmid:21586553
  67. 67. Winkelmann J, Czamara D, Schormair B, Knauf F, Schulte EC, Trenkwalder C, et al. Genome-wide association study identifies novel restless legs syndrome susceptibility loci on 2p14 and 16q12.1. PLoS genetics. 2011;7(7):e1002171. Epub 2011/07/23. pmid:21779176
  68. 68. Hargens TA, Kaleth AS, Edwards ES, Butner KL. Association between sleep disorders, obesity, and exercise: a review. Nature and science of sleep. 2013;5:27–35. Epub 2013/04/27. pmid:23620691
  69. 69. Droppelmann CA, Wang J, Campos-Melo D, Keller B, Volkening K, Hegele RA, et al. Detection of a novel frameshift mutation and regions with homozygosis within ARHGEF28 gene in familial amyotrophic lateral sclerosis. Amyotrophic lateral sclerosis & frontotemporal degeneration. 2013;14(5–6):444–51. Epub 2013/01/05.
  70. 70. Keller BA, Volkening K, Droppelmann CA, Ang LC, Rademakers R, Strong MJ. Co-aggregation of RNA binding proteins in ALS spinal motor neurons: evidence of a common pathogenic mechanism. Acta neuropathologica. 2012;124(5):733–47. Epub 2012/09/04. pmid:22941224
  71. 71. Zhao L, Gregoire F, Sul HS. Transient induction of ENC-1, a Kelch-related actin-binding protein, is required for adipocyte differentiation. The Journal of biological chemistry. 2000;275(22):16845–50. Epub 2000/05/29. pmid:10828068
  72. 72. Kristiansson K, Perola M, Tikkanen E, Kettunen J, Surakka I, Havulinna AS, et al. Genome-wide screen for metabolic syndrome susceptibility Loci reveals strong lipid gene contribution but no evidence for common genetic basis for clustering of metabolic syndrome traits. Circulation Cardiovascular genetics. 2012;5(2):242–9. Epub 2012/03/09. pmid:22399527
  73. 73. Kilpelainen TO, Laaksonen DE, Lakka TA, Herder C, Koenig W, Lindstrom J, et al. The rs1800629 polymorphism in the TNF gene interacts with physical activity on the changes in C-reactive protein levels in the Finnish Diabetes Prevention Study. Experimental and clinical endocrinology & diabetes: official journal, German Society of Endocrinology [and] German Diabetes Association. 2010;118(10):757–9. Epub 2010/04/03.
  74. 74. Lakka HM, Lakka TA, Rankinen T, Rice T, Rao DC, Leon AS, et al. The TNF-alpha G-308A polymorphism is associated with C-reactive protein levels: the HERITAGE Family Study. Vascular pharmacology. 2006;44(5):377–83. Epub 2006/04/04. pmid:16581306
  75. 75. Ma L, Robinson LN, Towle HC. ChREBP*Mlx is the principal mediator of glucose-induced gene expression in the liver. The Journal of biological chemistry. 2006;281(39):28721–30. Epub 2006/08/04. pmid:16885160
  76. 76. Uyeda K, Repa JJ. Carbohydrate response element binding protein, ChREBP, a transcription factor coupling hepatic glucose utilization and lipid synthesis. Cell metabolism. 2006;4(2):107–10. Epub 2006/08/08. pmid:16890538
  77. 77. Lopez I, Mak EC, Ding J, Hamm HE, Lomasney JW. A novel bifunctional phospholipase c that is regulated by Galpha 12 and stimulates the Ras/mitogen-activated protein kinase pathway. The Journal of biological chemistry. 2001;276(4):2758–65. Epub 2000/10/07. pmid:11022047
  78. 78. Hinkes B, Wiggins RC, Gbadegesin R, Vlangos CN, Seelow D, Nurnberg G, et al. Positional cloning uncovers mutations in PLCE1 responsible for a nephrotic syndrome variant that may be reversible. Nature genetics. 2006;38(12):1397–405. Epub 2006/11/07. pmid:17086182
  79. 79. Boardman-Pretty F, Smith AJ, Cooper J, Palmen J, Folkersen L, Hamsten A, et al. Functional Analysis of a Carotid Intima-Media Thickness Locus Implicates BCAR1 and Suggests a Causal Variant. Circulation Cardiovascular genetics. 2015;8(5):696–706. Epub 2015/08/16. pmid:26276885
  80. 80. Cristancho AG, Lazar MA. Forming functional fat: a growing understanding of adipocyte differentiation. Nature reviews Molecular cell biology. 2011;12(11):722–34. Epub 2011/09/29. pmid:21952300
  81. 81. Roy SK, Hu J, Meng Q, Xia Y, Shapiro PS, Reddy SP, et al. MEKK1 plays a critical role in activating the transcription factor C/EBP-beta-dependent gene expression in response to IFN-gamma. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(12):7945–50. Epub 2002/06/06. pmid:12048245